1
|
Wang MQ, Li ZK, Li LY, Fan XB, Shu J, Hu QB, Wang HJ. A non-solvatochromic fluorescent probe for imaging of lipid droplets in live cells and tissues. Talanta 2025; 290:127805. [PMID: 40010120 DOI: 10.1016/j.talanta.2025.127805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/06/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Lipid droplets (LDs) have recently attracted considerable attention owing to their crucial roles in both biological processes and disease pathogenesis. Visualization of LDs is fundamental for elucidating their roles in biological mechanisms and facilitating the early detection of diseases. Donor-acceptor (D-A) typed fluorescent probes have been extensively designed and utilized for the detection of LDs. However, such probes often exhibit a pronounced solvatochromic effect, leading to several limitations in detecting LDs, such as short excitation/emission wavelength, low specificity. Herein, we reported a non-solvatochromic D-A typed fluorescent probe S7 for LDs imaging in live cells and in vivo. S7 is polarity-dependent, which exhibits a very weak fluorescence in high-polar solvents owing to the photoinduced electron transfer (PET) mechanism but intense fluorescence in low-polarity environments without undergoing a solvatochromic blue shift. Except polarity, the fluorescent signal of S7 remains unaffected by factors such as viscosity, pH, ions, reactive oxygen species, reactive sulfur species, nucleic acids, proteins, and other biological molecules, allowing it to selectively light up LDs in live cells. Furthermore, this probe S7 exhibits an enhanced fluorescence intensity in tumor tissue when compared to normal tissue. This characteristic potentially provides an efficient and straightforward approach for tumor diagnosis.
Collapse
Affiliation(s)
- Ming-Qi Wang
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China.
| | - Ze-Kai Li
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Lu-Yu Li
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Xu-Bo Fan
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Jing Shu
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Qi-Bin Hu
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hai-Jiao Wang
- The Key Laboratory of Biomedical Material, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
2
|
Zhang Y, Luo PY, Tang YN, Wang J, Gao S, Fan YC, Wang K. Association between the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) and mortality in patients with metabolic dysfunction-associated steatotic liver disease (MASLD): data from the NHANES III (1988-1994). Nutr Metab (Lond) 2025; 22:46. [PMID: 40399925 PMCID: PMC12093885 DOI: 10.1186/s12986-025-00942-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The prognostic value of the non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) remains unclear. This study aimed to evaluate the associations between the NHHR and all-cause and cause-specific mortality in patients with MASLD. METHODS Data for this study were obtained from the National Health and Nutrition Examination Survey (NHANES III and the National Death Index (NDI). The NHHR was calculated according to the formula. The results of mortality associated with the NDI were recorded as of December 31, 2019. We used a multivariate Cox proportional hazard model and restricted cubic spline (RCS) regression to assess the associations between the NHHR and all-cause and cause-specific mortality. In addition, subgroup analyses were performed to explore the relationships between the NHHR and all-cause and cause-specific mortality. RESULTS This study included 3155 patients with a definite diagnosis of MASLD. A total of 1,381 (43.8%) patients with MASLD died, and 1,774 (56.2%) survived. Multivariate Cox proportional hazards model analysis showed that NHHR was not significantly associated with all-cause mortality in MASLD patients. The RCS curve showed a significant nonlinear trend between the NHHR and all-cause mortality in patients with MASLD. Subgroup analysis revealed that the NHHR was better suited to predict cardiovascular mortality in patients without advanced fibrosis. CONCLUSIONS Our study revealed the clinical value of the NHHR in the prediction of mortality in the MASLD population. The NHHR can be used as a biomarker for follow-up in people without advanced fibrosis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Peng-Yu Luo
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Yu-Na Tang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Jing Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, Shandong, 250012, China.
- Hepatology Institute of Shandong University, Jinan, 250012, China.
| |
Collapse
|
3
|
Fu Y, Peng X, Song H, Li X, Zhi Y, Tang J, Liu Y, Chen D, Li W, Zhang J, Ma J, He M, Mao Y, Zhao XY. Disrupted minor intron splicing activates reductive carboxylation-mediated lipogenesis to drive metabolic dysfunction-associated steatotic liver disease progression. J Clin Invest 2025; 135:e186478. [PMID: 40100939 PMCID: PMC12077890 DOI: 10.1172/jci186478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/17/2025] [Indexed: 03/20/2025] Open
Abstract
Aberrant RNA splicing is tightly linked to diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD). In this study, we revealed that minor intron splicing, a unique and conserved RNA processing event, is largely disrupted upon the progression of metabolic dysfunction-associated steatohepatitis (MASH) in mice and humans. We demonstrated that deficiency of minor intron splicing in the liver induced MASH transition upon obesity-induced insulin resistance and LXR activation. Mechanistically, inactivation of minor intron splicing led to minor intron retention of Insig1 and Insig2, resulting in premature termination of translation, which drove proteolytic activation of SREBP1c. This mechanism was conserved in patients with MASH. Notably, disrupted minor intron splicing activated glutamine reductive metabolism for de novo lipogenesis through induction of Idh1, which caused accumulation of ammonia in the liver, thereby initiating hepatic fibrosis upon LXR activation. Ammonia clearance or IDH1 inhibition blocked hepatic fibrogenesis and mitigated MASH progression. More importantly, overexpression of Zrsr1 restored minor intron retention and ameliorated the development of MASH, indicating that dysfunctional minor intron splicing is an emerging pathogenic mechanism that drives MASH progression. Additionally, our results suggest that reductive carboxylation flux triggered by minor intron retention in hepatocytes serves as a crucial checkpoint and potential target for MASH therapy.
Collapse
Affiliation(s)
- Yinkun Fu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
- Institute for Translational Medicine on Cell Fate and Disease, Shanghai Ninth People’s Hospital, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Peng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
| | - Hongyong Song
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
| | - Xiaoyun Li
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Yang Zhi
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Jieting Tang
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Yifan Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
| | - Ding Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
| | - Wenyan Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
| | - Jing Zhang
- Institute for Translational Medicine on Cell Fate and Disease, Shanghai Ninth People’s Hospital, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Ma
- Department of Endocrinology and Metabolism, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming He
- Institute for Translational Medicine on Cell Fate and Disease, Shanghai Ninth People’s Hospital, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Yimin Mao
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Xu-Yun Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education and
- Department of Endocrinology and Metabolism, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Zwartsen A, Zeilmaker M, de Boer WJ, Rorije E, van der Voet H. Uncertainties in the Extrapolation of In Vitro Data in Human Risk Assessment: A Case Study of qIVIVE for Imazalil Using the Monte Carlo Risk Assessment Platform. Chem Res Toxicol 2025. [PMID: 40368340 DOI: 10.1021/acs.chemrestox.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
New approach methodologies (NAMs) are promising for refining, reducing, and replacing animal experiments for hazard characterization. Quantitative in vitro-in vivo extrapolation (qIVIVE) is essential to extrapolate an in vitro-based point of departure to an in vitro-based human equivalent dose and subsequently to an in vitro-based health-based guidance or threshold value. The use of NAMs for hazard characterization leads to the need for various new extrapolations and linked uncertainties that preferably are quantified. Currently, qIVIVE is often performed without addressing these uncertainties. A clear description and, if possible, quantification of extrapolations and uncertainties when using NAMs for risk assessment will aid the regulatory implementation of NAMs for risk assessment. A case study of a qIVIVE-based assessment on the risk of liver steatosis from dietary exposure to imazalil is reported, using a human cell line in vitro test method as an example of a NAM to replace animal experiments. We consider the uncertainties related to the extrapolations from in vitro to in vivo effects, from in vitro nominal concentrations to in vitro intracellular concentrations, from in vitro concentrations to external doses (reverse dosimetry), from in vitro exposure durations to in vivo exposure situations, and from the average human to a sensitive individual. The case study addresses these uncertainties in a mainly quantitative approach, using available data and the Monte Carlo Risk Assessment platform.
Collapse
Affiliation(s)
- Anne Zwartsen
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Marco Zeilmaker
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Waldo J de Boer
- Wageningen University & Research (WUR) Biometris, 6708 PB Wageningen, The Netherlands
| | - Emiel Rorije
- National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | - Hilko van der Voet
- Wageningen University & Research (WUR) Biometris, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
5
|
Chen C, Wang L. Aging and metabolic dysfunction-associated steatotic liver disease: a bidirectional relationship. Front Med 2025:10.1007/s11684-025-1133-7. [PMID: 40316793 DOI: 10.1007/s11684-025-1133-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/09/2025] [Indexed: 05/04/2025]
Abstract
In recent years, aging and cellular senescence have triggered an increased interest in corresponding research fields. Evidence shows that the complex aging process is involved in the development of many chronic liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). In fact, aging has a tremendous effect on the liver, leading to a gradual decline in the metabolism, detoxification and immune functions of the liver, which in turn increases the risk of liver disease. These changes can be based on the aging of liver cells (hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells, and Kupffer cells). Similarly, patients with liver diseases exhibit increases in the aging phenotype and aging cells, often manifesting as faster physical functional decline, which is closely related to the promoting effect of liver disease on aging. This review summarizes the interplay between MASLD/MASH development and aging, aiming to reveal the complex relationships that exacerbate one another. Moreover, the corresponding schemes for delaying aging or treating diseases are discussed to provide a basis for the development of effective prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
6
|
Singh V, Chattopadhyay P, Fatima F, Singh P, Pandey R, Agrawal A, Roy SS. Generation and characterization of a chronic in vitro model to study the early stage of metabolic dysfunction-associated steatotic liver disease (MASLD). Biochim Biophys Acta Mol Basis Dis 2025; 1871:167886. [PMID: 40324734 DOI: 10.1016/j.bbadis.2025.167886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic and progressive liver disease with an increasing global burden that starts with an early stage of simple steatosis (MASL) which frequently progresses to liver cirrhosis and hepatocellular carcinoma (HCC). Despite its widespread occurrence, the MASL or steatotic stage, characterized by excessive fat accumulation in the liver and considered reversible and benign, has not been extensively studied. To study MASL effectively, it is imperative to have a clinically relevant model system that focuses solely on steatosis, in a progressive and time-dependent manner, recapitulating molecular changes associated with human disease. We established a chronic cellular model of MASL using a primary immortalized human hepatocyte cell line treated with a low dose mixture of fatty acids. This model mimics the pattern of chronic disease progression, shows minimal lipotoxicity, exhibits progressive lipid accumulation (from early to moderate steatosis), and demonstrates macrosteatosis, a hallmark of MASL. To determine whether this model recapitulates both morphological and molecular aspects of steatosis, we measured the expression of key genes and pathways found to be dysregulated in a recently available early MASL patient dataset as well as a non-human primate model of MASL. In support of the relevance of our model, we observed increased fatty acid uptake, lipogenesis, mitochondrial activity, metabolic rewiring, and autophagic alterations that significantly overlap with the pathological features of human and non-human primate MASL. In conclusion, we generate a relevant cellular model of steatosis that can serve as a robust platform for screening of existing chemical libraries to identify potent inhibitors of MASL as well as discovering novel therapeutic targets by mechanistically studying altered molecular signatures associating early stages of MASLD.
Collapse
Affiliation(s)
- Vandana Singh
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Partha Chattopadhyay
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Fabeha Fatima
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India
| | - Praveen Singh
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Rajesh Pandey
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; Academy of Scientific & Innovative Research, Ghaziabad 201002, India
| | - Anurag Agrawal
- Trivedi School of Biosciences, Ashoka University, Sonipat 131029, India
| | - Soumya Sinha Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India; Academy of Scientific & Innovative Research, Ghaziabad 201002, India.
| |
Collapse
|
7
|
Cunneely OP, Roberts A, Fargue S, Knight J, Assimos DG, Wood KD. Metabolic dysfunction associated steatotic liver and kidney stones: what is going on? Curr Opin Nephrol Hypertens 2025; 34:247-253. [PMID: 39882643 DOI: 10.1097/mnh.0000000000001062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
PURPOSE OF REVIEW Metabolic dysfunction associated steatotic liver disease (MASLD) is increasing throughout the world, affecting nearly one in three individuals. Kidney stone disease, which is also increasing, is associated with MASLD. Common risk factors for both, including obesity, diabetes, dyslipidemia, hypertension, and metabolic syndrome, are likely drivers of this association. We present here a review of the associations and possible interconnections between these two common disease processes. RECENT FINDINGS Epidemiological studies are discordant regarding the impact of sex on this association and on the impact of MASLD on incident stone risk. The nature of kidney stones is rarely taken into account.A favorable milieu for uric acid kidney stone formation may be created by a lower urine pH resulting from defective ammonium production associated with insulin resistance, common in MASLD.Endogenous oxalate synthesis, a major risk factor for calcium oxalate kidney stones, may be increased in MASLD via decline in the activity of enzymes involved in the detoxification of glyoxylate, the immediate precursor of oxalate. SUMMARY The nature of kidney stones associated with MASLD and factors driving this association remain to be elucidated. Potential mechanisms identified underlying this include an increase in the risk factors for both uric acid and calcium oxalate kidney stones.
Collapse
Affiliation(s)
- Owen P Cunneely
- Department of Urology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | | | |
Collapse
|
8
|
Wang Y, Hong S, Hudson H, Kory N, Kinch LN, Kozlitina J, Cohen JC, Hobbs HH. PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by inhibiting ATGL-mediated triglyceride hydrolysis. J Hepatol 2025; 82:871-881. [PMID: 39550037 DOI: 10.1016/j.jhep.2024.10.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND & AIMS PNPLA3(148M) (patatin-like phospholipase domain-containing protein 3) is the most impactful genetic risk factor for steatotic liver disease. A key unresolved issue is whether PNPLA3(148M) confers a loss- or gain-of-function. Here we test the hypothesis that PNPLA3 causes steatosis by sequestering ABHD5 (α/β hydrolase domain-containing protein 5), the cofactor of ATGL (adipose TG lipase), thus limiting mobilization of hepatic triglyceride (TG). METHODS We quantified and compared the physical interactions between ABHD5 and PNPLA3/ATGL in cultured hepatocytes using NanoBiT complementation assays and immunocytochemistry. Recombinant proteins purified from human cells were used to compare TG hydrolytic activities of PNPLA3 and ATGL in the presence or absence of ABHD5. Adenoviruses and adeno-associated viruses were used to express PNPLA3 in liver-specific Atgl-/- mice and to express ABHD5 in livers of Pnpla3M/M mice, respectively. RESULTS ABHD5 interacted preferentially with PNPLA3 relative to ATGL in cultured hepatocytes. No differences were seen in the strength of the interactions between ABHD5 with PNPLA3(WT) and PNPLA3(148M). In contrast to prior findings, we found that PNPLA3, like ATGL, is activated by ABHD5 in in vitro assays using purified proteins. PNPLA3(148M)-associated inhibition of TG hydrolysis required that ATGL be expressed and that PNPLA3 be located on lipid droplets. Finally, overexpression of ABHD5 reversed the hepatic steatosis in Pnpla3M/M mice. CONCLUSIONS These findings support the premise that PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by accumulating on lipid droplets and inhibiting ATGL-mediated lipolysis in an ABHD5-dependent manner. Our results predict that reducing, rather than increasing, PNPLA3 expression will be the best strategy to treat PNPLA3(148M)-associated steatotic liver disease. IMPACT AND IMPLICATIONS Steatotic liver disease (SLD) is a common complex disorder associated with both environmental and genetic risk factors. PNPLA3(148M) is the most impactful genetic risk factor for SLD and yet its pathogenic mechanism remains controversial. Herein, we provide evidence that PNPLA3(148M) promotes triglyceride (TG) accumulation by sequestering ABHD5, thus limiting its availability to activate ATGL. Although the substitution of methionine for isoleucine reduces the TG hydrolase activity of PNPLA3, the loss of enzymatic function is not directly related to the steatotic effect of the variant. It is the resulting accumulation of PNPLA3 on LDs that confers a gain-of-function by interfering with ATGL-mediated TG hydrolysis. These findings have implications for the design of potential PNPLA3(148M)-based therapies. Reducing, rather than increasing, PNPLA3 levels is predicted to reverse steatosis in susceptible individuals.
Collapse
Affiliation(s)
- Yang Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA.
| | - Sen Hong
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Hannah Hudson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA
| | - Nora Kory
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lisa N Kinch
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA
| | - Jonathan C Cohen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Center for Human Nutrition, UTSW, Dallas, TX 75390, USA
| | - Helen H Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA; The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA.
| |
Collapse
|
9
|
Zheng DX, Ju BW, Wen LM, Xiang XY, Xue TT, Dai W, Liu JY, Hu JP, Yang JH. Efficacy and mechanism of iridoid glycosides from Gentianella turkestanorum (Gand.) Holub on non-alcoholic steatohepatitis based on RNA sequencing. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119888. [PMID: 40311720 DOI: 10.1016/j.jep.2025.119888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/08/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Non-alcoholic steatohepatitis (NASH) is an important component of non-alcoholic fatty liver disease (NAFLD), which is the most common chronic liver disease worldwide. Gentianella turkestanorum (Gands.) Holub is a traditional monk medicine used for the treatment of hepatitis, and total iridoid glycosides from it (GTI) are the main active substances in the treatment of liver diseases. However, the role and mechanism of GTI in NASH remain unclear. AIM OF THE STUDY This study aimed to assess the effectiveness of GTI in treating NASH induced by a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) and clarify the underlying mechanisms. METHODS The efficacy of GTI against aberrant lipid metabolism was evaluated in vitro in L02 cells and in vivo in a C57BL/6 NASH mouse model. Based on these evaluations in L02 cells and C57BL/6 NASH mice, swertiamarine (SWT) was identified as a potential active component of GTI. RNA sequencing was performed to further explore the therapeutic effects of SWT on the liver. The analysis identified the targeting of key signalling pathways PPARα, p53, and MAPKp38. The therapeutic efficacy of SWT was validated using siRNAs or agonists against PPARα or MAPKp38. RESULTS In this study, a combination of ex vivo and in vivo experiments was used to ascertain the mitigating effects of GTI and SWT on NASH. The efficacy was correlated with PPARα, p53, and MAPKp38 pathways, as determined by RNA sequencing. In vitro and in vivo experiments demonstrated that SWT influenced the expression of PPARα, p-p53, Caspase3, Bax, Bcl2, and p-MAPKp38. However, the efficacy of SWT is counteracted when PPARα is inhibited with siRNAs or MAPKp38 is activated with DHC in vitro. CONCLUSIONS SWT, a potential active ingredient in GTI, may have a therapeutic effect on abnormal L02 lipid metabolism and NASH in mice by affecting the PPARα/MAPK/p53 signalling pathway. Therefore, SWT holds potential clinical application in the prevention and treatment of NASH.
Collapse
Affiliation(s)
- Dong-Xuan Zheng
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China.
| | - Bo-Wei Ju
- Department of Pharmacy, The Fifth affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Li-Mei Wen
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China; Department of Pharmacy, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China.
| | - Xue-Ying Xiang
- Department of Pharmacognosy, School of Public Health, Xinjiang Medical University, Urumqi, China.
| | - Tao-Tao Xue
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China.
| | - Wu Dai
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China; Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China.
| | - Jiang-Yun Liu
- College of Pharmaceutical Science, Soochow University, Suzhou, China.
| | - Jun-Ping Hu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China.
| | - Jian-Hua Yang
- Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China; Department of Pharmacy, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China.
| |
Collapse
|
10
|
Zhang M, Yuan W, Li C, Chen C, Liu X, Ma Z, Xiang Y, Chen G, Wang C, Li L, Wang L, Xu Z, Xu C. Resveratrol and N-acetylcystein reduce hepatic steatosis but enhance initiation and progression of hepatocellular carcinoma by inhibiting GST-pi-MAPK axis in mice. Front Pharmacol 2025; 16:1574039. [PMID: 40356978 PMCID: PMC12066552 DOI: 10.3389/fphar.2025.1574039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Accumulating evidence indicates that antioxidants promote tumor growth and metastasis after tumor onset in several cancer types. However, whether antioxidants prevent or accelerate hepatic tumorigenesis during steatosis remains unknown. Therefore, we investigated the effects of resveratrol (RES) and N-acetylcysteine (NAC) on hepatocellular carcinoma (HCC) development using two fatty liver mouse models. Methods High-fat diet (HFD) plus diethylnitrosamine (DEN)- and AKT/Ras-induced primary HCC mouse models were used. The weight, liver weight ratio and the number of HCC tumors were calculated and histological features of mouse HCC tissues were analyzed using immumohistochemical staining such as hematoxylin and eosin staining. Proteomic analysis was used to screen for differences in liver cancer progression between antioxidant-treated HCC and models. Protein inhibitor recovery experiments in mice and in vitro cells validate the targets screened by proteomic analysis. The expression of GST-pi, p-JNK and p-p38 signaling molecules in HCC were investigated using Western blotting. Results RES and NAC enhance HCC formation in both DEN/HFD and AKT/Ras mice. RES and NAC alleviate hepatosteatosis, and reduce ROS and DNA damage in mice. Proteomic analysis and protein inhibitor recovery assay demonstrated that GST-pi is a therapeutic target for antioxidant-induced hepatocellular carcinoma growth. Mechanistically, RES and NAC decreased p-JNK and p-p38, the two major mitogen-activated protein kinases, in HCC cells. Blockade of GST-pi abrogated the reduction in p-JNK and p-p38 levels and increased apoptosis of HCC cells. Conclusion Antioxidants may increase the incidence of HCC in a population with fatty liver, despite reduction in ROS production, by inhibiting GST-pi-MAPK axis.
Collapse
Affiliation(s)
- Mi Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Weigang Yuan
- Department of Clinical Laboratory, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Chun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chanyuan Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhilu Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifei Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guisha Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Chunxu Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhong Xu
- Department of Gastroenterology and Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Health Management Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuanrui Xu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Yang R, Bian H, Zhou Z, Yang Y, Wang X, Xu J, Zhong W, Zhu L. Underlying mechanisms of metabolic dysfunction-associated steatotic liver disease induced by 2-ethylhexyl diphenyl phosphate and its hydroxylated metabolite in zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138407. [PMID: 40300519 DOI: 10.1016/j.jhazmat.2025.138407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025]
Abstract
2-Ethylhexyl diphenyl phosphate (EHDPHP) is ubiquitous in various environmental media and organisms. Due to its susceptibility to biotransformation, its primary product 2-ethyl-5-hydroxyhexyl diphenyl phosphate (5-OH-EHDPHP) is almost at equal level in organisms. However, their hepatotoxicity remains unclear. In this study, adult zebrafish were exposed to 5, 35, or 245 µg/L of EHDPHP for 28 days. Distinct metabolic dysfunction-associated steatotic liver disease (MASLD) was observed in treated zebrafish, indicated by increased hepatic lipid levels (total cholesterol, triglycerides, nonesterified fatty acids, and fat droplets), steatosis (hepatic ballooning), and inflammation (tnf-α and il-6). Combined the in vitro hepatic cell test, molecular docking and molecular dynamics simulation, it was revealed that peroxisome proliferator-activated receptor gamma (pparγ) was upregulated upon EHDPHP exposure, thereby facilitating lipid synthesis and hepatic lipid accumulation. Notably, its main metabolite 5-OH-EHDPHP induced stronger hepatocyte toxicity and PPARγ transcription. Additionally, serious liver function damage was observed, with aspartate aminotransferase, alanine transaminase, albumin, and γ-glutamyl transferase levels markedly disrupted. This increases the risk of development of cardiovascular disease, hepatic cirrhosis or other chronic conditions. Collectively, the results demonstrate that EHDPHP may cause strong hepatic toxicities, which may be pounded by its hydroxylated metabolites.
Collapse
Affiliation(s)
- Rongyan Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Hanfei Bian
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Zhou Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yi Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Xiao Wang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Jingshu Xu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China.
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China.
| |
Collapse
|
12
|
Chen R, Chen T, Li X, Yu J, Lin M, Wen S, Zhang M, Chen J, Yi B, Zhong H, Li Z. SREBP2 as a central player in cancer progression: potential for targeted therapeutics. Front Pharmacol 2025; 16:1535691. [PMID: 40308757 PMCID: PMC12041066 DOI: 10.3389/fphar.2025.1535691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Recent studies have identified the reprogramming of lipid metabolism as a critical hallmark of malignancy. Enhanced cholesterol uptake and increased cholesterol biosynthesis significantly contribute to the rapid growth of tumors, with cholesterol also playing essential roles in cellular signaling pathways. Targeting cholesterol metabolism has emerged as a promising therapeutic strategy in oncology. The sterol regulatory element-binding protein-2 (SREBP2) serves as a primary transcriptional regulator of genes involved in cholesterol biosynthesis and is crucial for maintaining cholesterol homeostasis. Numerous studies have reported the upregulation of SREBP2 across various cancers, facilitating tumor progression. This review aims to provide a comprehensive overview of the structure, biological functions, and regulatory mechanisms of SREBP2. Furthermore, we summarize that SREBP2 plays a crucial role in various cancers and tumor microenvironment primarily by regulating cholesterol, as well as through several non-cholesterol pathways. We also particularly emphasize therapeutic agents targeting SREBP2 that are currently under investigation. This review seeks to enhance our understanding of SREBP2's involvement in cancer and provide theoretical references for cancer therapies that target SREBP2.
Collapse
Affiliation(s)
- Ruiqi Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tianyu Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiang Li
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junfeng Yu
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Lin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siqi Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Man Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinchi Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bei Yi
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huage Zhong
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Zhao Li
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
13
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
14
|
Luque-Urbano MR, Fernández-Ramos D, Lopitz-Otsoa F, Gutiérrez de Juan V, Bizkarguenaga M, Castro-Espadas L, Hermoso-Martínez U, Barbier-Torres L, Lu SC, Millet O, Mato JM. S-adenosylmethionine deficit disrupts very low-density lipoprotein metabolism promoting liver lipid accumulation in mice. J Lipid Res 2025; 66:100794. [PMID: 40180215 DOI: 10.1016/j.jlr.2025.100794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/20/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025] Open
Abstract
Hepatic deletion of methionine adenosyltransferase-1a (Mat1a) in mice reduces S-adenosylmethionine (SAMe), a key methyl donor essential for many biological processes, which promotes the development and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Hyperglycemia and reduced MAT1A expression, along with low SAMe levels, are common in MASLD patients. This study explores how Mat1a-knockout (KO) hepatocytes respond to prolonged high glucose conditions, focusing on glucose metabolism and lipid accumulation. Hepatocytes from methionine adenosyltransferase-1a-knockout (Mat1a-KO) mice were incubated in high glucose conditions overnight, allowing for analysis of key metabolic intermediates and gene expression related to glycolysis, gluconeogenesis, glyceroneogenesis, phospholipid synthesis, and very low density lipoprotein (VLDL) secretion. SAMe deficiency in Mat1a-KO hepatocytes led to reduced protein methyltransferase-1 activity, resulting in increased expression of glycolytic enzymes (glucokinase, phosphofructokinase, and pyruvate kinase) and decreased expression of gluconeogenic enzymes (phosphoenolpyruvate carboxykinase, fructose-1,6-bisphosphatase, and glucose-6-phosphatase). These alterations led to a reduction in dihydroxyacetone phosphate (DHAP), which subsequently inhibited mammalian target of rapamycin complex 1 (mTORC1) activity. This inhibition resulted in decreased phosphatidylcholine synthesis via the CDP-choline pathway and impaired VLDL secretion, ultimately causing lipid accumulation. Thus, under high glucose conditions, SAMe deficiency in hepatocytes depletes DHAP, inhibits mTORC1 activity, and promotes lipid buildup.
Collapse
Affiliation(s)
- María R Luque-Urbano
- Atlas Molecular Pharma, Derio, Spain; Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - David Fernández-Ramos
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Fernando Lopitz-Otsoa
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Virginia Gutiérrez de Juan
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Maider Bizkarguenaga
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Lia Castro-Espadas
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Uxue Hermoso-Martínez
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Oscar Millet
- Atlas Molecular Pharma, Derio, Spain; Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - José M Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
15
|
Osmani Z, Brouwer WP, Grashof DGB, Lim Y, Doukas M, Janssen HLA, van de Werken HJG, Boonstra A. Metabolic dysfunction-associated steatohepatitis reduces interferon and macrophage liver gene signatures in patients with chronic hepatitis B. J Hepatol 2025; 82:594-603. [PMID: 39490745 DOI: 10.1016/j.jhep.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND & AIMS Patients with chronic HBV infection and concomitant metabolic dysfunction-associated steatohepatitis (MASH) have been shown to develop more severe liver disease than patients with chronic HBV alone. However, our understanding of the underlying mechanisms is limited. Herein, we study how comorbid MASH impacts immune activity in the livers of patients with chronic HBV infection. METHODS Bulk RNA sequencing was performed on liver biopsies from patients with only MASH (n = 10), only HBeAg-negative chronic HBV (ENEG; n = 11), combined MASH/ENEG (n = 9) and healthy controls (n = 9). Biopsies with no or minimal fibrosis (≤F2) were selected to avoid confounding effects of fibrosis. We compared whole transcriptome data from patients with MASH/ENEG to those with ENEG alone to determine the impact of comorbid MASH on chronic hepatitis B. RESULTS There is a high degree of overlap of liver gene expression profiles in patients with only ENEG vs. those with only MASH compared to healthy controls, suggesting a largely shared mechanism of liver dysfunction and immune activity for these distinct conditions. In patients with ENEG, comorbid MASH was associated with significantly reduced interferon pathway activity (normalized enrichment score = 2.03, p.adj = 0.0251), the expression of interferon-stimulated genes (e.g., IFIT2, IFI27, IFITM1, IFI6), and macrophage gene signatures (e.g., MARCO, CD163, CD5L, CD63), when compared to patients with ENEG alone. CONCLUSIONS Transcriptomic profiling of the liver suggests that MASH negatively impacts interferon-stimulated gene expression and macrophage gene signatures in the livers of patients with ENEG, which may affect antiviral immune pathways, viral replication and inflammatory responses, resulting in an increased risk of advanced fibrosis in patients with chronic hepatitis B. Our study provides valuable insights for guiding future research aimed at developing effective, tailored strategies for managing patients with both conditions. IMPACT AND IMPLICATIONS In recent decades, obesity and associated health issues have reached epidemic levels, with steatotic liver disease affecting up to 30% of adults in developed countries, and this trend is also observed among patients with chronic hepatitis B. Given the high and rising prevalence of steatotic liver disease and its frequent co-occurrence in patients with chronic hepatitis B, it is essential to understand how conditions such as metabolic dysfunction-associated steatohepatitis (MASH) impact immune responses in the liver. This study provides unique insights into the impact of MASH on HBV antiviral immune activity in the livers of patients with chronic hepatitis B. The rising number of patients with both conditions affects treatment outcomes and highlights the urgent need for novel, tailored therapeutic strategies. Our study holds significant relevance for guiding future research on developing treatment strategies for patients with both MASH and chronic hepatitis B.
Collapse
Affiliation(s)
- Zgjim Osmani
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Willem Pieter Brouwer
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dwin G B Grashof
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Youkyung Lim
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michael Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands; Toronto General Hospital, University of Toronto, Canada
| | | | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
16
|
Peng H, Wang B, Yang W, Jia R, Luo Y, Chen W. Association Between Triglyceride and Incident Diabetes Mellitus: A Secondary Retrospective Analysis Based on a Chinese Cohort Study. J Multidiscip Healthc 2025; 18:1779-1790. [PMID: 40171237 PMCID: PMC11960458 DOI: 10.2147/jmdh.s510549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 04/03/2025] Open
Abstract
Background The association between triglyceride(TG) levels and the risk of diabetes mellitus (DM) continues to be a subject of considerable interest and debate within the scientific community. To date, there has been a lack of studies specifically examining this relationship within the Chinese population. This study seeks to elucidate the correlation between TG levels and the incidence of DM among the Chinese demographic. Methods This study constitutes a secondary analysis of a retrospective cohort investigation comprising 202,888 Chinese participants who were free of DM at baseline and were subsequently followed from 2010 to 2016. Cox regression method and sensitivity analyses were used to examine the relationship between TG levels and DM. To examine the potential non-linear relationship between TG levels and the incidence of DM, Cox proportional hazards regression incorporating cubic spline functions and smooth curve fitting was employed. Additionally, a two-piece Cox proportional hazards regression model was utilized to identify the inflection point at which TG levels influence the risk of developing DM. Results In participants with DM, baseline TG levels were elevated. After adjusting for confounding variables, baseline TG levels were positively associated with incident DM. (HR:1.25,95% CI:1.21-1.30,P<0.001). In addition, we conducted sensitivity analyses to ensure the results were robust. There was a 88% increase in DM risk from the top TG tertile to the bottom TG tertile.Our research discovered a significant link between TG and DM when TG levels were below 1.27 mmol/L (HR:2.35, 95% CI: 1.95-2.83,P < 0.001). Conclusion This study shows that TG was positively and non-linearly associated with the risk of DM after adjusting for other confounding factors.Below 1.27 mmol/L, increasing TG levels greatly heighten the risk of DM, whereas above this level, the risk is lower.
Collapse
Affiliation(s)
- Hui Peng
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Bin Wang
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Wei Yang
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Rui Jia
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Youlian Luo
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| | - Weifeng Chen
- Department of General Practice, Shenzhen second People’s Hospital (The First Affiliated of Shenzhen University), Shenzhen, Guangdong, 518000, People’s Republic of China
| |
Collapse
|
17
|
Askeland A, Rasmussen RW, Gjela M, Frøkjær JB, Højlund K, Mellergaard M, Handberg A. Non-invasive liver fibrosis markers are increased in obese individuals with non-alcoholic fatty liver disease and the metabolic syndrome. Sci Rep 2025; 15:10652. [PMID: 40148373 PMCID: PMC11950363 DOI: 10.1038/s41598-025-85508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/03/2025] [Indexed: 03/29/2025] Open
Abstract
The need for early non-invasive diagnostic tools for chronic liver fibrosis is growing, particularly in individuals with obesity, non-alcoholic fatty liver disease (NAFLD), and the metabolic syndrome (MetS) since prevalence of these conditions is increasing. This case-control study compared non-invasive liver fibrosis markers in obesity with NAFLD and MetS (NAFLD-MetS, n = 33), in obese (n = 28) and lean (n = 27) control groups. We used MRI (T1 relaxation times (T1) and liver stiffness), circulating biomarkers (CK18, PIIINP, and TIMP1), and algorithms (FIB-4 index, Forns score, FNI, and MACK3 score) to assess their potential in predicting liver fibrosis risk. We found that T1 (892 ± 81 ms vs. 818 ± 64 ms, p < 0.001), FNI (15 ± 12% vs. 9 ± 7%, p = 0.018), CK18 (166 ± 110 U/L vs. 113 ± 41 U/L, p = 0.019), and MACK3 (0.18 ± 0.15 vs. 0.05 ± 0.04, p < 0.001) were higher in the NAFLD-MetS group compared with the obese control group. Moreover, correlations were found between CK18 and FNI (r = 0.69, p < 0.001), CK18 and T1 (r = 0.41, p < 0.001), FNI and T1 (r = 0.33, p = 0.006), MACK3 and FNI (r = 0.79, p < 0.001), and MACK3 and T1 (r = 0.50, p < 0.001). We show that liver fibrosis markers are increased in obese individuals with NAFLD and MetS without clinical signs of liver fibrosis. More studies are needed to validate the use of these non-invasive biomarkers for early identification of liver fibrosis risk.
Collapse
Affiliation(s)
- Anders Askeland
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Mimoza Gjela
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
18
|
Li W, Shi X, Zhang D, Hu J, Zhao S, Ye S, Wang J, Liu X, Zhang Q, Wang Z, Zhang Y, Yan L. Adipose derived mesenchymal stem cell-seeded regenerated silk fibroin scaffolds reverse liver fibrosis in mice. J Mater Chem B 2025; 13:4201-4213. [PMID: 40059659 DOI: 10.1039/d5tb00275c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Liver fibrosis (LF) is an important process in the progression of chronic liver disease to cirrhosis. We have previously demonstrated that a regenerated silk fibroin scaffold loaded with adipose-derived stem cells (RSF + ADSCs) can repair acute liver injury. In this study, we established a chronic LF animal model using carbon tetrachloride (CCl4) and a high-fat diet. We then investigated the liver repair capacity after transplanting RSF + ADSC scaffolds and RSF scaffolds onto the liver surface of mice. Compared with the control group, the concentrations of ALT and AST in the serum were significantly reduced in the RSF and RSF + ADSC groups. HE staining and Masson trichrome staining revealed a decrease in the SAF score in both the RSF and RSF + ADSC groups. Meanwhile, the biomarkers of blood vessels and bile ducts, such as CD34, ERG, muc1, and CK19, were significantly elevated in the RSF + ADSC group. Finally, transcriptome analysis showed that the PPAR signaling pathway, which inhibits liver fibrosis, was significantly upregulated in both the RSF and RSF + ADSC groups. Our study suggests that, compared with RSF scaffolds alone, RSF + ADSCs have a significant repair effect on chronic LF in mice.
Collapse
Affiliation(s)
- Weilong Li
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Xiaonan Shi
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Daxu Zhang
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Jingjing Hu
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Shuo Zhao
- Department of Critical Care Medicine,Aerospace Central Hospital,Beijing,, PR China
| | - Shujun Ye
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Jingyi Wang
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| | - Xiaojiao Liu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, PR China.
| | - Qian Zhang
- School of nursing, Lanzhou University, Gansu 730000, PR China
| | - Zhanbo Wang
- Department of Pathology, Chinese PLA General Hospital, Beijing 100853, P. R. China.
| | - Yaopeng Zhang
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, PR China.
| | - Li Yan
- The Second Medical Center and National Clinical Research Center of Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, PR China.
| |
Collapse
|
19
|
Chen F, Sarver DC, Saqib M, Velez LM, Aja S, Seldin MM, Wong GW. Loss of CTRP10 results in female obesity with preserved metabolic health. eLife 2025; 13:RP93373. [PMID: 40126547 PMCID: PMC11932693 DOI: 10.7554/elife.93373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Obesity is a major risk factor for type 2 diabetes, dyslipidemia, cardiovascular disease, and hypertension. Intriguingly, there is a subset of metabolically healthy obese (MHO) individuals who are seemingly able to maintain a healthy metabolic profile free of metabolic syndrome. The molecular underpinnings of MHO, however, are not well understood. Here, we report that CTRP10/C1QL2-deficient mice represent a unique female model of MHO. CTRP10 modulates weight gain in a striking and sexually dimorphic manner. Female, but not male, mice lacking CTRP10 develop obesity with age on a low-fat diet while maintaining an otherwise healthy metabolic profile. When fed an obesogenic diet, female Ctrp10 knockout (KO) mice show rapid weight gain. Despite pronounced obesity, Ctrp10 KO female mice do not develop steatosis, dyslipidemia, glucose intolerance, insulin resistance, oxidative stress, or low-grade inflammation. Obesity is largely uncoupled from metabolic dysregulation in female KO mice. Multi-tissue transcriptomic analyses highlighted gene expression changes and pathways associated with insulin-sensitive obesity. Transcriptional correlation of the differentially expressed gene (DEG) orthologs in humans also shows sex differences in gene connectivity within and across metabolic tissues, underscoring the conserved sex-dependent function of CTRP10. Collectively, our findings suggest that CTRP10 negatively regulates body weight in females, and that loss of CTRP10 results in benign obesity with largely preserved insulin sensitivity and metabolic health. This female MHO mouse model is valuable for understanding sex-biased mechanisms that uncouple obesity from metabolic dysfunction.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Leandro M Velez
- Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marcus M Seldin
- Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
20
|
Tsutsumi T, Kawaguchi T, Fujii H, Kamada Y, Suzuki Y, Sawada K, Tatsuta M, Maeshiro T, Tobita H, Akahane T, Hasebe C, Kawanaka M, Kessoku T, Eguchi Y, Syokita H, Nakajima A, Kamada T, Yoshiji H, Sakugawa H, Morishita A, Masaki T, Ohmura T, Watanabe T, Yoda Y, Enomoto N, Ono M, Fuyama K, Okada K, Nishimoto N, Ito YM, Takahashi H, Charlton MR, Rinella ME, Sumida Y. Low HDL cholesterol levels in women and hypertriglyceridemia in men: predictors of MASLD onset in individuals without steatosis. J Gastroenterol 2025:10.1007/s00535-025-02242-y. [PMID: 40097845 DOI: 10.1007/s00535-025-02242-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Individuals with metabolic-associated steatotic liver disease (MASLD) have a worse prognosis compared to patients without steatosis, and its prevalence is increasing. However, detailed risk factors based on obesity and sex remain unclear. We aimed to investigate the impact of cardiometabolic risk factors (CMRFs) on the risk of MASLD in individuals without pre-existing SLD. METHODS SLD was diagnosed by ultrasonography. Non-SLD individuals were followed 65,657 person-years. Incidence rates of MASLD were assessed by Kaplan-Meier analysis. Furthermore, independent factors associated with the development of MASLD were identified using Cox regression analysis, stratified by four groups: obese men, non-obese men, obese women, and non-obese women. RESULTS The overall incidence rate of MASLD was 39.3/1,000 person-years. The cumulative incidence was highest in obese men, followed by obese women, non-obese men, and non-obese women. Two or more CMRFs increased the risk of MASLD in all groups. Low HDL cholesterol level was the strongest independent risk factor in both obese and non-obese women and hypertriglyceridemia for both obese and non-obese men. The impact of these CMRFs was stronger in non-obese individuals. (HR [95% CI]: women non-obese 1.9 [1.5-2.4], obese 1.4 [1.1-1.8]; men non-obese 2.3 [1.9-2.9], obese 1.5 [1.2-2.0]). CONCLUSIONS Multiple CMRFs are important to MASLD development, regardless of sex and obesity. In this Japanese cohort, low HDL cholesterol in women and hypertriglyceridemia in men were the most significant risk factors, especially among the non-obese group. These findings suggest that sex-specific CMRFs may play a role in the development of MASLD.
Collapse
Affiliation(s)
- Tsubasa Tsutsumi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA.
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Hideki Fujii
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abeno, Osaka, 545-8585, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Suzuki
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo-Shi, Yamanashi, 409-3898, Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1, Midorigaoka Higashi, Asahikawa-City, Hokkaido, 078-8510, Japan
| | - Miwa Tatsuta
- Department of Gastroenterology, KKR Takamatsu Hospital, 4-18 Tenjinmae, Takamatsu, Kagawa, 760-0018, Japan
| | - Tatsuji Maeshiro
- First Department of Internal Medicine, University of the Ryukyus Hospital, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Tobita
- Department of Hepatology, Shimane University Hospital, 89-1 Enya-Cho, Izumo, Shimane, 693-8501, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Uda City Hospital, 815 Haibarahagihara, Uda, Nara, 633-0253, Japan
| | - Chitomi Hasebe
- Department of Gastroenterology, Japanese Red Cross Asahikawa Hospital, Akebono-1Jo 1Chome 1-1, Asahikawa-City, Hokkaido, 070-0061, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, Kawasaki Medical School, Kawasaki Medical Center, 2-6-1, Nakayamashimo, Kita, Okayama, 700-8505, Japan
| | - Takaomi Kessoku
- Kanagawa Dental University Yokohama Clinic, Yokohama, Kanagawa, Japan
- Department of Palliative Medicine and Gastroenterology, International University Health and Welfare Narita Hospital, 4-3, Kimizunomori 4-Chome, Narita-Shi, Chiba, 286-8686, Japan
- Division of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Yuichiro Eguchi
- Loco Medical General Institute, Mikatsukicho Kanada, Ogi-Shi, Saga, 845-0032, Japan
| | - Hayashi Syokita
- Department of Gastroenterology, Northern OKINAWA Medical Center, 1712-3 Umusa, Nago, Okinawa, 905-0006, Japan
| | - Atsushi Nakajima
- Division of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9, Fukuura, Kanazawa-Ku, Yokohama, 236-0004, Japan
| | - Tomoari Kamada
- Department of General Internal Medicine 2, Kawasaki Medical School, Kawasaki Medical Center, 2-6-1, Nakayamashimo, Kita, Okayama, 700-8505, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Uda City Hospital, 815 Haibarahagihara, Uda, Nara, 633-0253, Japan
| | - Hiroshi Sakugawa
- Department of Gastroenterology, Heartlife Hospital, 208, Iju, Nakagusuku, Nakagami, Okinawa, 901-2492, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Takumi Ohmura
- Department of Health Care, Asahikawa-Kosei General Hospital, 24-111, 1 Jo-Dori, Asahikawa-City, Hokkaido, 078-8211, Japan
| | - Toshio Watanabe
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, 1-7, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshioki Yoda
- JA Yamanashi Koseiren Health Care Center, 1-26, Iida 1, Kofu, Yamanashi, 400-0035, Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, 1110, Shimokato, Chuo-Shi, Yamanashi, 409-3898, Japan
| | - Masafumi Ono
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Oaza Ikenobe Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Kanako Fuyama
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Naoki Nishimoto
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Yoichi M Ito
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Hirokazu Takahashi
- Liver Center, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Michael R Charlton
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA
| | - Mary E Rinella
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Chicago, 5841 South Maryland Ave., Chicago, IL, 60637, USA
| | - Yoshio Sumida
- Graduate School of Healthcare Management, International University of Healthcare and Welfare, 4-1-26, Akasaka, Minato-Ku, Tokyo, Japan
| |
Collapse
|
21
|
Bahrami G, Sajadimajd S, Kazemi F, Yarmohammadi F, Miraghaee SS. An oligosaccharide isolated from Rosa canina ameliorates lipid profile and liver damage in MASLD modeled rabbits: in vivo and in silico studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04027-9. [PMID: 40095053 DOI: 10.1007/s00210-025-04027-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is designated as the main hepatic evidence of metabolic syndrome. Over the past few decades, the use of herbal medications in the treatment of MASLD has been increasingly studied due to evidence of their potential therapeutic mechanisms, wide availability, and lower side effects. This study aimed to scrutinize the effect of an oligosaccharide isolated from Rosa canina on high-cholesterol diet-induced MASLD rabbits. Twenty-four New Zealand rabbits were categorized into three groups (eight rabbits in each group). The first group received only the standard diet. Others received 2% cholesterol for 120 days. Then, the rabbits were treated with 20 mg/kg of an isolated oligosaccharide daily by gavage for 60 days or 6 mg/kg of simvastatin as a standard. After 14-16 h of starvation, blood samples were collected to measure lipid profile and liver enzymes. In addition, histological sampling of the liver and thoracic aorta was done. Cholesterol and triglyceride were significantly decreased in the oligosaccharide-treated group (p < 0.05). Also, the activity of alanine aminotransferase, aspartate aminotransferase, and creatine phosphokinase decreased significantly. Lactate dehydrogenase activity was also decreased. Based on histopathological studies, the treatment with an isolated oligosaccharide prevented atherosclerotic changes and decreased liver injury. Our data suggest that an oligosaccharide isolated from Rosa canina possesses hypolipidemic and hepatoprotective activities which will be beneficial in MASLD.
Collapse
Affiliation(s)
- Gholamreza Bahrami
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran
| | - Farnoush Kazemi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Shahram Miraghaee
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
22
|
Zhang L, Du Y, Li Y, Wang T, Pan Y, Xue X, Mu X, Qiu J, Qian Y. Mitochondrial mechanism of florfenicol-induced nonalcoholic fatty liver disease in zebrafish using multi-omics technology. JOURNAL OF HAZARDOUS MATERIALS 2025; 486:136958. [PMID: 39724715 DOI: 10.1016/j.jhazmat.2024.136958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Florfenicol (FF), a third-generation chloramphenicol antibiotic widely used in food-producing animals, has become a "pseudopersistent" environmental contaminant, raising concerns about its potential ecological and human health impacts. However, its bioaccumulation behavior and hepatotoxic mechanisms remain poorly understood. This study aims to address these gaps with a 28-day exposure experiment in adult zebrafish at 0.05 and 0.5 mg/L FF. Multiomic analyses (metabolomics, lipidomics, and transcriptomics), combined with histological and mitochondrial function assessments, were employed. Higher bioaccumulation was observed at 0.05 mg/L, potentially due to metabolic saturation at higher concentrations. Histological analysis revealed significant hepatic steatosis (>5 % steatosis area), indicative of moderate nonalcoholic fatty liver disease (NAFLD). Multiomic data demonstrated global dysregulation in energy metabolism, including marked alterations in lipids (accumulation of toxic sphingolipids, excessive fatty acids, and acylglycerol), amino acids, tricarboxylic acid cycle intermediates, and nucleotides. Crucially, mitochondrial dysfunction was identified as a central mechanism, with impaired respiratory chain activities, adenosine triphosphate depletion, elevated reactive oxygen species, and oxidative stress promoting NAFLD progression. These findings highlight mitochondrial impairment and oxidative stress as key drivers of FF-induced hepatotoxicity, providing novel insights into its toxicological mechanisms and emphasizing the ecological risks posed by antibiotic pollution in aquatic systems.
Collapse
Affiliation(s)
- Lin Zhang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China; Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Yang Du
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yameng Li
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Tiancai Wang
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yecan Pan
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaofeng Xue
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Xiyan Mu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jing Qiu
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Yongzhong Qian
- Key Laboratory of Agri-food Quality and Safety of Ministry of Agriculture and Rural Affairs, Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
23
|
Meyer J, Teixeira AM, Richter S, Larner DP, Syed A, Klöting N, Matz-Soja M, Gaul S, Barnikol-Oettler A, Kiess W, Le Duc D, Penke M, Garten A. Sex differences in diet-induced MASLD - are female mice naturally protected? Front Endocrinol (Lausanne) 2025; 16:1567573. [PMID: 40162312 PMCID: PMC11949793 DOI: 10.3389/fendo.2025.1567573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Males suffer more often from profibrotic changes in liver than females. The underlying mechanism for this sex difference in the prevalence and manifestation of Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) is not yet completely known. We studied male and female mice that were induced to develop MASLD by consuming a "fast food" diet (FFD) and assessed metabolic phenotype as well as liver histology and compared them with mice fed with a matched control diet (CD). Our aim was to check for sex-specific differences in MASLD development in a mouse model of diet-induced profibrotic changes in the liver. Our results demonstrate a clear difference in body weight, fat distribution and changes in liver tissue for male and female mice fed with FFD. We found that female mice stored lipids mainly in subcutaneous and visceral adipose tissue while males increased ectopic lipid accumulation in the liver which resulted in hepatomegaly and increased transforming growth factor β 1 (Tgfb1) and collagen I (Col1a1) expression concomitant to fibrosis development. This was absent in female mice. Analysis of estrogen receptor -α (Esr1) and -β (Esr2) expression revealed an upregulation of Esr2 in livers of male FFD-fed mice whereas in female liver tissue a higher expression in Esr1 could be observed. This study supports Esr1 and Esr2 as potential targets to reverse negative effects of diet-induced profibrotic changes in the liver.
Collapse
Affiliation(s)
- Jana Meyer
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Ana Mendes Teixeira
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Sandy Richter
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Dean P. Larner
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Asifuddin Syed
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) belonging to Helmholtz Center Munich at the University and University Hospital, Leipzig, Germany
| | - Madlen Matz-Soja
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Susanne Gaul
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
- Klinik und Poliklinik für Kardiologie, University Hospital Leipzig, Leipzig University, Leipzig, Germany
| | - Anja Barnikol-Oettler
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig, Germany
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Melanie Penke
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research, University Hospital for Children and Adolescents, Leipzig University, Leipzig, Germany
| |
Collapse
|
24
|
Qaisar SA, Moludi J, Shahnazi N, Soleimani D, Pasdar Y. Relationship of tea consumption with hepatic steatosis and fibrosis: findings from a longitudinal RaNCD cohort. BMC Nutr 2025; 11:47. [PMID: 40033335 DOI: 10.1186/s40795-025-01032-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Tea, known for its high content of antioxidants and anti-inflammatory compounds such as catechins, is believed to support liver health. This study aimed to explore the relationship between tea consumption and hepatic steatosis and fibrosis. METHODS This longitudinal study involved 2,537 participants from the Ravanser Non-Communicable Disease (RaNCD) cohort, conducted from 2015 to 2023. Dietary intake was evaluated using a 118-item food frequency questionnaire (FFQ). The Fibrosis-4 (FIB-4) index and the Hepatic Steatosis Index (HSI) were utilized as predictive indicators for hepatic fibrosis and steatosis, respectively. RESULTS After adjusting for potential confounding factors, our findings indicated that tea consumption was not significantly associated with an increased risk of worsening hepatic steatosis or fibrosis (P-value > 0.05). However, participants who consumed more than 2.88 cups of tea per day had a 27% lower likelihood of experiencing improvement in hepatic steatosis compared to those who consumed less than 1.92 cups per day (Relative Risk: 0.73; 95% CI: 0.53-0.99; P-value: 0.046). CONCLUSION Our study suggests that higher tea consumption is not significantly linked to an elevated risk of worsening hepatic steatosis or fibrosis. However, it is noteworthy that individuals who consumed more tea were less likely to see improvements in hepatic steatosis. This finding highlights the need for further research to better understand the potential effects of tea on liver health.
Collapse
Affiliation(s)
- Shaimaa A Qaisar
- Garmin University Research Center, kalar. Sulymaniyah, Iraq
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jalal Moludi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Research Center of Oils and Fats, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Narges Shahnazi
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Davood Soleimani
- Nutrition Sciences and Food Technology Research Center, Health Institute, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Yahya Pasdar
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
25
|
Jung W, Kim MH, Yang JW, Kim DC, Lee JS, Lee JH, An HJ, Song DH. Boron-dipyrromethene Staining May Enhance Fat Detection in the MASLD Zebrafish Model: NGS-validated lncRNA Profiling. In Vivo 2025; 39:749-757. [PMID: 40010962 PMCID: PMC11884486 DOI: 10.21873/invivo.13879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 02/28/2025]
Abstract
BACKGROUND/AIM Metabolic dysfunction-associated steatotic liver disease (MASLD) is a serious global public health concern. Long non-coding RNAs (lncRNAs) have been identified as key contributors to MASLD pathogenesis. Zebrafish can be utilized to study the relationship between MASLD and lncRNAs because of their similarity to human genes. Oil Red O staining is a traditional method for confirming liver fatty changes; however, it has several limitations. This study aimed to evaluate the efficacy of boron-dipyrromethene (BODIPY) in detecting fatty changes in the liver. MATERIALS AND METHODS Liver tissues were collected from 30 zebrafish that were fed a BODIPY-containing high-cholesterol diet. Oil Red O and BODIPY staining were evaluated by two pathologists, and next-generation sequencing (NGS) was performed using liver tissues categorized into high fatty change (six liver tissues) and low fatty change (six liver tissues) groups. RESULTS BODIPY and Oil Red O staining of zebrafish liver sections correlated significantly (p=0.009). NGS identified eight differentially expressed lncRNAs with over a 10-fold difference between the high- and low-fatty acid change groups. Of these, three showed lncRNA-mRNA interaction networks linked to human disorders. CONCLUSION BODIPY staining is a reliable alternative to Oil Red O staining for assessing fatty changes in MASLD zebrafish models, particularly when examining frozen liver sections.
Collapse
Affiliation(s)
- Wookjae Jung
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Min Hye Kim
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Jung Wook Yang
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Dong Chul Kim
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jong Sil Lee
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jeong-Hee Lee
- Department of Pathology, Gyeongsang National University Hospital, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyo Jung An
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea;
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Dae Hyun Song
- Department of Pathology, Gyeongsang National University School of Medicine, Jinju, Republic of Korea;
- Institute of Medical Sciences, Gyeongsang National University, Jinju, Republic of Korea
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| |
Collapse
|
26
|
Wang L, Duan W, Ruan C, Liu J, Miyagishi M, Kasim V, Wu S. YY2-CYP51A1 signaling suppresses hepatocellular carcinoma progression by restraining de novo cholesterol biosynthesis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167658. [PMID: 39761760 DOI: 10.1016/j.bbadis.2025.167658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Lipid accumulation is a frequently observed characteristic of cancer. Lipid accumulation is closely related to tumor progression, metastasis, and drug resistance; however, the mechanism underlying lipid metabolic reprogramming in tumor cells is not fully understood. Yin yang 2 (YY2) is a C2H2‑zinc finger transcription factor that exerts tumor-suppressive effects. However, its involvement in tumor cell lipid metabolic reprogramming remains unclear. In the present study, we identified YY2 as a novel regulator of cholesterol metabolism. We showed that YY2 suppressed cholesterol accumulation in hepatocellular carcinoma (HCC) cells by downregulating the transcriptional activity of cytochrome P450 family 51 subfamily A member 1 (CYP51A1), a key enzyme in de novo cholesterol biosynthesis. Subsequently, through in vitro and in vivo experiments, we demonstrated that this downregulation is crucial for the YY2 tumor suppressive effect. Together, our findings unraveled a previously unprecedented regulation of HCC cells cholesterol metabolism, and eventually, their tumorigenic potential, through YY2 negative regulation on CYP51A1 expression. This study revealed a novel regulatory mechanism of lipid metabolic reprogramming in tumor cells and provided insights into the molecular mechanism underlying the YY2 the suppressive effect. Furthermore, our findings suggest a potential antitumor therapeutic strategy targeting cholesterol metabolic reprogramming using YY2.
Collapse
Affiliation(s)
- Lingxian Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Cao Ruan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingyi Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| |
Collapse
|
27
|
Song BG, Goh MJ, Kang W, Gwak GY, Paik YH, Choi MS, Lee JH, Sinn DH. Serum Ferritin Levels and Liver-Related Events in Individuals With Steatotic Liver Disease: A Longitudinal Cohort Study. Aliment Pharmacol Ther 2025; 61:491-500. [PMID: 39573902 DOI: 10.1111/apt.18402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Serum ferritin has been suggested as a potential biomarker associated with disease progression in metabolic dysfunction-associated steatotic liver disease (MASLD). AIMS We investigated the association between serum ferritin levels and liver-related events (LREs) in individuals with steatotic liver disease (SLD). METHODS This cohort study included 17,560 adults with SLD (MASLD [n = 15,744], MASLD with increased alcohol intake (MetALD) [n = 1103] and cryptogenic SLD [n = 713]) without LRE at baseline. A steatotic liver was diagnosed using ultrasound, and LRE was defined as the development of decompensation (ascites, variceal bleeding and hepatic encephalopathy) or hepatocellular carcinoma. Participants were categorised into high (≥ 300 μg/L for males, ≥ 200 μg/L for females) or normal to low (< 300 μg/L for males, < 200 μg/L for females) ferritin levels. RESULTS During 211,425 person-years of follow-up (median: 12.3 years), 74 incident LRE cases were identified, with 63 cases in MASLD, 10 in MetALD and 1 in cryptogenic SLD. The multivariable-adjusted hazard ratio (aHR) for LRE comparing individuals with high and normal-to-low ferritin level was 3.13 (95% confidence interval [CI] 1.89-5.18). Increased risk of LRE in individuals with high serum ferritin level compared to those with normal to low serum ferritin level was consistent across SLD subtypes (aHR 2.69, 95% CI 1.55-4.67 for MASLD; aHR 5.73, 95% CI 1.31-25.0 for MetALD), and SLD severity assessed by Fibrosis-4 (FIB-4) index (aHR 2.38, 95% CI 1.34-4.21 for FIB-4 ≥ 1.3; aHR 3.13, 95% CI 1.18-8.29 for FIB-4 < 1.3). CONCLUSIONS Serum ferritin levels correlated with the risk of LRE in patients with SLD.
Collapse
Affiliation(s)
- Byeong Geun Song
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Myung Ji Goh
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Wonseok Kang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Geum-Youn Gwak
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong-Han Paik
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Moon Seok Choi
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joon Hyeok Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Dong Hyun Sinn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Wang J, Wang Z, Yu Y, Cheng S, Wu J. Advances in research on metabolic dysfunction-associated steatotic liver disease. Life Sci 2025; 362:123362. [PMID: 39761743 DOI: 10.1016/j.lfs.2024.123362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
The global increase in obesity-related metabolic disorders has led to metabolic dysfunction-associated steatotic liver disease (MASLD) emerging as one of the most prevalent chronic liver disease worldwide. Despite growing concerns, the exact pathogenesis of MASLD remains unclear and no definitive treatments have been made available. Consequently, the need for comprehensive research on MASLD is more critical than ever. Gaining insight into the mechanisms of the disease can lay the groundwork for identifying new therapeutic targets and can facilitate the development of diagnostic tools that enable the early detection and intervention of MASLD. Research has discovered a multifactorial etiology for MASLD, suggesting that potential therapeutic strategies should be considered from a variety of perspectives. This review delves into the pathogenesis of MASLD, current diagnostic approaches, potential therapeutic targets, the status of clinical trials for emerging drugs, and the most promising treatment methods available today. With a focus on therapeutic targets, the aim is to offer fresh insights and guide for future research in the treatment of MASLD.
Collapse
Affiliation(s)
- Jiawang Wang
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yao Yu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Si Cheng
- Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| | - Jianping Wu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; Department of Pharmacology, Hubei University of Medicine, Shiyan 440070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| |
Collapse
|
29
|
Park JS, Lee J, Wang F, Ma H, Zhou Z, Lee YS, Oh K, Lee H, Sui G, Lee S, Yang YM, Lee JW, Ji YH, Park CW, Yoo HS, Hwang BY, Han SB, Song N, Oh S, Kim B, Seki E, Hong JT, Roh YS. A1AT dysregulation of metabolically stressed hepatocytes by Kupffer cells drives MASH and fibrosis. Exp Mol Med 2025; 57:450-465. [PMID: 39939782 PMCID: PMC11873038 DOI: 10.1038/s12276-025-01408-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/14/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is associated with the activation of Kupffer cells (KCs) and hepatic stellate cells, at which point a metabolically stressed hepatocyte becomes integral to the progression of the disease. We observed a significant reduction in the level of alpha-1-antitrypsin (A1AT), a hepatocyte-derived secreted factor, in both patients with MASH and mice fed a fast-food diet (FFD). KC-mediated hepatic inflammation, most notably IL-1β, led to the transcriptional inhibition of A1AT by HNF4α. In quintuple Serpina1a-e knockout mice, ablation of A1AT worsened MASH through increased activity of proteinase 3 (PR3), a proinflammatory protease produced by F4/80hi/CD11blow/TIM4-/CCR2+ monocyte-derived KCs (MoKCs). Conversely, A1AT restoration or PR3 inhibition mitigated MASH progression. A PR3-bound cytokine array identified IL-32 as a key factor associated with MASH. Combining IL-32 with SERPINA1, the gene encoding A1AT, synergistically predicted patients at risk of MASH through univariate logistic regression analysis. Furthermore, in vivo overexpression of IL-32γ alleviated MASH induced by FFD. However, additional knockout of A1AT increased PR3 activity, consequently abolishing the anti-MASH effects of IL-32γ. Blocking PR3-mediated IL-32γ cleavage via the V104A mutation sustained its protective actions, while the PR3-cleaved C-terminal fragment activated KCs. Additionally, after cleavage, the antifibrogenic effect of IL-32γ is lost, resulting in a failure to prevent the activation of hepatic stellate cells. This study highlights the critical role of hepatocyte-derived A1AT in the PR3/IL-32γ axis during MASH development. Strategies to correct A1AT dysregulation, such as A1AT supplementation or PR3 inhibition with sivelestat, may offer protection against the development and progression of MASH and fibrosis. Elevated hepatic IL-1β levels in MASH lead to the downregulation of A1AT via the transcription factor HNF4α, resulting in increased recruitment of proinflammatory MoKCs and heightened PR3 activity. PR3 cleaves IL-32γ, transforming it from an anti-inflammatory and antifibrogenic cytokine into a potent activator of KCs and failing to prevent HSC activation. This cascade amplifies liver inflammation and fibrosis, suggesting that targeting the A1AT/PR3/IL-32γ axis could be a strategy for treating MASH.
Collapse
Affiliation(s)
- Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California, San Diego, CA, USA
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Zixiong Zhou
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yong-Sun Lee
- Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, South Korea
| | - Kwangyeon Oh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Haram Lee
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Sangkyu Lee
- College of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Yoon Mee Yang
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Jang-Won Lee
- Research and Development Center, MediTake Co. Ltd., Suwon, South Korea
| | - Yong-Ha Ji
- Research and Development Center, MediTake Co. Ltd., Suwon, South Korea
| | - Chun-Woong Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Hwan-Soo Yoo
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Bang-Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Nan Song
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea
| | - Soohwan Oh
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Bumseok Kim
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, South Korea
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea.
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, South Korea.
| |
Collapse
|
30
|
Karin M, Kim JY. MASH as an emerging cause of hepatocellular carcinoma: current knowledge and future perspectives. Mol Oncol 2025; 19:275-294. [PMID: 38874196 PMCID: PMC11793012 DOI: 10.1002/1878-0261.13685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatocellular carcinoma is one of the deadliest and fastest-growing cancers. Among HCC etiologies, metabolic dysfunction-associated fatty liver disease (MAFLD) has served as a major HCC driver due to its great potential for increasing cirrhosis. The obesogenic environment fosters a positive energy balance and results in a continuous rise of obesity and metabolic syndrome. However, it is difficult to understand how metabolic complications lead to the poor prognosis of liver diseases and which molecular mechanisms are underpinning MAFLD-driven HCC development. Thus, suitable preclinical models that recapitulate human etiologies are essentially required. Numerous preclinical models have been created but not many mimicked anthropometric measures and the course of disease progression shown in the patients. Here we review the literature on adipose tissues, liver-related HCC etiologies and recently discovered genetic mutation signatures found in MAFLD-driven HCC patients. We also critically review current rodent models suggested for MAFLD-driven HCC study.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Ju Youn Kim
- Department of Molecular and Life ScienceHanyang University ERICAAnsanKorea
| |
Collapse
|
31
|
Song H, Lu J, Deng R. Polysaccharides from Tremella Fuciformis Enhance Glucose and Lipid Metabolism in HepG2 Cells Through Activating the AMPK Signaling Pathway. Chem Biodivers 2025; 22:e202401926. [PMID: 39319532 DOI: 10.1002/cbdv.202401926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 09/26/2024]
Abstract
Polysaccharides have gained substantial attention for their diverse biological activities. The present study was conucted to elucidate the effects and molecular mechanisms of Tremella fuciformis-derived polysaccharides (PTP-3a) on glucose and lipid metabolism in palmitic acid (PA) - treated HepG2 cells. Multiple parameters were assessed following PTP-3a treatment, including lipid accumulation, glycogen content, glucose consumption, and enzyme activities, including pyruvate kinase (PK) and hexokinase (HK). Additionally, the expression levels of genes associated with glucose and lipid metabolism was evaluated using western blot analysis. PTP-3a effectively inhibited lipid accumulation, promoted the glucose consumption, increased the amount of cellular glycogen, and enhanced PK and HK activities in PA-treated cells. Furthermore, PTP-3a induced a significant increase in the p-AMPK/AMPK ratio and the expression level of PPARa, while decreasing the expression levels of SREBP, FAS, ACC, and SOCS3. In conclusion, these findings suggested that PTP-3a exerted beneficial effects on glucose and lipid metabolism by activating the AMPK signaling pathway, resulting in the inhibition of lipogenesis, promotion of fatty acid oxidation, and enhancement of cellular glycogen synthesis and glycolysis. These findings hold clinical relevance and provide a foundation for potential treatments for non-alcoholic fatty liver disease (NAFLD) and and related metabolic disorders.
Collapse
Affiliation(s)
- Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics, /Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, 210023, China
| | - Jing Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, /Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, 210023, China
| | - Rou Deng
- College of Food Science and Engineering, Nanjing University of Finance and Economics, /Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, 210023, China
| |
Collapse
|
32
|
Jiang Y, Wei S, Shen S, Liu Y, Su W, Ding D, Zheng Z, Yu H, Zhang T, Yang Q, Zhao J, Shen Y, Fang X, Lin L, Xiao D, Cui A, Wan Q, Zhang Y, Li Y, Zhang C. Ethyl Lactate Ameliorates Hepatic Steatosis and Acute-on-Chronic Liver Injury in Alcohol-Associated Liver Disease by Inducing Fibroblast Growth Factor 21. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409516. [PMID: 39661730 PMCID: PMC11792039 DOI: 10.1002/advs.202409516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Aberrant upregulation of hepatic lipogenesis induced by chronic and excessive alcohol consumption is a critical driver of the progression of alcohol-associated liver disease (ALD), however, no effective approaches inhibiting lipogenesis are currently available for treating ALD patients. Moreover, little is known about whether and how nonethanol ingredients in alcoholic beverages regulate the pathogenesis of ALD. Here the discovery of a small molecule that activates the production and secretion of fibroblast growth factor 21 (FGF21) is reported. It is shown that the activator ethyl lactate, a nonethanol ingredient found in distilled liquors, ameliorates alcoholic hepatosteatosis, inflammation and acute-on-chronic liver injury by stimulating FGF21. In response to chronic-plus-binge ethanol feeding or fasting, ethyl lactate mimics lipogenesis lowering effects by stimulating FGF21 production through the NAD+-dependent deacetylase sirtuin 1 (SIRT1) signaling pathway. These ethyl lactate-mediated beneficial effects are abolished by inhibition of SIRT1 through injection of EX527. Importantly, FGF21 deficiency in hepatocytes blocks the downregulation of lipogenesis by ethyl lactate and exacerbates alcoholic steatosis, inflammation and liver injury. The regulatory mechanism is discussed during the pathophysiological conditions and suggests new lines of research into the therapeutic use of a foodborne small molecule ethyl lactate.
Collapse
Affiliation(s)
- Yang Jiang
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Shuang Wei
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Shiming Shen
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
| | - Yuxiao Liu
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Weitong Su
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Dong Ding
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Zengpeng Zheng
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Haokai Yu
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Tingting Zhang
- CAS Engineering Laboratory for NutritionShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Qiuli Yang
- CAS Engineering Laboratory for NutritionShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Jiuxiang Zhao
- CAS Engineering Laboratory for NutritionShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Yi Shen
- Sichuan Langjiu Co.LtdGulinSichuan646523China
| | - Xia Fang
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
- Department of Endocrinology and MetabolismMetabolic Vascular Disease Key Laboratory of Sichuan ProvinceThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuan646000China
| | - Liangcai Lin
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
| | - Dongguang Xiao
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
| | - Aoyuan Cui
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Qin Wan
- Department of Endocrinology and MetabolismMetabolic Vascular Disease Key Laboratory of Sichuan ProvinceThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuan646000China
| | | | - Yu Li
- CAS Key Laboratory of NutritionMetabolism and Food SafetyShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
- CAS Engineering Laboratory for NutritionShanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Cuiying Zhang
- State Key Laboratory of Food Nutrition and SafetyCollege of BiotechnologyTianjin University of Science and TechnologyTianjin300457China
| |
Collapse
|
33
|
Ma S, Xia E, Zhang M, Hu Y, Tian S, Zheng X, Li B, Ma G, Su R, Sun K, Fan Q, Yang F, Guo G, Guo C, Shang Y, Zhou X, Zhou X, Wang J, Han Y. Role of the FOXM1/CMA/ER stress axis in regulating the progression of nonalcoholic steatohepatitis. Clin Transl Med 2025; 15:e70202. [PMID: 39924645 PMCID: PMC11807764 DOI: 10.1002/ctm2.70202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND/AIMS The molecular mechanisms driving nonalcoholic steatohepatitis (NASH) progression are poorly understood. This research examines the involvement of chaperone-mediated autophagy (CMA) in NASH progression. METHODS Hepatic CMA activity was analysed in NASH mice and patients. Lysosome-associated membrane protein 2A (LAMP2A) was knocked down or overexpressed to assess the effects of hepatocyte-specific CMA on NASH progression. Mice received a high-fat diet or a methionine and choline-deficient diet to induce NASH. Palmitic acid was employed to mimic lipotoxicity-induced hepatocyte damage in vitro. The promoter activity of FOXM1 was evaluated via ChIP and dual-luciferase reporter assays. RESULTS Hepatic CMA activity was substantially low in NASH mice and patients. LAMP2A knockdown resulted in hepatocyte-specific CMA deficiency, which promoted fibrosis and hepatic inflammation in NASH mice. Both in vitro and in vivo, CMA deficiency also exacerbated hepatocyte damage and endoplasmic reticulum (ER) stress. Mechanistically, CMA deficiency in hepatocytes increased cholesterol accumulation by blocking the degradation of 3-hydroxy-3-methylglutaryl coenzyme A (HMGCR), a key cholesterol synthesis-related enzyme, and the accumulated cholesterol subsequently induced ER stress and hepatocyte damage. The restoration of hepatocyte-specific CMA activity effectively ameliorated diet-induced NASH and ER stress in vivo and in vitro. FOXM1 directly bound to LAMP2A promoter and negatively regulated its transcription. The upregulation of FOXM1 expression impaired CMA and enhanced ER stress, which in turn increased FOXM1 expression, resulting in a vicious cycle and promoting NASH development. CONCLUSIONS This study highlights the significance of the FOXM1/CMA/ER stress axis in NASH progression and proposes novel therapeutic targets for NASH. KEY POINTS Chaperone-mediated autophagy (CMA) deficiency in hepatocytes promotes hepatic inflammation and fibrosis in mice with nonalcoholic steatohepatitis (NASH) by inducing cholesterol accumulation and endoplasmic reticulum (ER) stress. Upregulated FOXM1 impairs CMA by suppressing the transcription of lysosome-associated membrane protein 2A (LAMP2A), a rate-limiting component of CMA. ER stress increases FOXM1 expression and cholesterol accumulation. FOXM1/CMA/ER stress axis forms a vicious circle and promotes the development of NASH.
Collapse
Affiliation(s)
- Shuoyi Ma
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Erzhuo Xia
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Miao Zhang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Yinan Hu
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Siyuan Tian
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xiaohong Zheng
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Bo Li
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Gang Ma
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Rui Su
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Keshuai Sun
- Department of GastroenterologyThe Air Force Hospital From Eastern Theater of PLANanjingChina
| | - Qingling Fan
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Fangfang Yang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Guanya Guo
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Changcun Guo
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Yulong Shang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xinmin Zhou
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xia Zhou
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Jingbo Wang
- Science and Technology Innovation Research InstituteTangdu Hospital, The Fourth Military Medical UniversityXi'anChina
| | - Ying Han
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
34
|
Queathem ED, Moazzami Z, Stagg DB, Nelson AB, Fulghum K, Hayir A, Seay A, Gillingham JR, d’Avignon DA, Han X, Ruan HB, Crawford PA, Puchalska P. Ketogenesis supports hepatic polyunsaturated fatty acid homeostasis via fatty acid elongation. SCIENCE ADVANCES 2025; 11:eads0535. [PMID: 39879309 PMCID: PMC11777252 DOI: 10.1126/sciadv.ads0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
Ketogenesis is a dynamic metabolic conduit supporting hepatic fat oxidation particularly when carbohydrates are in short supply. Ketone bodies may be recycled into anabolic substrates, but a physiological role for this process has not been identified. Here, we use mass spectrometry-based 13C-isotope tracing and shotgun lipidomics to establish a link between hepatic ketogenesis and lipid anabolism. Unexpectedly, mouse liver and primary hepatocytes consumed ketone bodies to support fatty acid biosynthesis via both de novo lipogenesis (DNL) and polyunsaturated fatty acid (PUFA) elongation. While an acetoacetate intermediate was not absolutely required for ketone bodies to source DNL, PUFA elongation required activation of acetoacetate by cytosolic acetoacetyl-coenzyme A synthetase (AACS). Moreover, AACS deficiency diminished free and esterified PUFAs in hepatocytes, while ketogenic insufficiency depleted PUFAs and increased liver triacylglycerols. These findings suggest that hepatic ketogenesis influences PUFA metabolism, representing a molecular mechanism through which ketone bodies could influence systemic physiology and chronic diseases.
Collapse
Affiliation(s)
- Eric D. Queathem
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Zahra Moazzami
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - David B. Stagg
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Kyle Fulghum
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Abdirahman Hayir
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alisha Seay
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jacob R. Gillingham
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - D. André d’Avignon
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xianlin Han
- Department of Medicine-Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter A. Crawford
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
35
|
Chen F, Sarver DC, Saqib M, Velez LM, Aja S, Seldin MM, Wong GW. Loss of CTRP10 results in female obesity with preserved metabolic health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.01.565163. [PMID: 37961647 PMCID: PMC10635050 DOI: 10.1101/2023.11.01.565163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Obesity is a major risk factor for type 2 diabetes, dyslipidemia, cardiovascular disease, and hypertension. Intriguingly, there is a subset of metabolically healthy obese (MHO) individuals who are seemingly able to maintain a healthy metabolic profile free of metabolic syndrome. The molecular underpinnings of MHO, however, are not well understood. Here, we report that CTRP10/C1QL2-deficient mice represent a unique female model of MHO. CTRP10 modulates weight gain in a striking and sexually dimorphic manner. Female, but not male, mice lacking CTRP10 develop obesity with age on a low-fat diet while maintaining an otherwise healthy metabolic profile. When fed an obesogenic diet, female Ctrp10 knockout (KO) mice show rapid weight gain. Despite pronounced obesity, Ctrp10 KO female mice do not develop steatosis, dyslipidemia, glucose intolerance, insulin resistance, oxidative stress, or low-grade inflammation. Obesity is largely uncoupled from metabolic dysregulation in female KO mice. Multi-tissue transcriptomic analyses highlighted gene expression changes and pathways associated with insulin-sensitive obesity. Transcriptional correlation of the differentially expressed gene (DEG) orthologous in humans also shows sex differences in gene connectivity within and across metabolic tissues, underscoring the conserved sex-dependent function of CTRP10. Collectively, our findings suggest that CTRP10 negatively regulates body weight in females, and that loss of CTRP10 results in benign obesity with largely preserved insulin sensitivity and metabolic health. This female MHO mouse model is valuable for understanding sex-biased mechanisms that uncouple obesity from metabolic dysfunction.
Collapse
Affiliation(s)
- Fangluo Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leandro M Velez
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcus M. Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, USA
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Feng Y, Mei W, Chen Q, Chen X, Ni Y, Lei M, Liu J. Probiotic Supplementation Alleviates Corticosterone-Induced Fatty Liver Disease by Regulating Hepatic Lipogenesis and Increasing Gut Microbiota Diversity in Broilers. Microorganisms 2025; 13:200. [PMID: 39858968 PMCID: PMC11767375 DOI: 10.3390/microorganisms13010200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/19/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Emerging evidence indicates a close relationship between gut microbiota and fatty liver disease. It has been suggested that gut microbiota modulation with probiotics ameliorates fatty liver disease in rodents and humans, yet it remains unclear whether the same results will also be obtained in poultry. The aim of this study was to investigate whether a mixture of probiotics supplemented after hatching can prevent CORT-induced fatty liver disease in broilers, and to determine how such effects, if any, are associated with hepatic de novo lipogenesis and gut microbiota composition. Ninety-six one-day-old green-legged chickens were divided into a control group (CON) and probiotic group (PB). At 28 days of age, fatty liver was induced in 16 broilers that were randomly selected from the CON or PB group. At the end of the experiment, broilers from four groups, (i) the control group (CON), (ii) corticosterone group (CORT), (iii) probiotic group (PB), and (iv) PB plus CORT group (CORT&PB), were slaughtered for sampling and analysis. The results showed that probiotic administration significantly prevented CORT-induced body weight loss (p < 0.05) but did not alleviate the weight loss of immune organs caused by CORT. Compared to CON, the broilers in the CORT group exhibited a significant increase in triglyceride (TG) levels in plasma and liver (p < 0.01), as well as severe hepatocytic steatosis and hepatocellular ballooning, which was accompanied by the upregulation of hepatic lipogenesis gene expression. However, probiotic supplementation markedly decreased the intrahepatic lipid accumulation and steatosis histological score, which was associated with the downregulation of sterol regulatory element-binding protein-1 (SREBP1) and acetyl-CoA carboxylase (ACC) mRNA (p < 0.05) and the expression of its protein (p = 0.06). The cecal microbiota composition was determined by 16S rRNA high-throughput sequencing. The results showed that CORT treatment induced distinct gut microbiota alterations with a decrease in microbial diversity and an increase in Proteobacteria abundance (p < 0.05). In contrast, probiotic supplementation increased the beta diversity, the community richness, and the diversity index (p > 0.05), as well as the abundance of Intestinimonas (p < 0.05). Our results indicate that CORT treatment induced severe fatty liver disease and altered the gut microbiota composition in broilers. However, post-hatching probiotic supplementation had a beneficial effect on alleviating fatty liver disease by regulating lipogenic gene expression and increasing gut microbiota diversity and the abundance of beneficial bacteria. We demonstrate for the first time that the supplementation of probiotics to chicks had a beneficial effect on preventing fatty liver disease through regulating lipogenic gene expression and improving the gut microbial balance. Thus, our results indicate that probiotics are a potential nutritional agent for preventing fatty liver disease in chickens.
Collapse
Affiliation(s)
- Yuyan Feng
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Wenqing Mei
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Qu Chen
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Xiaojing Chen
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Yingdong Ni
- Key Laboratory of Animal Physiologic and Biochemistry, College of Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Nanjing Agricultural University, Nanjing 210094, China; (W.M.); (Q.C.); (Y.N.)
| | - Mingming Lei
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
| | - Jie Liu
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing 210094, China; (Y.F.); (X.C.)
| |
Collapse
|
37
|
Khalafi M, Rosenkranz SK, Ghasemi F, Kheradmand S, Habibi Maleki A, Korivi M, Tsao JP. Efficacy of intermittent fasting on improving liver function in individuals with metabolic disorders: a systematic review and meta-analysis. Nutr Metab (Lond) 2025; 22:1. [PMID: 39762987 PMCID: PMC11706068 DOI: 10.1186/s12986-024-00885-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Intermittent fasting (IF) can be an effective dietary therapy for weight loss and improving cardiometabolic health. However, there is scant evidence regarding the role of IF on indicators of liver function, particularly in adults with metabolic disorders. Therefore, we performed a systematic review and meta-analysis to investigate the effects of IF on liver function in adults with metabolic disorders. METHODS Three primary electronic databases including PubMed, Web of Science, and Scopus, were searched from inception to September 2024 to identify original studies that used IF interventions with or without control groups in adults with metabolic disorders. Inclusion criteria were (1) studies of human participants with metabolic diseases, (2) interventions that evaluated the effects of IF, (3) with or without a control group, and (4) measured liver fat, liver steatosis, liver fibrosis, or liver enzymes, including alanine aminotransferase (ALT) and aspartate aminotransferase (AST) as primary outcomes. Standardized mean differences (SMD) and 95% confidence intervals were calculated using random effects models. Heterogeneity was assessed using the Cochran's Q statistic and I-squared statistic (I2). Publication bias was assessed using the visual inspection of funnel plots and Egger's tests. The risk of bias was assessed using the PEDro scale and the NIH quality assessment tool. RESULTS A total 21 studies involving 1,226 participants with metabolic disorders were included in the meta-analysis. Overall, IF effectively decreased liver fat with a large effect size [SMD: -1.22 (95% CI: -1.63 to -0.80), p = 0.001], liver steatosis with a medium effect size [SMD: -0.73 (95% CI: -1.12 to -0.35), p = 0.001], ALT with a small effect size [SMD: -0.44 (95% CI: -0.58 to -0.30), p = 0.001], and AST with a small effect size [SMD: -0.30 (95% CI: -0.49 to -0.11), p = 0.001], but not liver fibrosis [SMD: -0.28 (95% CI: -0.59 to 0.02), p = 0.07]. Subgroup analyses showed that IF decreased liver fat and ALT significantly, independent of IF mode, participant age, health status, weight status, and intervention duration. IF significantly decreased liver fibrosis in those with obesity; and decreased AST following 5:2 diets, in middle-aged adults, adults with obesity, and regardless of health status or intervention duration. CONCLUSIONS IF seems to be an effective dietary therapy for improving liver function in adults with metabolic disorders, and many of liver function-related benefits occur regardless of IF mode, intervention duration, or participant health status. LIMITATIONS Significant heterogeneity, small numbers of studies and inclusion of non-randomized trials or single-group pre-post trials were the main limitation of our meta-analysis. Further randomized clinical trials are needed to elucidate the effects of IF on liver function in adults with metabolic disorders.
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Sara K Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Faeghe Ghasemi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Guilan, Iran
| | - Shokoufeh Kheradmand
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Mazandaran, Babolsar, Iran
| | - Aref Habibi Maleki
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mallikarjuna Korivi
- Institute of Human Movement and Sports Engineering, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China.
| | - Jung-Piao Tsao
- Department of Sports Medicine, China Medical University, Taichung City, Taiwan.
| |
Collapse
|
38
|
Ueda H, Honda A, Miyazaki T, Morishita Y, Hirayama T, Iwamoto J, Ikegami T. High-fat/high-sucrose diet results in a high rate of MASH with HCC in a mouse model of human-like bile acid composition. Hepatol Commun 2025; 9:e0606. [PMID: 39670881 PMCID: PMC11637755 DOI: 10.1097/hc9.0000000000000606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/21/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Wild-type (WT) mice fed a conventional high-fat/high-sucrose diet (HFHSD) rarely develop metabolic dysfunction-associated steatohepatitis (MASH) with HCC. Because mouse bile acid (BA) is highly hydrophilic, we hypothesized that making it hydrophobic would lead to MASH with HCC. METHODS Eleven-week-old WT and Cyp2a12/Cyp2c70 double knockout (DKO) mice were divided into two groups, including one which was fed a normal chow diet, and one which was fed an HFHSD. Samples were collected after 15, 30, 47, and 58 weeks for histological, biochemical, and immunological analyses. RESULTS In the HFHSD group, body weight gain did not differ in WT versus DKO mice, although HFHSD-fed DKO mice exhibited markedly accelerated liver inflammation, fibrosis, and carcinogenesis. HFHSD upregulated lipogenesis and downregulated fatty acid oxidation in both WT and DKO mice, which increased liver lipid accumulation and lipotoxicity. However, the increase in reactive oxygen species production and carcinogenesis observed in DKO mice could not be explained by abnormal lipid metabolism alone. Regarding BA metabolism, DKO mice had a higher hydrophobicity index. They exhibited an age-associated increase in chenodeoxycholic acid (CDCA) levels because of CYP8B1 activity inhibition due to the farnesoid X receptor activation. HFHSD further downregulated CYP8B1, presumably by activating the Liver X receptor. Liver CDCA accumulation was associated with increased inflammation, reactive oxygen species production, and hepatocyte FGF15 induction. Moreover, in noncancerous liver tissues, HFHSD appeared to activate STAT3, an oncogenic transcription factor, which was enhanced by a CDCA-rich environment. CONCLUSIONS Here, we developed a new model of MASH with HCC using mice with human-like BA composition and found that HFHSD and elevated hepatic CDCA synergistically increased the risk of MASH with HCC.
Collapse
Affiliation(s)
- Hajime Ueda
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Akira Honda
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Teruo Miyazaki
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Yukio Morishita
- Diagnostic Pathology Division, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Takeshi Hirayama
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Junichi Iwamoto
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Tadashi Ikegami
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| |
Collapse
|
39
|
Alvarez HM, Lanfranconi MP, Hernández MA. Metabolism-lipid droplet-nucleic acid crosstalk to regulate lipid storage and other cellular processes in oleaginous Rhodococcus bacteria. Biol Cell 2025; 117:e2400094. [PMID: 39853774 DOI: 10.1111/boc.202400094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/14/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
Actinobacteria belonging to Mycobacterium and Rhodococcus genera are able to synthesize and intracellularly accumulate variable amounts of triacylglycerols (TAG) in the form of lipid droplets (LDs). The lipid storage capacity of LDs in cells is controlled by the balance between lipogenesis and lipolysis. The growth of LDs in bacterial cells may be directly promoted by TAG biosynthesis, whereas TAG degradation might result in the reduction of LD sizes and lipid storage capacity. Therefore, LD formation and turnover have to be precisely regulated to maintain a balanced lipid distribution, coupling gene regulation with the metabolic state of the cell. In eukaryotic cells, LDs have emerged as critical mediators of diverse cellular responses, including fatty acid trafficking and modulation of transcriptional programs. Recent studies performed in mycobacteria and rhodococci suggested the existence of similar crosstalk mechanisms between lipid metabolism, LDs, and gene expression regulation in cells. This review connects and organizes results of different studies in a comprehensive framework for providing evidence of "lipid metabolism-LDs-genomic DNA" crosstalk occurring in TAG-accumulating actinobacteria. We provide examples indicating that bacterial cells evolved sensing mechanisms that detect lipid metabolites changes as indicators of metabolic states, and adapt their transcriptional profiles through epigenetic-like mechanisms mediated by LD-associated proteins. Here, we describe the molecular interconnections of this coupling system and the main role of each component that integrates the information about the cellular metabolic state into the regulation of lipogenesis, LD formation and transcription in oleaginous bacteria.
Collapse
Affiliation(s)
- Héctor M Alvarez
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| | - Mariana P Lanfranconi
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| | - Martín A Hernández
- INBIOP (Instituto de Biociencias de la Patagonia), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Comodoro Rivadavia, Chubut, Argentina
| |
Collapse
|
40
|
Gopal P, Hu X, Robert ME, Zhang X. The evolving role of liver biopsy: Current applications and future prospects. Hepatol Commun 2025; 9:e0628. [PMID: 39774070 PMCID: PMC11717517 DOI: 10.1097/hc9.0000000000000628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Histopathologic evaluation of liver biopsy has played a longstanding role in the diagnosis and management of liver disease. However, the utility of liver biopsy has been questioned by some, given the improved imaging modalities, increased availability of noninvasive serologic tests, and development of artificial intelligence over the past several years. In this review, we discuss the current and future role of liver biopsy in both non-neoplastic and neoplastic liver diseases in the era of improved noninvasive laboratory, radiologic, and digital technologies.
Collapse
Affiliation(s)
- Purva Gopal
- Deparment of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiaobang Hu
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Marie E. Robert
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
41
|
Xiang M, Tian X, Wang H, Gan P, Zhang Q. Inappropriate Diet Exacerbates Metabolic Dysfunction-Associated Steatotic Liver Disease via Abdominal Obesity. Nutrients 2024; 16:4208. [PMID: 39683601 DOI: 10.3390/nu16234208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a refined categorization of non-alcoholic fatty liver disease (NAFLD), highlighting the intricate relationship between hepatic steatosis and metabolic dysfunction. Abdominal obesity (AO), a key diagnostic criterion for metabolic dysfunction, predominantly results from inappropriate diet and unhealthy dietary habits. To comprehensively investigate which dietary factors contribute to MASLD through AO and to understand the underlying biological mechanisms, we initially conducted a systematic review of meta-analysis articles in the PubMed database from the past decade, summarizing dietary factors that affect AO. Subsequently, we conducted targeted searches in the PubMed database for these dietary factors and provided a narrative review of the mechanisms of how these dietary factors lead to AO and how AO exacerbates MASLD. A diet characterized by excessive intake of energy, carbohydrates, fructose, or ultra-processed foods (UPFs) is considered inappropriate. Inappropriate diet leads to the formation of MASLD and AO by enhancing pathways such as de novo lipid synthesis (DNL) in the liver, insulin resistance (IR), gut-liver dysfunction, and inflammation. Dietary interventions for inappropriate diets can effectively intervene in and improve MASLD and AO. The mechanism of inappropriate diet on abdominal fat deposition is through excessive energy or the activation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD-1) to increase endocortisol secretion. Then, the excessive accumulation of visceral fat facilitates a rapid and augmented flux of free fatty acids (FFAs) to the liver and initiates a series of deleterious effects, including oxidative stress (OS), endoplasmic reticulum stress (ERS), activation of protein kinase C (PKC) pathways, and inflammation. Additionally, FFAs may mediate excessive lipid deposition and hepatocellular damage through the action of hormones. These pathways to liver damage exacerbate MASLD and progression to metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis. Furthermore, investigating other potential mechanisms by which AO may influence MASLD could offer new recommendations for the treatment guidelines of MASLD.
Collapse
Affiliation(s)
- Minghui Xiang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xiaoli Tian
- School of Public Health, Xinjiang Medical College, Ürümqi 830000, China
- School of Public Health, Xinjiang Second Medical College, Karamay 834000, China
| | - Hui Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Ping Gan
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511400, China
| | - Qian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| |
Collapse
|
42
|
Homsana A, Southisavath P, Kling K, Hattendorf J, Vorasane S, Paris DH, Sayasone S, Odermatt P, Probst-Hensch N. Steatotic liver disease among lean and non-lean individuals in Southern Lao PDR: a cross-sectional study of risk factors. Ann Med 2024; 56:2329133. [PMID: 38502916 PMCID: PMC10953781 DOI: 10.1080/07853890.2024.2329133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/24/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Steatotic liver disease (SLD) prevalence is rising worldwide, linked to insulin resistance and obesity. SLD prevalence can surpass 10% even among those with normal weight. In Lao People's Democratic Republic (Lao PDR), where Opisthorchis viverrini (OV) trematode infection and type 2 diabetes mellitus (T2DM) are common, infection related liver morbidity such as cholangiocarcinoma (CCA) is high, but data on SLD prevalence is lacking. The objective of this study was to estimate the prevalence and explore determinants of SLD in rural southern Lao PDR for lean and non-lean populations. METHOD A cross-sectional community-based study assessed SLD prevalence using abdominal ultrasonography (US). Factors investigated for association with SLD were identified by interview, serological tests (Hepatitis B surface antigen (HBsAg); lipids and HbA1c), anthropometrical measurements, and parasitological assessments (OV infection). Uni- and multivariable logistic regression analyses with SLD as endpoint were conducted separately for lean (body mass index (BMI) <23.0 kg/m2) and non-lean (BMI ≥ 23.0 kg/m2) participants. RESULT 2,826 participants were included. SLD prevalence was 27.1% (95% confidence interval (95% CI) 24.0%-30.4%), higher among non-lean (39.8%) than lean individuals (17.4%). Lean individuals with OV infection had a statistically significant association with lower odds of SLD (adjusted odds ratio (aOR) 0.49, 95% CI 0.33 - 0.73). T2DM showed a significant positive association with SLD in both lean (aOR 3.58, 95% CI 2.28 - 5.63) and non-lean individuals (aOR 3.31, 95% CI 2.31 - 4.74) while dyslipidemia was significantly associated only in the non-lean group (aOR 1.83, 95% CI 1.09 - 3.07). Females participants exhibited elevated odds of SLD in both lean (aOR 1.43, 95% CI 1.02 - 2.01) and non-lean SLD (aOR 1.50, 95% CI 1.12 - 2.01). CONCLUSION SLD prevalence is notably high among Laotian adults in rural areas, particularly in females and in non-lean individuals. Lean individuals with OV infection exhibited lower SLD prevalence. SLD was more prevalent in individuals with T2DM, independent of BMI. SLD adds to the burden of infection-related liver morbidity in Lao PDR.
Collapse
Affiliation(s)
- Anousin Homsana
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane Capital, Lao PDR
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Phonesavanh Southisavath
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane Capital, Lao PDR
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Department of Radiology, Mahosot Hospital, Ministry of Health, Vientiane Capital, Lao PDR
| | - Kerstin Kling
- Immunization Unit, Robert Koch Institute, Berlin, Germany
| | - Jan Hattendorf
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Savina Vorasane
- Department of Radiology, Mahosot Hospital, Ministry of Health, Vientiane Capital, Lao PDR
| | - Daniel Henry Paris
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Somphou Sayasone
- Lao Tropical and Public Health Institute, Ministry of Health, Vientiane Capital, Lao PDR
| | - Peter Odermatt
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
43
|
Yue P, Lv X, Cao H, Zou Y, You J, Luo J, Lu Z, Chen H, Liu Z, Zhong Z, Xiong Y, Fan X, Ye Q. Hypothermic oxygenated perfusion inhibits CLIP1-mediated TIRAP ubiquitination via TFPI2 to reduce ischemia‒reperfusion injury of the fatty liver. Exp Mol Med 2024; 56:2588-2601. [PMID: 39617791 DOI: 10.1038/s12276-024-01350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 12/28/2024] Open
Abstract
The use of fatty livers in liver transplantation has emerged as a crucial strategy to expand the pool of donor livers; however, fatty livers are more sensitive to ischemia‒reperfusion injury (IRI). Excessive congenital inflammatory responses are crucial in IRI. Hypothermic oxygenated perfusion (HOPE) is a novel organ preservation technique that may improve marginal donor liver quality by reducing the inflammatory response. Tissue factor pathway inhibitor-2 (TFPI2) and CAP-Gly domain-containing linker protein 1 (CLIP1) exhibit modulatory effects on the inflammatory response. However, the underlying mechanisms of HOPE in fatty liver and the effects of TFPI2 and CLIP1 in fatty liver IRI remain unclear. Here, we aimed to explore the impact of HOPE on the inflammatory response in a rat model of fatty liver IRI and the mechanisms of action of TFPI2 and CLIP1. HOPE significantly reduces liver injury, especially the inflammatory response, and alleviates damage to hepatocytes and endothelial cells. Mechanistically, HOPE exerts its effects by inhibiting TFPI2, and CLIP1 can rescue the damaging effects of TFPI2. Moreover, HOPE promoted the ubiquitination and subsequent degradation of Toll/interleukin-1 receptor domain-containing adapter protein (TIRAP) by regulating the binding of R24 of the KD1 domain of TFPI2 with CLIP1, thereby negatively regulating the TLR4/NF-κB-mediated inflammatory response and reducing IRI. Furthermore, TFPI2 expression increased and CLIP1 expression decreased following cold ischemia in human fatty livers. Overall, our results suggest that targeting the inflammatory response by modulating the TFPI2/CLIP1/TIRAP signaling pathway via HOPE represents a potential therapeutic approach to ameliorate IRI during fatty liver transplantation.
Collapse
Affiliation(s)
- Pengpeng Yue
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Xiaoyan Lv
- Department of Hematology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Hankun Cao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Yongkang Zou
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People's Hospital, 550002, Guiyang, China
| | - Jian You
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Jun Luo
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Zhongshan Lu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Hao Chen
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Yan Xiong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China
| | - Xiaoli Fan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China.
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071, Wuhan, China.
- The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, 410013, Changsha, China.
| |
Collapse
|
44
|
Huang YL, Zhang KY, Sun YL, Qian MB, Wang Z. The risk of hepatobiliary complications in Clonorchis and Opisthorchis infection: A systematic review and meta-analysis. Acta Trop 2024; 260:107457. [PMID: 39521195 DOI: 10.1016/j.actatropica.2024.107457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/16/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Clonorchis sinensis, Opisthorchis viverrini, and Opisthorchis felineus are the three important liver flukes, infecting approximately 25 million people worldwide. Despite the reporting of the carcinogenesis of these liver flukes, the comprehensive and systematic analysis of the pathogenicity of these parasites in hepatobiliary system is still not sufficient. We conducted a thorough systematic review and search for published articles in MEDLINE, Embase, Cochrane Library, China National Knowledge Infrastructure databases until early 2024. Cohort studies, case-control studies, and cross-sectional studies associated with C. sinensis, O. viverrini, or O. felineus infection were selected. Pooled risk ratio (RR), odds ratio (OR) and their 95 % confidence intervals (95 % CIs) were calculated to assess the risk of hepatobiliary complications due to these liver fluke infections. From a total of 6488 articles, 22 eligible studies and 34,367 participants were included for review. Our results showed C. sinensis, O. viverrini, and O. felineus infections were significantly associated with cholangiocarcinoma, with an overall OR of 4.24 (95 % CI: 3.33-5.39, P < 0.00001) and an overall RR of 10.43 (95 % CI: 2.90-37.47, P = 0.0003). The ORs for the association between cholangiocarcinoma and C. sinensis and O. viverrini infection were 4.49 (95 % CI:3.43-5.87, P < 0.00001) and 3.69 (95 % CI: 2.07-6.55, P < 0.00001) respectively. For the association between cholelithiasis and C. sinensis infection, the OR was 6.46 (95 % CI: 5.15-8.11, P < 0.00001). C. sinensis infection increased the risk of cholecystitis and cirrhosis, with the RR of 21.02 (95 % CI: 17.27-25.58) and an overall RR of 8.77 (95 % CI: 6.79-11.33, P < 0.00001) respectively. C. sinensis infection was also significantly associated with fatty liver, with an overall OR of 2.46 (95 % CI: 1.79-3.37, P < 0.00001). This comprehensive study, reviewing the largest dataset to date, provided an overall risk of hepatobiliary complications due to Clonorchis and Opisthorchis infections, and aids more systematic understanding for the pathogenicity of Opisthorchiidae family parasites.
Collapse
Affiliation(s)
- Yi-Lin Huang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kai-Yan Zhang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan-Lin Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Men-Bao Qian
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Centre for Tropical Diseases, NHC Key Laboratory of Parasite and Vector Biology, National Center for International Research on Tropical Diseases, Shanghai 200025, China.
| | - Zhaojun Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
45
|
Zhu Y, Shang L, Tang Y, Li Q, Ding L, Wang Y, Zhang T, Xie B, Ma J, Li X, Chen S, Yi X, Peng J, Liang Y, He A, Yan H, Zhu H, Zhang B, Zhu Y. Genome-Wide Profiling of H3K27ac Identifies TDO2 as a Pivotal Therapeutic Target in Metabolic Associated Steatohepatitis Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404224. [PMID: 39364706 PMCID: PMC11615751 DOI: 10.1002/advs.202404224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/13/2024] [Indexed: 10/05/2024]
Abstract
H3K27ac has been widely recognized as a representative epigenetic marker of active enhancer, while its regulatory mechanisms in pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) remain elusive. Here, a genome-wide comparative study on H3K27ac activities and transcriptome profiling in high fat diet (HFD)-induced MASLD model is performed. A significantly enhanced H3K27ac density with abundant alterations of regulatory transcriptome is observed in MASLD rats. Based on integrative analysis of ChIP-Seq and RNA-Seq, TDO2 is identified as a critical contributor for abnormal lipid accumulation, transcriptionally activated by YY1-promoted H3K27ac. Furthermore, TDO2 depletion effectively protects against hepatic steatosis. In terms of mechanisms, TDO2 activates NF-κB pathway to promote macrophages M1 polarization, representing a crucial event in MASLD progression. A bovine serum albumin nanoparticle is fabricated to provide sustained release of Allopurinol (NPs-Allo) for TDO2 inhibition, possessing excellent biocompatibility and desired targeting capacity. Venous injection of NPs-Allo robustly alleviates HFD-induced metabolic disorders. This study reveals the pivotal role of TDO2 and its underlying mechanisms in pathogenesis of MASLD epigenetically and genetically. Targeting H3K27ac-TDO2-NF-κB axis may provide new insights into the pathogenesis of abnormal lipid accumulation and pave the way for developing novel strategies for MASLD prevention and treatment.
Collapse
Affiliation(s)
- Yaling Zhu
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Limeng Shang
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Yunshu Tang
- Laboratory Animal Research CenterSchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Qiushuang Li
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Lin Ding
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Yi Wang
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Tiantian Zhang
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Bin Xie
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Jinhu Ma
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Xinyu Li
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Shuwen Chen
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Xinrui Yi
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Jin Peng
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Youfeng Liang
- Department of CardiologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230001China
| | - Anyuan He
- School of Life SciencesAnhui Medical UniversityHefeiAnhui230032China
| | - Hong Yan
- Department of PathologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Department of PathologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Huaqing Zhu
- Laboratory of Molecular Biology and Department of BiochemistryAnhui Medical UniversityHefeiAnhui230032China
| | - Buchun Zhang
- Department of CardiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
| | - Yong Zhu
- Department of PathophysiologySchool of Basic Medical SciencesAnhui Medical UniversityHefeiAnhui230032China
| |
Collapse
|
46
|
Feng G, He N, Gao J, Li XC, Zhang FN, Liu CC, Targher G, Byrne CD, Mi M, Zheng MH, Ye F. Causal relationship between key genes and metabolic dysfunction-associated fatty liver disease risk mediated by immune cells: A Mendelian randomization and mediation analysis. Diabetes Obes Metab 2024; 26:5590-5599. [PMID: 39228284 DOI: 10.1111/dom.15925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024]
Abstract
AIM Non-invasive diagnostics for metabolic dysfunction-associated fatty liver disease (MAFLD) remain challenging. We aimed to identify novel key genes as non-invasive biomarkers for MAFLD, elucidate causal relationships between biomarkers and MAFLD and determine the role of immune cells as potential mediators. MATERIALS AND METHODS Utilizing published transcriptome data of patients with biopsy-proven MAFLD, we applied linear models for microarray data, least absolute shrinkage and selector operation (LASSO) regressions and receiver operating characteristic (ROC) curve analyses to identify and validate biomarkers for MAFLD. Using the expression quantitative trait loci database and a cohort of 778 614 Europeans, we used Mendelian randomization to analyse the causal relationships between key biomarkers and MAFLD. Additionally, mediation analysis was performed to examine the involvement of 731 immunophenotypes in these relationships. RESULTS We identified 31 differentially expressed genes, and LASSO regression showed three hub genes, IGFBP2, PEG10, and P4HA1, with area under the receiver operating characteristic (AUROC) curve of 0.807, 0.772 and 0.791, respectively, for identifying MAFLD. The model of these three genes had an AUROC of 0.959 and 0.800 in the development and validation data sets, respectively. This model was also validated using serum-based enzyme-linked immunosorbent assay data from MAFLD patients and control subjects (AUROC: 0.819, 95% confidence interval: 0.736-0.902). PEG10 was associated with an increased MAFLD risk (odds ratio = 1.106, p = 0.032) via inverse variance-weighted analysis, and about 30% of this risk was mediated by the percentage of CD11c + CD62L- monocytes. CONCLUSIONS The MAFLD panels have good diagnostic accuracy, and the causal link between PEG10 and MAFLD was mediated by the percentage of CD11c + CD62L- monocytes.
Collapse
Affiliation(s)
- Gong Feng
- Department of Infectious Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Institute of General Practice, Xi'an Medical University, Xi'an, China
| | - Na He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jing Gao
- School of Medicine, Xiamen University, Xiamen, China
- Department of Emergency Medicine, Affiliated Hospital of Xizang Minzu University, Xianyang, China
| | - Xiao-Cheng Li
- Institute of General Practice, Xi'an Medical University, Xi'an, China
| | - Fen-Na Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Cheng-Cheng Liu
- Institute of General Practice, Xi'an Medical University, Xi'an, China
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton, UK
| | - Man Mi
- Institute of General Practice, Xi'an Medical University, Xi'an, China
| | - Ming-Hua Zheng
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Feng Ye
- Department of Infectious Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
47
|
Kathuria I, Prasad A, Sharma BK, Aithabathula RV, Ofosu-Boateng M, Gyamfi MA, Jiang J, Park F, Singh UP, Singla B. Nidogen 2 Overexpression Promotes Hepatosteatosis and Atherosclerosis. Int J Mol Sci 2024; 25:12782. [PMID: 39684493 DOI: 10.3390/ijms252312782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Clinical and genetic studies strongly support a significant connection between nonalcoholic fatty liver disease (NAFLD) and atherosclerotic cardiovascular disease (ASCVD) and identify ASCVD as the primary cause of death in NAFLD patients. Understanding the molecular factors and mechanisms regulating these diseases is critical for developing novel therapies that target them simultaneously. Our preliminary immunoblotting experiments demonstrated elevated expression of nidogen 2 (NID2), a basement membrane glycoprotein, in human atherosclerotic vascular tissues and murine steatotic livers. Therefore, we investigated the role of NID2 in regulating hepatosteatosis and atherosclerosis utilizing Western diet-fed Apoe-/- mice with/without NID2 overexpression. Quantitative real-time PCR confirmed increased NID2 mRNA expression in multiple organs (liver, heart, kidney, and adipose) of NID2-overexpressing mice. Male mice with NID2 overexpression exhibited higher liver and epididymal white adipose tissue mass, increased hepatic lipid accumulation, and fibrosis. Additionally, these mice developed larger atherosclerotic lesions in the whole aortas and aortic roots, with increased necrotic core formation. Mechanistic studies showed reduced AMPK activation in the livers of NID2-overexpressing mice compared with controls, without any effects on hepatic inflammation. In conclusion, these findings suggest that NID2 plays a deleterious role in both hepatosteatosis and atherosclerosis, making it a potential therapeutic target for these conditions.
Collapse
Affiliation(s)
- Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Aditi Prasad
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Bal Krishan Sharma
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Maxwell A Gyamfi
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
48
|
Stachowicz A, Czepiel K, Wiśniewska A, Stachyra K, Ulatowska-Białas M, Kuśnierz-Cabala B, Surmiak M, Majka G, Kuś K, Wood ME, Torregrossa R, Whiteman M, Olszanecki R. Mitochondria-targeted hydrogen sulfide donor reduces fatty liver and obesity in mice fed a high fat diet by inhibiting de novo lipogenesis and inflammation via mTOR/SREBP-1 and NF-κB signaling pathways. Pharmacol Res 2024; 209:107428. [PMID: 39303773 DOI: 10.1016/j.phrs.2024.107428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Metabolic diseases that include obesity and metabolic-associated fatty liver disease (MAFLD) are a rapidly growing worldwide public health problem. The pathogenesis of MAFLD includes abnormally increased lipogenesis, chronic inflammation, and mitochondrial dysfunction. Mounting evidence suggests that hydrogen sulfide (H2S) is an important player in the liver, regulating lipid metabolism and mitochondrial function. However, direct delivery of H2S to mitochondria has not been investigated as a therapeutic strategy in obesity-related metabolic disorders. Therefore, our aim was to comprehensively evaluate the influence of prolonged treatment with a mitochondria sulfide delivery molecule (AP39) on the development of fatty liver and obesity in a high fat diet (HFD) fed mice. Our results demonstrated that AP39 reduced hepatic steatosis in HFD-fed mice, which was corresponded with decreased triglyceride content. Furthermore, treatment with AP39 downregulated pathways related to biosynthesis of unsaturated fatty acids, lipoprotein assembly and PPAR signaling. It also led to a decrease in hepatic de novo lipogenesis by downregulating mTOR/SREBP-1/SCD1 pathway. Moreover, AP39 administration alleviated obesity in HFD-fed mice, which was reflected by reduced weight of mice and adipose tissue, decreased leptin levels in the plasma and upregulated expression of adipose triglyceride lipase in epididymal white adipose tissue (eWAT). Finally, AP39 reduced inflammation in the liver and eWAT measured as the expression of proinflammatory markers (Il1b, Il6, Tnf, Mcp1), which was due to downregulated mTOR/NF-κB pathway. Taken together, mitochondria-targeted sulfide delivery molecules could potentially provide a novel therapeutic approach to the treatment/prevention of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Aneta Stachowicz
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| | - Klaudia Czepiel
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Wiśniewska
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Kamila Stachyra
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Ulatowska-Białas
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Beata Kuśnierz-Cabala
- Department of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Surmiak
- II Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Grzegorz Majka
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna Kuś
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Mark E Wood
- School of Biosciences, University of Exeter, Exeter, UK
| | | | | | - Rafał Olszanecki
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
49
|
Reyes-Avendaño I, Villaseñor-Altamirano AB, Reyes-Jimenez E, Velazquez-Enriquez JM, Baltiérrez-Hoyos R, Piña-Vázquez C, Muriel P, Villa-Treviño S, Arellanes-Robledo J, Vásquez-Garzón VR. Identification of key markers for the stages of nonalcoholic fatty liver disease: An integrated bioinformatics analysis and experimental validation. Dig Liver Dis 2024; 56:1887-1896. [PMID: 38824040 DOI: 10.1016/j.dld.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/14/2024] [Accepted: 05/08/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND The identification of biomarkers for the early diagnosis of nonalcoholic fatty liver disease (NAFLD) is urgently needed. Here, we aimed to identify NAFLD biomarkers in the early stages of steatosis (SS) and nonalcoholic steatohepatitis (NASH) based on differential gene expression from bioinformatics data. METHODS A meta-analysis was performed from transcriptomic databases retrieved from public repositories containing data from biopsies of patients at various stages of NAFLD development. The status of the selected molecules was validated in the serum of patients with NAFLD by ELISA. RESULTS We identified 121 differentially expressed genes (DEGs) associated with SS and 402 associated with NASH. Gene Ontology (GO) enrichment revealed that the altered genes were primarily associated with dysfunction of primary cellular processes, and pathway analyses were mainly related to cholesterol metabolism. We identified ACSS2, PCSK9, and CYP7A1 as candidate biomarkers for SS and ANGPTL3, CD36, CYP51A1, FASN, FAS, FDFT1, and LSS as candidate biomarkers for NASH. CONCLUSIONS By experimental validation of bioinformatics data from patients with NAFLD, we identified promising biomarkers for detecting SS and NASH that might be useful for screening and diagnosing early NAFLD stages in humans.
Collapse
Affiliation(s)
- Itayetzi Reyes-Avendaño
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico
| | - Ana Beatriz Villaseñor-Altamirano
- International Laboratory for Human Genome Research, Laboratorio Internacional de Investigación sobre el Genoma Humano (LIIGH), Universidad Nacional Autónoma de México (UNAM), 3001 Boulevard Juriquilla 76230, Querétaro, Mexico
| | - Edilburga Reyes-Jimenez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico
| | - Juan Manuel Velazquez-Enriquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Cinvestav-IPN, 07360 Ciudad de México, Mexico
| | - Saul Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 07360, Mexico
| | - Jaime Arellanes-Robledo
- CONAHCYT-Instituto Nacional de Medicina Genómica, Laboratorio de Enfermedades Hepáticas, Instituto Nacional de Medicina Genómica, Periférico Sur No. 4809, Col. Arenal Tepepan, Alcaldía Tlalpan 14610 Ciudad de México, Mexico
| | - Verónica Rocío Vásquez-Garzón
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua 68020, Oaxaca, Mexico.
| |
Collapse
|
50
|
Jung H, Kyun ML, Kwon JI, Kim J, Kim JK, Park D, Lee YB, Moon KS. Amplified response of drug-induced liver fibrosis via immune cell co-culture in a 3D in vitro hepatic fibrosis model. Biomater Sci 2024. [PMID: 39483068 DOI: 10.1039/d4bm00874j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Liver fibrosis, a critical consequence of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition driven by inflammation. This process involves complex interactions among hepatocytes, hepatic stellate cells (HSCs), and Kupffer cells, the liver's resident macrophages. Kupffer cells are essential in initiating fibrosis through the release of pro-inflammatory cytokines that activate HSCs. Although various in vitro liver fibrosis models have been developed, there is a lack of models that include the immune environment of the liver to clarify the influence of immune cells on the progression of liver fibrosis. We developed an in vitro liver fibrosis model by co-culturing hepatocytes (HepaRG), a hepatic stellate cell line (LX-2), and macrophages (differentiated THP-1). The effects of liver fibrosis inducers, transforming growth factor-beta1 (TGF-β1) and methotrexate (MTX), on the inflammatory response and stellate cell activation were evaluated in this triple co-culture model. A triple co-culture condition was developed as a 3D in vitro model using gelatin methacrylate (GelMA), offering a more biomimetic environment and achieving liver fibrosis via immune cell activation associated ECM deposition. In this study, the developed triple co-culture model has the potential to elucidate cell progression roles in liver fibrosis and can be applied in drug screening and toxicity assessments targeting liver fibrosis.
Collapse
Affiliation(s)
- Hyewon Jung
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| | - Mi-Lang Kyun
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Ji-In Kwon
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Department of Food and Nutrition, University of Hannam, Daejeon, 34054, Republic of Korea
| | - Jeongha Kim
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Department of Food and Nutrition, University of Hannam, Daejeon, 34054, Republic of Korea
| | - Ju-Kang Kim
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Daeui Park
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
- Center for Biomimetic Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Yu Bin Lee
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| | - Kyoung-Sik Moon
- Center for Global Biopharmaceutical Research, Korea Institute of Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
- Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|