1
|
Xu C, Ezzi SHA, Zou X, Dong Y, Alhaskawi A, Zhou H, Kota VG, Abdulla MHAH, Abdalbary SA, Lu H. The role of TNF in metabolic disorders and liver diseases. Cytokine 2025; 190:156933. [PMID: 40174483 DOI: 10.1016/j.cyto.2025.156933] [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: 01/20/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Tumor necrosis factor (TNF) is identified as a pro-inflammatory cytokine critical to the pathology of liver disease. In the carbohydrate metabolism, TNF has been demonstrated to impede the insulin signaling pathway, thereby precipitating glucose intolerance and insulin resistance. In lipid metabolism, TNF upregulates genes implicated in fatty acid synthesis, resulting in increased lipid accumulation within the liver. In amino acid metabolism, TNF has shown to promote the gene expression for amino acid catabolism, leading to decreased protein synthesis. Additionally, TNF stimulates the production of other chemokines and inflammatory cytokines that can further exacerbate liver injury. Overall, TNF is crucial in developing liver diseases by disrupting various metabolic pathways in the liver, causing insulin resistance, lipid accumulation, and decreased protein synthesis. This review summarizes the present understanding of TNF's role in the regulation of carbohydrate, lipid and amino acid metabolism in liver disease together with its potential therapeutic implications.
Collapse
Affiliation(s)
- Chuze Xu
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Xiaodi Zou
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yanzhao Dong
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ahmad Alhaskawi
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Haiying Zhou
- Faculty of Medicine, The Chinese University of Hong Kong School of Biomedical Science, Hong Kong, China
| | | | | | - Sahar Ahmed Abdalbary
- Department of Orthopedic Physical Therapy, Faculty of Physical Therapy, Nahda University, Beni Suef, Egypt
| | - Hui Lu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University, Hangzhou, China; Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Marei M, El-Nikhely N, Sheta E, Ragab H, Wahid A, Saeed H, Rostom SAF. Biochemical and Molecular Studies on the Role of Celecoxib and Some Related Bipyrazoles in Mitigating Induced Liver Injury in Experimental Animals. Drug Des Devel Ther 2025; 19:3857-3882. [PMID: 40391176 PMCID: PMC12087607 DOI: 10.2147/dddt.s512058] [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: 12/13/2024] [Accepted: 05/01/2025] [Indexed: 05/21/2025] Open
Abstract
Introduction Liver fibrosis is a life-threatening disease that greatly impacts the morbidity and mortality of hepatic patients worldwide, resulting mainly as a consequence of hepatitis C, alcoholic and non-alcoholic fatty liver. COX-1 and COX-2 isozymes catalyze the synthesis of prostaglandins (PGs) and thromboxanes (TXs) from arachidonic acid causing inflammation. Owing to the scarcity of approved fibrolytic drugs available for human use, celecoxib (a selective COX-2 inhibitor) has been repurposed as a potential antifibrotic and fibrolytic agent in some chronic liver fibrosis models. Methods The present study aims to discover a non-invasive treatment for liver fibrosis through investigating the possible ability of three celecoxib-related bipyrazole compounds HR1-3 to reverse chemically induced liver fibrosis in rats using CCl4. This fibrolytic effect was verified by histopathological, immunohistochemical, biochemical and biomolecular assays. In addition, in silico computer-aided evaluation of the compounds' binding mode to certain molecular targets was performed, and the in silico physicochemical properties, drug likeness and pharmacokinetic parameters were predicted using web-based applications. Results The analogs HR1-3 could serve as novel therapeutic candidates for the mitigation of liver fibrosis that deserves further derivatizations and investigations. In particular, the fluorinated analog HR3 proved to be the most active member in this study when compared to celecoxib due to its distinguished histopathological and immunohistochemical investigation results, beside its antioxidant potential, as well as its reliable effects against some biomarkers, namely, MMP-9, TGF-β1, TIMP-1, IL-6 and TNF-α. Conclusion Based on the obtained results, the fluorinated analog HR3 could serve as a novel therapeutic candidate for the amelioration of liver fibrosis that deserves further derivatizations and investigations.
Collapse
Affiliation(s)
- Maram Marei
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21521, Egypt
| | - Nefertiti El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21521, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, 21521, Egypt
| | - Hanan Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21521, Egypt
| | - Sherif A F Rostom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
3
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
4
|
Vosough M, Shokouhian B, Sharbaf MA, Solhi R, Heidari Z, Seydi H, Hassan M, Devaraj E, Najimi M. Role of mitogens in normal and pathological liver regeneration. Hepatol Commun 2025; 9:e0692. [PMID: 40304568 PMCID: PMC12045551 DOI: 10.1097/hc9.0000000000000692] [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: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 05/02/2025] Open
Abstract
The liver has a unique ability to regenerate to meet the body's metabolic needs, even following acute or chronic injuries. The cellular and molecular mechanisms underlying normal liver regeneration have been well investigated to improve organ transplantation outcomes. Once liver regeneration is impaired, pathological regeneration occurs, and the underlying cellular and molecular mechanisms require further investigations. Nevertheless, a plethora of cytokines and growth factor-mediated pathways have been reported to modulate physiological and pathological liver regeneration. Regenerative mitogens play an essential role in hepatocyte proliferation. Accelerator mitogens in synergism with regenerative ones promote liver regeneration following hepatectomy. Finally, terminator mitogens restore the proliferating status of hepatocytes to a differentiated and quiescent state upon completion of regeneration. Chronic loss of hepatocytes, which can manifest in chronic liver disorders of any etiology, often has undesired structural consequences, including fibrosis, cirrhosis, and liver neoplasia due to the unregulated proliferation of remaining hepatocytes. In fact, any impairment in the physiological function of the terminator mitogens results in the progression of pathological liver regeneration. In the current review, we intend to highlight the updated cellular and molecular mechanisms involved in liver regeneration and discuss the impairments in central regulating mechanisms responsible for pathological liver regeneration.
Collapse
Affiliation(s)
- Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bahare Shokouhian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Amin Sharbaf
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Heidari
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ezhilarasan Devaraj
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| |
Collapse
|
5
|
Farbod Y, Kankouni H, Moini M, Fung S. Hepatitis B-Induced Hepatocellular Carcinoma: Understanding Viral Carcinogenesis and Disease Management. J Clin Med 2025; 14:2505. [PMID: 40217955 PMCID: PMC11989475 DOI: 10.3390/jcm14072505] [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: 02/28/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Hepatitis B virus (HBV) infection is a leading cause of chronic liver disease and liver cancer worldwide. Hepatocellular carcinoma (HCC) remains one of the major causes of cancer-related mortality globally. Effective prevention and management strategies for HBV infection are crucial in reducing liver-related complications, including HCC. HBV plays a distinct role in liver carcinogenesis, and there is growing knowledge about the factors contributing to its oncogenic potential. With advancements in HCC management, special attention must be given to the treatment of HBV infection in patients with HBV-induced HCC. In this review, we summarize current insights into the carcinogenic mechanisms of HBV and discuss the latest approaches to managing HBV-induced HCC.
Collapse
Affiliation(s)
- Yasamin Farbod
- Division of Gastroenterology and Hepatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Husain Kankouni
- Division of Gastroenterology and Hepatology, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Maryam Moini
- Division of Gastroenterology, McMaster University, Hamilton, ON L8S 2A5, Canada
| | - Scott Fung
- Department of Medicine, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
6
|
Fahim JR, Samy MN, Ibrahem ES, Fawzy MA, Saber EA, Kamel MS, Sugimoto S, Matsunami K, Attia EZ. Hepatoprotective potential of Ceiba chodatii Hassl. Against carbon tetrachloride-induced chronic liver damage supported with phytochemical investigation. Fitoterapia 2025; 182:106466. [PMID: 40058657 DOI: 10.1016/j.fitote.2025.106466] [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: 05/18/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Hepatic fibrosis is a major health concern that can develop into other life-threatening pathologies, with no fully effective treatments are available to date. Ceiba is a genus of multipurpose trees with diverse therapeutic applications, including liver ailments. Prior research has also unveiled the protecting role of Ceiba plants in chemical liver injuries via a number of in vitro and in vivo tests. Due to the crucial need for alternative therapies to prevent liver damage and stop its progress, the present work evaluates the protective effects of the total extract of Ceiba chodatii Hassl. flowers and its derived fractions (I-IV) against CCl4-induced chronic liver damage for the first time. The obtained results indicated the ability of C. chodatii flowers, particularly their chloroform- and ethyl acetate-soluble fractions (II and III), to alleviate liver damage in CCl4-intoxicated rats via normalizing high liver injury hallmarks (e.g., ALT, AST, albumin, and total bilirubin), preventing the build-up of malondialdehyde, enhancing the antioxidant capacity of hepatocytes, mitigating aberrant histopathological changes, and reducing extracellular matrix accumulation. Further mechanistic studies showed the aptitude of C. chodatii flowers to attenuate inflammatory, fibrotic, and apoptotic responses via counteracting the production of inflammatory cytokines (e.g., IL-6 and TNF-α), reducing the levels of cleaved caspase-3, and inhibiting JAK2/STAT3 and TGF-β/Smad signaling pathways. Interestingly, the liver-protecting actions of fractions II and III were also comparable to those of silymarin (50 mg/kg). Moreover, phytochemical investigation of C. chodatii flowers led to the isolation and identification of a group of flavonoid glycosides (1-10), with good antioxidant and liver supporting properties, suggesting their potential contribution to the anti-fibrotic properties of C. chodatii. These data highlight the multi-target hepatoprotective effects of C. chodatii and its potential as an alternative source to develop natural therapeutic agents against liver fibrosis.
Collapse
Affiliation(s)
- John Refaat Fahim
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Minia National University, 61111 New Minia, Egypt.
| | - Mamdouh Nabil Samy
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Minia National University, 61111 New Minia, Egypt
| | - Engy Saadalah Ibrahem
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt; Department of Biochemistry, Faculty of Pharmacy, Minia National University, 61111 New Minia, Egypt
| | - Entesar Ali Saber
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, 61519 Minia, Egypt; Department of Medical Science, Histology and Cell Biology, Deraya University, 61111 New Minia, Egypt
| | - Mohamed Salah Kamel
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Sachiko Sugimoto
- Department of Pharmacognosy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8553 Hiroshima, Japan
| | - Katsuyoshi Matsunami
- Department of Pharmacognosy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, 734-8553 Hiroshima, Japan
| | - Eman Zekry Attia
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Minia National University, 61111 New Minia, Egypt
| |
Collapse
|
7
|
Robinson GI, Gerasymchuk M, Zanikov T, Gojani EG, Asghari S, Groves A, Haselhorst L, Nandakumar S, Stahl C, Cruz C, Cameron M, Zahoruiko Y, Li D, Rodriguez-Juarez R, Snelling A, Hudson D, Fiselier A, Kovalchuk O, Kovalchuk I. LPS-Induced Liver Inflammation Is Inhibited by Psilocybin and Eugenol in Mice. Pharmaceuticals (Basel) 2025; 18:451. [PMID: 40283890 PMCID: PMC12030523 DOI: 10.3390/ph18040451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Liver inflammatory diseases are a major global health burden and are often exacerbated by inflammation driven by lipopolysaccharides (LPS) through toll-like receptor 4 signaling. This study evaluates the anti-inflammatory effects of psilocybin and eugenol in an LPS-induced liver inflammation model in C57BL/6J mice. Methods: Mice were treated with psilocybin (0.88 mg/kg) and/or eugenol (17.59 mg/kg) either before (pre-treatment) or after (post-treatment) LPS injection. Results: Psilocybin and eugenol, individually and in combination, significantly reduced the LPS-induced mRNA levels of pro-inflammatory cytokines, with post-treatment administration exhibiting stronger effects than pre-treatment. Psilocybin alone displayed the most pronounced anti-inflammatory response, especially for IL-1β, IL-6, and MCP-1, while its combination with eugenol in 1:50 ratio demonstrated similar results, with strongly reduced COX-2 and TNF-α. Histological analysis revealed improved nuclear circularity and reduced inflammatory infiltration in the treatment groups. Eugenol alone showed potential adverse effects, including increased MCP-1 and GM-CSF, but this was mitigated by the co-administration of psilocybin. Conclusions: These findings highlight psilocybin and its combination with eugenol as promising therapies for hepatic inflammation, suggesting their application in treating acute and chronic liver diseases. Future research should explore their long-term effects, the mechanisms underlying their anti-inflammatory actions, and their therapeutic efficacy in humans.
Collapse
Affiliation(s)
- Gregory Ian Robinson
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Marta Gerasymchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Timur Zanikov
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Esmaeel Ghasemi Gojani
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Shima Asghari
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Alyssa Groves
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Lucie Haselhorst
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Institute for Medical Nutrition Science, Universität zu Lübeck, 23562 Lübeck, Germany
| | - Sanjana Nandakumar
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Cora Stahl
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- Department of Medicine, Medical Sciences, and Nutrition, King’s College, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Ceejay Cruz
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Mackenzie Cameron
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Yeva Zahoruiko
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Rocio Rodriguez-Juarez
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Alex Snelling
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Darryl Hudson
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Anna Fiselier
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- GoodCap Pharmaceuticals, 520 3rd Avenue SW, Suite 1900, Calgary, AB T2P 0R3, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada (M.G.)
| |
Collapse
|
8
|
Żurawek D, Pydyn N, Major P, Szade K, Trzos K, Kuś E, Pośpiech E, Małczak P, Radkowiak D, Budzyński A, Chłopicki S, Jura J, Kotlinowski J. Diosmetin alleviates TNFα-induced liver inflammation by improving liver sinusoidal endothelial cell dysfunction. Biomed Pharmacother 2025; 183:117843. [PMID: 39813788 DOI: 10.1016/j.biopha.2025.117843] [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: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Sterile inflammation contributes to the development of many liver diseases including non-alcoholic fatty liver disease. Tumor necrosis factor alpha (TNFα) is a key cytokine driving liver inflammation primarily through pro-inflammatory activation of liver sinusoidal endothelial cells (LSEC). The knowledge of whether modulating LSEC activation can alleviate liver inflammation is scarce. This study aims to establish and validate an animal model mimicking LSEC dysfunction observed in obese patients with elevated plasma levels of TNFα, and explore whether vasoactive flavonoid diosmetin could serve as a therapeutic agent for liver inflammation by modulation of LSEC dysfunction. Obese patients with elevated plasma levels of TNFα, LSEC dysfunction and liver inflammation had also reduced Mcpip1 expression in peripheral blood mononuclear cells. Mcpip1 is a protein that negatively regulates the levels of pro-inflammatory cytokines. To model this, we generated mice with Mcpip1 knock-out in myeloid cells (Mcpip1fl/flLysMCre), which displayed systemic and liver inflammation like that observed in patients. Diosmetin treatment efficiently reduced TNFα-dependent LSEC activation in vitro and in vivo, and reduced liver inflammation in Mcpip1fl/flLysMCre mice without affecting systemic inflammation. Diosmetin's effects may stem from inhibiting NF-κB pathway in TNFα-activated endothelial cells. Our findings demonstrate that the Mcpip1fl/flLysMCre mouse model is useful for studying new anti-inflammatory therapies for the liver. We show that diosmetin, a vasoactive flavonoid used in the clinic to treat chronic venous insufficiency, also has strong anti-inflammatory properties in the liver. These results indicate that diosmetin has the potential to be further investigated as a supportive therapy for liver inflammation in humans.
Collapse
Affiliation(s)
- Dariusz Żurawek
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Gronostajowa 7, Kraków 30-387, Poland; Douglas Mental Health University Institute, McGill University, 6875 LaSalle Boulevard, Montreal, QC H4H 1R3, Canada.
| | - Natalia Pydyn
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Gronostajowa 7, Kraków 30-387, Poland
| | - Piotr Major
- Jagiellonian University Medical College, 2nd Department of General Surgery, Jakubowskiego 2, Kraków 30-688, Poland
| | - Krzysztof Szade
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Laboratory of Stem Cell Biology, Gronostajowa 7, Kraków 30-387, Poland
| | - Katarzyna Trzos
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Gronostajowa 7, Kraków 30-387, Poland
| | - Edyta Kuś
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics, Bobrzyńskiego 14, Kraków 30-348, Poland
| | - Ewelina Pośpiech
- Department of Genomics and Forensic Genetics, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-204, Szczecin, Poland
| | - Piotr Małczak
- Jagiellonian University Medical College, 2nd Department of General Surgery, Jakubowskiego 2, Kraków 30-688, Poland
| | - Dorota Radkowiak
- Jagiellonian University Medical College, 2nd Department of General Surgery, Jakubowskiego 2, Kraków 30-688, Poland
| | - Andrzej Budzyński
- Jagiellonian University Medical College, 2nd Department of General Surgery, Jakubowskiego 2, Kraków 30-688, Poland
| | - Stefan Chłopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics, Bobrzyńskiego 14, Kraków 30-348, Poland
| | - Jolanta Jura
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Gronostajowa 7, Kraków 30-387, Poland
| | - Jerzy Kotlinowski
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of General Biochemistry, Gronostajowa 7, Kraków 30-387, Poland.
| |
Collapse
|
9
|
Barbhuiya PA, Ahmed A, Dutta PP, Sen S, Pathak MP. Mitigating Metabolic Dysfunction-associated Steatotic Liver Disease (MASLD): The Role of Bioactive Phytoconstituents in Indian Culinary Spices. Curr Nutr Rep 2025; 14:20. [PMID: 39841356 DOI: 10.1007/s13668-024-00598-w] [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] [Accepted: 11/11/2024] [Indexed: 01/30/2025]
Abstract
PURPOSE OF REVIEW The term metabolic dysfunction-associated steatotic liver disease (MASLD) refers to a group of progressive steatotic liver conditions that include metabolic dysfunction-associated steatohepatitis (MASH), which has varying degrees of liver fibrosis and may advance to cirrhosis, and independent hepatic steatosis. MASLD has a complex underlying mechanism, with patients exhibiting diverse causes and phases of the disease. India has a pool prevalence of MASLD of 38.6% in adults. In 2023, the term NAFLD has been redefined and changed to MASLD. Currently, there are no drugs approved by the FDA for the treatment of MASLD. This study investigates the potential of bioactive phytoconstituents present in spices as a therapeutic approach for MASLD. Moreover, it offers comprehensive data on several pre-clinical studies of bioactive phytoconstituents derived from spices that primarily focus on treating obesity-associated MASLD. RECENT FINDINGS Spices include a high amount of bioactive chemicals and several research have indicated their diverse pharmacological activities. Bioactive phytoconstituents from common Indian spices like cinnamic acid, eugenol, curcumin, allicin, 6-gingerols, capsaicin, piperine, eucalyptol, trigonelline, and linalool have been reported to exhibit anti-MASLD effects both in-vivo and in-vitro. Bioactive phytoconstituents from different culinary species of India have shown promising potential against MASLD in pre-clinical status. Further clinical studies on a large scale would be beneficial for paving the path to the development of a new drug which is the need of time.
Collapse
Affiliation(s)
- Pervej Alom Barbhuiya
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Ameena Ahmed
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Rahman Institute of Pharmaceutical Sciences and Research, Tepesia, Sonapur, Assam, India, PIN - 782402
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026.
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026.
| |
Collapse
|
10
|
Houtzager JHE, van Stalborch AM, Hofstee C, van Gulik TM, van Buul JD. Protection of liver sinusoidal endothelial cells using different preservation solutions. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2025; 7:e240004. [PMID: 39812559 PMCID: PMC11825184 DOI: 10.1530/vb-24-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 01/16/2025]
Abstract
Donor liver preservation methods and solutions have evolved over the last years. Liver sinusoidal endothelial cell (LSEC) barrier function and integrity during preservation are crucial for outcomes of liver transplantation. Therefore, the present study aimed to determine optimal preservation of LSEC barrier function and integrity using different preservation solutions. Human umbilical vein endothelial cells (HUVECs) and LSECs were incubated in either University of Wisconsin machine perfusion solution (UW-MPS), histidine-tryptophan-ketoglutarate, or endothelial cell growth medium 2 (EGM2) (as a gold standard for cell culturing). Endothelial integrity was assessed by measurement of cellular morphology and expression of membrane proteins: PECAM-1, ICAM-1 and Fc-gamma receptor CD32b (FcΥRCD32b). Endothelial barrier function was measured by electric cell-substrate impedance sensing. Cellular response to inflammatory stimuli with tumor necrosis factor-alpha (TNF-α) was tested by studying trans-endothelial migration (TEM) under flow conditions. Differences in these parameters were analyzed between the different preservation solutions. PECAM-1 expression was high for all preservation solutions in HUVECs and LSECs. ICAM-1 expression was increased in both LSECs and HUVECs in all preservation solutions plus TNF-α. UW reduced PECAM-1 expression, whereas EGM2 medium promoted barrier function in LSECs and HUVECs, and monolayer recovery after wounding was best achieved in cells incubated in EGM2. LSECs and HUVECs incubated with EGM2 plus TNF-α both supported neutrophil adhesion and TEM, but much less to none when incubated in UW plus TNF-α. Overall, EGM2 showed the best results in preserving endothelial barrier function for both HUVECs and LSECs.
Collapse
Affiliation(s)
- Julia H E Houtzager
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Molecular Cell Biology Lab., Department Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Anne-Marieke van Stalborch
- Molecular Cell Biology Lab., Department Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
- Vascular Cell Biology Lab., Department Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Charlotte Hofstee
- Molecular Cell Biology Lab., Department Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab., Department Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
- Vascular Cell Biology Lab., Department Medical Biochemistry, Amsterdam UMC, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Lee J, Hong SW, Kim MJ, Lim YM, Moon SJ, Kwon H, Park SE, Rhee EJ, Lee WY. Sodium-glucose cotransporter 2 inhibitors ameliorate ER stress-induced pro-inflammatory cytokine expression by inhibiting CD36 in NAFLD progression in vitro. Biochem Biophys Res Commun 2024; 735:150620. [PMID: 39265364 DOI: 10.1016/j.bbrc.2024.150620] [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: 06/28/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024]
Abstract
Sodium-dependent glucose cotransporter-2 (SGLT2) inhibitors, antidiabetic drugs that reduce blood sugar levels by inhibiting glucose reabsorption in the renal proximal tubules, also ameliorate nonalcoholic fatty liver disease (NAFLD). This study aimed to examine the effects of SGLT2 inhibition on hepatic steatosis and nonalcoholic steatohepatitis (NASH) using an in vitro model of NAFLD progression. HepG2 cells and a coculture of Hepa1c1c7 and Raw 264.7 cells were treated with 400 μM palmitic acid (PA), followed by treatment with or without 10 μM empagliflozin and dapagliflozin. In HepG2 cells, PA increased hepatic lipid accumulation, the expression of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), exocytosis mediators (VAMP3 and SNAP23), and ER stress markers (GRP78, PERK, IRE1α, ATF6, ATF4, and CHOP), and the gene and protein expression of CD36. SGLT2 inhibitors reversed the effects of PA. SGLT2 inhibition via siRNA reduced proinflammatory-cytokine gene expression in thapsigargin-treated HepG2 cells. Transfection with CD36 siRNA reversed the elevated ATF4 and CHOP expression in PA-treated HepG2 cells. SGLT2 inhibition via an SGTL2 inhibitor and SGLT2 siRNA reduced CD36, Tnf-α, Il-6, Il-1β, Vamp2, Snap23, Atf4, and Chop expression in the PA-treated Hepa1c1c7-Raw 264.7 cell coculture and suppressed Tnf-α release in the Hepa1c1c7-Raw 264.7 cell coculture treated with lipopolysaccharide and PA. These findings indicate that SGLT2 inhibitors inhibited NAFLD progression by reducing hepatic lipid accumulation and inflammation.
Collapse
Affiliation(s)
- Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Min-Jeong Kim
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Yu-Mi Lim
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Sun Joon Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea
| | - Eun-Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| | - Won-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, #29 Seamunan-ro, Jongro-Ku, Seoul, 03181, Republic of Korea.
| |
Collapse
|
12
|
Lee YP, Chiu CC, Chang YC, Chen YH, Wu WK, Wu MS, Chuang HL. Co-exposure to different bacterial species' lipopolysaccharides with the NASH diet exacerbates NASH and liver fibrosis progression in mice. Clin Res Hepatol Gastroenterol 2024; 48:102470. [PMID: 39317267 DOI: 10.1016/j.clinre.2024.102470] [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: 02/11/2024] [Revised: 09/16/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND AND AIM With the obesity epidemic, nonalcoholic fatty liver disease (NAFLD) has become a public health concern, but its progression mechanism remains unclear. Experimental models mimicking human NAFLD/steatohepatitis (NASH) are crucial. This study simulates gut microbiota imbalance effects on NASH and liver fibrosis. METHODS We used different bacterial sources of lipopolysaccharide (LPS), including Escherichia coli (GEC) and Salmonella abortus equi (GSE), combined with a Gubra Amylin NASH (GAN) diet to induce NASH and liver fibrosis. RESULTS The GSE group showed significantly higher serum alanine aminotransferase, hydroxyproline, CD68-positive cells, α-smooth muscle actin, glial fibrillary acidic protein, and TNF-α, COL1A1, TGF-β, and NLRP3 expressions compared to the the GAN group. The GSE group also had higher Erysipelotrichaceae, Akkermansiaceae, and Bacteroidaceae family numbers. CONCLUSIONS The GAN diet with LPS treatment successfully induced NASH and fibrosis making this model useful for preclinical NASH drug testing.
Collapse
Affiliation(s)
- Yen-Peng Lee
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Chien-Chao Chiu
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Yung-Chi Chang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Yi-Hsun Chen
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Wei-Kai Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan.
| |
Collapse
|
13
|
Elazab ST, Hsu WH. Ferulic acid ameliorates concanavalin A-induced hepatic fibrosis in mice via suppressing TGF-β/smad signaling. Toxicol Appl Pharmacol 2024; 492:117099. [PMID: 39260469 DOI: 10.1016/j.taap.2024.117099] [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: 06/08/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIM Hepatic fibrosis, one of the main reasons for death globally, is a serious complication of chronic liver disorders. However, the available therapies for liver fibrosis are limited, ineffective, and often associated with adverse events. Hence, seeking for a novel, effective therapy is warranted. Our objective was to investigate the potential efficacy of ferulic acid (FA), a phenolic phytochemical, at different doses in hindering the progress of concanavalin A (Con A)-induced hepatic fibrosis and explore the involved mechanisms. METHODS Thirty-six mice were assorted into 6 groups (n = 6): Group I (control); group II received FA (20 mg/kg/day orally for 4 weeks); group III received Con A (6 mg/kg/week/i.v.) for 4 weeks; groups IV, V, and VI received Con A and were offered FA at 5, 10, and 20 mg/kg/day, respectively. RESULTS The data showed the palliative effect of FA against Con A-induced fibrosis in a dose-dependent manner. This was obvious from the recovery of liver markers and hepatic architecture with the regression of fibrosis in FA-treated mice. FA abolished Con A-mediated oxidative insults and promoted the antioxidant enzyme activities, which run through the Nrf2/HO-1 signaling. Additionally, FA suppressed Con A-induced increase in NF-kB and IL-β levels, and TNF-α immune-expression. The anti-fibrotic effect of FA was evident from the drop in TGF-β, smad3 levels, α-SMA expression, and hydroxyproline content. CONCLUSION FA attenuated Con A-induced liver fibrosis through stimulating Nrf2 signaling, suppressing NF-kB, and inhibiting the TGF-β/smad3 signaling pathway. Thus FA can be considered as a promising therapy for combating liver fibrosis.
Collapse
Affiliation(s)
- Sara T Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Walter H Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa 50011, USA.
| |
Collapse
|
14
|
Sun Z, Wei Y, Xu Y, Jiao J, Duan X. The use of traditional Chinese medicine in the treatment of non-alcoholic fatty liver disease: A review. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2024; 12:100475. [DOI: 10.1016/j.prmcm.2024.100475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Wang Y, Stoess C, Holzmann G, Mogler C, Stupakov P, Altmayr F, Schulze S, Wang B, Steffani M, Friess H, Hüser N, Holzmann B, Hartmann D, Laschinger M. Signalling of the neuropeptide calcitonin gene-related peptide (CGRP) through RAMP1 promotes liver fibrosis via TGFβ1/Smad2 and YAP pathways. Exp Cell Res 2024; 442:114193. [PMID: 39103072 DOI: 10.1016/j.yexcr.2024.114193] [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: 02/01/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
The liver is innervated by primary sensory nerve fibres releasing the neuropeptide calcitonin gene-related peptide (CGRP). Elevated plasma levels of CGRP have been found in patients with liver fibrosis or cirrhosis. We hypothesised that signalling of CGRP and its receptors might regulate liver fibrosis and propose a novel potential target for the treatment. In this study, hepatic expression of CGRP and its receptor component, the receptor activity-modifying protein 1 (RAMP1), was dramatically increased in diseased livers of patients. In a murine liver fibrosis model, deficiency of RAMP1 resulted in attenuated fibrogenesis characterized by less collagen deposition and decreased activity of hepatic stellate cells (HSC). Mechanistically, activity of the TGFβ1 signalling core component Smad2 was severely impaired in the absence of RAMP1, and Yes-associated protein (YAP) activity was found to be diminished in RAMP1-deficient liver parenchyma. In vitro, stimulation of the HSC line LX-2 cells with CGRP induces TGFβ1 production and downstream signalling as well as HSC activation documented by increased α-SMA expression and collagen synthesis. We further demonstrate in LX-2 cells that CGRP promotes YAP activation and its nuclear translocation subsequent to TGFβ1/Smad2 signals. These data support a promotive effect of CGRP signalling in liver fibrosis via stimulation of TGFβ1/Smad2 and YAP activity.
Collapse
Affiliation(s)
- Yang Wang
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany; Department of Hepato-Pancreato-Biliary Center, Zhongda Hospital, Southeast University School of Medicine, Dingjia Road 87, 210009, Nanjing, China
| | - Christian Stoess
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Gabriela Holzmann
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Carolin Mogler
- Technical University of Munich, TUM School of Medicine and Health, Institute of Pathology, Trogerstr. 18, 81675, Munich, Germany
| | - Pavel Stupakov
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Felicitas Altmayr
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Sarah Schulze
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Baocai Wang
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany; University Hospital of Tübingen, Department of General, Visceral and Transplantation Surgery, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany; The M3 Research Center, Eberhard Karls University, Otfried-Müller-Str. 37, 72076 Tübingen, Germany
| | - Marcella Steffani
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Helmut Friess
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Norbert Hüser
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Bernhard Holzmann
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany
| | - Daniel Hartmann
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany; University Hospital of Tübingen, Department of General, Visceral and Transplantation Surgery, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany; The M3 Research Center, Eberhard Karls University, Otfried-Müller-Str. 37, 72076 Tübingen, Germany
| | - Melanie Laschinger
- Technical University of Munich, TUM School of Medicine and Health, Department of Surgery, Klinikum rechts der Isar, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
16
|
Barbhuiya PA, Sen S, Pathak MP. Ameliorative role of bioactive phytoconstituents targeting obesity associated NAFLD by modulation of inflammation and lipogenesis pathways: a comprehensive review. PHYTOCHEMISTRY REVIEWS 2024; 23:969-996. [DOI: 10.1007/s11101-023-09912-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2025]
|
17
|
Tagliaferro M, Marino M, Basile V, Pocino K, Rapaccini GL, Ciasca G, Basile U, Carnazzo V. New Biomarkers in Liver Fibrosis: A Pass through the Quicksand? J Pers Med 2024; 14:798. [PMID: 39201990 PMCID: PMC11355846 DOI: 10.3390/jpm14080798] [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: 06/08/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Chronic liver diseases (CLD) stem from various causes and lead to a gradual progression that ultimately may result in fibrosis and eventually cirrhosis. This process is typically prolonged and asymptomatic, characterized by the complex interplay among various cell types, signaling pathways, extracellular matrix components, and immune responses. With the prevalence of CLD increasing, diagnoses are often delayed, which leads to poor prognoses and in some cases, the need for liver transplants. Consequently, there is an urgent need for the development of novel, non-invasive methods for the diagnosis and monitoring of CLD. In this context, serum biomarkers-safer, repeatable, and more acceptable alternatives to tissue biopsies-are attracting significant research interest, although their clinical implementation is not yet widespread. This review summarizes the latest advancements in serum biomarkers for detecting hepatic fibrogenesis and advocates for concerted efforts to consolidate current knowledge, thereby providing patients with early, effective, and accessible diagnoses that facilitate personalized therapeutic strategies.
Collapse
Affiliation(s)
- Marzia Tagliaferro
- Dipartimento di Patologia Clinica, Ospedale Santa Maria Goretti, A.U.S.L. Latina, 04100 Latina, Italy; (M.T.); (V.C.)
| | - Mariapaola Marino
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.M.); (G.L.R.)
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Krizia Pocino
- Clinical Pathology Unit, San Pietro Fatebenefratelli Hospital, 00189 Rome, Italy;
| | - Gian Ludovico Rapaccini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.M.); (G.L.R.)
| | - Gabriele Ciasca
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Umberto Basile
- Dipartimento di Patologia Clinica, Ospedale Santa Maria Goretti, A.U.S.L. Latina, 04100 Latina, Italy; (M.T.); (V.C.)
| | - Valeria Carnazzo
- Dipartimento di Patologia Clinica, Ospedale Santa Maria Goretti, A.U.S.L. Latina, 04100 Latina, Italy; (M.T.); (V.C.)
| |
Collapse
|
18
|
Kumar V, Kumar R, Gurusubramanian G, Rathore SS, Roy VK. Morin hydrate ameliorates Di-2-ethylhexyl phthalate (DEHP) induced hepatotoxicity in a mouse model via TNF-α and NF-κβ signaling. 3 Biotech 2024; 14:181. [PMID: 38911474 PMCID: PMC11189377 DOI: 10.1007/s13205-024-04012-8] [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: 02/01/2024] [Accepted: 05/18/2024] [Indexed: 06/25/2024] Open
Abstract
Di-(2-ethylhexyl) phthalic acid (DEHP) pollutes the environment, and posing a significant risk to human and animal health. Consequently, a successful preventative strategy against DEHP-induced liver toxicity needs to be investigated. Morin hydrate (MH), a flavanol compound, possesses toxic preventive attributes against various environmental pollutants. However, the effects of MH have not been investigated against DEHP-induced liver toxicity. Female Swiss albino mice were divided into four groups: control, DEHP (orally administered with 500 mg/kg, DEHP plus MH 10 mg/kg, and DEHP plus MH 100 mg/kg for 14 days. The results showed that the MH treatment ameliorated the DEHP-induced liver dysfunctions by decreasing the alanine transaminase (ALT), aspartate aminotransferase (AST), total bilirubin, liver histoarchitecture, fibrosis, and markers of oxidative stress. Furthermore, DEHP increased apoptosis, increased active caspase 3 and decreased B cell lymphoma-2 (Bcl-2) expression. However, the MH treatment showed a differential effect on these proteins; a lower dose increased, and a higher dose decreased the expression. Thus, a lower dose of MH could be involved in the disposal of damaged hepatocytes. Expression of Estrogen receptors alpha (ERα) also showed a similar trend with active caspase 3. Furthermore, the expression of Tumor necrosis factor alpha (TNF-α) and Nuclear factor-κβ (NF-κβ) were up-regulated by DEHP treatment, and MH treatment down-regulated the expression of these two inflammatory markers. Since this down-regulation of TNF-α and NF-κβ coincides with improved liver functions against DEHP-induced toxicity, it can be concluded that MH-mediated liver function involves the singling of TNF-α and NF-κβ.
Collapse
Affiliation(s)
- Vikash Kumar
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar 845401 India
| | - Rahul Kumar
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar 845401 India
| | | | - Saurabh Singh Rathore
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, Bihar 845401 India
| | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram 796 004 India
| |
Collapse
|
19
|
Xu M, Warner C, Duan X, Cheng Z, Jeyarajan AJ, Li W, Wang Y, Shao T, Salloum S, Chen PJ, Yu X, Chung RT, Lin W. HIV coinfection exacerbates HBV-induced liver fibrogenesis through a HIF-1α- and TGF-β1-dependent pathway. J Hepatol 2024; 80:868-881. [PMID: 38311121 PMCID: PMC11102332 DOI: 10.1016/j.jhep.2024.01.026] [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: 06/20/2023] [Revised: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND & AIMS Persons with chronic HBV infection coinfected with HIV experience accelerated progression of liver fibrosis compared to those with HBV monoinfection. We aimed to determine whether HIV and its proteins promote HBV-induced liver fibrosis in HIV/HBV-coinfected cell culture models through HIF-1α and TGF-β1 signaling. METHODS The HBV-positive supernatant, purified HBV viral particles, HIV-positive supernatant, or HIV viral particles were directly incubated with cell lines or primary hepatocytes, hepatic stellate cells, and macrophages in mono or 3D spheroid coculture models. Cells were incubated with recombinant cytokines and HIV proteins including gp120. HBV sub-genomic constructs were transfected into NTCP-HepG2 cells. We also evaluated the effects of inhibitor of HIF-1α and HIV gp120 in a HBV carrier mouse model that was generated via hydrodynamic injection of the pAAV/HBV1.2 plasmid into the tail vein of wild-type C57BL/6 mice. RESULTS We found that HIV and HIV gp120, through engagement with CCR5 and CXCR4 coreceptors, activate AKT and ERK signaling and subsequently upregulate hypoxia-inducible factor-1α (HIF-1α) to increase HBV-induced transforming growth factor-β1 (TGF-β1) and profibrogenic gene expression in hepatocytes and hepatic stellate cells. HIV gp120 exacerbates HBV X protein-mediated HIF-1α expression and liver fibrogenesis, which can be alleviated by inhibiting HIF-1α. Conversely, TGF-β1 upregulates HIF-1α expression and HBV-induced liver fibrogenesis through the SMAD signaling pathway. HIF-1α small-interfering RNA transfection or the HIF-1α inhibitor (acriflavine) blocked HIV-, HBV-, and TGF-β1-induced fibrogenesis. CONCLUSIONS Our findings suggest that HIV coinfection exacerbates HBV-induced liver fibrogenesis through enhancement of the positive feedback between HIF-1α and TGF-β1 via CCR5/CXCR4. HIF-1α represents a novel target for antifibrotic therapeutic development in HBV/HIV coinfection. IMPACT AND IMPLICATIONS HIV coinfection accelerates the progression of liver fibrosis compared to HBV monoinfection, even among patients with successful suppression of viral load, and there is no sufficient treatment for this disease process. In this study, we found that HIV viral particles and specifically HIV gp120 promote HBV-induced hepatic fibrogenesis via enhancement of the positive feedback between HIF-1α and TGF-β1, which can be ameliorated by inhibition of HIF-1α. These findings suggest that targeting the HIF-1α pathway can reduce liver fibrogenesis in patients with HIV and HBV coinfection.
Collapse
Affiliation(s)
- Min Xu
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Charlotte Warner
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaoqiong Duan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan 610052, China; Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Zhimeng Cheng
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andre J Jeyarajan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wenting Li
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, China
| | - Yongtao Wang
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Tuo Shao
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Shadi Salloum
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pei-Jer Chen
- Graduate Institute of Microbiology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Xu Yu
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA 02129, USA
| | - Raymond T Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
20
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Joven Araus A, Wang H, Simon A, Yun MH, Del Rio-Tsonis K. Macrophages modulate fibrosis during newt lens regeneration. Stem Cell Res Ther 2024; 15:141. [PMID: 38745238 PMCID: PMC11094960 DOI: 10.1186/s13287-024-03740-1] [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: 11/15/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Previous studies have suggested that macrophages are present during lens regeneration in newts, but their role in the process is yet to be elucidated. METHODS Here we generated a transgenic reporter line using the newt, Pleurodeles waltl, that traces macrophages during lens regeneration. Furthermore, we assessed early changes in gene expression during lens regeneration using two newt species, Notophthalmus viridescens and Pleurodeles waltl. Finally, we used clodronate liposomes to deplete macrophages during lens regeneration in both species and tested the effect of a subsequent secondary injury after macrophage recovery. RESULTS Macrophage depletion abrogated lens regeneration, induced the formation of scar-like tissue, led to inflammation, decreased iris pigment epithelial cell (iPEC) proliferation, and increased rates of apoptosis in the eye. Some of these phenotypes persisted throughout the last observation period of 100 days and could be attenuated by exogenous FGF2 administration. A distinct transcript profile encoding acute inflammatory effectors was established for the dorsal iris. Reinjury of the newt eye alleviated the effects of macrophage depletion, including the resolution of scar-like tissue, and re-initiated the regeneration process. CONCLUSIONS Together, our findings highlight the importance of macrophages for facilitating a pro-regenerative environment in the newt eye by regulating fibrotic responses, modulating the overall inflammatory landscape, and maintaining the proper balance of early proliferation and late apoptosis of the iPECs.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maximina H Yun
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA.
- Center for Visual Sciences at, Miami University, Oxford, OH, USA.
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
21
|
Lombardo D, Franzè MS, Caminiti G, Pollicino T. Hepatitis Delta Virus and Hepatocellular Carcinoma. Pathogens 2024; 13:362. [PMID: 38787214 PMCID: PMC11124437 DOI: 10.3390/pathogens13050362] [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: 03/22/2024] [Revised: 04/14/2024] [Accepted: 04/20/2024] [Indexed: 05/25/2024] Open
Abstract
The hepatitis D virus (HDV) is a compact, enveloped, circular RNA virus that relies on hepatitis B virus (HBV) envelope proteins to initiate a primary infection in hepatocytes, assemble, and secrete new virions. Globally, HDV infection affects an estimated 12 million to 72 million people, carrying a significantly elevated risk of developing cirrhosis, liver failure, and hepatocellular carcinoma (HCC) compared to an HBV mono-infection. Furthermore, HDV-associated HCC often manifests at a younger age and exhibits more aggressive characteristics. The intricate mechanisms driving the synergistic carcinogenicity of the HDV and HBV are not fully elucidated but are believed to involve chronic inflammation, immune dysregulation, and the direct oncogenic effects of the HDV. Indeed, recent data highlight that the molecular profile of HCC associated with HDV is unique and distinct from that of HBV-induced HCC. However, the question of whether the HDV is an oncogenic virus remains unanswered. In this review, we comprehensively examined several crucial aspects of the HDV, encompassing its epidemiology, molecular biology, immunology, and the associated risks of liver disease progression and HCC development.
Collapse
Affiliation(s)
| | | | | | - Teresa Pollicino
- Department of Clinical and Experimental Medicine, University Hospital of Messina, 98124 Messina, Italy; (D.L.); (M.S.F.); (G.C.)
| |
Collapse
|
22
|
Sayaf K, Battistella S, Russo FP. NLRP3 Inflammasome in Acute and Chronic Liver Diseases. Int J Mol Sci 2024; 25:4537. [PMID: 38674122 PMCID: PMC11049922 DOI: 10.3390/ijms25084537] [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: 01/29/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) is an intracellular complex that upon external stimuli or contact with specific ligands, recruits other components, forming the NLRP3 inflammasome. The NLRP3 inflammasome mainly mediates pyroptosis, a highly inflammatory mode of regulated cell death, as well as IL-18 and IL-1β production. Acute and chronic liver diseases are characterized by a massive influx of pro-inflammatory stimuli enriched in reactive oxygen species (ROS) and damage-associated molecular patterns (DAMPs) that promote the assemblage and activation of the NLRP3 inflammasome. As the major cause of inflammatory cytokine storm, the NLRP3 inflammasome exacerbates liver diseases, even though it might exert protective effects in regards to hepatitis C and B virus infection (HCV and HBV). Here, we summarize the current knowledge concerning NLRP3 inflammasome function in both acute and chronic liver disease and in the post liver transplant setting, focusing on the molecular mechanisms involved in NLRP3 activity.
Collapse
Affiliation(s)
- Katia Sayaf
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
| | - Sara Battistella
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, 35128 Padua, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padua, Italy; (K.S.); (S.B.)
- Gastroenterology and Multivisceral Transplant Unit, Padua University Hospital, 35128 Padua, Italy
| |
Collapse
|
23
|
Jafari Z, Sadeghi S, Dehaghi MM, Bigham A, Honarmand S, Tavasoli A, Hoseini MHM, Varma RS. Immunomodulatory activities and biomedical applications of melittin and its recent advances. Arch Pharm (Weinheim) 2024; 357:e2300569. [PMID: 38251938 DOI: 10.1002/ardp.202300569] [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: 10/06/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Melittin (MLT), a peptide containing 26 amino acids, is a key constituent of bee venom. It comprises ∼40%-60% of the venom's dry weight and is the main pricing index for bee venom, being the causative factor of pain. The unique properties of MLT extracted from bee venom have made it a very valuable active ingredient in the pharmaceutical industry as this cationic and amphipathic peptide has propitious effects on human health in diverse biological processes. It has the ability to strongly impact the membranes of cells and display hemolytic activity with anticancer characteristics. However, the clinical application of MLT has been limited by its severe hemolytic activity, which poses a challenge for therapeutic use. By employing more efficient mechanisms, such as modifying the MLT sequence, genetic engineering, and nano-delivery systems, it is anticipated that the limitations posed by MLT can be overcome, thereby enabling its wider application in therapeutic contexts. This review has outlined recent advancements in MLT's nano-delivery systems and genetically engineered cells expressing MLT and provided an overview of where the MLTMLT's platforms are and where they will go in the future with the challenges ahead. The focus is on exploring how these approaches can overcome the limitations associated with MLT's hemolytic activity and improve its selectivity and efficacy in targeting cancer cells. These advancements hold promise for the creation of innovative and enhanced therapeutic approaches based on MLT for the treatment of cancer.
Collapse
Affiliation(s)
- Zohreh Jafari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Mirzarazi Dehaghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy (IPCB-CNR), Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
| | - Shokouh Honarmand
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Afsaneh Tavasoli
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Mostafa Haji Molla Hoseini
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rajender S Varma
- Department of Chemistry, Centre of Excellence for Research in Sustainable Chemistry, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
24
|
Stauffer WT, Goodman AZ, Bobardt M, Ure DR, Foster RT, Gallay P. Mice lacking cyclophilin B, but not cyclophilin A, are protected from the development of NASH in a diet and chemical-induced model. PLoS One 2024; 19:e0298211. [PMID: 38427624 PMCID: PMC10906846 DOI: 10.1371/journal.pone.0298211] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/20/2024] [Indexed: 03/03/2024] Open
Abstract
Cyclophilins are a diverse family of peptidyl-prolyl isomerases (PPIases) of importance in a variety of essential cellular functions. We previously reported that the pan-cyclophilin inhibitor drug reconfilstat (CRV431) decreased disease in mice under the western-diet and carbon tetrachloride (CCl4) non-alcoholic steatohepatitis (NASH) model. CRV431 inhibits several cyclophilin isoforms, among which cyclophilin A (CypA) and B (CypB) are the most abundant. It is not known whether simultaneous inhibition of multiple cyclophilin family members is necessary for the observed therapeutic effects or if loss-of-function of one is sufficient. Identifying the responsible isoform(s) would enable future fine-tuning of drug treatments. Features of human liver fibrosis and complete NASH can be reliably replicated in mice by administration of intraperitoneal CCl4 alone or CCl4 in conjunction with high sugar, high cholesterol western diet, respectively. Here we show that while wild-type (WT) and Ppia-/- CypA KO mice develop severe NASH disease features under these models, Ppib-/- CypB KO mice do not, as measured by analysis of picrosirius red and hematoxylin & eosin-stained liver sections and TNFα immuno-stained liver sections. Cyclophilin inhibition is a promising and novel avenue of treatment for diet-induced NASH. In this study, mice without CypB, but not mice without CypA, were significantly protected from the development of the characteristic features of NASH. These data suggest that CypB is necessary for NASH disease progression. Further investigation is necessary to determine whether the specific role of CypB in the endoplasmic reticulum secretory pathway is of significance to its effect on NASH development.
Collapse
Affiliation(s)
- Winston T. Stauffer
- Department of Immunology & Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Asha Z. Goodman
- Department of Immunology & Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Michael Bobardt
- Department of Immunology & Microbiology, Scripps Research, La Jolla, California, United States of America
| | - Daren R. Ure
- Hepion Pharmaceuticals, Edison, New Jersey, United States of America
| | - Robert T. Foster
- Hepion Pharmaceuticals, Edison, New Jersey, United States of America
| | - Philippe Gallay
- Department of Immunology & Microbiology, Scripps Research, La Jolla, California, United States of America
| |
Collapse
|
25
|
Elzainy A, El Sadik A, Altowayan WM. Comparison between the Regenerative and Therapeutic Impacts of Bone Marrow Mesenchymal Stem Cells and Adipose Mesenchymal Stem Cells Pre-Treated with Melatonin on Liver Fibrosis. Biomolecules 2024; 14:297. [PMID: 38540717 PMCID: PMC10968153 DOI: 10.3390/biom14030297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The distinctive feature of liver fibrosis is the progressive replacement of healthy hepatic cells by the extracellular matrix protein, which is abundant in collagen I and III, with impaired matrix remodeling. The activation of myofibroblastic cells enhances the fibrogenic response of complex interactions of hepatic stellate cells, fibroblasts, and inflammatory cells to produce the excessive deposition of the extracellular protein matrix. This process is activated by multiple fibrogenic mediators and cytokines, such as TNF-α and IL-1β, accompanied with a decrease in the anti-fibrogenic factor NF-κβ. Mesenchymal stem cells (MSCs) represent a promising therapy for liver fibrosis, allowing for a more advanced regenerative influence when cultured with extrinsic or intrinsic proliferative factors, cytokines, antioxidants, growth factors, and hormones such as melatonin (MT). However, previous studies showed conflicting findings concerning the therapeutic effects of adipose (AD) and bone marrow (BM) MSCs; therefore, the present work aimed to conduct a comparative and comprehensive study investigating the impact of MT pre-treatment on the immunomodulatory, anti-inflammatory, and anti-apoptotic effects of AD- and BM-MSCs and to critically analyze whether MT-pre-treated AD-MSCs and BM-MSCs reveal equal or different therapeutic and regenerative potentials in a CCl4-injured liver experimental rat model. MATERIALS AND METHODS Six groups of experimental rats were used, with ten rats in each group: group I (control group), group II (CCl4-treated group), group III (CCl4- and BM-MSC-treated group), group IV (CCl4 and MT-pre-treated BM-MSC group), group V (CCl4- and AD-MSC-treated group), and group VI (CCl4 and MT-pre-treated AD-MSC group). Liver function tests and the gene expression of inflammatory, fibrogenic, apoptotic, and proliferative factors were analyzed. Histological and immunohistochemical changes were assessed. RESULTS The present study compared the ability of AD- and BM-MSCs, with and without MT pre-treatment, to reduce hepatic fibrosis. Both types of MSCs improved hepatocyte function by reducing the serum levels of ALT, aspartate aminotransferase (AST), alkaline phosphatase (AKP), and total bilirubin (TBIL). In addition, the changes in the hepatocellular architecture, including the hepatocytes, liver sinusoids, central veins, portal veins, biliary ducts, and hepatic arteries, showed a decrease in hepatocyte injury and cholestasis with a reduction in inflammation, apoptosis, and necrosis of the hepatic cells, together with an inhibition of liver tissue fibrosis. These results were augmented by an analysis of the expression of the pro-inflammatory cytokines TNFα and IL-1β, the anti-fibrogenic factor NF-κβ, the apoptotic factor caspase-3, and the proliferative indicators antigen Ki-67 and proliferating cell nuclear antigen (PCNA). These findings were found to be statistically significant, with the restoration of normal parameters in the rats that received AD-MSCs pre-treated with MT, denoting optimal regenerative and therapeutic effects. CONCLUSIONS AD-MSCs pre-treated with MT are the preferred choice in improving hepatic fibrosis and promoting the therapeutic and regenerative ability of liver tissue. They represent a very significant tool for future stem cell use in the tissue regeneration strategy for the treatment of liver diseases.
Collapse
Affiliation(s)
- Ahmed Elzainy
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (A.E.); (A.E.S.)
- Department of Anatomy and Embryology, College of Medicine, Cairo University, Cairo 11956, Egypt
| | - Abir El Sadik
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (A.E.); (A.E.S.)
- Department of Anatomy and Embryology, College of Medicine, Cairo University, Cairo 11956, Egypt
| | - Waleed Mohammad Altowayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
26
|
Zhu B, Wu H, Li KS, Eisa-Beygi S, Singh B, Bielenberg DR, Huang W, Chen H. Two sides of the same coin: Non-alcoholic fatty liver disease and atherosclerosis. Vascul Pharmacol 2024; 154:107249. [PMID: 38070759 DOI: 10.1016/j.vph.2023.107249] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 02/03/2024]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) and atherosclerosis remain high, which is primarily due to widespread adoption of a western diet and sedentary lifestyle. NAFLD, together with advanced forms of this disease such as non-alcoholic steatohepatitis (NASH) and cirrhosis, are closely associated with atherosclerotic-cardiovascular disease (ASCVD). In this review, we discussed the association between NAFLD and atherosclerosis and expounded on the common molecular biomarkers underpinning the pathogenesis of both NAFLD and atherosclerosis. Furthermore, we have summarized the mode of function and potential clinical utility of existing drugs in the context of these diseases.
Collapse
Affiliation(s)
- Bo Zhu
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Hao Wu
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Kathryn S Li
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Bandana Singh
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Diane R Bielenberg
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolic Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, United States of America
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
27
|
Cates WT, Denbeigh JM, Salvagno RT, Kakar S, van Wijnen AJ, Eaton C. Inflammatory Markers Involved in the Pathogenesis of Dupuytren's Contracture. Crit Rev Eukaryot Gene Expr 2024; 34:1-35. [PMID: 38912961 DOI: 10.1615/critreveukaryotgeneexpr.2024052889] [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: 06/25/2024]
Abstract
Dupuytren's disease is a common fibroproliferative disease that can result in debilitating hand deformities. Partial correction and return of deformity are common with surgical or clinical treatments at present. While current treatments are limited to local procedures for relatively late effects of the disease, the pathophysiology of this connective tissue disorder is associated with both local and systemic processes (e.g., fibrosis, inflammation). Hence, a better understanding of the systemic circulation of Dupuytren related cytokines and growth factors may provide important insights into disease progression. In addition, systemic biomarker analysis could yield new concepts for treatments of Dupuytren that attenuate circulatory factors (e.g., anti-inflammatory agents, neutralizing antibodies). Progress in the development of any disease modifying biologic treatment for Dupuytren has been hampered by the lack of clinically useful biomarkers. The characterization of nonsurgical Dupuytren biomarkers will permit disease staging from diagnostic and prognostic perspectives, as well as allows evaluation of biologic responses to treatment. Identification of such markers may transcend their use in Dupuytren treatment, because fibrotic biological processes fundamental to Dupuytren are relevant to fibrosis in many other connective tissues and organs with collagen-based tissue compartments. There is a wide range of potential Dupuytren biomarker categories that could be informative, including disease determinants linked to genetics, collagen metabolism, as well as immunity and inflammation (e.g., cytokines, chemokines). This narrative review provides a broad overview of previous studies and emphasizes the importance of inflammatory mediators as candidate circulating biomarkers for monitoring Dupuytren's disease.
Collapse
Affiliation(s)
- William T Cates
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet M Denbeigh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | | |
Collapse
|
28
|
Baig TA, Haniffa HM, Siddiqui H, Shah SF, Jabeen A. A new acyl derivative of sulfadimethoxine inhibits phagocyte oxidative burst and ameliorates inflammation in a mice model of zymosan-induced generalised inflammation. Inflammopharmacology 2023; 31:3303-3316. [PMID: 37971604 DOI: 10.1007/s10787-023-01372-0] [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: 06/07/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023]
Abstract
Chronic inflammation and oxidative stress play a pivotal role in the pathophysiology of most challenging illnesses, including cancer, Alzheimer's, cardiovascular and autoimmune diseases. The present study aimed to investigate the anti-inflammatory potential of a new sulfadimethoxine derivative N-(4-(N-(2,6-dimethoxypyrimidin-4-yl) sulfamoyl) phenyl) dodecanamide (MHH-II-32). The compound was characterised by applying 1H-, 13C-NMR, EI-MS and HRFAB-MS spectroscopic techniques. The compound inhibited zymosan-induced oxidative bursts from whole blood phagocytes and isolated polymorphonuclear cells with an IC50 value of (2.5 ± 0.4 and 3.4 ± 0.3 µg/mL), respectively. Furthermore, the inhibition of nitric oxide with an IC50 (3.6 ± 2.2 µg/mL) from lipopolysaccharide-induced J774.2 macrophages indicates its in vitro anti-inflammatory efficacy. The compound did not show toxicity towards normal fibroblast cells. The observational findings, gross anatomical analysis of visceral organs and serological tests revealed the non-toxicity of the compound at the highest tested intraperitoneal (IP) dose of 100 mg/kg in acute toxicological studies in Balb/c mice. The compound treatment (100 mg/kg) (SC) significantly (P < 0.001) downregulated the mRNA expression of inflammatory markers TNF-α, IL-1β, IL-2, IL-13, and NF-κB, which were elevated in zymosan-induced generalised inflammation (IP) in Balb/c mice while upregulated the expression of anti-inflammatory cytokine IL-10, which was reduced in zymosan-treated mice. No suppressive effect was observed at the dose of 25 mg/kg. Ibuprofen was taken as a standard drug. The results revealed that the new acyl derivative of sulfadimethoxine has an immunomodulatory effect against generalised inflammatory response with non-toxicity both in vitro and in vivo, and has therapeutic potential for various chronic inflammatory illnesses.
Collapse
Affiliation(s)
- Tariq Ahmad Baig
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Haroon M Haniffa
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Department of Chemical Sciences, Faculty of Applied Sciences, South Eastern University, Sammanthurai, 32200, Sri Lanka
| | - Hina Siddiqui
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Syeda Farah Shah
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Almas Jabeen
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
29
|
Wang B, Kaufmann B, Mogler C, Zhong S, Yin Y, Cheng Z, Schmid RM, Friess H, Hüser N, von Figura G, Hartmann D. Hepatocellular Brg1 promotes CCl4-induced liver inflammation, ECM accumulation and fibrosis in mice. PLoS One 2023; 18:e0294257. [PMID: 38033027 PMCID: PMC10688683 DOI: 10.1371/journal.pone.0294257] [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: 08/14/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Hepatic fibrosis is a progressive pathological process involving the exhaustion of hepatocellular regenerative capacity and ultimately leading to the development of cirrhosis and even hepatocellular carcinoma. Brg1, the core subunit of the SWI/SNF chromatin-remodeling complex, was recently identified as important for liver regeneration. This study investigates the role of Brg1 in hepatic fibrosis development. METHODS Hepatocyte-specific Brg1 knockout mice were generated and injected with carbon tetrachloride (CCl4) for 4, 6, 8, and 12 weeks to induce liver fibrosis. Afterwards, liver fibrosis and liver damage were assessed. RESULTS Brg1 expression was significantly increased in the fibrotic liver tissue of wild-type mice, as compared to that of untreated wild-type mice. The livers of the Brg1 knockout animals showed reduced liver inflammation, extracellular matrix accumulation, and liver fibrosis. TNF-α and NF-κB-mediated inflammatory response was reduced in Brg1 knockout animals. CONCLUSION Brg1 promotes the progression of liver fibrosis in mice and may therefore be used as a potential therapeutic target for treating patients with liver fibrosis due to chronic injury.
Collapse
Affiliation(s)
- Baocai Wang
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Department of General Surgery, The Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Benedikt Kaufmann
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Suyang Zhong
- Department of Medicine II, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Yuhan Yin
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Zhangjun Cheng
- Department of General Surgery, The Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Roland M. Schmid
- Department of Medicine II, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Norbert Hüser
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Guido von Figura
- Department of Medicine II, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel Hartmann
- Department of Surgery, TUM School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
30
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquive EL, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. RESEARCH SQUARE 2023:rs.3.rs-3603645. [PMID: 38045376 PMCID: PMC10690311 DOI: 10.21203/rs.3.rs-3603645/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Methods Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Results Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Conclusions Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Maximina H Yun
- Dresden University of Technology: Technische Universitat Dresden
| | | |
Collapse
|
31
|
Sayaf K, Zanotto I, Gabbia D, Alberti D, Pasqual G, Zaramella A, Fantin A, De Martin S, Russo FP. Sex Drives Functional Changes in the Progression and Regression of Liver Fibrosis. Int J Mol Sci 2023; 24:16452. [PMID: 38003640 PMCID: PMC10671597 DOI: 10.3390/ijms242216452] [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: 09/29/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Liver fibrosis is a common and reversible feature of liver damage associated with many chronic liver diseases, and its onset is influenced by sex. In this study, we investigated the mechanisms of liver fibrosis and regeneration, focusing on understanding the mechanistic gaps between females and males. We injected increasing doses of carbon tetrachloride into female and male mice and maintained them for a washout period of eight weeks to allow for liver regeneration. We found that male mice were more prone to developing severe liver fibrosis as a consequence of early chronic liver damage, supported by the recruitment of a large number of Ly6Chigh MoMφs and neutrophils. Although prolonged liver damage exacerbated the fibrosis in mice of both sexes, activated HSCs and Ly6Chigh MoMφs were more numerous and active in the livers of female mice than those of male mice. After eight weeks of washout, only fibrotic females reported no activated HSCs, and a phenotype switching of Ly6Chigh MoMφs to anti-fibrogenic Ly6Clow MoMφs. The early stages of liver fibrosis mostly affected males rather than females, while long-term chronic liver damage was not influenced by sex, at least for liver fibrosis. Liver repair and regeneration were more efficient in females than in males.
Collapse
Affiliation(s)
- Katia Sayaf
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
| | - Ilaria Zanotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35128 Padova, Italy (D.G.); (S.D.M.)
| | - Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35128 Padova, Italy (D.G.); (S.D.M.)
| | - Dafne Alberti
- Laboratory of Synthetic Immunology, Department of Surgery Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy (G.P.)
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy (G.P.)
- Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Alice Zaramella
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, University of Padova, 35128 Padova, Italy;
| | - Alberto Fantin
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, University of Padova, 35128 Padova, Italy;
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35128 Padova, Italy (D.G.); (S.D.M.)
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
| |
Collapse
|
32
|
Mohammed OS, Attia HG, Mohamed BMSA, Elbaset MA, Fayed HM. Current investigations for liver fibrosis treatment: between repurposing the FDA-approved drugs and the other emerging approaches. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2023; 26:11808. [PMID: 38022905 PMCID: PMC10662312 DOI: 10.3389/jpps.2023.11808] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Long-term liver injuries lead to hepatic fibrosis, often progressing into cirrhosis, liver failure, portal hypertension, and hepatocellular carcinoma. There is currently no effective therapy available for liver fibrosis. Thus, continuous investigations for anti-fibrotic therapy are ongoing. The main theme of anti-fibrotic investigation during recent years is the rationale-based selection of treatment molecules according to the current understanding of the pathology of the disease. The research efforts are mainly toward repurposing current FDA-approved drugs targeting etiological molecular factors involved in developing liver fibrosis. In parallel, investigations also focus on experimental small molecules with evidence to hinder or reverse the fibrosis. Natural compounds, immunological, and genetic approaches have shown significant encouraging effects. This review summarizes the efficacy and safety of current under-investigation antifibrosis medications targeting various molecular targets, as well as the properties of antifibrosis medications, mainly in phase II and III clinical trials.
Collapse
Affiliation(s)
- Omima S. Mohammed
- Department of Microbiology, College of Medicine, Najran University, Najran, Saudi Arabia
| | - Hany G. Attia
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Bassim M. S. A. Mohamed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Marawan A. Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| | - Hany M. Fayed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
33
|
Tilg H, Adolph TE, Tacke F. Therapeutic modulation of the liver immune microenvironment. Hepatology 2023; 78:1581-1601. [PMID: 37057876 DOI: 10.1097/hep.0000000000000386] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Inflammation is a hallmark of progressive liver diseases such as chronic viral or immune-mediated hepatitis, alcohol-associated liver disease, and NAFLD. Preclinical and clinical studies have provided robust evidence that cytokines and related cellular stress sensors in innate and adaptive immunity orchestrate hepatic disease processes. Unresolved inflammation and liver injury result in hepatic scarring, fibrosis, and cirrhosis, which may culminate in HCC. Liver diseases are accompanied by gut dysbiosis and a bloom of pathobionts, fueling hepatic inflammation. Anti-inflammatory strategies are extensively used to treat human immune-mediated conditions beyond the liver, while evidence for immunomodulatory therapies and cell therapy-based strategies in liver diseases is only emerging. The development and establishment of novel immunomodulatory therapies for chronic liver diseases has been dampened by several clinical challenges, such as invasive monitoring of therapeutic efficacy with liver biopsy in clinical trials and risk of DILI in several studies. Such aspects prevented advancements of novel medical therapies for chronic inflammatory liver diseases. New concepts modulating the liver immune environment are studied and eagerly awaited to improve the management of chronic liver diseases in the future.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
34
|
Bogahawaththa S, Kawamura T, Bandaranayake U, Hirakawa T, Yamada G, Ishino H, Hirohashi T, Kawaguchi SI, Wijesundera KK, Wijayagunawardane MPB, Ishimaru K, Kodithuwakku SP, Tsujita T. Identification and mechanistic investigation of ellagitannins from Osbeckia octandra that attenuate liver fibrosis via the TGF-β/SMAD signaling pathway. Biosci Biotechnol Biochem 2023; 87:1295-1309. [PMID: 37580142 DOI: 10.1093/bbb/zbad114] [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: 06/20/2023] [Accepted: 08/04/2023] [Indexed: 08/16/2023]
Abstract
Fibrosis is a major problem in chronic liver disease with limited treatment options due to its complex nature. Herbal medicines are often used as an alternative. The aim of this study was to investigate the therapeutic potential of Osbeckia octandra and to identify its active compounds and regulatory pathways. The effects of crude leaf suspension and boiled leaf extract were investigated in an animal model, and the extract was found to be the more effective treatment. Three major bioactive compounds, pedunculagin, casuarinin, and gallic acid, were isolated from the extract using the hepatic stellate cell line, LX-2-based antifibrotic effect evaluation system. The results showed that all these compounds ameliorated LX-2 in fibrotic state. This inhibitory mechanism was confirmed through the TGF-β/SMAD signaling pathway. Collectively, the presence of these compounds in O. octandra suggests its potential as a treatment for liver fibrosis.
Collapse
Affiliation(s)
- Sudarma Bogahawaththa
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Tomoaki Kawamura
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Udari Bandaranayake
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- Department of Functional Material Science, Graduate School of Science and Engineering, Saitama University, Shimo-Okubo 225, Sakura-ku, Saitama, 338-8570, Japan
| | - Tomoaki Hirakawa
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Goki Yamada
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| | - Hana Ishino
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Tsuzumi Hirohashi
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Shin-Ichi Kawaguchi
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Center for Education and Research in Agricultural Innovation, Faculty of Agriculture, Saga University, Karatsu, Saga, 847-0021, Japan
| | - Kavindra K Wijesundera
- Department of Veterinary Pathobiology, Faculty of Veterinary Medicine and Animal Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | | | - Kanji Ishimaru
- Department of Biological Resource Sciences, Faculty of Agriculture, Saga University, 1 Honjo, Saga, 840-8502, Japan
| | - Suranga P Kodithuwakku
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- ERA Chair COMBIVET, Institute of veterinary Medicine and Animal Science, Estonian University of Life Science, Tartu, 51014, Estonia
| | - Tadayuki Tsujita
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, 890-0065, Japan
- Laboratory of Biochemistry, Department of Advanced Lifesciences and Food Chemistry, Faculty of Agriculture, Saga University, Saga, 840-8502, Japan
| |
Collapse
|
35
|
Pratim Das P, Medhi S. Role of inflammasomes and cytokines in immune dysfunction of liver cirrhosis. Cytokine 2023; 170:156347. [PMID: 37639845 DOI: 10.1016/j.cyto.2023.156347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Liver cirrhosis develops as a result of persistent inflammation and liver injury. The prolonged inflammation triggers the buildup of fibrous tissue and regenerative nodules within the liver, leading to the distortion of the hepatic vascular structure and impaired liver function. Cirrhosis disrupts the ability of liver function to maintain homeostasis and hepatic immunosurveillance which causes immunological dysfunction in the body. In pathological conditions, the production of cytokines in the liver is carefully regulated by various cells in response to tissue stimulation. Cytokines and inflammasomes are the key regulators and systematically contribute to the development of cirrhosis which involves an inflammatory response. However, the crosstalk role of different cytokines in the cirrhosis progression is poorly understood. Tumour necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), and interferon-gamma (IFN-γ), among others, are proinflammatory cytokines that contribute to liver cell necrosis, which in turn causes the development of fibrosis. While IL-10 exhibits a potent anti-inflammatory effect on the liver by inhibiting immune cell activation and neutralizing pro-inflammatory cytokine activity. Inflammasomes have also been implicated in the profibrotic processes of liver cirrhosis, as well as the production of chemokines such as CCL2/MCP-1. It is evident that inflammasomes have a role in the proinflammatory response seen in chronic liver illnesses. In conclusion, cirrhosis significantly impacts the immune system, leading to immunological dysfunction and alterations in both innate and acquired immunity. Proinflammatory cytokines like TNF-α, IL-1β, IL-6, and IFNγ are upregulated in cirrhosis, contributing to liver cell necrosis and fibrosis development. Managing cytokine-mediated inflammation and fibrosis is a key therapeutic approach to alleviate portal hypertension and its associated liver complications. This review attempted to focus largely on the role of immune dysfunction mediated by different cytokines and inflammasomes involved in the progression, regulation and development of liver cirrhosis.
Collapse
Affiliation(s)
- Partha Pratim Das
- Dept. of Bioengineering & Technology, Gauhati University, Assam 781014, India
| | - Subhash Medhi
- Dept. of Bioengineering & Technology, Gauhati University, Assam 781014, India.
| |
Collapse
|
36
|
Linillos-Pradillo B, Paredes SD, Ortiz-Cabello M, Schlumpf M, Lichtensteiger W, Vara E, Tresguerres JAF, Rancan L. Activation of NLRP3 Inflammasome in Liver of Long Evans Lactating Rats and Its Perinatal Effects in the Offspring after Bisphenol F Exposure. Int J Mol Sci 2023; 24:14129. [PMID: 37762434 PMCID: PMC10532117 DOI: 10.3390/ijms241814129] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The liver is the organ responsible for the metabolism and detoxification of BPF, the BPA analogue that is replacing it in plastic-based products. It is not known whether BPF can trigger inflammatory responses via the NLRP3 inflammasome, which plays a major role in the development of liver disease. The aim of this study was to evaluate nitrosative stress species (RNS) and NLRP3 inflammasome activation in the liver of lactating dams after BPF exposure. Moreover, it was studied whether this effect could also be observed in the liver of female and male offspring at postnatal day 6 (PND6). 36 Long Evans rats were randomly distributed according to oral treatment into three groups: Control, BPF-low dose (LBPF; 0.0365 mg/kg b.w./day) group and BPF-high dose (HBPF; 3.65 mg/kg b.w./day) group. The levels of nitrosative stress-inducing proteins (eNOS, iNOS, HO-1d), NLRP3 inflammasome components (NLRP3, PyCARD, CASP1) and proinflammatory cytokines (IL-1β, IL-18, IFN-γ and TNF-α) were measured by gene and protein expression in the liver of lactating dams and in female and male PND6 offspring. Lactating dams treated with LBPF showed a significant increase in iNOS and HO-1d, activation of NLRP3 components (NLRP3, PyCARD, CASP1) and promoted the release of proinflammatory cytokines such as IL-1β, IL-18, IFN-γ and TNF-α. Similar effects were found in female and male PND6 offspring after perinatal exposure. LBPF oral administration and perinatal exposure caused an increase of nitrosative stress markers and proinflammatory cytokines. Also, NLRP3 inflammasome activation was significantly increased in in the liver of lactating dams and PND6 offspring.
Collapse
Affiliation(s)
- Beatriz Linillos-Pradillo
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (B.L.-P.); (M.O.-C.); (E.V.)
| | - Sergio D. Paredes
- Department of Physiology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (S.D.P.); (J.A.F.T.)
| | - María Ortiz-Cabello
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (B.L.-P.); (M.O.-C.); (E.V.)
| | - Margret Schlumpf
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland; (M.S.); (W.L.)
| | - Walter Lichtensteiger
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland; (M.S.); (W.L.)
| | - Elena Vara
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (B.L.-P.); (M.O.-C.); (E.V.)
| | - Jesús A. F. Tresguerres
- Department of Physiology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (S.D.P.); (J.A.F.T.)
| | - Lisa Rancan
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (B.L.-P.); (M.O.-C.); (E.V.)
| |
Collapse
|
37
|
Yin X, Li YS, Ye SZ, Zhang T, Zhang YW, Xi Y, Tang HB. Promotion Effect of Coexposure to a High-Fat Diet and Nano-Diethylnitrosamine on the Progression of Fatty Liver Malignant Transformation into Liver Cancer. Int J Mol Sci 2023; 24:14162. [PMID: 37762463 PMCID: PMC10531889 DOI: 10.3390/ijms241814162] [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: 08/25/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Overconsumption of high-fat foods increases the risk of fatty liver disease (FLD) and liver cancer with long pathogenic cycles. It is also known that the intake of the chemical poison nitrosamine and its nanopreparations can promote the development of liver injuries, such as FLD, and hepatic fibrosis, and significantly shorten the formation time of the liver cancer cycle. The present work confirmed that the coexposure of a high-fat diet (HFD) and nano-diethylnitrosamine (nano-DEN) altered the tumor microenvironment and studied the effect of this coexposure on the progression of fatty liver malignant transformation into liver cancer. Gene transcriptomics and immunostaining were used to evaluate the tumor promotion effect of the coexposure in mice. After coexposure treatment, tumor nodules were obviously increased, and inflammation levels were elevated. The liver transcriptomics analysis showed that the expression levels of inflammatory, fatty, and fibrosis-related factors in the coexposed group were increased in comparison with the nano-DEN- and high-fat-alone groups. The Kyoto Encyclopedia of Genes and Genomes (KEGG) results showed that coexposure aggravated the high expression of genes related to the carcinomatous pathway and accelerated the formation of the tumor microenvironment. The immunohistochemical staining results showed that the coexposure significantly increased the abnormal changes in proteins related to inflammation, proliferation, aging, and hypoxia in mouse liver tissues. The coexposure of high fat and nano-DEN aggravated the process of steatosis and carcinogenesis. In conclusion, the habitual consumption of pickled foods containing nitrosamines in a daily HFD significantly increases the risk of liver pathology lesions progressing from FLD to liver cancer.
Collapse
Affiliation(s)
- Xin Yin
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-Central Minzu University, No. 182, Minyuan Road, Wuhan 430074, China; (X.Y.); (Y.-S.L.); (T.Z.); (Y.-W.Z.)
| | - Yu-Sang Li
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-Central Minzu University, No. 182, Minyuan Road, Wuhan 430074, China; (X.Y.); (Y.-S.L.); (T.Z.); (Y.-W.Z.)
| | - Sha-Zhou Ye
- Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 315211, China;
| | - Ting Zhang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-Central Minzu University, No. 182, Minyuan Road, Wuhan 430074, China; (X.Y.); (Y.-S.L.); (T.Z.); (Y.-W.Z.)
| | - Yi-Wen Zhang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-Central Minzu University, No. 182, Minyuan Road, Wuhan 430074, China; (X.Y.); (Y.-S.L.); (T.Z.); (Y.-W.Z.)
| | - Yang Xi
- Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, No. 818 Fenghua Road, Jiangbei District, Ningbo 315211, China;
| | - He-Bin Tang
- Lab of Hepatopharmacology and Ethnopharmacology, School of Pharmaceutical Sciences, South-Central Minzu University, No. 182, Minyuan Road, Wuhan 430074, China; (X.Y.); (Y.-S.L.); (T.Z.); (Y.-W.Z.)
| |
Collapse
|
38
|
Hassan GS, Flores Molina M, Shoukry NH. The multifaceted role of macrophages during acute liver injury. Front Immunol 2023; 14:1237042. [PMID: 37736102 PMCID: PMC10510203 DOI: 10.3389/fimmu.2023.1237042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
The liver is situated at the interface of the gut and circulation where it acts as a filter for blood-borne and gut-derived microbes and biological molecules, promoting tolerance of non-invasive antigens while driving immune responses against pathogenic ones. Liver resident immune cells such as Kupffer cells (KCs), a subset of macrophages, maintain homeostasis under physiological conditions. However, upon liver injury, these cells and others recruited from circulation participate in the response to injury and the repair of tissue damage. Such response is thus spatially and temporally regulated and implicates interconnected cells of immune and non-immune nature. This review will describe the hepatic immune environment during acute liver injury and the subsequent wound healing process. In its early stages, the wound healing immune response involves a necroinflammatory process characterized by partial depletion of resident KCs and lymphocytes and a significant infiltration of myeloid cells including monocyte-derived macrophages (MoMFs) complemented by a wave of pro-inflammatory mediators. The subsequent repair stage includes restoring KCs, initiating angiogenesis, renewing extracellular matrix and enhancing proliferation/activation of resident parenchymal and mesenchymal cells. This review will focus on the multifaceted role of hepatic macrophages, including KCs and MoMFs, and their spatial distribution and roles during acute liver injury.
Collapse
Affiliation(s)
- Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
39
|
Vachliotis ID, Polyzos SA. The Role of Tumor Necrosis Factor-Alpha in the Pathogenesis and Treatment of Nonalcoholic Fatty Liver Disease. Curr Obes Rep 2023; 12:191-206. [PMID: 37407724 PMCID: PMC10482776 DOI: 10.1007/s13679-023-00519-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE OF REVIEW To summarize experimental and clinical evidence on the association between tumor necrosis factor-α (TNF-α) and nonalcoholic fatty liver disease (NAFLD) and discuss potential treatment considerations. RECENT FINDINGS Experimental evidence suggests that TNF-α is a cytokine with a critical role in the pathogenesis of NAFLD. Although, the production of TNF-α may be an early event during the course of nonalcoholic fatty liver (NAFL), TNF-α may play a more substantial role in the pathogenesis of nonalcoholic steatohepatitis (NASH) and NAFLD-associated fibrosis. Moreover, TNF-α may potentiate hepatic insulin resistance, thus interconnecting inflammatory with metabolic signals and possibly contributing to the development of NAFLD-related comorbidities, including cardiovascular disease, hepatocellular carcinoma, and extra-hepatic malignancies. In clinical terms, TNF-α is probably associated with the severity of NAFLD; circulating TNF-α gradually increases from controls to patients with NAFL, and then, to patients with NASH. Given this potential association, various therapeutic interventions (obeticholic acid, peroxisome proliferator-activated receptors, sodium-glucose co-transporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, probiotics, synbiotics, rifaximin, vitamin E, pentoxifylline, ursodeoxycholic acid, fibroblast growth factor-21, n-3 polyunsaturated fatty acids, statins, angiotensin receptor blockers) have been evaluated for their effect on TNF-α and NAFLD. Interestingly, anti-TNF biologics have shown favorable metabolic and hepatic effects, which may open a possible therapeutic window for the management of advanced NAFLD. The potential key pathogenic role of TNF-α in NAFLD warrants further investigation and may have important diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Ilias D. Vachliotis
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Second Department of Internal Medicine, 424 General Military Hospital, Thessaloniki, Greece
| | - Stergios A. Polyzos
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
40
|
Younis MA, Sato Y, Elewa YHA, Harashima H. Reprogramming activated hepatic stellate cells by siRNA-loaded nanocarriers reverses liver fibrosis in mice. J Control Release 2023; 361:592-603. [PMID: 37579975 DOI: 10.1016/j.jconrel.2023.08.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
We report on a novel strategy for treating liver fibrosis through reprogramming activated Hepatic Stellate Cells (aHSCs) into quiescent Hepatic Stellate Cells (qHSCs) using siRNA-loaded lipid nanoparticles (LNPs). The in vivo screening of an array of molecularly-diverse ionizable lipids identified two candidates, CL15A6 and CL15H6, with a high siRNA delivery efficiency to aHSCs. Optimization of the composition and physico-chemical properties of the LNPs enabled the ligand-free, selective, and potent siRNA delivery to aHSCs post intravenous administration, with a median effective siRNA dose (ED50) as low as 0.08 mg/Kg. The biosafety of the LNPs was confirmed by escalating the dose to 50-fold higher than the ED50 or by chronic administration. The recruitment of the novel LNPs for the simultaneous knockdown of Hedgehog (Hh) and Transforming Growth Factor Beta 1 (TGFβ1) signaling pathways using an siRNA cocktail enabled the reversal of liver fibrosis and the restoration of the normal liver function in mice. Analysis of the key transcription factors in aHSCs suggested that the reprogramming of aHSCs into qHSCs mediated the therapeutic outcomes. The scalable ligand-free platform developed in this study as well as the novel therapeutic strategy reported herein are promising for clinical translation.
Collapse
Affiliation(s)
- Mahmoud A Younis
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; Graduate School of Veterinary Medicine, Hokkaido University, Kita-18, Nishi-9, Kita-ku, Sapporo 060-0818, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
41
|
Mondal SA, Mann SN, van der Linden C, Sathiaseelan R, Kamal M, Das S, Bubak MP, Logan S, Miller BF, Stout MB. Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice. Sci Rep 2023; 13:9841. [PMID: 37330610 PMCID: PMC10276872 DOI: 10.1038/s41598-023-37007-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases. Dietary interventions can reverse metabolic declines and slow aging but remaining compliant is difficult. 17α-estradiol (17α-E2) treatment improves metabolic parameters and slows aging in male mice without inducing significant feminization. We recently reported that estrogen receptor α is required for the majority of 17α-E2-mediated benefits in male mice, but that 17α-E2 also attenuates fibrogenesis in liver, which is regulated by estrogen receptor β (ERβ)-expressing hepatic stellate cells (HSC). The current studies sought to determine if 17α-E2-mediated benefits on systemic and hepatic metabolism are ERβ-dependent. We found that 17α-E2 treatment reversed obesity and related systemic metabolic sequela in both male and female mice, but this was partially blocked in female, but not male, ERβKO mice. ERβ ablation in male mice attenuated 17α-E2-mediated benefits on hepatic stearoyl-coenyzme A desaturase 1 (SCD1) and transforming growth factor β1 (TGF-β1) production, which play critical roles in HSC activation and liver fibrosis. We also found that 17α-E2 treatment suppresses SCD1 production in cultured hepatocytes and hepatic stellate cells, indicating that 17α-E2 directly signals in both cell-types to suppress drivers of steatosis and fibrosis. We conclude that ERβ partially controls 17α-E2-mediated benefits on systemic metabolic regulation in female, but not male, mice, and that 17α-E2 likely signals through ERβ in HSCs to attenuate pro-fibrotic mechanisms.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Shivani N Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Carl van der Linden
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Matthew P Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
| | - Sreemathi Logan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Chapman S212, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
42
|
Mansouri RA, Ahmed AM, Alshaibi HF, Al-Bazi MM, Banjabi AA, Alsufiani HM, Aloqbi AA, Aboubakr EM. A new cirrhotic animal protocol combining carbon tetrachloride with methotrexate to address limitations of the currently used chemical-induced models. Front Pharmacol 2023; 14:1201583. [PMID: 37397479 PMCID: PMC10308223 DOI: 10.3389/fphar.2023.1201583] [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: 04/06/2023] [Accepted: 05/31/2023] [Indexed: 07/04/2023] Open
Abstract
Background: Chemically induced cirrhotic animal models are commonly used. However, they have limitations such as high mortalities and low yield of cirrhotic animals that limit their uses. Aims: To overcome limitations of the chemically induced cirrhotic animal model via combined administration of methotrexate (MTX) with CCl4 and decrease their commonly used doses depending on the proposed synergetic cirrhotic effect. Methods: Rats were divided into six groups: normal (4 weeks), normal (8 weeks), MTX, CCl4 (4 weeks), CCl4 (8 weeks), and MTX + CCl4 (4 weeks) groups. Animals' hepatic morphology and histopathological characterization were explored. Hepatic Bcl2 and NF-κB-p65 tissue contents were determined using the immunostaining technique, and hepatic tissue damage, oxidative status, and inflammatory status biochemical parameters were determined. Results: CCl4 + MTX combined administration produced prominent cirrhotic liver changes, further confirmed by a substantial increase in oxidative stress and inflammatory parameters, whereas mortalities were significantly lower than in other treated groups. Conclusion: The present study introduced a new model that can significantly improve the major limitations of chemically induced cirrhotic animal models with new pathological features that mimic human cirrhosis. Compared to other chemically induced methods, the present model can save time, cost, and animal suffering.
Collapse
Affiliation(s)
- Rasha A. Mansouri
- Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M. Ahmed
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, South Valley University, Qena, Egypt
| | - Huda F. Alshaibi
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Embryonic Stem Cell Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maha M. Al-Bazi
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abeer A. Banjabi
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hadeil Muhanna Alsufiani
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Akram Ahmed Aloqbi
- Department of Biology, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Esam M. Aboubakr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Qena, Egypt
| |
Collapse
|
43
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543633. [PMID: 37333184 PMCID: PMC10274724 DOI: 10.1101/2023.06.04.543633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Andras Simon
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Maximina H Yun
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| |
Collapse
|
44
|
Zhang Z, Ji C, Wang D, Wang M, She X, Song D, Xu X, Zhang D. Maresin1: A multifunctional regulator in inflammatory bone diseases. Int Immunopharmacol 2023; 120:110308. [PMID: 37192551 DOI: 10.1016/j.intimp.2023.110308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Inflammation plays a crucial role in the physical response to danger signals, the elimination of toxic stimuli, and the restoration of homeostasis. However, dysregulated inflammatory responses lead to tissue damage, and chronic inflammation can disrupt osteogenic-osteoclastic homeostasis, ultimately leading to bone loss. Maresin1 (MaR1), a member of the specialized pro-resolving mediators (SPMs) family, has been found to possess significant anti-inflammatory, anti-allergic, pro-hemolytic, pro-healing, and pain-relieving properties. MaR1 is synthesized by macrophages (Mφs) and omega-3 fatty acids, and it may have the potential to promote bone homeostasis and treat inflammatory bone diseases. MaR1 has been found to stimulate osteoblast proliferation through leucine-rich repeat G protein-coupled receptor 6 (LGR6). It also activates Mφ phagocytosis and M2-type polarization, which helps to control the immune system. MaR1 can regulate T cells to exert anti-inflammatory effects and inhibit neutrophil infiltration and recruitment. In addition, MaR1 is involved in antioxidant signaling, including nuclear factor erythroid 2-related factor 2 (NRF2). It has also been found to promote the autophagic behavior of periodontal ligament stem cells, stimulate Mφs against pathogenic bacteria, and regulate tissue regeneration and repair. In summary, this review provides new information and a comprehensive overview of the critical roles of MaR1 in inflammatory bone diseases, indicating its potential as a therapeutic approach for managing skeletal metabolism and inflammatory bone diseases.
Collapse
Affiliation(s)
- Zhanwei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | | | - Maoshan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xiao She
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Dawei Song
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
45
|
Moreno-Lanceta A, Medrano-Bosch M, Simón-Codina B, Barber-González M, Jiménez W, Melgar-Lesmes P. PPAR-γ Agonist GW1929 Targeted to Macrophages with Dendrimer-Graphene Nanostars Reduces Liver Fibrosis and Inflammation. Pharmaceutics 2023; 15:pharmaceutics15051452. [PMID: 37242695 DOI: 10.3390/pharmaceutics15051452] [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: 03/29/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Macrophages play essential roles during the progression of chronic liver disease. They actively participate in the response to liver damage and in the balance between fibrogenesis and regression. The activation of the PPARγ nuclear receptor in macrophages has traditionally been associated with an anti-inflammatory phenotype. However, there are no PPARγ agonists with high selectivity for macrophages, and the use of full agonists is generally discouraged due to severe side effects. We designed dendrimer-graphene nanostars linked to a low dose of the GW1929 PPARγ agonist (DGNS-GW) for the selective activation of PPARγ in macrophages in fibrotic livers. DGNS-GW preferentially accumulated in inflammatory macrophages in vitro and attenuated macrophage pro-inflammatory phenotype. The treatment with DGNS-GW in fibrotic mice efficiently activated liver PPARγ signaling and promoted a macrophage switch from pro-inflammatory M1 to anti-inflammatory M2 phenotype. The reduction of hepatic inflammation was associated with a significant reduction in hepatic fibrosis but did not alter liver function or hepatic stellate cell activation. The therapeutic antifibrotic utility of DGNS-GW was attributed to an increased expression of hepatic metalloproteinases that allowed extracellular matrix remodeling. In conclusion, the selective activation of PPARγ in hepatic macrophages with DGNS-GW significantly reduced hepatic inflammation and stimulated extracellular matrix remodeling in experimental liver fibrosis.
Collapse
Affiliation(s)
- Alazne Moreno-Lanceta
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | - Blanca Simón-Codina
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
| | | | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
46
|
Gole L, Liu F, Ong KH, Li L, Han H, Young D, Marini GPL, Wee A, Zhao J, Rao H, Yu W, Wei L. Quantitative image-based collagen structural features predict the reversibility of hepatitis C virus-induced liver fibrosis post antiviral therapies. Sci Rep 2023; 13:6384. [PMID: 37076590 PMCID: PMC10115775 DOI: 10.1038/s41598-023-33567-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/14/2023] [Indexed: 04/21/2023] Open
Abstract
The novel targeted therapeutics for hepatitis C virus (HCV) in last decade solved most of the clinical needs for this disease. However, despite antiviral therapies resulting in sustained virologic response (SVR), a challenge remains where the stage of liver fibrosis in some patients remains unchanged or even worsens, with a higher risk of cirrhosis, known as the irreversible group. In this study, we provided novel tissue level collagen structural insight into early prediction of irreversible cases via image based computational analysis with a paired data cohort (of pre- and post-SVR) following direct-acting-antiviral (DAA)-based treatment. Two Photon Excitation and Second Harmonic Generation microscopy was used to image paired biopsies from 57 HCV patients and a fully automated digital collagen profiling platform was developed. In total, 41 digital image-based features were profiled where four key features were discovered to be strongly associated with fibrosis reversibility. The data was validated for prognostic value by prototyping predictive models based on two selected features: Collagen Area Ratio and Collagen Fiber Straightness. We concluded that collagen aggregation pattern and collagen thickness are strong indicators of liver fibrosis reversibility. These findings provide the potential implications of collagen structural features from DAA-based treatment and paves the way for a more comprehensive early prediction of reversibility using pre-SVR biopsy samples to enhance timely medical interventions and therapeutic strategies. Our findings on DAA-based treatment further contribute to the understanding of underline governing mechanism and knowledge base of structural morphology in which the future non-invasive prediction solution can be built upon.
Collapse
Affiliation(s)
- Laurent Gole
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
| | - Feng Liu
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, No. 11, Xi Zhimen South Street, Beijing, 100044, People's Republic of China
| | - Kok Haur Ong
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| | - Longjie Li
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| | - Hao Han
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
| | - David Young
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
| | - Gabriel Pik Liang Marini
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore
- Bioinformatics Institute, A*STAR, Singapore, Singapore
| | - Aileen Wee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, National University Hospital, Singapore, Singapore
| | - Jingmin Zhao
- Department of Pathology, The Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Huiying Rao
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, No. 11, Xi Zhimen South Street, Beijing, 100044, People's Republic of China.
| | - Weimiao Yu
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos Building, Singapore, 138673, Singapore.
- Bioinformatics Institute, A*STAR, Singapore, Singapore.
| | - Lai Wei
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, No. 11, Xi Zhimen South Street, Beijing, 100044, People's Republic of China.
- Department of Hepatobiliary and Pancreatic Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| |
Collapse
|
47
|
Abstract
During the past 30 years, several advances have been made allowing for safer and more effective treatment of patients with liver cancer. This report reviews recent advances in radiation therapy for primary liver cancers including hepatocellular carcinoma and intrahepatic cholangiocarcinoma. First, studies focusing on liver stereotactic body radiation therapy (SBRT) are reviewed focusing on lessons learned and knowledge gained from early pioneering trials. Then, new technologies to enhance SBRT treatments are explored including adaptive therapy and MRI-guided and biology-guided radiation therapy. Finally, treatment with Y-90 transarterial radioembolization is reviewed with a focus on novel approaches focused on personalized therapy.
Collapse
|
48
|
Mondal SA, Mann SN, van der Linden C, Sathiaseelan R, Kamal M, Das S, Bubak MP, Logan S, Miller BF, Stout MB. Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.25.534216. [PMID: 36993459 PMCID: PMC10055366 DOI: 10.1101/2023.03.25.534216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Metabolic dysfunction underlies several chronic diseases. Dietary interventions can reverse metabolic declines and slow aging but remaining compliant is difficult. 17α-estradiol (17α-E2) treatment improves metabolic parameters and slows aging in male mice without inducing significant feminization. We recently reported that estrogen receptor α is required for the majority of 17α-E2-mediated benefits in male mice, but that 17α-E2 also attenuates fibrogenesis in liver, which is regulated by estrogen receptor β (ERβ)-expressing hepatic stellate cells (HSC). The current studies sought to determine if 17α-E2-mediated benefits on systemic and hepatic metabolism are ERβ-dependent. We found that 17α-E2 treatment reversed obesity and related systemic metabolic sequela in both male and female mice, but this was partially blocked in female, but not male, ERβKO mice. ERβ ablation in male mice attenuated 17α-E2-mediated benefits on hepatic stearoyl-coenyzme A desaturase 1 (SCD1) and transforming growth factor β1 (TGF-β1) production, which play critical roles in HSC activation and liver fibrosis. We also found that 17α-E2 treatment suppresses SCD1 production in cultured hepatocytes and hepatic stellate cells, indicating that 17α-E2 directly signals in both cell-types to suppress drivers of steatosis and fibrosis. We conclude that ERβ partially controls 17α-E2-mediated benefits on systemic metabolic regulation in female, but not male, mice, and that 17α-E2 likely signals through ERβ in HSCs to attenuate pro-fibrotic mechanisms.
Collapse
Affiliation(s)
- Samim Ali Mondal
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shivani N. Mann
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Carl van der Linden
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Roshini Sathiaseelan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Snehasis Das
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Matthew P. Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sreemathi Logan
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
49
|
Baldini F, Diab F, Serale N, Zeaiter L, Portincasa P, Diaspro A, Vergani L. Adipocyte-hepatocyte crosstalk in cellular models of obesity: Role of soluble factors. Life Sci 2023; 317:121464. [PMID: 36731646 DOI: 10.1016/j.lfs.2023.121464] [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: 09/07/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023]
Abstract
Hepatic steatosis is often a consequence of obesity. Adipose tissue is an important endocrine regulator of metabolic homeostasis in the body. In obesity, adipocytes become hypertrophic and develop an inflammatory phenotype, altering the panel of secreted adipokines. Moreover, excess fatty acids are, in part, released by adipocytes and delivered to the liver. These multiple pathways of adipose-liver crosstalk contribute to the development and progression of liver disease: TNFα induces hepatocyte dysfunction, excess of circulating fatty acids promotes hepatic steatosis and inflammation, whilst adipokines mediate and exacerbate liver injury. In this study, we investigated in vitro the effects and mechanisms of the crosstalk between adipocytes and hepatocytes, as a function of the different adipocyte status (mature vs hypertrophic) being mediated by soluble factors. We employed the conditioned medium method to test how mature and hypertrophic adipocytes distinctively affect the liver, leading to metabolic dysfunction. The media collected from adipocytes were characterized by high triglyceride content and led to lipid accumulation and fat-dependent dysfunction in hepatocytes. The present findings seem to suggest that, in addition to triglycerides, other soluble mediators, cytokines, are released by mature and hypertrophic adipocytes and influence the metabolic status of liver cells. Understanding the precise factors involved in the pathogenesis and pathophysiology of NAFLD in obesity will provide important insights into the mechanisms responsible for the metabolic complications of obesity, paving the way for new possible approaches.
Collapse
Affiliation(s)
- Francesca Baldini
- Nanoscopy, Istituto Italiano Tecnologia, Via Enrico Melen 83, 16152 Genova, Italy; Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Genova, Italy.
| | - Farah Diab
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Genova, Italy.
| | - Nadia Serale
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari, Medical School, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Lama Zeaiter
- Nanoscopy, Istituto Italiano Tecnologia, Via Enrico Melen 83, 16152 Genova, Italy; Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Genova, Italy.
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari, Medical School, Piazza Giulio Cesare 11, 70124 Bari, Italy.
| | - Alberto Diaspro
- Nanoscopy, Istituto Italiano Tecnologia, Via Enrico Melen 83, 16152 Genova, Italy; Department of Physics (DIFILAB), University of Genoa, Via Dodecaneso 33, 16146 Genoa, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6 - Torre di Francia, 16149 Genova, Italy.
| | - Laura Vergani
- Department of Earth, Environment and Life Sciences (DISTAV), University of Genova, Corso Europa 26, 16132, Genova, Italy.
| |
Collapse
|
50
|
Yusuf HR, Musa SA, Agbon AN, Eze ED, Okesina AA, Onanuga I, Pius T, Archibong V, Diaz MEF, Ochieng JJ, Kusiima N, Sunday BY, Usman IM. Hepatoprotective potential of Tamarindus indica following prenatal aluminum exposure in Wistar rat pups. Toxicol Rep 2023; 10:376-381. [PMID: 36926661 PMCID: PMC10014219 DOI: 10.1016/j.toxrep.2023.03.002] [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: 01/13/2023] [Revised: 02/12/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023] Open
Abstract
Over time, the use of plant-derived agents in the management of various human health conditions has gained a lot of attention. The study assessed the hepatoprotective potential of ethyl acetate fraction Tamarindus indica leaves (EFTI) during prenatal aluminum chloride exposure. Pregnant rats were divided into 5 groups (n = 4); Group I rats were administered 2 ml kg-1 of distilled water (negative control), Group II rats received only 200 mg kg-1 aluminum chloride (positive control), Group III rats were administered 200 mg kg-1 aluminum chloride and 400 mg kg-1 EFTI, Group IV rats were administered 200 mg kg-1 aluminum chloride and 800 mg kg-1 EFTI, Group V rats were administered 200 mg kg-1 aluminum chloride and 300 mg kg-1 Vit E (comparative control). On postnatal day 1, the pups were euthanized, and liver tissues were harvested for the biochemical study (tissue levels of malondialdehyde, caspase-3, tumor necrosis factor-alpha, aspartate aminotransferase, alkaline phosphatase, and alanine aminotransferases) and the liver histological examination. The administration of EFTI was marked with significant improvement in the tissue levels of malondialdehyde, caspase-3, tumor necrosis factor-alpha, aspartate aminotransferase, alkaline phosphatase, and alanine aminotransferases. There was a marked improvement in histopathological changes associated with prenatal aluminum chloride exposure. In conclusion, the administration of EFTI was protective during prenatal aluminum chloride exposure of the liver in Wistar rats, and is mediated by the anti-lipid peroxidative, antiapoptotic, and anti-inflammatory activity of EFTI.
Collapse
Affiliation(s)
- Helen Ruth Yusuf
- Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | | | | | | | | | - Ismail Onanuga
- Department of Human Anatomy, Kampala International University, Tanzania
| | - Theophilus Pius
- Department of Medical Laboratory, Kampala International University, Uganda
| | | | | | - Juma John Ochieng
- Department of Human Anatomy, Kampala International University, Uganda
| | - Nicholas Kusiima
- Department of Medical Laboratory, Kampala International University, Uganda
| | - Bot Yakubu Sunday
- Department of Medical Laboratory, Kampala International University, Uganda
| | - Ibe Michael Usman
- Department of Human Anatomy, Kampala International University, Uganda
| |
Collapse
|