1
|
Liu L, Ye F, Jiang Y, Liu W, He D, He W, Gao X, Liu H, Liao J, He B, He F. SIRT5 promotes the osteo-inductive potential of BMP9 by stabilizing the HIF-1α protein in mouse embryonic fibroblasts. Genes Dis 2025; 12:101563. [PMID: 40322223 PMCID: PMC12049830 DOI: 10.1016/j.gendis.2025.101563] [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: 07/15/2024] [Revised: 12/27/2024] [Accepted: 01/12/2025] [Indexed: 05/08/2025] Open
Abstract
Bone morphogenetic protein 9 (BMP9) exhibits remarkable osteogenic potential. However, the intricate mechanisms driving this function of BMP9 remain elusive. This study endeavors to investigate the potential role of sirtuin 5 (SIRT5) in enhancing BMP9's osteogenic capacity and decipher the underlying molecular pathways. To achieve this aim, we employed real-time PCR, western blotting, histochemical staining, and a cranial defect repair model to assess the impact of SIRT5 on BMP9-mediated osteogenesis. We utilized real-time PCR, western blotting, immunofluorescent staining, and immunoprecipitation assay to explore the associated mechanisms. Our results revealed that SIRT5 significantly up-regulated BMP9-induced osteogenic markers, while SIRT5 knockdown reduced their expression. Concurrently, hypoxia-inducible factor 1 subunit alpha (HIF-1α) level was increased by SIRT5, but reduced by SIRT5 knockdown. Notably, HIF-1α potentiated the SIRT5's ability to strengthen BMP9's osteogenic potential, whereas HIF-1α silencing reduced this effect, which was confirmed by bone defect repair assay. The acetylation and malonylation levels of HIF-1α were reduced by SIRT5, which may enhance its stability to promote BMP9's osteogenic effect. Conversely, SIRT5 knockdown reversed these effects and promoted the degradation of HIF-1α. Collectively, our results demonstrated that the BMP9's osteogenic potential could be promoted by SIRT5, potentially through stabilizing HIF-1α by reducing its acetylation and malonylation modification. This discovery may offer a novel strategy to accelerate bone tissue engineering by enhancing osteogenic differentiation, and it also sheds light on the possible mechanisms underlying BMP9-mediated osteogenic differentiation.
Collapse
Affiliation(s)
- Lu Liu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Fanglin Ye
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Wenting Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Dongmei He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Wenge He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Bone and Soft Tissue Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
- Department of Orthropetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiang Gao
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Orthropetics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hang Liu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
- Department of Orthropetics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junyi Liao
- Department of Orthropetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Baicheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Fang He
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
2
|
Zheng K, Dai L, Zhang S, Zhao Y, Li W, Gao Y, Mang Y, Jiao L, Tang Y, Ran J. Unraveling the Heterogeneity of CD8+ T-Cell Subsets in Liver Cirrhosis: Implications for Disease Progression. Gut Liver 2025; 19:410-426. [PMID: 38623058 PMCID: PMC12070210 DOI: 10.5009/gnl230345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 04/17/2024] Open
Abstract
Background/Aims : Liver cirrhosis involves chronic inflammation and progressive fibrosis. Among various immune cells, CD8+ T cells are considered a major contributor to hepatic inflammation and fibrosis. However, the exact molecular pathways governing CD8+ T-cell-mediated effects in cirrhosis remain unclear. Methods : This study analyzed transcriptomic and single-cell sequencing data to elucidate CD8+ T-cell heterogeneity and implications in cirrhosis. Results : Weighted gene co-expression analysis of bulk RNA-seq data revealed an association between cirrhosis severity and activated T-cell markers like HLA and chemokine genes. Furthermore, single-cell profiling uncovered eight CD8+ T-cell subtypes, notably, effector memory (Tem) and exhausted (Tex) T cells. Tex cells, defined by PDCD1, LAG3, and CXCL13 expression, were increased in cirrhosis, while Tem cells were decreased. Lineage tracing and differential analysis highlighted CXCL13+ Tex cells as a terminal, exhausted subtype of cells with roles in PD-1 signaling, glycolysis, and T-cell regulation. CXCL13+ Tex cells displayed T-cell exhaustion markers like PDCD1, HAVCR2, TIGIT, and TNFRSF9. Functional analysis implicated potential roles of these cells in immunosuppression. Finally, a CXCL13+ Tex-cell gene signature was found that correlated with cirrhosis severity and poorer prognosis of liver cancer. Conclusions : In summary, this comprehensive study defines specialized CD8+ T-cell subpopulations in cirrhosis, with CXCL13+ Tex cells displaying an exhausted phenotype associated with immune dysregulation and advanced disease. Key genes and pathways regulating these cells present potential therapeutic targets.
Collapse
Affiliation(s)
- Kepu Zheng
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Leiyang Dai
- Inspection Department of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Shengning Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yingpeng Zhao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Wang Li
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yang Gao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yuanyi Mang
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Lingfeng Jiao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yu Tang
- Kunming Medical University, Kunming, China
| | - Jianghua Ran
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
3
|
Owida HA, Saleh RO, Mohammad SI, Vasudevan A, Roopashree R, Kashyap A, Nanda A, Ray S, Hussein A, Yasin HA. Deciphering the role of circular RNAs in cancer progression under hypoxic conditions. Med Oncol 2025; 42:191. [PMID: 40314834 DOI: 10.1007/s12032-025-02727-z] [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: 02/23/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
Hypoxia, characterized by reduced oxygen levels, plays a pivotal role in cancer progression, profoundly influencing tumor behavior and therapeutic responses. A hallmark of solid tumors, hypoxia drives significant metabolic adaptations in cancer cells, primarily mediated by hypoxia-inducible factor-1α (HIF-1α), a key transcription factor activated in low-oxygen conditions. This hypoxic environment promotes epithelial-mesenchymal transition (EMT), enhancing cancer cell migration, metastasis, and the development of cancer stem cell-like properties, which contribute to therapy resistance. Moreover, hypoxia modulates the expression of circular RNAs (circRNAs), leading to their accumulation in the tumor microenvironment. These hypoxia-responsive circRNAs regulate gene expression and cellular processes critical for cancer progression, making them promising candidates for diagnostic and prognostic biomarkers in various cancers. This review delves into the intricate interplay between hypoxic circRNAs, microRNAs, and RNA-binding proteins, emphasizing their role as molecular sponges that modulate gene expression and signaling pathways involved in cell proliferation, apoptosis, and metastasis. It also explores the relationship between circRNAs and the tumor microenvironment, particularly how hypoxia influences their expression and functional dynamics. Additionally, the review highlights the potential of circRNAs as diagnostic and prognostic tools, as well as their therapeutic applications in innovative cancer treatments. By consolidating current knowledge, this review underscores the critical role of circRNAs in cancer biology and paves the way for future research aimed at harnessing their unique properties for clinical advancements. Specifically, this review examines the biogenesis, expression patterns, and mechanistic actions of hypoxic circRNAs, focusing on their ability to act as molecular sponges for microRNAs and their interactions with RNA-binding proteins. These interactions impact key signaling pathways related to tumor growth, metastasis, and drug resistance, offering new insights into the complex regulatory networks governed by circRNAs under hypoxic stress.
Collapse
Affiliation(s)
- Hamza Abu Owida
- Department of Medical Engineering, Faculty of Engineering, Al-Ahliyya Amman University, Amman, Jordan
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Suleiman Ibrahim Mohammad
- Research Follower, INTI International University, 71800, Negeri Sembilan, Malaysia.
- Electronic Marketing and Social Media, Economic and Administrative Sciences, Zarqa University, Zarqa, Jordan.
| | - Asokan Vasudevan
- Faculty of Business and Communications, INTI International University, 71800, Negeri Sembilan, Malaysia
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Ahmed Hussein
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
4
|
Jalali P, Shahmoradi A, Samii A, Mazloomnejad R, Hatamnejad MR, Saeed A, Namdar A, Salehi Z. The role of autophagy in cancer: from molecular mechanism to therapeutic window. Front Immunol 2025; 16:1528230. [PMID: 40248706 PMCID: PMC12003146 DOI: 10.3389/fimmu.2025.1528230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Autophagy is a cellular degradation process that plays a crucial role in maintaining metabolic homeostasis under conditions of stress or nutrient deprivation. This process involves sequestering, breaking down, and recycling intracellular components such as proteins, organelles, and cytoplasmic materials. Autophagy also serves as a mechanism for eliminating pathogens and engulfing apoptotic cells. In the absence of stress, baseline autophagy activity is essential for degrading damaged cellular components and recycling nutrients to maintain cellular vitality. The relationship between autophagy and cancer is well-established; however, the biphasic nature of autophagy, acting as either a tumor growth inhibitor or promoter, has raised concerns regarding the regulation of tumorigenesis without inadvertently activating harmful aspects of autophagy. Consequently, elucidating the mechanisms by which autophagy contributes to cancer pathogenesis and the factors determining its pro- or anti-tumor effects is vital for devising effective therapeutic strategies. Furthermore, precision medicine approaches that tailor interventions to individual patients may enhance the efficacy of autophagy-related cancer treatments. To this end, interventions aimed at modulating the fate of tumor cells by controlling or inducing autophagy substrates necessitate meticulous monitoring of these mediators' functions within the tumor microenvironment to make informed decisions regarding their activation or inactivation. This review provides an updated perspective on the roles of autophagy in cancer, and discusses the potential challenges associated with autophagy-related cancer treatment. The article also highlights currently available strategies and identifies questions that require further investigation in the future.
Collapse
Affiliation(s)
- Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amir Samii
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Radman Mazloomnejad
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Afshin Namdar
- Program in Cell Biology, The Hospital for Sick Children Peter Gilgan Centre for Research and Learning, Toronto, ON, United States
| | - Zahra Salehi
- Department of Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Chaikin CA, Thakkar AV, Steffeck AWT, Pfrender EM, Hung K, Zhu P, Waldeck NJ, Nozawa R, Song W, Futtner CR, Quattrocelli M, Bass J, Ben-Sahra I, Peek CB. Control of circadian muscle glucose metabolism through the BMAL1-HIF axis in obesity. Proc Natl Acad Sci U S A 2025; 122:e2424046122. [PMID: 40127275 PMCID: PMC12002348 DOI: 10.1073/pnas.2424046122] [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: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Disruptions of circadian rhythms are widespread in modern society and lead to accelerated and worsened symptoms of metabolic syndrome. In healthy mice, the circadian clock factor BMAL1 is required for skeletal muscle function and metabolism. However, the importance of muscle BMAL1 in the development of metabolic diseases, such as diet-induced obesity (DIO), remains unclear. Here, we demonstrate that skeletal muscle-specific BMAL1-deficient mice exhibit worsened glucose tolerance upon high-fat diet feeding, despite no evidence of increased weight gain. Metabolite profiling from Bmal1-deficient muscles revealed impaired glucose utilization specifically at early steps in glycolysis that dictate the switch between anabolic and catabolic glucose fate. We provide evidence that this is due to abnormal control of the nutrient stress-responsive hypoxia-inducible factor (HIF) pathway. Genetic HIF1α stabilization in muscle Bmal1-deficient mice restores glucose tolerance and expression of 217/736 dysregulated genes during DIO, including glycolytic enzymes. Together, these data indicate that during DIO, skeletal muscle BMAL1 is an important regulator of HIF-driven glycolysis and metabolic flexibility, which influences the development of high-fat-diet-induced glucose intolerance.
Collapse
Affiliation(s)
- Claire A. Chaikin
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Abhishek V. Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Adam W. T. Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Eric M. Pfrender
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Kaitlyn Hung
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Nathan J. Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Rino Nozawa
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Weimin Song
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Christopher R. Futtner
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Clara B. Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL60611
- Department of Medicine, Division of Endocrinology, Metabolism and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| |
Collapse
|
6
|
Nie T, Nepovimova E, Wu Q. Circadian rhythm, hypoxia, and cellular senescence: From molecular mechanisms to targeted strategies. Eur J Pharmacol 2025; 990:177290. [PMID: 39863143 DOI: 10.1016/j.ejphar.2025.177290] [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: 10/25/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Cellular senescence precipitates a decline in physiological activities and metabolic functions, often accompanied by heightened inflammatory responses, diminished immune function, and impaired tissue and organ performance. Despite extensive research, the mechanisms underpinning cellular senescence remain incompletely elucidated. Emerging evidence implicates circadian rhythm and hypoxia as pivotal factors in cellular senescence. Circadian proteins are central to the molecular mechanism governing circadian rhythm, which regulates homeostasis throughout the body. These proteins mediate responses to hypoxic stress and influence the progression of cellular senescence, with protein Brain and muscle arnt-like 1 (BMAL1 or Arntl) playing a prominent role. Hypoxia-inducible factor-1α (HIF-1α), a key regulator of oxygen homeostasis within the cellular microenvironment, orchestrates the transcription of genes involved in various physiological processes. HIF-1α not only impacts normal circadian rhythm functions but also can induce or inhibit cellular senescence. Notably, HIF-1α may aberrantly interact with BMAL1, forming the HIF-1α-BMAL1 heterodimer, which can instigate multiple physiological dysfunctions. This heterodimer is hypothesized to modulate cellular senescence by affecting the molecular mechanism of circadian rhythm and hypoxia signaling pathways. In this review, we elucidate the intricate relationships among circadian rhythm, hypoxia, and cellular senescence. We synthesize diverse evidence to discuss their underlying mechanisms and identify novel therapeutic targets to address cellular senescence. Additionally, we discuss current challenges and suggest potential directions for future research. This work aims to deepen our understanding of the interplay between circadian rhythm, hypoxia, and cellular senescence, ultimately facilitating the development of therapeutic strategies for aging and related diseases.
Collapse
Affiliation(s)
- Tong Nie
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
7
|
Li L, Zhao Y, Ding Y, Guo L, Dai R, Chen A, Duan G. Forsythia suspensa leaf fermented tea extracts attenuated oxidative stress in mice via the Ref-1/HIF-1α signal pathway and modulation of gut microbiota. Sci Rep 2025; 15:4106. [PMID: 39900709 PMCID: PMC11790883 DOI: 10.1038/s41598-025-87182-6] [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: 09/05/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
Forsythia suspensa leaf fermented tea (FSLFT) is made from tender buds of Forsythia suspensa collected in spring. The main active components of FSLFT include forsythiaside, forsythia ester glycoside, rutin, and forsythia flavonoids, which have antibacterial, antioxidant, liver-protective, and immune-regulatory effects. Oxidative stress can trigger excessive apoptosis in intestinal epithelial cells, leading to dysfunction of the small intestinal mucosa and impaired intestinal absorption. This study focused on Kunming mice as research subjects and used hydrogen peroxide as an inducer to investigate the antioxidant and anti-inflammatory effects of FSLFT in vivo, as well as its regulatory effects on the intestinal microbiota of mice. The aim of this study was to establish a theoretical foundation for the functional study of Forsythia suspensa leaves and provide specific recommendations for their growth and application. The results showed that H2O2 treatment led to an increase in oxidative levels in mice. FSLFT has been shown to have antioxidant effects via the Redox Factor-1(Ref-1)/ hypoxia-inducible factor-1 alpha (HIF-1α) pathway, reduce inflammation caused by hydrogen peroxide through the Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway, and protect mouse colons from oxidative stress by repairing gut microbiota imbalance and increasing microbial diversity and abundance. These findings establish a theoretical basis for studying the functional properties of FSLFT.
Collapse
Affiliation(s)
- Lijuan Li
- Shanxi Provincial Department, Municipal Key Laboratory Cultivation Base for Quality Enhancement and Utilization of Shangdang Chinese Medicinal Materials, Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | | | - Yuxin Ding
- Shanxi Agriculture University, Taigu, China
| | - Lanze Guo
- Shanxi Agriculture University, Taigu, China
| | - Ruiyao Dai
- Shanxi Agriculture University, Taigu, China
| | - Aixiang Chen
- Shanxi Provincial Department, Municipal Key Laboratory Cultivation Base for Quality Enhancement and Utilization of Shangdang Chinese Medicinal Materials, Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Guofeng Duan
- Shanxi Provincial Department, Municipal Key Laboratory Cultivation Base for Quality Enhancement and Utilization of Shangdang Chinese Medicinal Materials, Changzhi Medical College, Changzhi, 046000, Shanxi, China.
| |
Collapse
|
8
|
Sum H, Brewer AC. The Impact of Modifiable Risk Factors on the Endothelial Cell Methylome and Cardiovascular Disease Development. FRONT BIOSCI-LANDMRK 2025; 30:26082. [PMID: 39862076 PMCID: PMC7617538 DOI: 10.31083/fbl26082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 01/27/2025]
Abstract
Cardiovascular disease (CVD) is the most prevalent cause of mortality and morbidity in the Western world. A common underlying hallmark of CVD is the plaque-associated arterial thickening, termed atherosclerosis. Although the molecular mechanisms underlying the aetiology of atherosclerosis remain unknown, it is clear that both its development and progression are associated with significant changes in the pattern of DNA methylation within the vascular cell wall. The endothelium is the major regulator of vascular homeostasis, and endothelial cell dysfunction (ED) is considered an early marker for atherosclerosis. Thus, it is speculated that changes in DNA methylation within endothelial cells may, in part, be causal in ED, leading to atherosclerosis and CVD generally. This review will evaluate the extensive evidence that environmental risk factors, known to be associated with atherosclerosis, such as diabetes, metabolic disorder, smoking, hypertension and hypercholesterolaemia etc. can affect the methylome of the endothelium and consequently act to alter gene transcription and function. Further, the potential mechanisms whereby such risk factors might impact upon the activities and/or specificities of the epigenetic writers and erasers which determine the methylome [the DNA methyl transferases (DNMTs) and Ten Eleven translocases (TETs)] are considered here. Notably, the TET proteins are members of the 2-oxoglutarate-dependent dioxygenase superfamily which require molecular oxygen (O2) and α-ketoglutarate (α-KG) as substrates and iron-2+ (Fe II) as a cofactor. This renders their activities subject to modulation by hypoxia, metabolic flux and cellular redox. The potential significance of this, with respect to the impact of modifiable risk factors upon the activities of the TETs and the methylome of the endothelium is discussed.
Collapse
Affiliation(s)
- Hashum Sum
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, SE5 9NULondon, UK
| | - Alison C. Brewer
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, SE5 9NULondon, UK
| |
Collapse
|
9
|
Lotsios NS, Keskinidou C, Karagiannis SP, Papavassiliou KA, Papavassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE, Vassiliou AG. Expression and Regulation of Hypoxia-Inducible Factor Signalling in Acute Lung Inflammation. Cells 2024; 14:29. [PMID: 39791730 PMCID: PMC11719729 DOI: 10.3390/cells14010029] [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: 12/12/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are central regulators of gene expression in response to oxygen deprivation, a common feature in critical illnesses. The significant burden that critical illnesses place on global healthcare systems highlights the need for a deeper understanding of underlying mechanisms and the development of innovative treatment strategies. Among critical illnesses, impaired lung function is frequently linked to hypoxic conditions. This review focuses on the expression and regulation of HIF signalling in experimental models of acute lung injury (ALI) and clinical studies in critically ill patients with acute respiratory distress syndrome (ARDS). We explore the potential dual role of HIF signalling in acute lung inflammation. Furthermore, its role in key biological processes and its potential prognostic significance in clinical scenarios are discussed. Finally, we explore recent pharmacological advancements targeting HIF signalling, which have emerged as promising alternatives to existing therapeutic approaches, potentially enabling more effective management strategies.
Collapse
Affiliation(s)
- Nikolaos S. Lotsios
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Chrysi Keskinidou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Sotirios P. Karagiannis
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Chest Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Stylianos E. Orfanos
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| | - Alice G. Vassiliou
- First Department of Critical Care Medicine, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (N.S.L.); (C.K.); (S.P.K.); (A.K.); (I.D.); (S.E.O.)
| |
Collapse
|
10
|
Zhang X, Zhang H, Dong Q, Qin Y, Cao Y, Zhu H, Ma Z, Li Z, Rao Z, Ning P, Tian Z, Xia Y, Yang P, Wang Z. Bypassing Ca 2+ Influx for Antimetastasis Photodynamic Therapy via Robust Nucleus-Targeted Near-Infrared Cyanines. NANO LETTERS 2024; 24:15817-15826. [PMID: 39584561 DOI: 10.1021/acs.nanolett.4c04789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Hypoxia-induced tumor metastasis severely hinders the efficacy of photodynamic therapy (PDT) in cancer treatment. Current strategies predominantly offer palliative suppression of the HIF-1α pathway, emphasizing the urgent need for innovative PDT approaches to prevent metastasis from the outset. Our study revealed that typical PDT triggers an increase in cytoplasmic Ca2+ levels, activating HIF-1α, and that reducing Ca2+ levels can, in turn, mitigate metastasis. Considering cytoplasm's role in Ca2+ storage and regulation, we propose that PDT-induced metastasis can be addressed at its source by precise intracellular localization of photosensitizers (PSs). We developed near-infrared (NIR) cyanine PSs with inherent nucleus targeting capabilities. These PSs effectively inhibit cytoplasmic Ca2+ elevation and reduce HIF-1α activity upon irradiation, achieving remarkable antimetastatic effects in 4T1 tumors. Consequently, our findings highlight the pivotal role of Ca2+ in PDT-induced metastasis and provide a robust approach for circumventing metastasis from the outset using new nucleus-targeting organic PSs.
Collapse
Affiliation(s)
- Xianghan Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Huaicong Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Qunyan Dong
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yuan Qin
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yutian Cao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Haixing Zhu
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zimeng Ma
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zehua Li
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhiping Rao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Pengbo Ning
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zuhong Tian
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yuqiong Xia
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Peng Yang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
11
|
Li Y, Wu S, Huang J, Zhao L. Integrated miRNA-seq and functional analyses reveal the regulatory role of sha-miR-92a_L + 2R + 4 via targeting vegfaa in rainbow trout (Oncorhynchus mykiss) responding to acute hypoxia and reoxygenation stress. BMC Genomics 2024; 25:1163. [PMID: 39623322 PMCID: PMC11610304 DOI: 10.1186/s12864-024-11019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 11/08/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Hypoxia negatively affects the behavior, growth, reproduction and survival of fish, causing serious economic losses to aquaculture. Rainbow trout (Oncorhynchus mykiss), an important economic fish worldwide, belongs to a hypoxia-sensitive fish species, however, little is known about the regulatory mechanism of microRNAs (miRNAs) under hypoxia stress. RESULTS Rainbow trout were subjected to hypoxia stress for 3 h (H3h_L), 12 h (H12h_L), 24 h (H24h_L) and 3 h reoxygenation (R3h_L) to systemically evaluate the changes of miRNA expression profiles in liver, and functions of sha-miR-92a_L + 2R + 4 were investigated. We found 17, 144, 57 and 55 differentially expressed (DE) miRNAs in the H3h_L vs. control (N_L), H12h_L vs. N_L, H24h_L vs. N_L and R3h_L vs. N_L comparisons, respectively. Enrichment analysis revealed that the targets of DE miRNAs were significantly enriched in HIF signaling pathway, VEGF signaling pathway, FoxO signaling pathway and glycolysis/gluconeogenesis. Through miRNA-mRNA interaction and weighted gene co-expression network analysis (WGCNA), five key DE miRNAs (sha-miR-92a_L + 2R + 4, ssa-miR-128-3p, ssa-miR-101b-3p_R + 1, ola-miR-199a-5p_R + 2 and tni-miR-199_1ss18CG) were identified, which can target at least two hypoxia-responsive genes, such as vegfaa, ho, glut1a and junb. Functional analysis found that sha-miR-92a_L + 2R + 4 directly regulated vegfaa expression by targeting its 3'-UTR, overexpression of sha-miR-92a_L + 2R + 4 significantly decreased vegfaa expression in rainbow trout liver cells, while opposite results were obtained after transfection of sha-miR-92a_L + 2R + 4 inhibitor. Furthermore, overexpressed sha-miR-92a_L + 2R + 4 promoted rainbow trout liver cell proliferation and inhibited apoptosis. CONCLUSION This study deepens our understanding of the crucial roles of miRNAs under hypoxia stress in rainbow trout. These results can contribute to devise strategies for improving rainbow trout survival rate and aquaculture production during hypoxia stress and help speeding up the selective breeding of hypoxia-tolerant rainbow trout.
Collapse
Affiliation(s)
- Yongjuan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
- College of Science, Gansu Agricultural University, Lanzhou, 730070, China
| | - Shenji Wu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jinqiang Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Lu Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
12
|
Suhail Y, Liu Y, Du W, Afzal J, Qiu X, Atiq A, Vera-Licona P, Agmon E, Kshitiz. Oscillatory hypoxia induced gene expression predicts low survival in human breast cancer patients. Mol Carcinog 2024; 63:2305-2315. [PMID: 39150154 PMCID: PMC11905232 DOI: 10.1002/mc.23810] [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: 04/27/2024] [Revised: 07/08/2024] [Accepted: 08/03/2024] [Indexed: 08/17/2024]
Abstract
Hypoxia is one of the key factors in the tumor microenvironment regulating nearly all steps in the metastatic cascade in many cancers, including in breast cancer. The hypoxic regions can however be dynamic with the availability of oxygen fluctuating or oscillating. The canonical response to hypoxia is relayed by transcription factor Hypoxia-Inducible Factor 1 (HIF-1), which is stabilized in hypoxia and acts as the master regulator of a large number of downstream genes. However, HIF-1 transcriptional activity can also fluctuate either due to unstable hypoxia, or by lactate mediated noncanonical degradation of HIF-1. Our understanding of how oscillatory hypoxia or HIF-1 activity specifically influences cancer malignancy is very limited. Here, using MDA-MB-231 cells as a model of triple negative breast cancer characterized by severe hypoxia, we measured the gene expression changes induced specifically by oscillatory hypoxia. We found that oscillatory hypoxia can specifically regulate gene expression differently, and at times opposite to stable hypoxia. Using the Cancer Genome Atlas RNAseq data of human cancer samples, we show that the oscillatory specific gene expression signature in MDA-MB-231 is enriched in most human cancers, and prognosticates low survival in breast cancer patients. In particular, we found that oscillatory hypoxia, unlike stable hypoxia, induces unfolded protein folding response in cells resulting in gene expression predicting reduced survival.
Collapse
Affiliation(s)
- Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Junaid Afzal
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Xihua Qiu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Amina Atiq
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Paola Vera-Licona
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, Connecticut, USA
| | - Eran Agmon
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, Connecticut, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
13
|
Li Z, Yin B, Xu Y, Wang C, Li X, Lu S, Ke S, Qian B, Yu H, Bai M, Li Z, Zhou Y, Jiang H, Ma Y. Von Hippel-Lindau deficiency protects the liver against ischemia/reperfusion injury through the regulation of hypoxia-inducible factor 1α and 2α. Hepatol Commun 2024; 8:e0567. [PMID: 39585306 PMCID: PMC11596652 DOI: 10.1097/hc9.0000000000000567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/25/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Ischemia and reperfusion (I/R)-induced liver injury contributes to morbidity and mortality during hepatic surgery or liver transplantation. As a pivotal regulator of cancer and inflammation, the role of Von Hippel-Lindau (VHL) in hepatic I/R injury remains undetermined. METHODS We investigated the role of VHL in hepatic I/R injury by generating VHL conditional knockout (VHL-KO) mice. The downstream mechanisms of VHL were confirmed, and the role of HIF-2α in hepatic I/R injury was further investigated. RESULTS In this study, we discovered that VHL upregulation was associated with hepatic I/R injury in a mouse model. VHL gene knockout (VHL-KO) and overexpression (Ad-VHL) mice demonstrated that VHL aggravated liver injury, increased inflammation, and accelerated cell death in hepatic I/R injury. The VHL protein (pVHL) regulates a crucial control mechanism by targeting HIFα subunits for ubiquitin-mediated degradation. In vitro and in vivo studies demonstrated that VHL interacted with and repressed hypoxia-inducible factor 1α (HIF-1α) and hypoxia-inducible factor 2α (HIF-2α) expression during hepatic I/R injury. Notably, the inhibition of HIF-1α or 2α, as well as the concurrent inhibition of HIF-1α and 2α, abrogated the protective effect of VHL-KO. The severe stabilization of HIF-1α or 2α, as well as the simultaneous overexpression of HIF-1α and 2α, compensated for the detrimental effect of VHL. CONCLUSIONS Thus, we identified the VHL-HIF-1α/HIF-2α axis as an indispensable pathway that may be a novel target for mediating hepatic I/R injury.
Collapse
Affiliation(s)
- Zihao Li
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Yin
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanan Xu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Hepatopancreatobiliary Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaoqun Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xinglong Li
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shounan Lu
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanjia Ke
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Hepatobiliary Surgery, Peking University People’s Hospital, Beijing, China
| | - Baolin Qian
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongjun Yu
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miaoyu Bai
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhongyu Li
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongzhi Zhou
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongchi Jiang
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Ma
- Department of Minimally Invasive Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
14
|
Szymanska M, Basavaraja R, Meidan R. A tale of two endothelins: the rise and fall of the corpus luteum. Reprod Fertil Dev 2024; 37:RD24158. [PMID: 39680472 DOI: 10.1071/rd24158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Endothelins are small 21 amino acid peptides that interact with G-protein-coupled receptors. They are highly conserved across species and play important roles in vascular biology as well as in disease development and progression. Endothelins, mainly endothelin-1 and endothelin-2, are intricately involved in ovarian function and metabolism. These two peptides differ only in two amino acids but are encoded by different genes, which suggests an independent regulation and a cell-specific mode of expression. This review aims to comprehensively discuss the distinct regulation and roles of endothelin-1 and endothelin-2 regarding corpus luteum function throughout its life span.
Collapse
Affiliation(s)
- Magdalena Szymanska
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; and Present address: Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Olsztyn, Poland
| | - Raghavendra Basavaraja
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; and Present address: Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
15
|
Alsawaf Y, Maksimovic I, Zheng J, Zhang S, Vuckovic I, Dzeja P, Macura S, Irazabal MV. A brief harvesting-freezing delay significantly alters the kidney metabolome and leads to false positive and negative results. Am J Physiol Renal Physiol 2024; 327:F697-F711. [PMID: 39205659 PMCID: PMC11563588 DOI: 10.1152/ajprenal.00131.2024] [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: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Abnormalities in distinct metabolic pathways have been associated with the pathogenesis and progression of many forms of kidney disease. Metabolomics analyses can be used to determine organ-specific metabolic fingerprints and, ideally, should represent the metabolic state of the organ at the exact moment the sample is harvested. However, conventional harvesting methods depend on posteuthanasia tissue harvest, which results in ischemia conditions and metabolome changes that could potentially introduce artifacts into the final studies. We recently optimized a modified clamp-freezing technique for rodent kidney harvesting and freezing, significantly reducing ischemia and freezing times and granting a closer snapshot of in vivo metabolism. In this study, we characterized and compared the metabolome of kidneys harvested using our modified approach versus traditional techniques to determine which metabolites are preferentially affected by a brief lapse of ischemia and freezing delay and which are more stable. We used Sprague-Dawley rats as a model of wild-type (WT) kidneys and PCK [polycystic kidney disease (PKD)] rats as a model of chronic kidney disease kidneys. Finally, we compared the metabolic profile of clamp-frozen and delayed WT and PKD kidneys to determine which metabolic changes are most likely observed in vivo in PKD and which could be subjected to false positive or negative results. Our data indicate that a short harvesting-freezing delay is sufficient to impart profound metabolic changes in WT and PKD kidneys, leading to false positive and negative differences when comparing these genotypes. In addition, we identified a group of metabolites that were more stable. Interestingly, while the delay had a similar effect between WT and PKD, there were notable differences. The data obtained indicate that the quick clamp-freezing technique for kidney metabolomics provides a more accurate interpretation of the in vivo metabolic changes associated with the disease state. NEW & NOTEWORTHY Our study shows that a brief harvesting-freezing delay associated with organ collection and freezing can significantly alter the kidney metabolic profile of both male and female wild-type and a genetic model of chronic kidney disease. Importantly, given that the effect of this delay differs among genotypes, it is not safe to assume that equally delaying harvesting-freezing in wild-type and polycystic kidney disease kidneys adequately controls this effect, ultimately leading to false positive and negative results among different renal diseases.
Collapse
Affiliation(s)
- Yahya Alsawaf
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Igor Maksimovic
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Jamie Zheng
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| | - Song Zhang
- Metabolomics Core, Mayo Clinic, Rochester, Minnesota, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Ivan Vuckovic
- Metabolomics Core, Mayo Clinic, Rochester, Minnesota, United States
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Slobodan Macura
- Department of Biochemistry, Mayo Clinic, Rochester, Minnesota, United States
| | - Maria V Irazabal
- Mayo Translational PKD Center, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
16
|
Anvar MT, Rashidan K, Arsam N, Rasouli-Saravani A, Yadegari H, Ahmadi A, Asgari Z, Vanan AG, Ghorbaninezhad F, Tahmasebi S. Th17 cell function in cancers: immunosuppressive agents or anti-tumor allies? Cancer Cell Int 2024; 24:355. [PMID: 39465401 PMCID: PMC11514949 DOI: 10.1186/s12935-024-03525-9] [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: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
T helper (Th) 17 cells, a distinct subset of Th lymphocytes, are known for their prominent interleukin (IL)-17 production and other pro-inflammatory cytokines. These cells exhibit remarkable plasticity, allowing them to exhibit different phenotypes in the cancer microenvironment. This adaptability enables Th17 cells to promote tumor progression by immunosuppressive activities and angiogenesis, but also mediate anti-tumor immune responses through employing immune cells in tumor setting or even by directly converting toward Th1 phenotype and producing interferon-gamma (IFN-γ). This dual role of Th17 cells in cancer makes it a double-edged sword in encountering cancer. In this review, we aim to elucidate the complexities of Th17 cell function in cancer by summarizing recent studies and, ultimately, to design novel therapeutic strategies, especially targeting Th17 cells in the tumor milieu, which could pave the way for more effective cancer treatments.
Collapse
Affiliation(s)
- Milad Taghizadeh Anvar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimiya Rashidan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Arsam
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Yadegari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Asgari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Ghorbani Vanan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Alharthi SB, Alsubai AH, Almalki SK, El-Shehawi AM, Eldebsy AM, Alsoliman AA, Alharthi RF, Morsi M, Alharthi D, Mutabaqani RA. Comparative Analysis of Gene Expression at Sea Level and High Altitude: A Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) Approach. Cureus 2024; 16:e72489. [PMID: 39600785 PMCID: PMC11592035 DOI: 10.7759/cureus.72489] [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] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
This research studies the gene expression in response to different oxygen environments and looks at high vs low oxygen environments. Tracking down the activity of some of these genes, namely VHLEL, VEGF and HIF-1α, quantitative real-time polymerase chain reaction (PCR) analysis was done for the study group at sea level in Jeddah and at high altitude in Taif city. It has been found that these genes are much more active in higher altitudes which indicates that there is a biological mechanism that makes those specific sites more oversized for the issue of low oxygen. This knowledge is beneficial as it helps in understanding how people grow to live in high-altitude regions. This is positive, especially in justifying the use of the methods in treatment of altitude sickness and other related diseases. Although these findings bring some hope, it would be equally important to include more participants in future studies in order to consolidate our findings and gain deeper understanding of physiological adaptation in low oxygen. This research work has potential significant contribution to the medical profession under conditions of similar environment.
Collapse
Affiliation(s)
- Siraj B Alharthi
- Molecular Diagnostic Unit, Al Hada Armed Forces Hospital, Taif, SAU
- Biological Sciences, King Abdulaziz University, Jeddah, SAU
| | | | - Saad K Almalki
- Science Department, Shorouq Al Mamlakah International School, Taif, SAU
| | | | - Ahmed M Eldebsy
- Science Department, Shorouq Al Mamlakah International School, Taif, SAU
| | | | - Rasha F Alharthi
- Molecular Diagnostic Unit, Al Hada Armed Forces Hospital, Taif, SAU
| | - Mohamed Morsi
- Biological Sciences, King Abdulaziz University, Jeddah, SAU
| | - Dalia Alharthi
- Molecular Diagnostic Unit, Al Hada Armed Forces Hospital, Taif, SAU
| | | |
Collapse
|
18
|
Moore LG, Lorca RA, Gumina DL, Wesolowski SR, Reisz JA, Cioffi-Ragan D, Houck JA, Banerji S, Euser AG, D'Alessandro A, Hobbins JC, Julian CG. Maternal AMPK pathway activation with uterine artery blood flow and fetal growth maintenance during hypoxia. Am J Physiol Heart Circ Physiol 2024; 327:H778-H792. [PMID: 39028630 PMCID: PMC11482288 DOI: 10.1152/ajpheart.00193.2024] [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: 03/28/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
High-altitude (HA) hypoxia lowers uterine artery (UtA) blood flow during pregnancy and birth weight. Adenosine monophosphate kinase (AMPK) activation has selective, uteroplacental vasodilator effects that lessen hypoxia-associated birth weight reductions. In this study, we determined the relationship between AMPK-pathway gene expression and metabolites in the maternal circulation during HA pregnancy as well as with the maintenance of UtA blood flow and birth weight at HA. Residents at HA (2,793 m) versus low altitude (LA; 1,640 m) had smaller UtA diameters at weeks 20 and 34, lower UtA blood flow at week 20, and lower birth weight babies. At week 34, women residing at HA versus women residing at LA had decreased expression of upstream and downstream AMPK-pathway genes. Expression of the α1-AMPK catalytic subunit, PRKAA1, correlated positively with UtA diameter and blood flow at weeks 20 (HA) and 34 (LA). Downstream AMPK-pathway gene expression positively correlated with week 20 fetal biometry at both altitudes and with UtA diameter and birth weight at LA. Reduced gene expression of AMPK activators and downstream targets in women residing at HA versus women residing at LA, together with positive correlations between PRKAA1 gene expression, UtA diameter, and blood flow suggest that greater sensitivity to AMPK activation at midgestation at HA may help offset later depressant effects of hypoxia on fetal growth.NEW & NOTEWORTHY Fetal growth restriction (FGR) is increased and uterine artery (UtA) blood flow is lower at high altitudes (HA) but not all HA pregnancies have FGR. Here we show that greater UtA diameter and blood flow at week 20 are positively correlated with higher expression of the gene encoding the α1-catalytic subunit of AMP protein kinase, PRKAA1, suggesting that increased AMPK activation may help to prevent the detrimental effects of chronic hypoxia on fetal growth.
Collapse
Affiliation(s)
- Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Diane L Gumina
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- The University of Colorado John C. Hobbins Perinatal Center, Denver, Colorado, United States
| | - Stephanie R Wesolowski
- Division of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Darleen Cioffi-Ragan
- The University of Colorado John C. Hobbins Perinatal Center, Denver, Colorado, United States
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Julie A Houck
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Sarah Banerji
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Anna G Euser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - John C Hobbins
- The University of Colorado John C. Hobbins Perinatal Center, Denver, Colorado, United States
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Colleen G Julian
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
19
|
Racca AC, Nardi S, Flores-Martin J, Genti-Raimondi S, Panzetta-Dutari GM. KLF6 negatively regulates HIF-1α in extravillous trophoblasts under hypoxia. Placenta 2024; 156:38-45. [PMID: 39244791 DOI: 10.1016/j.placenta.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION HIF-1α, the master regulator of hypoxia cellular response, is stabilized under low oxygen levels and degraded in the presence of oxygen but its transcription, translation, and degradation are tightly regulated by numerous pathways. KLF6 is a transcription factor involved in proliferation, differentiation, and apoptosis in several cell systems. Under hypoxia it is upregulated in a HIF-1α-dependent manner in extravillous trophoblasts. Considering the importance of hypoxia modulation of EVT behavior through HIF1-α we aimed to study whether KLF6 modulates HIF-1α expression in HTR8/SVneo cells. METHODS HTR8/SVneo cells were cultured in a 1 % oxygen chamber or in 3D format where a spontaneous oxygen gradient is generated. qRT-PCR and Western blot were performed to analyze mRNA and protein expression, respectively. SiRNA, shRNA, or plasmids were used to down- or up-regulate gene expression. Wound healing assay was performed under hypoxia to evaluate migration. The NFκB pathway was modulated with dominant negative mutants and a chemical inhibitor. Cobalt chloride was used to block HIF-1α degradation. RESULTS KLF6 up- and down-regulation in HTR8/SVneo cells exposed to acute hypoxia revealed a negative regulation on HIF-1α. KLF6 silencing led to a partially HIF-1α-dependent increase in MMP9 and VEGF. The NF-κB pathway and HIF-1α degradation were involved in KLF6-dependent HIF-1α regulation. HTR8/SVneo-3D culture showed that KLF6 negatively regulates HIF-1α in a microenvironment with naturally generated hypoxia. DISCUSSION Present results reveal that KLF6 contributes to a fine tune modulation of HIF-1α level under hypoxia. Thus, sustaining a HIF-1α homeostatic level, KLF6 might contribute to control EVT adaptation to hypoxia.
Collapse
Affiliation(s)
- Ana C Racca
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina.
| | - Sofía Nardi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Jésica Flores-Martin
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Susana Genti-Raimondi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Graciela M Panzetta-Dutari
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| |
Collapse
|
20
|
Jan-ngam V, Boontha S, Tubsuwan A, Wongpalee SP, Fanhchaksai K, Tantiworawit A, Charoenkwan P, Khamphikham P. Genetic modifications of EGLN1 reactivate HbF production in β 0-thalassemia/HbE. Heliyon 2024; 10:e38020. [PMID: 39381253 PMCID: PMC11459010 DOI: 10.1016/j.heliyon.2024.e38020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Reactivation of fetal hemoglobin (HbF, α2γ2) potentially alleviates clinical presentation in β-thalassemia. Prolyl hydroxylase domain enzymes (PHDs) play roles in the canonical oxygen-sensing pathway and maintain the stability of cellular hypoxia-inducible factor α (HIF-α) in response to low oxygen levels or hypoxia. Pharmacological inhibition of PHDs has been shown to increase HbF production in erythroid progenitors derived from healthy donors. Here, we demonstrated the relationship between PHD2, the main PHD isoform, and clinical phenotypes in β0-thalassemia/HbE disease. Although the targeted sequencing annotated several common variants within EGLN1, the gene encoding PHD2, none of these variants were located in the functional domains of PHD2 and were irrelevant to the clinical phenotypes. CRISPR-mediated EGLN1 modifications at the functional regions; however, led to significantly reduce PHD2 expression and increase HbF expression levels in severe β-thalassemia erythroblasts. Moreover, these beneficial phenotypes were independent to the two well-known HbF regulators including BCL11A and GATA1. Our findings introduce an additional mechanism for HbF regulation in β-thalassemia and propose that targeting the canonical oxygen-sensing pathway, particularly PHD2 functional domains, might offer a promising therapeutic strategy to β-thalassemia diseases.
Collapse
Affiliation(s)
- Varit Jan-ngam
- Master of Science Program in Medical Technology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Siriraj Boontha
- Bachelor of Science Program in Medical Technology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Alisa Tubsuwan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Somsakul Pop Wongpalee
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kanda Fanhchaksai
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pinyaphat Khamphikham
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Hematology and Health Technology Research Center, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
21
|
Chettouh-Hammas N, Grillon C. Physiological skin oxygen levels: An important criterion for skin cell functionality and therapeutic approaches. Free Radic Biol Med 2024; 222:259-274. [PMID: 38908804 DOI: 10.1016/j.freeradbiomed.2024.06.015] [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: 04/29/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The skin is made up of different layers with various gradients, which maintain a complex microenvironment, particularly in terms of oxygen levels. However, all types of skin cells are cultured in conventional incubators that do not reproduce physiological oxygen levels. Instead, they are cultured at atmospheric oxygen levels, a condition that is far removed from physiology and may lead to the generation of free radicals known to induce skin ageing. This review aims to summarize the current literature on the effect of physiological oxygen levels on skin cells, highlight the shortcomings of current in vitro models, and demonstrate the importance of respecting skin oxygen levels. We begin by clarifying the terminology used about oxygen levels and describe the specific distribution of oxygen in the skin. We review and discuss how skin cells adapt their oxygen consumption and metabolism to oxygen levels environment, as well as the changes that are induced, particularly, their redox state, life cycle and functions. We examine the effects of oxygen on both simple culture models and more complex reconstructed skin models. Finally, we present the implications of oxygen modulation for a more therapeutic approach.
Collapse
Affiliation(s)
- Nadira Chettouh-Hammas
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| | - Catherine Grillon
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| |
Collapse
|
22
|
Du W, Novin A, Liu Y, Afzal J, Liu S, Suhail Y, Kshitiz. Stable and oscillatory hypoxia differentially regulate invasibility of breast cancer associated fibroblasts. MECHANOBIOLOGY IN MEDICINE 2024; 2:100070. [PMID: 40365532 PMCID: PMC12074668 DOI: 10.1016/j.mbm.2024.100070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
As local regions in the tumor outstrip their oxygen supply, hypoxia can develop, affecting not only the cancer cells, but also other cells in the microenvironment, including cancer associated fibroblasts (CAFs). Hypoxia is also not necessarily stable over time, and can fluctuate or oscillate. Hypoxia Inducible Factor-1 is the master regulator of cellular response to hypoxia, and can also exhibit oscillations in its activity. To understand how stable, and fluctuating hypoxia influence breast CAFs, we measured changes in gene expression in CAFs in normoxia, hypoxia, and oscillatory hypoxia, as well as measured change in their capacity to resist, or assist breast cancer invasion. We show that hypoxia has a profound effect on breast CAFs causing activation of key pathways associated with fibroblast activation, but reduce myofibroblast activation and traction force generation. We also found that oscillatory hypoxia, while expectedly resulted in a "sub-hypoxic" response in gene expression, it resulted in specific activation of pathways associated with actin polymerization and actomyosin maturation. Using traction force microscopy, and a nanopatterned stromal invasion assay, we show that oscillatory hypoxia increases contractile force generation vs stable hypoxia, and increases heterogeneity in force generation response, while also additively enhancing invasibility of CAFs to MDA-MB-231 invasion. Our data show that stable and unstable hypoxia can regulate many mechnobiological characteristics of CAFs, and can contribute to transformation of CAFs to assist cancer dissemination and onset of metastasis.
Collapse
Affiliation(s)
- Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Ashkan Novin
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Junaid Afzal
- Department of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Shaofei Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
- NEAG Comprehensive Cancer Center, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
23
|
Fry LG, Washam CL, Roys H, Bowlin AK, Venugopal G, Bird JT, Byrum SD, Weinkopff T. HIF-α signaling regulates the macrophage inflammatory response during Leishmania major infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.605844. [PMID: 39253467 PMCID: PMC11383058 DOI: 10.1101/2024.08.27.605844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cutaneous leishmaniasis (CL) contributes significantly to the global burden of neglected tropical diseases, with 12 million people currently infected with Leishmania parasites. CL encompasses a range of disease manifestations, from self-healing skin lesions to permanent disfigurations. Currently there is no vaccine available, and many patients are refractory to treatment, emphasizing the need for new therapeutic targets. Previous work demonstrated macrophage HIF-α-mediated lymphangiogenesis is necessary to achieve efficient wound resolution during murine L. major infection. Here, we investigate the role of macrophage HIF-α signaling independent of lymphangiogenesis. We sought to determine the relative contributions of the parasite and the host-mediated inflammation in the lesional microenvironment to myeloid HIF-α signaling. Because HIF-α activation can be detected in infected and bystander macrophages in leishmanial lesions, we hypothesize it is the host's inflammatory response and microenvironment, rather than the parasite, that triggers HIF-α activation. To address this, macrophages from mice with intact HIF-α signaling (LysM Cre ARNT f/+ ) or mice with deleted HIF-α signaling (LysM Cre ARNT f/f ) were subjected to RNASequencing after L. major infection and under pro-inflammatory stimulus. We report that L. major infection alone is enough to induce some minor HIF-α-dependent transcriptomic changes, while infection with L. major in combincation with pro-inflammatory stimuli induces numerous transcriptomic changes that are both dependent and independent of HIF-α signaling. Additionally, by coupling transcriptomic analysis with several pathway analyses, we found HIF-α suppresses pathways involved in protein translation during L. major infection in a pro-inflammatory environment. Together these findings show L. major induces a HIF-α-dependent transcriptomic program, but HIF-α only suppresses protein translation in a pro-inflammatory environment. Thus, this work indicates the host inflammatory response, rather than the parasite, largely contributes to myeloid HIF-α signaling during Leishmania infection.
Collapse
|
24
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [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: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
25
|
Shokr SM, Kahlert S, Kluess J, Hradsky J, Dänicke S, Rothkötter HJ, Nossol C. Modeling of culture conditions by culture system, glucose and propionic acid and their impact on metabolic profile in IPEC-J2. PLoS One 2024; 19:e0307411. [PMID: 39024309 PMCID: PMC11257281 DOI: 10.1371/journal.pone.0307411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 07/04/2024] [Indexed: 07/20/2024] Open
Abstract
The microbiological environment and their corresponding secreted metabolite spectrum are an essential modulator of the enterocyte function, effecting the whole organism. Intestinal porcine jejunal epithelial cell line (IPEC-J2) is an established in vitro model for differentiation of enterocytes in different cell culture models. An improved oxygen supply seems to be the main reason for differentiation in an air-liquid-interface culture, but this has not yet been conclusively clarified. In this context, the nutrition of the cell and its influence on the metabolism is also of crucial importance. The interest in short-chain fatty acids (SCFAs) has grown steadily in recent years due to their clinical relevance in certain diseases such as multiple sclerosis and other inflammatory diseases, but not much is known of FFAR2 and FFAR3 (free fatty acid receptor 2 and 3) in pigs. We want to address the questions: 1. about the distribution of FFAR2 and FFAR3 in vivo and in vitro in sus scrofa 2. whether there is an influence of propionic acid, glucose content and cultivation on metabolism of enterocytes? The morphological analysis of FFAR2 and FFAR3 in vivo was investigated through immunostaining of frozen sections of the porcine gut segments jejunum, ileum and colon. Both receptors are expressed along the gut and were found in the smooth muscle cells of the tunica muscularis and lamina muscularis mucosae. Furthermore, a high expression of FFAR2 and a low expression of FFAR3 in the enteric nerve system was also observed in jejunum, ileum and colon of sus scrofa. In addition, FFAR2 and FFAR3 within the vessels was investigated. FFAR3 showed a strong expression on endothelial cells of veins and lymphatic vessels but was not detectable on arteries. Furthermore, we demonstrate for the first time, FFAR2 and FFAR3 in IPEC-J2 cells on RNA- and protein level, as well as with confocal microscopy. In addition, ENO1 and NDUFA4 were investigated on RNA-level in IPEC-J2 cells as 2 important genes, which play an essential role in metabolism. Here, NDUFA4 is detected in the model animal sus scrofa as well as in the porcine cell line IPEC-J2. A potential impact of propionic acid and/or glucose and/or cultivation method on the metabolism of the cells was tested with the Seahorse analyzer. Here, a significant higher ECAR was observed in the SMC than in the OCR. In summary, we were able to show that the cultivation system appears to have a greater influence than the medium composition or nutrition of the cells. However, this can be modulated by incubation time or combination of different SCFAs.
Collapse
Affiliation(s)
- Shirko Marcel Shokr
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Stefan Kahlert
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Johannes Hradsky
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Sven Dänicke
- Friedrich-Loeffler Institute, Braunschweig, Germany
| | | | - Constanze Nossol
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
26
|
Tao B, Gong W, Xu C, Ma Z, Mei J, Chen M. The relationship between hypoxia and Alzheimer's disease: an updated review. Front Aging Neurosci 2024; 16:1402774. [PMID: 39086755 PMCID: PMC11288848 DOI: 10.3389/fnagi.2024.1402774] [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: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and the most prevalent form of dementia. The main hallmarks for the diagnosis of AD are extracellular amyloid-beta (Aβ) plaque deposition and intracellular accumulation of highly hyperphosphorylated Tau protein as neurofibrillary tangles. The brain consumes more oxygen than any other organs, so it is more easily to be affected by hypoxia. Hypoxia has long been recognized as one of the possible causes of AD and other neurodegenerative diseases, but the exact mechanism has not been clarified. In this review, we will elucidate the connection between hypoxia-inducible factors-1α and AD, including its contribution to AD and its possible protective effects. Additionally, we will discuss the relationship between oxidative stress and AD as evidence show that oxidative stress acts on AD-related pathogenic factors such as mitochondrial dysfunction, Aβ deposition, inflammation, etc. Currently, there is no cure for AD. Given the close association between hypoxia, oxidative stress, and AD, along with current research on the protective effects of antioxidants against AD, we speculate that antioxidants could be a potential therapeutic approach for AD and worth further study.
Collapse
Affiliation(s)
- Borui Tao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Wei Gong
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengyuan Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhihui Ma
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinyu Mei
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Suhail Y, Liu Y, Du W, Afzal J, Qiu X, Atiq A, Vera-Licona P, Agmon E, Kshitiz. Oscillatory Hypoxia Induced Unfolded Protein Folding Response Gene Expression Predicts Low Survival in Human Breast Cancer Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577274. [PMID: 38328204 PMCID: PMC10849661 DOI: 10.1101/2024.01.25.577274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Hypoxia is one of the key factors in the tumor microenvironment regulating nearly all steps in the metastatic cascade in many cancers, including in breast cancer. The hypoxic regions can however be dynamic with the availability of oxygen fluctuating or oscillating. The canonical response to hypoxia is relayed by transcription factor HIF-1, which is stabilized in hypoxia and acts as the master regulator of a large number of downstream genes. However, HIF-1 transcriptional activity can also fluctuate either due to unstable hypoxia, or by lactate mediated non-canonical degradation of HIF-1. Our understanding of how oscillatory hypoxia or HIF-1 activity specifically influence cancer malignancy is very limited. Here, using MDA-MB-231 cells as a model of triple negative breast cancer characterized by severe hypoxia, we measured the gene expression changes induced specifically by oscillatory hypoxia. We found that oscillatory hypoxia can specifically regulate gene expression differently, and at times opposite to stable hypoxia. Using The Cancer Genome Atlas (TCGA) RNAseq data of human cancer samples, we show that the oscillatory specific gene expression signature in MDA-MB-231 is enriched in most human cancers, and prognosticate low survival in breast cancer patients. In particular, we found that oscillatory hypoxia, unlike stable hypoxia, induces unfolded protein folding response (UPR) in cells resulting in gene expression predicting reduced survival.
Collapse
|
28
|
Schmidt K, Thatcher A, Grobe A, Broussard P, Hicks L, Gu H, Ellies LG, Sears DD, Kalachev L, Kroll E. The combined treatment with ketogenic diet and metformin slows tumor growth in two mouse models of triple negative breast cancer. TRANSLATIONAL MEDICINE COMMUNICATIONS 2024; 9:21. [PMID: 39574543 PMCID: PMC11580796 DOI: 10.1186/s41231-024-00178-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/22/2024] [Indexed: 11/24/2024]
Abstract
Background Many tumors contain hypoxic microenvironments caused by inefficient tumor vascularization. Hypoxic tumors have been shown to resist conventional cancer therapies. Hypoxic cancer cells rely on glucose to meet their energetic and anabolic needs to fuel uncontrolled proliferation and metastasis. This glucose dependency is linked to a metabolic shift in response to hypoxic conditions. Methods To leverage the glucose dependency of hypoxic tumor cells, we assessed the effects of a mild reduction in systemic glucose by controlling both dietary carbohydrates with a ketogenic diet and endogenous glucose production by using metformin on two mouse models of triple-negative breast cancer (TNBC). Results Here, we showed that animals with TNBC treated with the combination regimen of ketogenic diet and metformin (a) had their tumor burden lowered by two-thirds, (b) displayed 38% slower tumor growth, and (c) showed 36% longer latency, compared to the animals treated with a ketogenic diet or metformin alone. As a result, lowering systemic glucose by this combined dietary and pharmacologic approach improved overall survival in our mouse TNBC models by 31 days, approximately equivalent to 3 years of life extension in human terms. Conclusion This preclinical study demonstrates that reducing systemic glucose by combining a ketogenic diet and metformin significantly inhibits tumor proliferation and increases overall survival. Our findings suggest a possible treatment for a broad range of hypoxic and glycolytic tumor types that can augment existing treatment options to improve patient outcomes.
Collapse
Affiliation(s)
- Karen Schmidt
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Amber Thatcher
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Albert Grobe
- Silverlake Research Corporation, Missoula, MT, USA
| | - Pamela Broussard
- College of Humanities and Sciences, University of Montana, Missoula, MT, USA
| | - Linda Hicks
- College of Humanities and Sciences, University of Montana, Missoula, MT, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Lesley G Ellies
- Department of Pathology, University of California San Diego, San Diego, CA, USA
| | - Dorothy D. Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Leonid Kalachev
- Department of Mathematical Sciences, University of Montana, Missoula, MT, USA
| | - Eugene Kroll
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Present address: Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
29
|
Maida CD, Norrito RL, Rizzica S, Mazzola M, Scarantino ER, Tuttolomondo A. Molecular Pathogenesis of Ischemic and Hemorrhagic Strokes: Background and Therapeutic Approaches. Int J Mol Sci 2024; 25:6297. [PMID: 38928006 PMCID: PMC11203482 DOI: 10.3390/ijms25126297] [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: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke represents one of the neurological diseases most responsible for death and permanent disability in the world. Different factors, such as thrombus, emboli and atherosclerosis, take part in the intricate pathophysiology of stroke. Comprehending the molecular processes involved in this mechanism is crucial to developing new, specific and efficient treatments. Some common mechanisms are excitotoxicity and calcium overload, oxidative stress and neuroinflammation. Furthermore, non-coding RNAs (ncRNAs) are critical in pathophysiology and recovery after cerebral ischemia. ncRNAs, particularly microRNAs, and long non-coding RNAs (lncRNAs) are essential for angiogenesis and neuroprotection, and they have been suggested to be therapeutic, diagnostic and prognostic tools in cerebrovascular diseases, including stroke. This review summarizes the intricate molecular mechanisms underlying ischemic and hemorrhagic stroke and delves into the function of miRNAs in the development of brain damage. Furthermore, we will analyze new perspectives on treatment based on molecular mechanisms in addition to traditional stroke therapies.
Collapse
Affiliation(s)
- Carlo Domenico Maida
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
- Molecular and Clinical Medicine Ph.D. Programme, University of Palermo, 90133 Palermo, Italy
| | - Rosario Luca Norrito
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Salvatore Rizzica
- Department of Internal Medicine, S. Elia Hospital, 93100 Caltanissetta, Italy;
| | - Marco Mazzola
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| | - Elisa Rita Scarantino
- Division of Geriatric and Intensive Care Medicine, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50134 Florence, Italy;
| | - Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, University of Palermo, 90133 Palermo, Italy; (R.L.N.); (M.M.); (A.T.)
| |
Collapse
|
30
|
Odeh D, Oršolić N, Adrović E, Bilandžić N, Sedak M, Žarković I, Lesar N, Balta V. The Impact of the Combined Effect of Inhalation Anesthetics and Iron Dextran on Rats' Systemic Toxicity. Int J Mol Sci 2024; 25:6323. [PMID: 38928030 PMCID: PMC11203443 DOI: 10.3390/ijms25126323] [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: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Disruption of any stage of iron homeostasis, including uptake, utilization, efflux, and storage, can cause progressive damage to peripheral organs. The health hazards associated with occupational exposure to inhalation anesthetics (IA) in combination with chronic iron overload are not well documented. This study aimed to investigate changes in the concentration of essential metals in the peripheral organs of rats after iron overload in combination with IA. The aim was also to determine how iron overload in combination with IA affects tissue metal homeostasis, hepcidin-ferritin levels, and MMP levels according to physiological, functional, and tissue features. According to the obtained results, iron accumulation was most pronounced in the liver (19×), spleen (6.7×), lungs (3.1×), and kidneys (2.5×) compared to control. Iron accumulation is associated with elevated heavy metal levels and impaired essential metal concentrations due to oxidative stress (OS). Notably, the use of IA increases the iron overload toxicity, especially after Isoflurane exposure. The results show that the regulation of iron homeostasis is based on the interaction of hepcidin, ferritin, and other proteins regulated by inflammation, OS, free iron levels, erythropoiesis, and hypoxia. Long-term exposure to IA and iron leads to the development of numerous adaptation mechanisms in response to toxicity, OS, and inflammation. These adaptive mechanisms of iron regulation lead to the inhibition of MMP activity and reduction of oxidative stress, protecting the organism from possible damage.
Collapse
Affiliation(s)
- Dyana Odeh
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Nada Oršolić
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Emanuela Adrović
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Nina Bilandžić
- Laboratory for Determination of Residues, Croatian Veterinary Institute, Savska cesta 143, 10000 Zagreb, Croatia
| | - Marija Sedak
- Laboratory for Determination of Residues, Croatian Veterinary Institute, Savska cesta 143, 10000 Zagreb, Croatia
| | - Irena Žarković
- Laboratory for Analysis of Veterinary Medicinal Products, Croatian Veterinary Institute, Savska cesta 143, 10000 Zagreb, Croatia
| | - Nikola Lesar
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Vedran Balta
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| |
Collapse
|
31
|
Altınönder İ, Kaya M, Yentür SP, Çakar A, Durmuş H, Yegen G, Özkan B, Parman Y, Sawalha AH, Saruhan-Direskeneli G. Thymic gene expression analysis reveals a potential link between HIF-1A and Th17/Treg imbalance in thymoma associated myasthenia gravis. J Neuroinflammation 2024; 21:126. [PMID: 38734662 PMCID: PMC11088784 DOI: 10.1186/s12974-024-03095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/07/2024] [Indexed: 05/13/2024] Open
Abstract
Myasthenia gravis (MG) is an immune-mediated disease frequently associated with thymic changes. Increased T helper 17 (Th17) cell activity and dysfunctional regulatory T (Treg) cells have been demonstrated in subgroups of MG. On the other hand, hypoxia-inducible factor 1 (HIF-1) has been shown to regulate the Th17/Treg balance by inducing Th17 differentiation while attenuating Treg development. To identify the underlying mechanisms of different thymic pathologies in MG development, we evaluated thymic samples from thymoma-associated myasthenia gravis (TAMG), MG with hyperplasia (TFH-MG) and thymoma without MG (TOMA) patients. Differential gene expression analysis revealed that TAMG and TFH-MG cells are associated with different functional pathways. A higher RORC/FOXP3 ratio provided evidence for Th17/Treg imbalance in TAMG potentially related to increased HIF1A. The hypoxic microenvironment in thymoma may be a driver of TAMG by increasing HIF1A. These findings may lead to new therapeutic approaches targeting HIF1A in the development of TAMG.
Collapse
Affiliation(s)
- İlayda Altınönder
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Mustafa Kaya
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Sibel P Yentür
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Arman Çakar
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Hacer Durmuş
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Gülçin Yegen
- Department of Thoracic Surgery, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Berker Özkan
- Department of Pathology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Yeşim Parman
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, 34093, Turkey
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
32
|
Nan Y, Wu X, Luo Q, Chang W, Zhao P, Zhang L, Liu Z. OTUB2 silencing promotes ovarian cancer via mitochondrial metabolic reprogramming and can be synthetically targeted by CA9 inhibition. Proc Natl Acad Sci U S A 2024; 121:e2315348121. [PMID: 38701117 PMCID: PMC11087800 DOI: 10.1073/pnas.2315348121] [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: 09/05/2023] [Accepted: 03/27/2024] [Indexed: 05/05/2024] Open
Abstract
Ovarian cancer is an aggressive gynecological tumor characterized by a high relapse rate and chemoresistance. Ovarian cancer exhibits the cancer hallmark of elevated glycolysis, yet effective strategies targeting cancer cell metabolic reprogramming to overcome therapeutic resistance in ovarian cancer remain elusive. Here, we revealed that epigenetic silencing of Otubain 2 (OTUB2) is a driving force for mitochondrial metabolic reprogramming in ovarian cancer, which promotes tumorigenesis and chemoresistance. Mechanistically, OTUB2 silencing destabilizes sorting nexin 29 pseudogene 2 (SNX29P2), which subsequently prevents hypoxia-inducible factor-1 alpha (HIF-1α) from von Hippel-Lindau tumor suppressor-mediated degradation. Elevated HIF-1α activates the transcription of carbonic anhydrase 9 (CA9) and drives ovarian cancer progression and chemoresistance by promoting glycolysis. Importantly, pharmacological inhibition of CA9 substantially suppressed tumor growth and synergized with carboplatin in the treatment of OTUB2-silenced ovarian cancer. Thus, our study highlights the pivotal role of OTUB2/SNX29P2 in suppressing ovarian cancer development and proposes that targeting CA9-mediated glycolysis is an encouraging strategy for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yabing Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| | - Xiaowei Wu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA02215
| | - Qingyu Luo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Wan Chang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| | - Pengfei Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing100850, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100021, China
| |
Collapse
|
33
|
Wang S, He Y, Wang J, Luo E. Re-exploration of immunotherapy targeting EMT of hepatocellular carcinoma: Starting from the NF-κB pathway. Biomed Pharmacother 2024; 174:116566. [PMID: 38631143 DOI: 10.1016/j.biopha.2024.116566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/15/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancies worldwide, and its high morbidity and mortality have brought a heavy burden to the global public health system. Due to the concealment of its onset, the limitation of treatment, the acquisition of multi-drug resistance and radiation resistance, the treatment of HCC cannot achieve satisfactory results. Epithelial mesenchymal transformation (EMT) is a key process that induces progression, distant metastasis, and therapeutic resistance to a variety of malignant tumors, including HCC. Therefore, targeting EMT has become a promising tumor immunotherapy method for HCC. The NF-κB pathway is a key regulatory pathway for EMT. Targeting this pathway has shown potential to inhibit HCC infiltration, invasion, distant metastasis, and therapeutic resistance. At present, there are still some controversies about this pathway and new ideas of combined therapy, which need to be further explored. This article reviews the progress of immunotherapy in improving EMT development in HCC cells by exploring the mechanism of regulating EMT.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - Yan He
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China
| | - Jun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - En Luo
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China.
| |
Collapse
|
34
|
Ubaid S, Kashif M, Laiq Y, Nayak AK, Kumar V, Singh V. Targeting HIF-1α in sickle cell disease and cancer: unraveling therapeutic opportunities and risks. Expert Opin Ther Targets 2024; 28:357-373. [PMID: 38861226 DOI: 10.1080/14728222.2024.2367640] [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: 02/10/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION HIF-1α, a key player in medical science, holds immense significance in therapeutic approaches. This review delves into its complex dynamics, emphasizing the delicate balance required for its modulation. HIF-1α stands as a cornerstone in medical research, its role extending to therapeutic strategies. This review explores the intricate interplay surrounding HIF-1α, highlighting its critical involvement and the necessity for cautious modulation. AREAS COVERED In sickle cell disease (SCD), HIF-1α's potential to augment fetal hemoglobin (HbF) production and mitigate symptoms is underscored. Furthermore, its role in cancer is examined, particularly its influence on survival in hypoxic tumor microenvironments, angiogenesis, and metastasis. The discussion extends to the intricate relationship between HIF-1α modulation and cancer risks in SCD patients, emphasizing the importance of balancing therapeutic benefits and potential hazards. EXPERT OPINION Managing HIF-1α modulation in SCD patients requires a nuanced approach, considering therapeutic potential alongside associated risks, especially in exacerbating cancer risks. An evolutionary perspective adds depth, highlighting adaptations in populations adapted to low-oxygen environments and aligning cancer cell metabolism with primitive cells. The role of HIF-1α as a therapeutic target is discussed within the context of complex cancer biology and metabolism, acknowledging varied responses across diverse cancers influenced by intricate evolutionary adaptations.
Collapse
Affiliation(s)
- Saba Ubaid
- Department of Biochemistry, King George's Medical University, Lucknow, India
| | - Mohammad Kashif
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Yusra Laiq
- Department of Biotechnology, Era University, Lucknow, India
| | | | - Vipin Kumar
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Vivek Singh
- Department of Biochemistry, King George's Medical University, Lucknow, India
| |
Collapse
|
35
|
Monaci S, Coppola F, Filippi I, Falsini A, Carraro F, Naldini A. Targeting hypoxia signaling pathways in angiogenesis. Front Physiol 2024; 15:1408750. [PMID: 38725568 PMCID: PMC11079266 DOI: 10.3389/fphys.2024.1408750] [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: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Oxygen (O2) supply is constantly maintained by the vascular network for a proper tissue oxygenation. Hypoxia is the result of an increased O2 demand and/or decreased supply and is common in both physiological conditions and human diseases. Angiogenesis is one of the adaptive responses to hypoxia and is mainly regulated by the hypoxia-inducible factors, HIFs. These heterodimeric transcription factors are composed of one of three O2-dependent α subunits (HIF-1, HIF-2, and HIF-3) and a constitutively expressed O2-insensitive subunit (HIF-1β). Among them HIF-1α is the most characterized and its activity is tightly controlled. Under hypoxia, its intracellular accumulation triggers the transcription of several genes, involved in cell survival/proliferation, autophagy, apoptosis, cell metabolism, and angiogenesis. HIF pathway is also modulated by specific microRNAs (miRNAs), thus resulting in the variation of several cellular responses, including alteration of the angiogenic process. The pro-angiogenic activity of HIF-1α is not restricted to endothelial cells, as it also affects the behavior of other cell types, including tumor and inflammatory/immune cells. In this context, exosomes play a crucial role in cell-cell communication by transferring bio-active cargos such as mRNAs, miRNAs, and proteins (e.g., VEGFA mRNA, miR210, HIF-1α). This minireview will provide a synopsis of the multiple factors able to modulate hypoxia-induced angiogenesis especially in the tumor microenvironment context. Targeting hypoxia signaling pathways by up-to-date approaches may be relevant in the design of therapeutic strategies in those pathologies where angiogenesis is dysregulated.
Collapse
Affiliation(s)
- Sara Monaci
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Federica Coppola
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Irene Filippi
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Alessandro Falsini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Fabio Carraro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Antonella Naldini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
36
|
Papanikolaou NA, Kakavoulia M, Ladias C, Papavassiliou AG. The ras-related protein RAB22A interacts with hypoxia-inducible factor 1-alpha (HIF-1α) in MDA-MB-231 breast cancer cells in hypoxia. Mol Biol Rep 2024; 51:564. [PMID: 38647725 DOI: 10.1007/s11033-024-09516-3] [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: 02/24/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Recent studies suggest that hypoxia-inducible factor 1-alpha (HIF-1α) and the small GTPase protein Ras-related protein Rab-22 A (RAB22A) may be colocalized in the cytoplasm and that as a conequence they may enhance the formation of microvesicles in breast cancer cells under hypoxia. Therefore, we sought to determine whether these two proteins are present in intracellular complexes in breast carcinoma cells. METHODS AND RESULTS Evaluation using molecular docking indicated that HIF-1α and RAB22A interact with each other. Co-immunoprecipitation of endogenous or ectopically expressed HIF-1α and RAB22A proteins in MDA-MB-231 breast cancer cells or HEK-293T cells demonstrated that endogenous HIF-1α and RAB22A can form an intracellular complex; however, transiently expressed HIF-1α and RAB22A failed to interact. Investigating RAB22A and HIF-1α interactions in various cancer cell lines under hypoxia may shed light on their roles in cancer cell survival and progression through regulation of intracellular trafficking by HIF-1α under hypoxic conditions. CONCLUSIONS Our study is the first to reveal the potential involvement of HIF-1α in intracellular trafficking through physical interactions with the small GTPase protein RAB22A. We discuss the implications of our work on the role of exosomes and microvesicles in tumor invasiveness.
Collapse
Affiliation(s)
- Nikolaos A Papanikolaou
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Macedonia, Greece.
| | - Maria Kakavoulia
- Laboratory of Biological Chemistry, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Macedonia, Greece
| | - Christos Ladias
- Department of Biological Applications and Technology, University of Ioannina, 45110, Ioannina, Epirus, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| |
Collapse
|
37
|
Lemaitre T, Cornu M, Schwalen F, Since M, Kieffer C, Voisin-Chiret AS. Molecular glue degraders: exciting opportunities for novel drug discovery. Expert Opin Drug Discov 2024; 19:433-449. [PMID: 38240114 DOI: 10.1080/17460441.2024.2306845] [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: 10/06/2023] [Accepted: 01/15/2024] [Indexed: 01/25/2024]
Abstract
INTRODUCTION Molecular Glue Degraders (MGDs) is a concept that refers to a class of compounds that facilitate the interaction between two proteins or molecules within a cell. These compounds act as bridge that enhances specific Protein-Protein Interactions (PPIs). Over the past decade, this technology has gained attention as a potential strategy to target proteins that were traditionally considered undruggable using small molecules. AREAS COVERED This review presents the concept of cellular homeostasis and the balance between protein synthesis and protein degradation. The concept of protein degradation is concerned with molecular glues, which form part of the broader field of Targeted Protein Degradation (TPD). Next, pharmacochemical strategies for the rational design of MGDs are detailed and illustrated by examples of Ligand-Based (LBDD), Structure-Based (SBDD) and Fragment-Based Drug Design (FBDD). EXPERT OPINION Expanding the scope of what can be effectively targeted in the development of treatments for diseases that are incurable or resistant to conventional therapies offers new therapeutic options. The treatment of microbial infections and neurodegenerative diseases is a major societal challenge, and the discovery of MGDs appears to be a promising avenue. Combining different approaches to discover and exploit a variety of innovative therapeutic agents will create opportunities to treat diseases that are still incurable.
Collapse
Affiliation(s)
| | - Marie Cornu
- Normandie University, UNICAEN, CERMN, Caen, France
| | - Florian Schwalen
- Normandie University, UNICAEN, CERMN, Caen, France
- Department of Pharmacy, Caen University Hospital, Caen, France
| | - Marc Since
- Normandie University, UNICAEN, CERMN, Caen, France
| | | | | |
Collapse
|
38
|
Du W, Novin A, Liu Y, Afzal J, Liu S, Suhail Y, Kshitiz. Stable and Oscillatory Hypoxia Differentially Regulate Invasibility of Breast Cancer Associated Fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586706. [PMID: 38585723 PMCID: PMC10996662 DOI: 10.1101/2024.03.26.586706] [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: 04/09/2024]
Abstract
As local regions in the tumor outstrip their oxygen supply, hypoxia can develop, affecting not only the cancer cells, but also other cells in the microenvironment, including cancer associated fibroblasts (CAFs). Hypoxia is also not necessarily stable over time, and can fluctuate or oscillate. Hypoxia Inducible Factor-1 is the master regulator of cellular response to hypoxia, and can also exhibit oscillations in its activity. To understand how stable, and fluctuating hypoxia influence breast CAFs, we measured changes in gene expression in CAFs in normoxia, hypoxia, and oscillatory hypoxia, as well as measured change in their capacity to resist, or assist breast cancer invasion. We show that hypoxia has a profound effect on breast CAFs causing activation of key pathways associated with fibroblast activation, but reduce myofibroblast activation and traction force generation. We also found that oscillatory hypoxia, while expectedly resulted in a "sub-hypoxic" response in gene expression, it resulted in specific activation of pathways associated with actin polymerization and actomyosin maturation. Using traction force microscopy, and a nanopatterned stromal invasion assay, we show that oscillatory hypoxia increases contractile force generation vs stable hypoxia, and increases heterogeneity in force generation response, while also additively enhancing invasibility of CAFs to MDA-MB-231 invasion. Our data show that stable and unstable hypoxia can regulate many mechnobiological characteristics of CAFs, and can contribute to transformation of CAFs to assist cancer dissemination and onset of metastasis.
Collapse
Affiliation(s)
- Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Ashkan Novin
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
| | - Junaid Afzal
- Department of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Shaofei Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT, USA
- Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT, USA
- NEAG Comprehensive Cancer Center, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
39
|
Hai Y, Ren K, Zhang Y, Yang L, Cao H, Yuan X, Su L, Li H, Feng X, Liu D. HIF-1α serves as a co-linker between AD and T2DM. Biomed Pharmacother 2024; 171:116158. [PMID: 38242039 DOI: 10.1016/j.biopha.2024.116158] [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: 09/24/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD)-related brain deterioration is linked to the type 2 diabetes mellitus (T2DM) features hyperglycemia, hyperinsulinemia, and insulin resistance. Hypoxia as a common risk factor for both AD and T2DM. Hypoxia-inducible factor-1 alpha (HIF-1α) acts as the main regulator of the hypoxia response and may be a key target in the comorbidity of AD and T2DM. HIF-1α expression is closely related to hyperglycemia, insulin resistance, and inflammation. Tissue oxygen consumption disrupts HIF-1α homeostasis, leading to increased reactive oxygen species levels and the inhibition of insulin receptor pathway activity, causing neuroinflammation, insulin resistance, abnormal Aβ deposition, and tau hyperphosphorylation. HIF-1α activation also leads to the deposition of Aβ by promoting the abnormal shearing of amyloid precursor protein and inhibiting the degradation of Aβ, and it promotes tau hyperphosphorylation by activating oxidative stress and the activation of astrocytes, which further exasperates AD. Therefore, we believe that HIF-α has great potential as a target for the treatment of AD. Importantly, the intracellular homeostasis of HIF-1α is a more crucial factor than its expression level.
Collapse
Affiliation(s)
- Yang Hai
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China.
| | - Ke Ren
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Yarong Zhang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Lili Yang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Haoshi Cao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xianxia Yuan
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Linling Su
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Hailong Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Xiaoli Feng
- Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Key Laboratory of Dunhuang Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China
| | - Dongling Liu
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Northwest Collaborative Innovation Center for Traditional Chinese Medicine, Lanzhou 730000, Gansu Province, PR China; Gansu Pharmaceutical Industry Innovation Research Institute, Lanzhou 730000, Gansu Province, PR China.
| |
Collapse
|
40
|
Linawati L, Sitam S, Mulyawan W, Purba A, Syawqie A, Handharyani E, Subiakto Y, Amaliya A. Effect of Intermittent Hypobaric Hypoxia Exposure on HIF-1α, VEGF, and Angiogenesis in the Healing Process of Post-Tooth Extraction Sockets in Rats. Eur J Dent 2024; 18:304-313. [PMID: 37295455 PMCID: PMC10959591 DOI: 10.1055/s-0043-1768639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the effect of intermittent hypobaric hypoxia (IHH) exposure on the expression of hypoxia-induced factor-1α (HIF-1α) messenger RNA (mRNA), vascular endothelial growth factor-a (VEGF-a) mRNA, and angiogenesis after tooth extraction in rats. MATERIALS AND METHODS On 45 male Sprague-Dawley rats were performed the removal of the maxillary left first molar, and then they were randomly divided into 9 groups, namely: 4 groups that were exposed to IHH for 30 minutes every day in the Hypobaric Chamber at an altitude of 18,000 feet, with 1 time hypobaric hypoxia (HH), 3 times HH, 5 times HH, and 7 times HH; 4 normoxia groups that were terminated on days 1, 3, 5, and 7 after tooth extraction; and the 1 control group. Real-time polymerase chain reaction measured the molecular changes in the socket tissue after tooth extraction in rats to evaluate the expression of HIF-1α mRNA and VEGF mRNA. Histological changes with hematoxylin and eosin staining were noted to evaluate the amount of angiogenesis in the socket after tooth extraction. Molecular and histological parameters were calculated at the end of each experiment on days 0, 1, 3, 5, and 7 after tooth extraction, which exhibited the improvement phase of the wound-healing process. RESULTS Increases in the expression of HIF-1α mRNA, VEGF mRNA, and angiogenesis were found in the IHH group compared with the normoxia group and the control group. The expression of HIF-1α mRNA increased significantly (p < 0.05) in the group after one time HH exposure on day 1, then decreased in the IHH group (three times HH exposure, five times HH exposure, and seven times HH exposure) approaching the control group. The expression of VEGF mRNA and angiogenesis began to increase after one time HH exposure on day 1, and increased again after three times HH exposure on day 3, then increased even more after five times HH exposure on day 5, and increased very significantly (**p < 0.05) after seven times HH exposure on day 7. It showed that repeated or intermittent exposure to HH conditions induced a protective response that made cells adapt under hypoxia conditions. CONCLUSION IHH exposure accelerates the socket healing of post-tooth extraction, which is proven by changes in HIF-1α mRNA expression and increase in VEGF mRNA expression as stimuli for angiogenesis in post-tooth extraction sockets under hypobaric hypoxic condition, which also stimulates the formation of new blood vessels, thereby increasing blood supply and accelerating wound healing.
Collapse
Affiliation(s)
- Linawati Linawati
- Doctoral Degree Study Program in Military Dentistry Science, Dental Faculty, Universitas Padjadjaran, Bandung, Indonesia
| | - Suhardjo Sitam
- Department of Radiology, Dental Faculty, Universitas Padjadjaran, Bandung, Indonesia
| | - Wawan Mulyawan
- Department of Community Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ambrosius Purba
- Division of Physiology, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Achmad Syawqie
- Department of Oral Biology, Dental Faculty, Universitas Padjadjaran, Bandung, Indonesia
| | - Ekowati Handharyani
- Department of Veterinary Clinic Reproduction and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor, Indonesia
| | - Yuli Subiakto
- Military Pharmacy Faculty, Universitas Pertahanan, Jakarta, Indonesia
| | - Amaliya Amaliya
- Departement of Periodontology, Dental Faculty, Universitas Padjadjaran, Bandung, Indonesia
- Centre for Military Dentistry Research, Dental Faculty, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
41
|
Wang X, Wang Z, He J. Similarities and Differences of Vascular Calcification in Diabetes and Chronic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:165-192. [PMID: 38222032 PMCID: PMC10788067 DOI: 10.2147/dmso.s438618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Presently, the mechanism of occurrence and development of vascular calcification (VC) is not fully understood; a range of evidence suggests a positive association between diabetes mellitus (DM) and VC. Furthermore, the increasing burden of central vascular disease in patients with chronic kidney disease (CKD) may be due, at least in part, to VC. In this review, we will review recent advances in the mechanisms of VC in the context of CKD and diabetes. The study further unveiled that VC is induced through the stimulation of pro-inflammatory factors, which in turn impairs endothelial function and triggers similar mechanisms in both disease contexts. Notably, hyperglycemia was identified as the distinctive mechanism driving calcification in DM. Conversely, in CKD, calcification is facilitated by mechanisms including mineral metabolism imbalance and the presence of uremic toxins. Additionally, we underscore the significance of investigating vascular alterations and newly identified molecular pathways as potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiabo Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
42
|
Yasan GT, Gunel-Ozcan A. Hypoxia and Hypoxia Mimetic Agents As Potential Priming Approaches to Empower Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:33-54. [PMID: 36642875 DOI: 10.2174/1574888x18666230113143234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSC) exhibit self-renewal capacity and multilineage differentiation potential, making them attractive for research and clinical application. The properties of MSC can vary depending on specific micro-environmental factors. MSC resides in specific niches with low oxygen concentrations, where oxygen functions as a metabolic substrate and a signaling molecule. Conventional physical incubators or chemically hypoxia mimetic agents are applied in cultures to mimic the original low oxygen tension settings where MSC originated. This review aims to focus on the current knowledge of the effects of various physical hypoxic conditions and widely used hypoxia-mimetic agents-PHD inhibitors on mesenchymal stem cells at a cellular and molecular level, including proliferation, stemness, differentiation, viability, apoptosis, senescence, migration, immunomodulation behaviors, as well as epigenetic changes.
Collapse
Affiliation(s)
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
43
|
He YZG, Wang YX, Ma JS, Li RN, Wang J, Lian TY, Zhou YP, Yang HP, Sun K, Jing ZC. MicroRNAs and their regulators: Potential therapeutic targets in pulmonary arterial hypertension. Vascul Pharmacol 2023; 153:107216. [PMID: 37699495 DOI: 10.1016/j.vph.2023.107216] [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: 12/31/2022] [Revised: 08/26/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by pulmonary arterial remodeling. Despite that current combination therapy has shown improvement in morbidity and mortality, a better deciphering of the underlying pathological mechanisms and novel therapeutic targets is urgently needed to combat PAH. MicroRNA, the critical element in post-transcription mechanisms, mediates cellular functions mainly by tuning downstream target gene expression. Meanwhile, upstream regulators can regulate miRNAs in synthesis, transcription, and function. In vivo and in vitro studies have suggested that miRNAs and their regulators are involved in PAH. However, the miRNA-related regulatory mechanisms governing pulmonary vascular remodeling and right ventricular dysfunction remain elusive. Hence, this review summarized the controversial roles of miRNAs in PAH pathogenesis, focused on different miRNA-upstream regulators, including transcription factors, regulatory networks, and environmental stimuli, and finally proposed the prospects and challenges for the therapeutic application of miRNAs and their regulators in PAH treatment.
Collapse
Affiliation(s)
- Yang-Zhi-Ge He
- Center for bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yi-Xuan Wang
- Laboratory Department of Qingzhou People's Hospital, Qingzhou 262500, Shandong, China
| | - Jing-Si Ma
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Ruo-Nan Li
- Department of School of Pharmacy, Henan University, Kaifeng 475100, Henan, China
| | - Jia Wang
- Department of Medical Laboratory, Weifang Medical University, Weifang 261053, Shandong, China
| | - Tian-Yu Lian
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China
| | - Hao-Pu Yang
- Tsinghua University School of Medicine, Beijing 100084, China
| | - Kai Sun
- Medical Science Research Center, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing 100730, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex, Severe and Rare Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
44
|
Rosell-Garcia T, Rivas-Muñoz S, Kin K, Romero-Albillo V, Alcaraz S, Fernandez-Tornero C, Rodriguez-Pascual F. Multimerization of HIF enhances transcription of target genes containing the hypoxia ancillary sequence. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194963. [PMID: 37499936 DOI: 10.1016/j.bbagrm.2023.194963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Transcriptional activity of the hypoxia inducible factor (HIF) relies on the formation of a heterodimer composed of an oxygen-regulated α-subunit and a stably expressed β-subunit. Heterodimeric HIF activates expression by binding to RCGTG motifs within promoters of hypoxia-activated genes. Some hypoxia targets also possess an adjacent HIF ancillary sequence (HAS) reported to increase transcription but whose function remains obscure. Here, we investigate the contribution of the HAS element to the hypoxia response and its mechanism of action, using the HAS-containing prolyl 4-hydroxylase subunit α1 (P4HA1) as a gene model in NIH/3T3 mouse embryonic fibroblasts and HEK293 human embryonic kidney cells. Our HIF overexpression experiments demonstrate that the HAS motif is essential for full induction by hypoxia and that the presence of the tandem HAS/HIF, as opposed to HIF-only sequences, provides HIF proteins with the capacity to form complexes of stoichiometry beyond the classical heterodimer, likely tetramers, to cooperatively potentiate hypoxia-induced transcription. We also provide evidence of the crucial role played by the Fα helix of the PAS-B domain of the HIF1β subunit to support the interaction between heterodimers. Functional analysis showed that human genes containing the HAS/HIF motifs are better responders to hypoxia, and their promoters are enriched for specific transcription factor binding sites. Gene ontology enrichment revealed a predominance of HAS/HIF in genes primarily related to tissue formation and development. Our findings add an extra level of regulation of the hypoxia/HIF signaling through multimerization of HIF proteins on regulatory elements containing the HAS/HIF motifs.
Collapse
Affiliation(s)
- Tamara Rosell-Garcia
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Sergio Rivas-Muñoz
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Koryu Kin
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Spain
| | - Verónica Romero-Albillo
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Silvia Alcaraz
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | | | - Fernando Rodriguez-Pascual
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain.
| |
Collapse
|
45
|
Sharma D, Khan H, Kumar A, Grewal AK, Dua K, Singh TG. Pharmacological modulation of HIF-1 in the treatment of neuropsychiatric disorders. J Neural Transm (Vienna) 2023; 130:1523-1535. [PMID: 37740098 DOI: 10.1007/s00702-023-02698-3] [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: 05/08/2023] [Accepted: 09/06/2023] [Indexed: 09/24/2023]
Abstract
Hypoxia-inducible factor 1 has been identified as an important therapeutic target in psychiatric illnesses. Hypoxia is a condition in which tissues do not receive enough oxygen, resulting in less oxidative energy production. HIF-1, the master regulator of molecular response to hypoxia, is destabilized when oxygen levels fall. HIF-1, when activated, increases the gene transcription factors that promote adaptive response and longevity in hypoxia. HIF-regulated genes encode proteins involved in cell survival, energy metabolism, angiogenesis, erythropoiesis, and vasomotor control. Multiple genetic and environmental variables contribute to the pathophysiology of psychiatric disease. This review focuses on the most recent findings indicating the role of oxygen deprivation in CNS damage, with strong attention on HIF-mediated pathways. Several pieces of evidence suggested that, in the case of hypoxia, induction and maintenance of HIF-1 target genes may help reduce nerve damage. Major new insights into the molecular mechanisms that control HIF's sensitivity to oxygen are used to make drugs that can change the way HIF works as a therapeutic target for some CNS diseases.
Collapse
Affiliation(s)
- Diksha Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amit Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
46
|
Yoo S, Choi S, Kim I, Kim IS. Hypoxic regulation of extracellular vesicles: Implications for cancer therapy. J Control Release 2023; 363:201-220. [PMID: 37739015 DOI: 10.1016/j.jconrel.2023.09.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Extracellular vesicles (EVs) play a pivotal role in intercellular communication and have been implicated in cancer progression. Hypoxia, a pervasive hallmark of cancer, is known to regulate EV biogenesis and function. Hypoxic EVs contain a specific set of proteins, nucleic acids, lipids, and metabolites, capable of reprogramming the biology and fate of recipient cells. Enhancing the intrinsic therapeutic efficacy of EVs can be achieved by strategically modifying their structure and contents. Moreover, the use of EVs as drug delivery vehicles holds great promise for cancer treatment. However, various hurdles must be overcome to enable their clinical application as cancer therapeutics. In this review, we aim to discuss the current knowledge on the hypoxic regulation of EVs. Additionally, we will describe the underlying mechanisms by which EVs contribute to cancer progression in hypoxia and outline the progress and limitations of hypoxia-related EV therapeutics for cancer.
Collapse
Affiliation(s)
- Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Sanga Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea; Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, South Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, South Korea; Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute Science and Technology, Seoul 02792, South Korea.
| |
Collapse
|
47
|
Liu S, Yan W, Lv Q, Yang L, Miao Y, Hu Y, Wei Z. 3, 3'-diindolylmethane, a natural aryl hydrocarbon receptor agonist, alleviates ulcerative colitis by enhancing "glycolysis-lactate-STAT3″ and TIP60 signals-mediated Treg differentiation. Mol Immunol 2023; 163:147-162. [PMID: 37793204 DOI: 10.1016/j.molimm.2023.09.009] [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: 03/29/2023] [Revised: 07/26/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Aryl hydrocarbon receptor (AhR) plays an important role in the occurrence and development of ulcerative colitis (UC). In this study, the effect and mechanism of 3, 3'-diindolylmethane (DIM), the classical AhR agonist, on UC was investigated from the angle of recovering the balance of Th17/Treg. METHODS The in vivo colitis model was established in mice by using dextran sulfate sodium, and CD4+ T cells were used to simulate the in vitro differentiation of Treg and Th17 cells. The proportions and related factors of Th17 and Treg cells were measured using flow cytometry, Q-PCR and western blotting. The glycolysis was evaluated by examining the glucose uptake, glucose consumption and lactate production using kits or immunofluorescence. The activation of AhR was detected by western blotting and the XRE-luciferase reporter gene. The co-immunoprecipitation, transfection or other methods were selected to investigate and identify the signaling molecular pathway. RESULTS DIM significantly attenuated symptoms of colitis mice by rebuilding the balance of Th17/Treg in anoxic colons. In hypoxia, a more potent promotion of Treg differentiation was showed by DIM relative to normoxia, and siFoxp3 prevented DIM-suppressed Th17 differentiation. DIM repressed the excessive glycolysis in hypoxia evidenced by down-regulated glucose uptake, lactate production, Glut1 and HK2 levels. Interestingly, IL-10, the function-related factor of Treg cells, showed the feedback effect of DIM-suppressed glycolysis. Besides, 2-deoxy-D-glucose, HK2 plasmid and IL-10 antibody prevented increase of DIM on the expression of Foxp3 at the transcriptional level and subsequent Treg differentiation through the lactate-STAT3 pathway, and reasons for the direct improvement of DIM on Foxp3 protein was attributed to promoting the formation of HIF-1α/TIP60 complexes as well as subsequent acetylation and protein stability. Finally, AhR dependence and mechanisms for DIM-improved Treg differentiation in vitro and in vivo were well confirmed by using plasmids or inhibitors. CONCLUSIONS DIM enhances activation of AhR and subsequent "glycolysis-lactate-STAT3″ and TIP60 signals-mediated Treg differentiation.
Collapse
Affiliation(s)
- Shukun Liu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Wenxin Yan
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Qi Lv
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ling Yang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yumeng Miao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yuxiao Hu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
48
|
Liu R, Liu Z, Chen H, He S, Wang S, Dai J, Li X. Ginkgolide K delays the progression of osteoarthritis by regulating YAP to promote the formation of cartilage extracellular matrix. Phytother Res 2023; 37:5205-5222. [PMID: 37527970 DOI: 10.1002/ptr.7953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/12/2023] [Accepted: 07/02/2023] [Indexed: 08/03/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease characterized by cartilage wear and degradation. Ginkgolide K (GK) is a natural compound extracted from Ginkgo biloba leaves and possesses anti-inflammatory and anti-apoptotic effects. We found that the biological characteristics of GK were highly consistent with those of OA medications. This study aimed to determine and verify the therapeutic effect of GK on OA and mechanism of its therapeutic effect. For the in vivo experiment, OA rats were regularly injected in the articular cavity with GK, and the curative effects were observed after 4 and 8 weeks. For the in vitro experiment, we treated OA chondrocytes with different concentrations of GK and then detected the related indices of OA. Through the in vivo and in vitro experiments, we found that GK could promote the production of major components of the cartilage extracellular matrix. Transcriptome sequencing revealed that GK may activate hypoxia-inducible factor 1 alpha via the hypoxia signaling pathway, which, in turn, activates yes-associated protein and inhibits apoptosis of OA chondrocytes. GK has a therapeutic effect on OA and, therefore, has the potential to be developed into a new drug for OA treatment.
Collapse
Affiliation(s)
- Ruizhou Liu
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| | | | - Hui Chen
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| | - Shiping He
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| | - Shihan Wang
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| | - Jihang Dai
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| | - Xiaolei Li
- Clinical Medical Collage of Yangzhou University, North Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
49
|
Ye Z, Wei Y, Jiang L, Zhang Y. Oxygenolytic sulfoquinovose degradation by an iron-dependent alkanesulfonate dioxygenase. iScience 2023; 26:107803. [PMID: 37731605 PMCID: PMC10507154 DOI: 10.1016/j.isci.2023.107803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/05/2023] [Accepted: 08/29/2023] [Indexed: 09/22/2023] Open
Abstract
Sulfoquinovose (6-deoxy-6-sulfo-D-glucose, SQ), the polar head group of sulfolipids in plants, is abundant in nature. Many bacteria degrade SQ through pathways termed sulfoglycolysis producing C3 or C2 sulfonates, while certain bacteria degrade SQ through direct oxygenolytic cleavage of the SQ C-S bond, catalyzed by a flavin-dependent alkanesulfonate monooxygenase (sulfo-ASMO pathway). Here we report bioinformatics and biochemical studies revealing an alternative mechanism for oxygenolytic cleavage of the SQ C-S bond, catalyzed by an iron and α-ketoglutarate-dependent alkanesulfonate dioxygenase (SqoD, sulfo-ASDO pathway). In both the ASMO and ASDO pathways, the product 6-dehydroglucose is reduced to glucose by NAD(P)H-dependent SquF. Marinomonas ushuaiensis, a marine bacterium, which harbors the sulfo-ASDO gene cluster is shown utilizing SQ as a carbon source for growth, accompanied by increased transcription of SqoD. The sulfo-ASDO pathway highlights the range of microbial strategies for degradation of this ubiquitous sulfo-sugar, with potential implications for sulfur recycling in different biological environments.
Collapse
Affiliation(s)
- Zonghua Ye
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Department of Chemistry, Tianjin University, Tianjin 300072, P.R.China
| | - Yifeng Wei
- Singapore Institute of Food and Biotechnology Innovation, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Li Jiang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Department of Chemistry, Tianjin University, Tianjin 300072, P.R.China
| | - Yan Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Department of Chemistry, Tianjin University, Tianjin 300072, P.R.China
| |
Collapse
|
50
|
Abdel Rahman DE, Fouad MA, Mohammed ER, El-Zoheiry HH, Abdelrasheed Allam H. Novel VEGFR-2 inhibitors as antiangiogenic and apoptotic agents via paracrine and autocrine cascades: Design, synthesis, and biological evaluation. Bioorg Chem 2023; 139:106678. [PMID: 37354661 DOI: 10.1016/j.bioorg.2023.106678] [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: 04/18/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
Appertaining to its paracrine and autocrine signaling loops, VEGFR-2 succeeded in grabbing attention as one of the leading targets in cancer treatment. Based on the foregoing and our comprehensive studies regarding pharmacophoric features and activity of sorafenib, novel phenylpyridazinone based VEGFR-2 inhibitors 4, 6a-e, 7a,b, 9a,b, 12a-c, 13a,b, 14a,b, 15a,b, and 17a-d were optimized. An assortment of biological assays was conducted to assess the antiangiogenic and apoptotic activities of the synthesized derivatives. In vitro VEGFR-2 kinase assay verified the inhibitory activity of the synthesized derivatives with IC50 values from 49.1 to 418.0 nM relative to the reference drug sorafenib (IC50 = 81.8 nM). Antiproliferative activity against HUVECs revealed that compounds 2-{2-[2-(6-oxo-3-phenylpyridazin-1(6H)-yl)acetyl]hydrazineyl}-N-(p-tolyl)acetamide (12c) and 2-[(5-mercapto-4-methyl-4H-1,2,4-triazol-3-yl)methyl]-6-phenylpyridazin-3(2H)-one (13a) possessed superior activity (IC50 values = 11.5 and 12.3 nM, respectively) in comparison to sorafenib (IC50 = 23.2 nM). For the purpose of appraising their antiproliferative effect, derivatives 12c and 13a were exposed to cell cycle analysis, apoptotic, cell invasion and migration assays in addition to determination of VEGFR-2 in protein level. Moreover, cytotoxicity as well as selectivity index against WI-38 cell line was measured to examine safety of derivatives 12c and 13a. After that, molecular docking study was executed on the top five compounds in the in vitro VEGFR-2 kinase assay 6d, 12c, 13a, 14a and 17c to get a deep perception on binding mode of the synthesized compounds and correlate the design strategy with biological results. Finally, physicochemical, pharmacokinetic properties, and drug-likeness studies were performed on the top five derivative in in vitro VEGFR-2 kinase assay.
Collapse
Affiliation(s)
- Doaa E Abdel Rahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Marwa A Fouad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt; Pharmaceutical Chemistry Department, School of Pharmacy, New Giza University, Newgiza, km 22 Cairo-Alexandria Desert Road, Cairo, Egypt
| | - Eman R Mohammed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| | - Haidy H El-Zoheiry
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt.
| | - Heba Abdelrasheed Allam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo 11562, Egypt
| |
Collapse
|