1
|
Ashraf A, Zechmann B, Bruce ED. Hypoxia-inducible factor 1α modulates acrolein-induced cellular damage in bronchial epithelial cells. Toxicology 2025; 515:154158. [PMID: 40252947 DOI: 10.1016/j.tox.2025.154158] [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: 02/18/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Acrolein, a highly reactive α,β-unsaturated aldehyde, is a widespread environmental pollutant. It is generated during the incomplete combustion of materials such as tobacco smoke, petrol, coal, forest fires, and plastics, as well as from the overheating of frying oils. Acrolein is known to induce cellular damage and oxidative stress. This study investigates the critical role of hypoxia-inducible factor 1α (HIF-1α), which is a transcription factor required to regulate cell survival and angiogenesis, in protecting bronchial epithelial cells from acrolein-induced cytotoxicity and DNA damage under normoxic and hypoxic conditions. To our knowledge, no prior study has comprehensively evaluated the effects of HIF-1α on cellular responses to acrolein under normoxic and hypoxic conditions in vitro. Therefore, the goal of this study was to explore how silencing HIF-1α influences cellular responses to acrolein, and our study focused on changes in cytotoxicity, metabolic activity, DNA damage, and oxidative stress using the BEAS-2B cell line. We observed enhanced cell damage and reduced viability in cells exposed to acrolein when silenced with HIF-1α, particularly in hypoxic environments. While results indicate that silencing HIF-1α significantly increases cytotoxicity and DNA damage under hypoxia compared to normoxic conditions, oxidative stress indicator levels did not rise noticeably under hypoxia following HIF-1α silencing. This research warrants further investigation to indicate the importance of HIF-1α in adapting to environmental and hypoxic stressors, which are commonly found in chronic lung diseases and ischemic conditions.
Collapse
Affiliation(s)
- Asha Ashraf
- Department of Environmental Science, Baylor University, 101 Bagby Ave, Waco, TX 76706, USA
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, TX, USA
| | - Erica D Bruce
- Department of Environmental Science, Baylor University, 101 Bagby Ave, Waco, TX 76706, USA.
| |
Collapse
|
2
|
Wu F, Deng Y, Sokolov EP, Falfushynska H, Glänzer A, Xie L, Sokolova IM. Nanopollutants (nZnO) amplify hypoxia-induced cellular stress in a keystone marine bivalve, Mytilus edulis. ENVIRONMENTAL RESEARCH 2025; 274:121346. [PMID: 40058547 DOI: 10.1016/j.envres.2025.121346] [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: 01/05/2025] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Zinc oxide nanoparticles (nZnO) are increasingly utilized in industrial, medical, and personal care products, particularly as the main ingredient in sunscreens, raising concerns about their environmental impact, especially in coastal ecosystems. The Baltic Sea, experiencing severe eutrophication, faces persistent hypoxia due to excessive nutrient runoff and limited water exchange. Simultaneously, coastal pollution from industrial and urban activities introduces nZnO, a highly biotoxic nanopollutant. The combined effects of hypoxia and nZnO contamination may amplify environmental stress, yet their interactions remain insufficiently studied. This study investigates the combined effects of nZnO exposure and fluctuating dissolved oxygen regimes (specifically short- and long-term hypoxia and subsequent reoxygenation) on Mytilus edulis, a sentinel species in these ecosystems. By assessing a range of cellular and molecular markers, including oxidative stress, oxygen sensing, protein quality control, stress response, apoptosis, and inflammation, we show that nZnO exacerbates hypoxia-induced oxidative stress, delaying redox recovery and prolonging oxidative damage during reoxygenation. Specifically, nZnO exposure maintains elevated LPO and PC levels after reoxygenation, indicating prolonged oxidative imbalance. While M. edulis typically recovers from hypoxia-induced stress, nZnO disrupts this process by impairing antioxidant defenses, prolonging HIF-1α activation, and dysregulating p53, JNK, and p38 expression, thereby interfering with normal hypoxia-reoxygenation response. Additionally, nZnO alters HSP70, Lon protease, and caspase-3 regulation, disrupting protein-folding and apoptotic pathways. These findings suggest a synergistic interaction between nZnO and hypoxia, heightening the organism's vulnerability to environmental stress and suggesting risks for marine organisms in nanoparticle-polluted, hypoxia-prone coastal regions.
Collapse
Affiliation(s)
- Fangli Wu
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Minjiang University, Fuzhou, China
| | - Yuqing Deng
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China
| | - Eugene P Sokolov
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Halina Falfushynska
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Faculty of Economics, Anhalt University of Applied Sciences, 06406, Köthen, Germany; ENERTRAG SE, Gut Dauerthal, Dauerthal, 17291, Germany
| | - Aneka Glänzer
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Lingtian Xie
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China
| | - Inna M Sokolova
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.
| |
Collapse
|
3
|
Li J, Li Z, Li X, Li Z, Song Y, Yuan L, Wang Y, Yan R, Lai F, Wang J, Xiao W. MYLIP attenuates hypoxia tolerance by inducing K27-linked polyubiquitination and subsequent proteasomal degradation of HIF-α. Commun Biol 2025; 8:774. [PMID: 40399570 DOI: 10.1038/s42003-025-08200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
Hypoxia tolerance is mainly controlled by the hypoxia signaling pathway and HIF-1α/2α serve as master regulators in this pathway. Here we identify MYLIP, an E3 ubiquitin ligase thought to specifically target lipoprotein receptors, as a downstream target of HIF-2α and a negative regulator of both HIF-1α and HIF-2α. MYLIP interacts with HIF-1α/2α and catalyzes K27-linked polyubiquitination at lysine 118/442 (HIF-1α) or lysine 117 (HIF-2α). This modification induces proteasomal degradation of HIF-1α, resulting in inhibition of hypoxia signaling. Furthermore, Mylip-deficient bluntsnout bream, zebrafish and mice are more tolerant to hypoxia. These findings reveal a role for MYLIP in regulating hypoxia signaling and identify a target for the development of fish strains with high hypoxia tolerance for the benefit of the aquaculture industry.
Collapse
Affiliation(s)
- Jun Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Zhi Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Xiong Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Ziyi Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Yanan Song
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Le Yuan
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Yanyi Wang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Runkun Yan
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Fuxiang Lai
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China
| | - Jing Wang
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China.
| | - Wuhan Xiao
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences; Hubei Hongshan Laboratory, Wuhan, 430072, P. R. China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.
- The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, P. R. China.
| |
Collapse
|
4
|
De Domenico S, La Banca V, D'Amico S, Nicolai S, Peschiaroli A. Defining the transcriptional routes controlling lncRNA NEAT1 expression: implications in cellular stress response, inflammation, and differentiation. Discov Oncol 2025; 16:768. [PMID: 40369379 PMCID: PMC12078918 DOI: 10.1007/s12672-025-02510-6] [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: 01/29/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
NEAT1 (Nuclear Enriched Abundant Transcript 1) is a long non-coding RNA playing a critical role in both physiological and pathological settings by directly modulating a variety of biological events, including transcriptional regulation, RNA processing, and chromatin remodeling. Multiple evidence demonstrated that different transcription factors and signaling pathways modulate biological processes by tightly regulating NEAT1 expression. These regulatory mechanisms act at different levels, allowing cells to rapidly modulate NEAT1 expression and dynamically respond to sudden changes in cellular conditions. In this review, we summarize and discuss the transcriptional routes controlling NEAT1 expression, emphasizing recent evidence showing the pivotal role of NEAT1 in regulating important biological processes, such as cellular stress response, inflammation, and cell differentiation.
Collapse
Affiliation(s)
- Sara De Domenico
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Veronica La Banca
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Silvia D'Amico
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Sara Nicolai
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| | - Angelo Peschiaroli
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133, Rome, Italy.
| |
Collapse
|
5
|
Cinkornpumin JK, Kwon SY, Prandstetter AM, Maxian T, Sirois J, Goldberg J, Zhang J, Saini D, Dasgupta P, Jeyarajah MJ, Renaud SJ, Paul S, Haider S, Pastor WA. Hypoxia and loss of GCM1 expression prevent differentiation and contact inhibition in human trophoblast stem cells. Stem Cell Reports 2025; 20:102481. [PMID: 40280139 DOI: 10.1016/j.stemcr.2025.102481] [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: 10/21/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
During the first stages of embryonic development, the placenta develops under very low oxygen tension (∼1%-2% O2), so we sought to determine the regulatory role of oxygen in human trophoblast stem cells (hTSCs). We find that low oxygen promotes hTSC self-renewal but inhibits differentiation to syncytiotrophoblast (STB) and extravillous trophoblast (EVT). The transcription factor GCM1 (glial cell missing transcription factor 1) is downregulated in low oxygen, and concordantly, there is substantial reduction of GCM1-regulated genes in hypoxic conditions. Knockout of GCM1 in hTSC likewise impaired EVT and STB formation. Treatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor reported to reduce GCM1 protein levels likewise counteracts spontaneous or directed differentiation. Additionally, chromatin immunoprecipitation of GCM1 showed binding near key genes upregulated upon differentiation including the contact inhibition factor CDKN1C. Loss of GCM1 resulted in downregulation of CDKN1C and corresponding loss of contact inhibition, implicating GCM1 in regulation of this critical process.
Collapse
Affiliation(s)
| | - Sin Young Kwon
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anna-Maria Prandstetter
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Jacinthe Sirois
- Department of Biochemistry, McGill University, Montreal, QC, Canada; The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - James Goldberg
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Joy Zhang
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Deepak Saini
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, Kansas, USA
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, Kansas, USA; Institute for Reproduction and Developmental Sciences, University of Kansas, Kansas City, Kansas, USA; Department of Obstetrics and Gynecology, University of Kansas, Kansas City, Kansas, USA
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - William A Pastor
- Department of Biochemistry, McGill University, Montreal, QC, Canada; The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Kuzuoglu-Ozturk D, Nguyen HG, Xue L, Figueredo E, Subramanyam V, Liu I, Bonitto K, Noronha A, Dabrowska A, Cowan JE, Oses-Prieto JA, Burlingame AL, Worland ST, Carroll PR, Ruggero D. Small-molecule RNA therapeutics to target prostate cancer. Cancer Cell 2025; 43:841-855.e8. [PMID: 40118049 DOI: 10.1016/j.ccell.2025.02.027] [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: 05/05/2024] [Revised: 11/20/2024] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Tuning protein expression by targeting RNA structure using small molecules is an unexplored avenue for cancer treatment. To understand whether this vulnerability could be therapeutically targeted in the most lethal form of prostate cancer, castration-resistant prostate cancer (CRPC), we use a clinical small molecule, zotatifin, that targets the RNA helicase and translation factor eukaryotic initiation factor 4A (eIF4A). Zotatifin represses tumorigenesis in patient-derived and xenograft models and prolonged survival in vivo alongside hormone therapy. Genome-wide transcriptome, translatome, and proteomic analysis reveals two important translational targets: androgen receptor (AR), a key oncogene in CRPC, and hypoxia-inducible factor 1A (HIF1A), an essential cancer modulator in hypoxia. We solve the structure of the 5' UTRs of these oncogenic mRNAs and strikingly observe complex structural remodeling of these select mRNAs by this small molecule. Remarkably, tumors treated with zotatifin become more sensitive to anti-androgen therapy and radiotherapy. Therefore, "translatome therapy" provides additional strategies to treat the deadliest cancers.
Collapse
MESH Headings
- Male
- Humans
- Animals
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Mice
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/pathology
- Xenograft Model Antitumor Assays
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Eukaryotic Initiation Factor-4A/genetics
- Eukaryotic Initiation Factor-4A/antagonists & inhibitors
- Eukaryotic Initiation Factor-4A/metabolism
- Thiohydantoins/pharmacology
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- 5' Untranslated Regions
- RNA, Messenger/genetics
- RNA, Messenger/chemistry
- RNA, Messenger/metabolism
- Prostatic Neoplasms/genetics
Collapse
Affiliation(s)
- Duygu Kuzuoglu-Ozturk
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Hao G Nguyen
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Lingru Xue
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Emma Figueredo
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Vishvak Subramanyam
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Isabelle Liu
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Kenya Bonitto
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Tetrad Graduate Program, University of California, San Francisco, CA, USA
| | - Ashish Noronha
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Adrianna Dabrowska
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Janet E Cowan
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | | | - Peter R Carroll
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
7
|
Ito-Harashima S, Miura N. Compartmentation of multiple metabolic enzymes and their preparation in vitro and in cellulo. Biochim Biophys Acta Gen Subj 2025; 1869:130787. [PMID: 40058614 DOI: 10.1016/j.bbagen.2025.130787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Compartmentalization of multiple enzymes in cellulo and in vitro is a means of controlling the cascade reaction of metabolic enzymes. The compartmentation of enzymes through liquid-liquid phase separation may facilitate the reversible control of biocatalytic cascade reactions, thereby reducing the transcriptional and translational burden. This has attracted attention as a potential application in bioproduction. Recent research has demonstrated the existence and regulatory mechanisms of various enzyme compartments within cells. Mounting evidence suggests that enzyme compartmentation allows in vitro and in vivo regulation of cellular metabolism. However, the comprehensive regulatory mechanisms of enzyme condensates in cells and ideal organization of cellular systems remain unknown. This review provides an overview of the recent progress in multiple enzyme compartmentation in cells and summarizes strategies to reconstruct multiple enzyme assemblies in vitro and in cellulo. By examining parallel examples, we have evaluated the consensus and future perspectives of enzyme condensation.
Collapse
Affiliation(s)
- Sayoko Ito-Harashima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 599-8531, Japan
| | - Natsuko Miura
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai 599-8531, Japan.
| |
Collapse
|
8
|
Pan D, Chen P, Zhang H, Zhao Q, Fang W, Ji S, Chen T. Mitochondrial quality control: A promising target of traditional Chinese medicine in the treatment of cardiovascular disease. Pharmacol Res 2025; 215:107712. [PMID: 40154932 DOI: 10.1016/j.phrs.2025.107712] [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: 01/17/2025] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cardiovascular disease remains the leading cause of death globally, and drugs for new targets are urgently needed. Mitochondria are the primary sources of cellular energy, play crucial roles in regulating cellular homeostasis, and are tightly associated with pathological processes in cardiovascular disease. In response to physiological signals and external stimuli in cardiovascular disease, mitochondrial quality control, which mainly includes mitophagy, mitochondrial dynamics, and mitochondrial biogenesis, is initiated to meet cellular requirements and maintain cellular homeostasis. Traditional Chinese Medicine (TCM) has been shown to have pharmacological effects on alleviating cardiac injury in various cardiovascular diseases, including myocardial ischemia/reperfusion, myocardial infarction, and heart failure, by regulating mitochondrial quality control. Recently, several molecular mechanisms of TCM in the treatment of cardiovascular disease have been elucidated. However, mitochondrial quality control by TCM for treating cardiovascular disease has not been investigated. In this review, we aim to decipher the pharmacological effects and molecular mechanisms of TCM in regulating mitochondrial quality in various cardiovascular diseases. We also present our perspectives regarding future research in this field.
Collapse
Affiliation(s)
- Deng Pan
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Pengfei Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhao
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Wei Fang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Siyan Ji
- Stomatology Department of Qiqihar Medical College School, Heilongjiang, China
| | - Tielong Chen
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
9
|
Sánchez-Bayuela T, Peral-Rodrigo M, Parra-Izquierdo I, López J, Gómez C, Montero O, Pérez-Riesgo E, San Román JA, Butcher JT, Sánchez Crespo M, García-Rodríguez C. Inflammation via JAK-STAT/HIF-1α Drives Metabolic Changes in Pentose Phosphate Pathway and Glycolysis That Support Aortic Valve Cell Calcification. Arterioscler Thromb Vasc Biol 2025. [PMID: 40308196 DOI: 10.1161/atvbaha.124.322375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Inflammation and metabolic reprogramming are hallmarks of cardiovascular disorders, wherein myocardiocytes switch from fatty acids to glucose to yield energy. This has also been found in the myocardium of patients with calcific aortic valve disease, a prevalent disease exhibiting features of inflammatory disease that lacks pharmacological treatments. Therefore, we posited that the analysis of proinflammatory and metabolic mechanisms might give cues to disclose therapeutic targets. METHODS The metabolic analysis of aortic valve interstitial cells (VIC) explanted from human valves was performed by Seahorse real-time cell metabolic analysis, fluxomics using ultra-performance liquid chromatography/mass spectrometry, quantitative polymerase chain reaction, metabolite quantitation, and loss-of-function experiments with gene silencing and pharmacological approaches. Findings were validated in quiescent VIC, 3-dimensional porcine VIC-valve endothelial cell cocultures, as well as in valve leaflets and VIC from human patients. RESULTS The hyperglycolytic program present in calcific aortic valve disease was replicated in control/nonstenotic VIC by cytokine exposure and enhanced by pathogen-associated molecular patterns. Inflammatory stimuli increased fluxes in glycolysis, tricarboxylic acid cycle, and the pentose phosphate pathway. Inflamed VIC exhibited increased glycolytic ATP production and lactate secretion, as well as changes in redox state and metabolic gene profile, that is, upregulation of glycolytic enzyme expression and downregulation of G6PD (glucose-6-phosphate dehydrogenase), the rate-limiting enzyme of the oxidative phase of pentose phosphate pathway. Notably, these alterations were replicated in quiescent VIC and 3-dimensional VIC-valve endothelial cell cocultures and are observed in diseased valves from patients. Strikingly, metabolic rewiring in control VIC was required for inflammation-triggered calcification and differentiation. A Food and Drug Administration-approved JAK (Janus kinase) inhibitor blunted these changes, whose major drivers are the JAK-STAT system, HIF (hypoxia-inducible factor)-1α, and NF-κB (nuclear factor-κB). CONCLUSIONS Inflammation reprograms VIC metabolism to support calcification by downregulating oxidative phase of pentose phosphate pathway and enhancing glycolytic flux and oxidative stress. These findings parallel the metabolic profile of stenotic VIC and provide novel therapeutic clues.
Collapse
Affiliation(s)
- Tania Sánchez-Bayuela
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Mirian Peral-Rodrigo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Iván Parra-Izquierdo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Javier López
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain (J.L., J.A.S.R.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV) (J.L., J.A.S.R., C.G.-R.)
| | - Cristina Gómez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Olimpio Montero
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Enrique Pérez-Riesgo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - J Alberto San Román
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain (J.L., J.A.S.R.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV) (J.L., J.A.S.R., C.G.-R.)
| | - Jonathan T Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY (J.T.B.)
| | - Mariano Sánchez Crespo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
| | - Carmen García-Rodríguez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Universidad de Valladolid y Consejo Superior de Investigaciones Científicas (CSIC), Spain (T.S.-B., M.P.-R., I.P.-I., C.G., O.M., E.P.-R., M.S.C., C.G.-R.)
- CIBER de Enfermedades Cardiovasculares (CIBERCV) (J.L., J.A.S.R., C.G.-R.)
| |
Collapse
|
10
|
Lynch-Miller M, Lockow S, Dümmer K, Henneck T, Olmer R, Jaboreck MC, Mergani AO, Wandrey M, Branitzki-Heinemann K, Brogden G, Naim HY, Martin U, Schulz C, Talbot SR, Meurer M, Baumgärtner W, von Köckritz-Blickwede M. Characterization of 3D human pulmonary epithelial model morphology and oxygen status under normoxia and hypoxia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119980. [PMID: 40315920 DOI: 10.1016/j.bbamcr.2025.119980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/05/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Infection generates localized hypoxia in affected tissue, inducing cellular survival responses and modulating inflammatory processes. Consideration of oxygen status as a parameter in in vitro infection research is therefore vital to the generation of physiologically relevant data within the 3R context. In this study, we characterize the culture morphology and oxygenation of liquid-liquid interface (LLI) permanent bronchial epithelial (Calu-3), classical air-liquid interface (cALI) Calu-3, and cALI human primary bronchial epithelial cell (hBEC) models under the normoxic conditions within standard incubators, commonly employed in in vitro work. We compare the normoxic state of these models to their hypoxic state to assess changes in the airway epithelial environment in response to oxygen deprivation, and the extent to which select hypoxia responses can be observed at the molecular level. Additional juxtapositions are drawn between Calu-3 LLI and cALI models and Calu-3 conventional monolayer (CM) and inverted air-liquid interface (iALI) models, due to their relevance for basic and specialized research, respectively. Epithelial complexity was observed to vary amongst the filter-based models, and all models were found to exhibit characteristic extracellular oxygen depletion patterns under normoxia. Importantly, the extracellular oxygen contents of Calu-3 LLI, cALI, and CM models significantly decreased during normoxic incubation. Specific hypoxia responses through stabilization of HIF-1α, HIF-2α, and/or HIF-3α and alteration of ACE2 protein levels differed in response to both culture format and cell type. Therefore, while all models examined provide valuable opportunities for in vitro exploration, variation in their morphological, physiological, and molecular characteristics necessitates careful consideration during experimental design.
Collapse
Affiliation(s)
- Maura Lynch-Miller
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Sandra Lockow
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katrin Dümmer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Timo Henneck
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Mark-Christian Jaboreck
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - AhmedElmontaser O Mergani
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Madita Wandrey
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Katja Branitzki-Heinemann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Graham Brogden
- Institute of Experimental Virology, TWINCORE, Center for Experimental and Clinical Infection Research Hannover, Hannover, Germany
| | - Hassan Y Naim
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Steven R Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Marita Meurer
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine, Foundation, Hannover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Foundation, Hannover, Germany.
| |
Collapse
|
11
|
Tu T, Peng Z, Zhang L, Yang J, Guo K, Tang X, Ye J, Zhang F, Huang A, Yu J, Huang C, Zhang H, Wang D, Peng J, Jiang Y. Neuroinflammation and hypoxia promote astrocyte phenotypic transformation and propel neurovascular dysfunction in brain arteriovenous malformation. J Neuroinflammation 2025; 22:124. [PMID: 40301964 PMCID: PMC12042495 DOI: 10.1186/s12974-025-03442-2] [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: 01/31/2025] [Accepted: 04/11/2025] [Indexed: 05/01/2025] Open
Abstract
Brain arteriovenous malformation (BAVM) is a complex cerebrovascular disease characterized by an abnormal high-flow vascular network, which increases the risk of hemorrhage, particularly in young individuals. Endothelial dysfunction has traditionally been considered the primary cause, while the contributions of the microenvironment and glial cells have not been fully explored. Astrocytes, as a key component of the central nervous system, play a crucial role in regulating neurovascular function, maintaining the integrity of the blood-brain barrier, and ensuring neural homeostasis. However, under the pathological conditions of BAVM, the phenotypic changes in astrocytes and their role in disease progression remain poorly understood. In our study, we emphasized the critical role of neuroinflammation and hypoxia in the progression of BAVM within its pathological microenvironment. Specifically, reactive astrocytes undergo phenotypic changes under these pathological conditions, significantly promoting vascular instability. Moreover, nitric oxide (NO) produced by BAVM endothelial cells activates signaling pathways that stabilize HIF-1α in astrocytes, initiating a "hypoxic" gene program under normoxic conditions. Furthermore, we discovered that COX-2, a direct target gene of HIF-1α, is upregulated in the BAVM microenvironment. These changes promoted endothelial dysfunction and vascular fragility, creating a vicious cycle that exacerbates hemorrhage risk. The application of COX-2 inhibitors significantly reduced neuroinflammation, stabilized blood vessels, and decreased hemorrhage risk. Our findings highlighted the crucial interaction between the BAVM microenvironment and astrocytes in driving disease progression, suggesting that COX-2 could be a potential therapeutic target for stabilizing BAVM vessels and reducing hemorrhagic events.
Collapse
Affiliation(s)
- Tianqi Tu
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- Department of Neurosurgery and Shandong Key Laboratory of Brain Health and Function Remodeling, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Zhenghong Peng
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lihan Zhang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jieru Yang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Kecheng Guo
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xiaogang Tang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiasen Ye
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Fan Zhang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - An Huang
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiaxing Yu
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, China
| | - Changren Huang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing, China
| | - Donghai Wang
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China.
- Department of Neurosurgery and Shandong Key Laboratory of Brain Health and Function Remodeling, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
- Department of Neurosurgery, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou, China), Cheeloo Hospital of Shandong University, Jinan, Shandong, China.
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China.
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China.
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital Southwest Medical University, No. 25 of Taiping Street, Luzhou, 646000, Sichuan, China.
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, China.
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China.
- Institute of Brain Science, Southwest Medical University, Luzhou, China.
| |
Collapse
|
12
|
Ye Y, Wang Y, Xu Q, Liu J, Yang Z, Wuren T, Ge RL. In vitro study: HIF-1α-dependent glycolysis enhances NETosis in hypoxic conditions. Front Immunol 2025; 16:1583587. [PMID: 40356921 PMCID: PMC12066692 DOI: 10.3389/fimmu.2025.1583587] [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: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Hypoxia plays a pivotal role in modulating immune responses, especially in neutrophils, which are essential components of the innate immune system. Hypoxia-inducible factor (HIF)-1α, a key transcription factor in hypoxic adaptation, regulates cellular metabolism and inflammatory responses. However, the impact of HIF-1α-dependent glycolysis on the formation of neutrophil extracellular traps (known as NETosis) under hypoxic conditions remains unclear. Methods We employed two established neutrophil models, neutrophils isolated from human whole blood and DMSO-induced dHL-60 cells, to explore the role of HIF-1α in regulating glycolysis and its influence on NETosis under hypoxic conditions. We utilized western blotting, immunofluorescence staining, ELISA, and flow cytometry to evaluate the expression of key glycolytic enzymes and NETosis markers under hypoxia. Additionally, the effects of inhibiting HIF-1α with LW6 and blocking the glycolytic pathway with Bay-876 were investigated. Results HIF-1α-dependent glycolysis, through the upregulation of key glycolytic enzymes, significantly enhances NETosis under hypoxic conditions. Pharmacological inhibition of HIF-1α with LW6 and glycolytic blockade with Bay-876 markedly reduced NETosis, underscoring the crucial role of metabolic reprogramming in neutrophil function during hypoxia. Conclusion This study provides novel insights into the interplay between metabolic reprogramming and NETosis in response to hypoxic stress. We identify HIF-1α-dependent glycolysis as a key driver of NETs formation, advancing our understanding of the mechanisms underlying hypoxia-related inflammatory diseases. These findings also suggest that targeting metabolic pathways may offer potential therapeutic strategies for modulating immune responses in hypoxia-associated disorders.
Collapse
Affiliation(s)
- Yi Ye
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
| | - Yanjun Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Department of Geriatrics, Qinghai University Affiliated Hospital, Xining, China
| | - Qiying Xu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Department of Gynecology, Qinghai University Affiliated Hospital, Xining, China
| | - Juanli Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Department of Critical Care Medicine, Qinghai Provincial People's Hospital, Xining, China
| | - Ziqi Yang
- Medical College of Qinghai University, Xining, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
| |
Collapse
|
13
|
Yuan G, Wang J, Qiu S, Zhu Y, Cheng Q, Li L, Sha J, Yang X, Yuan Y. Improving in vitro induction efficiency of human primordial germ cell-like cells using N2B27 or NAC-based medium. J Biomed Res 2025; 39:1-14. [PMID: 40204653 DOI: 10.7555/jbr.38.20240433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Primordial germ cells (PGCs), the precursors of oocytes or spermatozoa, are highly pluripotent. In recent years, the in vitro induction of human primordial germ cell-like cells (hPGCLCs) has advanced significantly. However, the stability and efficacy of obtaining hPGCLCs in vitro still require further improvement. In the current study, we identified a novel induction system by using Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F-12) as the basal medium supplemented with B27 and N2 (referred to as N2B27) in combination with four cytokines: bone morphogenetic protein 4 (BMP4), stem cell factor (SCF), epidermal growth factor (EGF), and leukemia inhibitory factor (LIF). The hPGCLCs induced under these conditions closely resemble PGCs from 4 to 5-week-old embryos at the transcriptome level. Compared with traditional GK15 (GMEM supplemented with 15% Knockout™ SR)-based induction conditions, the N2B27 system significantly increased the speed and efficacy of hPGCLC induction. RNA sequencing analysis revealed that this improvement resulted from an increased cell capacity to cope with hypoxic stress and avoid apoptosis. The N2B27 medium promoted an increase in mitochondrial activity, enabling cells to better cope with hypoxic stress while also reducing the production of reactive oxygen species. Moreover, by gradient concentration experiments, we demonstrated that addition of the common antioxidant N-acetyl-L-cysteine at an optimized concentration further enhanced the efficiency of PGCLC induction under GK15 conditions. Thus, our study established an optimized induction system that enhances the efficiency of hPGCLC differentiation by improving cellular resilience to hypoxic stress and apoptosis.
Collapse
Affiliation(s)
- Gege Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiachen Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Shuangshuang Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yunfei Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Women and Children's Healthcare Hospital, Nanjing, Jiangsu 211100, China
| | - Laihua Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029 China
| | - Yan Yuan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
14
|
Warden AS, Sharma N, Hutchens S, Liu C, Haggerty NR, Gurol KC, Jursa T, Smith DR, Dayne Mayfield R, Mukhopadhyay S. Elevated brain manganese induces motor disease by upregulating the kynurenine pathway of tryptophan metabolism. Proc Natl Acad Sci U S A 2025; 122:e2423628122. [PMID: 40244671 PMCID: PMC12036984 DOI: 10.1073/pnas.2423628122] [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: 11/13/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
Elevated brain levels of the essential metals manganese (Mn), copper, or iron induce motor disease. However, mechanisms of metal-induced motor disease are unclear and treatments are lacking. Elucidating the mechanisms of Mn-induced motor disease is particularly important because occupational and environmental Mn overexposure is a global public health problem. To address this, here we combined unbiased transcriptomics and metabolomics with functional studies in a mouse model of human environmental Mn exposure. Transcriptomics unexpectedly revealed that Mn exposure up-regulated expression of metabolic pathways in the brain and liver. Notably, genes in the kynurenine pathway of tryptophan metabolism, which produces neuroactive metabolites that impact neurological function, were up-regulated by Mn. Subsequent unbiased metabolomics revealed that Mn treatment altered kynurenine pathway metabolites in the brain and liver. Functional experiments then demonstrated that pharmacological inhibition of the first and rate-limiting step of the kynurenine pathway fully rescued Mn-induced motor deficits. Finally, elevated Mn directly activates hypoxia-inducible factor (HIF) transcription factors, and additional mechanistic assays identified a role for HIF1, but not HIF2, in regulating expression of hepatic kynurenine pathway genes under physiological or Mn exposure conditions, suggesting that Mn-induced HIF1 activation may contribute to the dysregulation of the kynurenine pathway in Mn toxicity. These findings (1) identify the upregulation of the kynurenine pathway by elevated Mn as a fundamental mechanism of Mn-induced motor deficits; (2) provide a pharmacological approach to treat Mn-induced motor disease; and (3) should broadly advance understanding of the general principles underlying neuromotor deficits caused by metal toxicity.
Collapse
Affiliation(s)
- Anna S. Warden
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Nishant Sharma
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Steven Hutchens
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Chunyi Liu
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Noah R. Haggerty
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Kerem C. Gurol
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Thomas Jursa
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Donald R. Smith
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA95064
| | - Roy Dayne Mayfield
- Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX78712
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
15
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
16
|
Alonaizan R, Purnama U, Malandraki-Miller S, Gunadasa-Rohling M, Lewis A, Smart N, Carr C. MicroRNA-210 Enhances Cell Survival and Paracrine Potential for Cardiac Cell Therapy While Targeting Mitophagy. J Funct Biomater 2025; 16:147. [PMID: 40278255 PMCID: PMC12028018 DOI: 10.3390/jfb16040147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The therapeutic potential of presumed cardiac progenitor cells (CPCs) in heart regeneration has garnered significant interest, yet clinical trials have revealed limited efficacy due to challenges in cell survival, retention, and expansion. Priming CPCs to survive the hostile hypoxic environment may be key to enhancing their regenerative capacity. We demonstrate that microRNA-210 (miR-210), known for its role in hypoxic adaptation, significantly improves CPC survival by inhibiting apoptosis through the downregulation of Casp8ap2, a ~40% reduction in caspase activity, and a ~90% decrease in DNA fragmentation. Contrary to the expected induction of Bnip3-dependent mitophagy by hypoxia, miR-210 did not upregulate Bnip3, indicating a distinct anti-apoptotic mechanism. Instead, miR-210 reduced markers of mitophagy and increased mitochondrial biogenesis and oxidative metabolism, suggesting a role in metabolic reprogramming. Furthermore, miR-210 enhanced the secretion of paracrine growth factors from CPCs, with a ~1.6-fold increase in the release of stem cell factor and of insulin growth factor 1, which promoted in vitro endothelial cell proliferation and cardiomyocyte survival. These findings elucidate the multifaceted role of miR-210 in CPC biology and its potential to enhance cell-based therapies for myocardial repair by promoting cell survival, metabolic adaptation, and paracrine signalling.
Collapse
Affiliation(s)
- Rita Alonaizan
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
- King Faisal Specialist Hospital & Research Centre, Riyadh 12713, Saudi Arabia
| | - Ujang Purnama
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | | | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Andrew Lewis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| | - Carolyn Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK (C.C.)
| |
Collapse
|
17
|
Liu B, Wang Y, Zhu J, Huang H, Chen R, Yu F, Zhou G, Ba Y. Quercetin Simultaneously Treats Skeletal Fluorosis and Kashin-Beck Disease by Modulating HIF-1 and Ferroptosis Signaling Pathways. Biol Trace Elem Res 2025:10.1007/s12011-025-04604-2. [PMID: 40183961 DOI: 10.1007/s12011-025-04604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
Excessive fluoride exposure can lead to skeletal fluorosis (SF), and selenium deficiency is one of the important pathogenic factors of Kashin-Beck disease (KBD). Although the pathogenic factors of these two diseases vary, there are many similarities in their pathogenic mechanisms on skeletal and articular cartilage lesions. There are currently no specific drugs for either disease, and investigating their shared pathogenic mechanisms may facilitate the development of new drugs for the treatment. This study found through bioinformatics technology that the HIF-1 signaling pathway and ferroptosis pathway might exert significant effects in both SF and KBD. Targeted small molecule drug prediction was conducted for the above two signaling pathways, and quercetin was screened as the best candidate therapeutic drug. Meanwhile, molecular docking and molecular dynamics simulations once again validated our screening results. In summary, quercetin may alleviate the symptoms of SF and KBD by regulating the HIF-1 signaling pathway and the ferroptosis pathway. In other words, it can attain the objective of treating two diseases simultaneously with one drug. This will provide new theoretical references for the treatment of comorbidity.
Collapse
Affiliation(s)
- Bin Liu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Jingyuan Zhu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Hui Huang
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Ruiqin Chen
- Jinshui District Center for Disease Control and Prevention, Zhengzhou, Henan, 450053, P.R. China
| | - Fangfang Yu
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Guoyu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China
| | - Yue Ba
- Department of Occupational and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, Henan, 450001, P.R. China.
| |
Collapse
|
18
|
Sekine K, Tokunaka K, Tomura A, Sugihara H, Saijo Y, Shin Y, Hayashi T, Morita M, Imamura Y. Production of non-triple-helical collagen polypeptides under hypoxia and the implication for tumour. J Biochem 2025; 177:287-295. [PMID: 39756403 DOI: 10.1093/jb/mvae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/18/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025] Open
Abstract
Non-triple-helical collagen polypeptides (NTHs) are alternative gene products lacking the typical collagen triple-helical structure. This study investigated NTH production in tumour cells and tissues. NTH α1(IV) was detected in various human tumour cell lines and extracted from human lung cancer tissues and tumours in mice. NTH production was significantly affected by serum concentration and occurred under hypoxic or hypoxia-mimetic conditions, even with sufficient ascorbic acid. This suggests NTHs are produced under physiological hypoxia, potentially contributing to tumour angiogenesis. NTH production generally coincided with hypoxia-inducible factor-1α (HIF-1α) accumulation, except with cobalt chloride, indicating HIF-1α is not directly involved in NTH α1(IV) production. NTH electrophoretic mobility on SDS-PAGE was higher under hypoxia or deferoxamine treatment, likely due to suppressed lysyl hydroxylase 3 activity. This study demonstrates NTH production in tumour cells and tissues under hypoxia, suggesting their association with tumour angiogenesis and potential as therapeutic targets.
Collapse
Affiliation(s)
- Kosuke Sekine
- Graduate School of Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
| | - Kazuhiro Tokunaka
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., 3-31-12, Shimo, Kita-ku, Tokyo 115-0042, Japan
| | - Arihiro Tomura
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., 3-31-12, Shimo, Kita-ku, Tokyo 115-0042, Japan
| | - Hidemitsu Sugihara
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., 3-31-12, Shimo, Kita-ku, Tokyo 115-0042, Japan
| | - Yuki Saijo
- Graduate School of Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
| | - Yongchol Shin
- Graduate School of Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
| | - Toshihiko Hayashi
- Nippi Research Institute of Biomatrix, 520-11, Kuwabara,Toride, Ibaraki-ken 302-0017, Japan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, P. R. China
| | - Makoto Morita
- Pharmaceutical Research Laboratories, Nippon Kayaku Co., Ltd., 3-31-12, Shimo, Kita-ku, Tokyo 115-0042, Japan
| | - Yasutada Imamura
- Graduate School of Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
- Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakano, Hachioji, Tokyo 192-0015, Japan
| |
Collapse
|
19
|
Kanuri B, Maremanda KP, Chattopadhyay D, Essop MF, Lee MKS, Murphy AJ, Nagareddy PR. Redefining Macrophage Heterogeneity in Atherosclerosis: A Focus on Possible Therapeutic Implications. Compr Physiol 2025; 15:e70008. [PMID: 40108774 DOI: 10.1002/cph4.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/22/2025]
Abstract
Atherosclerosis is a lipid disorder where modified lipids (especially oxidized LDL) induce macrophage foam cell formation in the aorta. Its pathogenesis involves a continuum of persistent inflammation accompanied by dysregulated anti-inflammatory responses. Changes in the immune cell status due to differences in the lesional microenvironment are crucial in terms of plaque development, its progression, and plaque rupture. Ly6Chi monocytes generated through both medullary and extramedullary cascades act as one of the major sources of plaque macrophages and thereby foam cells. Both monocytes and monocyte-derived macrophages also participate in pathological events in atherosclerosis-associated multiple organ systems through inter-organ communications. For years, macrophage phenotypes M1 and M2 have been shown to perpetuate inflammatory and resolution responses; nevertheless, such a dualistic classification is too simplistic and contains severe drawbacks. As the lesion microenvironment is enriched with multiple mediators that possess the ability to activate macrophages to diverse phenotypes, it is obvious that such cells should demonstrate substantial heterogeneity. Considerable research in this regard has indicated the presence of additional macrophage phenotypes that are exclusive to atherosclerotic plaques, namely Mox, M4, Mhem, and M(Hb) type. Furthermore, although the concept of macrophage clusters has come to the fore in recent years with the evolution of high-dimensional techniques, classifications based on such 'OMICS' approaches require extensive functional validation as well as metabolic phenotyping. Bearing this in mind, the current review provides an overview of the status of different macrophage populations and their role during atherosclerosis and also outlines possible therapeutic implications.
Collapse
Affiliation(s)
- Babunageswararao Kanuri
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Krishna P Maremanda
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - Dipanjan Chattopadhyay
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Man Kit Sam Lee
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Section of Cardiovascular Diseases, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, Oklahoma, USA
| |
Collapse
|
20
|
Zhang S, Wang YS, Li Y, To KI, Zhang ET, Jin YH. Annexin A2 binds the 3'-UTR of H2AX mRNA and regulates histone-H2AX-derived hypoxia-inducible factor 1-alpha activation. Cell Signal 2025; 132:111781. [PMID: 40164417 DOI: 10.1016/j.cellsig.2025.111781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Annexin A2 (Anxa2), a multifunctional protein with RNA-binding capabilities, is frequently overexpressed in various tumors, and its expression is highly correlated with malignant progression. In this study, we demonstrate for the first time that Anxa2 was co-expressed with glycolytic genes, suggesting its potential role as a regulator of glycolysis. RNA-protein interaction assay revealed that Anxa2 interacted with 3'-UTR of H2AX mRNA and protected it from miRNA-mediated degradation. Up-regulated Histone-H2AX enhances the expression of glycolytic genes including GLUT1, HK2, PGK1, ENO1, PKM2, GAPDH and LDHA via stabilizing hypoxia-inducible factor 1-alpha (HIF1α), thereby accelerating lactic acid production and secretion. (20S) G-Rh2, a natural compound targeting Anxa2, significantly interfered the Anxa2-H2AX mRNA interaction, and inhibited subsequent glycolysis progression. We propose that Anxa2 acts as a novel regulator in glycolysis via enhancing H2AX expression, and (20S) G-Rh2 may exert its anti-cancer activity by targeting Anxa2-H2AX-HIF1α-glycolysis axis in human hepatoma HepG2 cells.
Collapse
Affiliation(s)
- Shiyin Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yu-Shi Wang
- Department of Criminal Science and Technology, Jilin Police College, Changchun 130117, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Kwang-Il To
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - En-Ting Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
21
|
Zhou EH, Zhou TJ, Wang XT, Zhang JY, Guan J, Yin SK, Huang WJ, Yi HL, Zou JY. Identifying and validating immunological biomarkers in obstructive sleep apnea through bioinformatics analysis. Sci Rep 2025; 15:9746. [PMID: 40118992 PMCID: PMC11928569 DOI: 10.1038/s41598-025-93915-4] [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: 08/15/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
Obstructive sleep apnea (OSA) is a prevalent sleep disorder characterized by disrupted breathing patterns and dysfunctions in multiple organ systems. Although studies support a close correlation between OSA and immune function, the broader implications and specific manifestations remain unclear. Therefore, it is pressingly needed to identify potential immune-related markers and elucidate underlying immunological mechanisms of OSA. OSA-related datasets (GSE38792) and immune-related genes were downloaded from the GEO and ImmPort databases and intersected to obtain differentially expressed immune-related genes (DEIRGs). GO, KEGG, and GSEA were employed to explore the biological functions of DEIRGs. Immune cells and immune regulation were analyzed by CIBERSORT. The ROC curve was constructed to assess the accuracy of each DEIRG. The co-regulatory networks of transcription factors, microRNAs, and drugs were built using the NetworkAnalyst database and visualized by Cytoscape. The levels of DEIRGs in clinical samples were validated by RT-qPCR. GO, KEGG, and GSEA revealed that DEGs were mainly enriched in negative regulation of immune response and antigen processing and presentation in OSA. IL33, IL10RB, ANGPTL1, EIF2AK2, SEM1, IFNA16, SLC40A1, FCER1G, IL1R1, TNFRSF17, and ERAP2 were identified as DEIRGs among 175 differentially expressed genes in OSA. Memory B cells, mast cells resting, and dendritic cells resting were the predominant immune cells related to DEIRGs. The co-regulatory network contained 128 miRNAs, 40 transcription factors, and 172 drugs/compounds. Finally, IL33, EIF2AK2, IL10RB, and ANGPTL1 were also upregulated in clinical OSA samples. The present study identified potential immune-related biomarkers and systematically elucidated underlying immunological mechanisms of OSA. These findings provide novel insights into the diagnosis, mechanism research, and management strategies for future studies.
Collapse
Affiliation(s)
- En-Hui Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Tian-Jiao Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Ting Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yu Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Jian Guan
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Shan-Kai Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Jun Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China.
| | - Hong-Liang Yi
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China.
| | - Jian-Yin Zou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yishan Road, Shanghai, 200233, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
- Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
22
|
Murao M, Fukazawa T, Bhawal UK, Tewari N, Shime N, Hirohashi N, Tanimoto K. Differential Effects of the Prolyl-Hydroxylase Inhibitor on the Cellular Response to Radiation. Int J Mol Sci 2025; 26:2742. [PMID: 40141384 PMCID: PMC11943049 DOI: 10.3390/ijms26062742] [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: 01/16/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The prolyl-hydroxylase inhibitor (PHI), used effectively in several countries for the treatment of renal anemia, activates the multifunctional hypoxia-inducible factors (HIFs). While hypoxic conditions in tumors are known to affect the response to radiation therapy, the effect of PHI on the radiation response of cancer cells has not been determined. Hypoxic pretreatment increased the radiation sensitivity of A549 lung adenocarcinoma cells, whereas hypoxic culture after irradiation decreased the radiation sensitivity of HSC2 oral squamous cell carcinoma cells. Treatment of PC9 lung adenocarcinoma and HSC2 cells with the PHI FG-4592 significantly increased radiation resistance, whereas A549 and TIG3 lung fibroblast cells tended to be sensitized, suggesting cell type-specific differential effects of PHI. Quantitative RT-PCR analyses revealed that the basal and radiation-inducible expressions of DEC2, BAX, and BCL2 may be related to PHI-mediated radiation responses. Knock-down experiments showed that silencing of DEC2 sensitized both A549 and PC9 cells under PHI-treated conditions. On the other hand, silencing of p53, which regulates BAX/BCL2, desensitized A549 cells expressing wild-type p53, but not PC9 cells, with mutant-type p53, to irradiation, regardless of whether PHI was treated or not. Taken together, PHI modifies radiation responses in a cell type-specific manner, possibly through DEC2 signaling.
Collapse
Affiliation(s)
- Masaki Murao
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (M.M.); (N.H.)
- Radiation Disaster Medicine Support Center, Hiroshima University, Hiroshima 734-8553, Japan
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Takahiro Fukazawa
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 734-8553, Japan;
- Division of Medical Research Support, Advanced Research Support Center, Ehime University, Ehime 791-0204, Japan
| | - Ujjal K. Bhawal
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai 600077, India;
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Nitesh Tewari
- Division of Pediatric and Preventive Dentistry, Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Nobuyuki Hirohashi
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (M.M.); (N.H.)
- Radiation Disaster Medicine Support Center, Hiroshima University, Hiroshima 734-8553, Japan
| | - Keiji Tanimoto
- Department of Radiation Disaster Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan; (M.M.); (N.H.)
| |
Collapse
|
23
|
Li L, Hammerlindl H, Shen SQ, Bao F, Hammerlindl S, Altschuler SJ, Wu LF. A phenopushing platform to identify compounds that alleviate acute hypoxic stress by fast-tracking cellular adaptation. Nat Commun 2025; 16:2684. [PMID: 40102413 PMCID: PMC11920246 DOI: 10.1038/s41467-025-57754-1] [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: 06/25/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
Severe acute hypoxic stress is a major contributor to the pathology of human diseases, including ischemic disorders. Current treatments focus on managing consequences of hypoxia, with few addressing cellular adaptation to low-oxygen environments. Here, we investigate whether accelerating hypoxia adaptation could provide a strategy to alleviate acute hypoxic stress. We develop a high-content phenotypic screening platform to identify compounds that fast-track adaptation to hypoxic stress. Our platform captures a high-dimensional phenotypic hypoxia response trajectory consisting of normoxic, acutely stressed, and chronically adapted cell states. Leveraging this trajectory, we identify compounds that phenotypically shift cells from the acutely stressed state towards the adapted state, revealing mTOR/PI3K or BET inhibition as strategies to induce this phenotypic shift. Importantly, our compound hits promote the survival of liver cells exposed to ischemia-like stress, and rescue cardiomyocytes from hypoxic stress. Our "phenopushing" platform offers a general, target-agnostic approach to identify compounds and targets that accelerate cellular adaptation, applicable across various stress conditions.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Heinz Hammerlindl
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Susan Q Shen
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Feng Bao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Sabrina Hammerlindl
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Steven J Altschuler
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
24
|
Arias CF, Acosta FJ, Bertocchini F, Fernández-Arias C. Redefining the role of hypoxia-inducible factors (HIFs) in oxygen homeostasis. Commun Biol 2025; 8:446. [PMID: 40089642 PMCID: PMC11910619 DOI: 10.1038/s42003-025-07896-1] [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: 05/09/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are key regulators of intracellular oxygen homeostasis. The marked increase in HIFs activity in hypoxia as compared to normoxia, together with their transcriptional control of primary metabolic pathways, motivated the widespread view of HIFs as responsible for the cell's metabolic adaptation to hypoxic stress. In this work, we suggest that this prevailing model of HIFs regulation is misleading. We propose an alternative model focused on understanding the dynamics of HIFs' activity within its physiological context. Our model suggests that HIFs would not respond to but rather prevent the onset of hypoxic stress by regulating the traffic of electrons between catabolic substrates and oxygen. The explanatory power of our approach is patent in its interpretation of the Warburg effect, the tendency of tumor cells to favor anaerobic metabolism over respiration, even in fully aerobic conditions. This puzzling behavior is currently considered as an anomalous metabolic deviation. Our model predicts the Warburg effect as the expected homeostatic response of tumor cells to the abnormal increase in metabolic demand that characterizes malignant phenotypes. This alternative perspective prompts a redefinition of HIFs' function and underscores the need to explicitly consider the cell's metabolic activity in understanding its responses to changes in oxygen availability.
Collapse
Affiliation(s)
- Clemente F Arias
- Grupo Interdisciplinar de Sistemas Complejos de Madrid (GISC), 28040, Madrid, Spain.
| | - Francisco J Acosta
- Departamento de Ecología, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | | | - Cristina Fernández-Arias
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| |
Collapse
|
25
|
Jiang M, Huang J, Guo X, Fu W, Peng L, Wang Y, Liu W, Liu J, Zhou L, Xiao Y. HIF-3α/PPAR-γ Regulates Hypoxia Tolerance by Altering Glycolysis and Lipid Synthesis in Blunt Snout Bream ( Megalobrama amblycephala). Int J Mol Sci 2025; 26:2613. [PMID: 40141255 PMCID: PMC11942064 DOI: 10.3390/ijms26062613] [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: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Hypoxic stress causes cell damage and serious diseases in organisms, especially in aquatic animals. It is important to elucidate the changes in metabolic function caused by hypoxia and the mechanisms underlying these changes. This study focuses on the low oxygen tolerance feature of a new blunt snout bream strain (GBSBF1). Our data show that GBSBF1 has a different lipid and carbohydrate metabolism pattern than wild-type bream, with altering glycolysis and lipid synthesis. In GBSBF1, the expression levels of phd2 and vhl genes are significantly decreased, while the activation of HIF-3α protein is observed to have risen significantly. The results indicate that enhanced HIF-3α can positively regulate gpd1ab and gpam through PPAR-γ, which increases glucose metabolism and reduces lipolysis of GBSBF1. This research is beneficial for creating new aquaculture strains with low oxygen tolerance traits.
Collapse
Affiliation(s)
- Minggui Jiang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jing Huang
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Xing Guo
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
| | - Wen Fu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Liangyue Peng
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Wenbin Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Jinhui Liu
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Y.W.); (L.Z.)
| | - Yamei Xiao
- College of Life Sciences, Hunan Normal University, Changsha 410081, China; (M.J.); (J.H.); (X.G.); (W.F.); (L.P.); (W.L.); (J.L.)
- Engineering Research Center of Polyploid Fish Reproduction and Breeding of the State Education Ministry, Changsha 410081, China
| |
Collapse
|
26
|
Qin J, Wang R, Liang W, Man Z, Li W, An Y, Chen H. Adipose-Derived Stem Cell Specific Affinity Peptide-Modified Adipose Decellularized Scaffolds for Promoting Adipogenesis. ACS Biomater Sci Eng 2025; 11:1705-1720. [PMID: 39969077 DOI: 10.1021/acsbiomaterials.4c02161] [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] [Indexed: 02/20/2025]
Abstract
Adipose-derived stem cells (ADSCs) are known to promote angiogenesis and adipogenesis. However, their limited ability to efficiently target and integrate into specific tissues poses a major challenge for ADSC-based therapies. In this study, we identified a seven-amino acid peptide sequence (P7) with high specificity for ADSCs using phage display technology. P7 was then covalently conjugated to decellularized adipose-derived matrix (DAM), creating an "ADSC homing device" designed to recruit ADSCs both in vitro and in vivo. The P7-conjugated DAM significantly enhanced ADSC adhesion and proliferation in vitro. After being implanted into rat subcutaneous tissue, immunofluorescence staining after 14 days revealed that P7-conjugated DAM recruited a greater number of ADSCs, promoting angiogenesis and adipogenesis in the surrounding tissue. Moreover, CD206 immunostaining at 14 days indicated that P7-conjugated DAM facilitated the polarization of macrophages to the M2 phenotype at the implantation site. These findings demonstrate that the P7 peptide has a high affinity for ADSCs, and its conjugation with DAM significantly improves ADSC recruitment in vivo. This approach holds great potential for a wide range of applications in material surface modification.
Collapse
Affiliation(s)
- Jiahang Qin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Ruoxi Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| |
Collapse
|
27
|
Albendea-Gomez T, Mendoza-Tamajon S, Castro-Mecinas R, Escobar B, Ferreira Rocha S, Urra-Balduz S, Nicolas-Avila JA, Oliver E, Villalba-Orero M, Martin-Puig S. Vascular HIF2 Signaling Prevents Cardiomegaly, Alveolar Congestion, and Capillary Remodeling During Chronic Hypoxia. Arterioscler Thromb Vasc Biol 2025; 45:e78-e98. [PMID: 39846162 DOI: 10.1161/atvbaha.124.321780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Hypoxia is associated with the onset of cardiovascular diseases including cardiac hypertrophy and pulmonary hypertension. HIF2 (hypoxia inducible factor 2) signaling in the endothelium mediates pulmonary arterial remodeling and subsequent elevation of the right ventricular systolic pressure during chronic hypoxia. Thus, novel therapeutic opportunities for pulmonary hypertension based on specific HIF2 inhibitors have been proposed. Nevertheless, HIF2 relevance beyond the pulmonary endothelium or in the cardiac adaptation to hypoxia remains elusive. Wt1 (Wilms tumor 1) lineage contributes to the heart and lung vascular compartments, including pericytes, endothelial cells, and smooth muscle cells. METHODS Here, we describe the response to chronic hypoxia of a novel HIF2 mutant mouse model in the Wt1 lineage (Hif2/Wt1 cKO [conditional knockout]), characterizing structural and functional aspects of the heart and lungs by means of classical histology, immunohistochemistry, flow cytometry, echocardiography, and lung ultrasound analysis. RESULTS Hif2/Wt1 cKO is protected against pulmonary remodeling and increased right ventricular systolic pressure induced by hypoxia, but displays alveolar congestion, inflammation, and hemorrhages associated with microvascular instability. Furthermore, lack of HIF2 in the Wt1 lineage leads to cardiomegaly, capillary remodeling, right and left ventricular hypertrophy, systolic dysfunction, and left ventricular dilation, suggesting pulmonary-independent cardiac direct roles of HIF2 in hypoxia. These structural defects are partially restored upon reoxygenation, while cardiac functional parameters remain altered. CONCLUSIONS Our results indicate that cardiopulmonary HIF2 signaling prevents excessive vascular proliferation during chronic hypoxia and define novel protective roles of HIF2 to warrant stable microvasculature and organ function.
Collapse
MESH Headings
- Animals
- Signal Transduction
- Vascular Remodeling
- Hypoxia/metabolism
- Hypoxia/physiopathology
- Hypoxia/complications
- Hypoxia/genetics
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/deficiency
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Disease Models, Animal
- Mice, Knockout
- Ventricular Function, Right
- Hypertrophy, Right Ventricular/prevention & control
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/pathology
- Chronic Disease
- Cardiomegaly/prevention & control
- Cardiomegaly/physiopathology
- Cardiomegaly/metabolism
- Cardiomegaly/genetics
- Cardiomegaly/pathology
- Cardiomegaly/etiology
- Pulmonary Alveoli/blood supply
- Pulmonary Alveoli/metabolism
- Pulmonary Alveoli/pathology
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Capillaries/physiopathology
- Capillaries/metabolism
- Capillaries/pathology
- Ventricular Remodeling
- Male
- Mice
- Mice, Inbred C57BL
- Transcription Factors
Collapse
Affiliation(s)
- Teresa Albendea-Gomez
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- School of Medicine, Universidad Francisco de Vitoria, Madrid, Spain (T.A.-G., S.M.-P.)
| | - Susana Mendoza-Tamajon
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
| | - Rosana Castro-Mecinas
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
| | - Beatriz Escobar
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Mouse Genome Editing Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain (B.E.)
| | - Susana Ferreira Rocha
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
| | - Sonia Urra-Balduz
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
| | - Jose Angel Nicolas-Avila
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Cardiovascular Research Institute & Department of Microbiology and Immunology, University of California San Francisco (J.A.N.-A.)
| | - Eduardo Oliver
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), Madrid, Spain (E.O.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (E.O.)
| | - Maria Villalba-Orero
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- Department of Animal Medicine and Surgery, Universidad Complutense de Madrid, Madrid, Spain (M.V.-O.)
| | - Silvia Martin-Puig
- Metabolic and Immune Diseases Department, Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain (T.A.-G., S.M.-T., R.C.-M., S.U.-B., S.M.-P.)
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Carlos III (CNIC), Madrid, Spain (T.A.-G., S.M.-T., B.E., S.F.R., J.A.N.-A., E.O., M.V.-O., S.M.-P.)
- School of Medicine, Universidad Francisco de Vitoria, Madrid, Spain (T.A.-G., S.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Spain (S.M.-P.)
| |
Collapse
|
28
|
Tharayil JS, Kandettu A, Chakrabarty S. The curious case of mitochondrial sirtuin in rewiring breast cancer metabolism: Mr Hyde or Dr Jekyll? Biochim Biophys Acta Mol Basis Dis 2025; 1871:167691. [PMID: 39864670 DOI: 10.1016/j.bbadis.2025.167691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Mammalian sirtuins are class III histone deacetylases involved in the regulation of multiple biological processes including senescence, DNA repair, apoptosis, proliferation, caloric restriction, and metabolism. Among the mammalian sirtuins, SIRT3, SIRT4, and SIRT5 are localized in the mitochondria and collectively termed the mitochondrial sirtuins. Mitochondrial sirtuins are NAD+-dependent deacetylases that play a central role in cellular metabolism and function as epigenetic regulators by performing post-translational modification of cellular proteins. Several studies have identified the role of mitochondrial sirtuins in age-related pathologies and the rewiring of cancer metabolism. Mitochondrial sirtuins regulate cellular functions by contributing to post-translational modifications, including deacetylation, ADP-ribosylation, demalonylation, and desuccinylation of diverse cellular proteins to maintain cellular homeostasis. Here, we review and discuss the structure and function of the mitochondrial sirtuins and their role as metabolic regulators in breast cancer. Altered breast cancer metabolism may promote tumor progression and has been an essential target for therapy. Further, we discuss the potential role of targeting mitochondrial sirtuin and its impact on breast cancer progression using sirtuin inhibitors and activators as anticancer agents.
Collapse
Affiliation(s)
- Jesline Shaji Tharayil
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Amoolya Kandettu
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
29
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
30
|
Sun XQ, Li X, Li YQ, Lu XY, Liu XN, Cui LW, Wang G, Zhang M, Li C, Wang W. Qishen Granules Modulate Metabolism Flexibility Against Myocardial Infarction via HIF-1 α-Dependent Mechanisms in Rats. Chin J Integr Med 2025; 31:215-227. [PMID: 39305458 DOI: 10.1007/s11655-024-3667-y] [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] [Accepted: 05/20/2024] [Indexed: 02/26/2025]
Abstract
OBJECTIVE To assess the cardioprotective effect and impact of Qishen Granules (QSG) on different ischemic areas of the myocardium in heart failure (HF) rats by evaluating its metabolic pattern, substrate utilization, and mechanistic modulation. METHODS In vivo, echocardiography and histology were used to assess rat cardiac function; positron emission tomography was performed to assess the abundance of glucose metabolism in the ischemic border and remote areas of the heart; fatty acid metabolism and ATP production levels were assessed by hematologic and biochemical analyses. The above experiments evaluated the cardioprotective effect of QSG on left anterior descending ligation-induced HF in rats and the mode of energy metabolism modulation. In vitro, a hypoxia-induced H9C2 model was established, mitochondrial damage was evaluated by flow cytometry, and nuclear translocation of hypoxia-inducible factor-1 α (HIF-1 α) was observed by immunofluorescence to assess the mechanism of energy metabolism regulation by QSG in hypoxic and normoxia conditions. RESULTS QSG regulated the pattern of glucose and fatty acid metabolism in the border and remote areas of the heart via the HIF-1 α pathway, and improved cardiac function in HF rats. Specifically, QSG promoted HIF-1 α expression and entry into the nucleus at high levels of hypoxia (P<0.05), thereby promoting increased compensatory glucose metabolism; while reducing nuclear accumulation of HIF-1 α at relatively low levels of hypoxia (P<0.05), promoting the increased lipid metabolism. CONCLUSIONS QSG regulates the protein stability of HIF-1 α, thereby coordinating energy supply balance between the ischemic border and remote areas of the myocardium. This alleviates the energy metabolism disorder caused by ischemic injury.
Collapse
Affiliation(s)
- Xiao-Qian Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yan-Qin Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiang-Yu Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
- Guangzhou University of Chinese Medicine, Guangdong, 510006, China
| | - Xiang-Ning Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ling-Wen Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Gang Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Man Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chun Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Beijing Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Guangzhou University of Chinese Medicine, Guangdong, 510006, China.
| |
Collapse
|
31
|
Thapa R, Marianesan AB, Rekha A, Ganesan S, Kumari M, Bhat AA, Ali H, Singh SK, Chakraborty A, MacLoughlin R, Gupta G, Dua K. Hypoxia-inducible factor and cellular senescence in pulmonary aging and disease. Biogerontology 2025; 26:64. [PMID: 40011266 PMCID: PMC11865175 DOI: 10.1007/s10522-025-10208-z] [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: 01/04/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Cellular senescence and hypoxia-inducible factor (HIF) signaling are crucial in pulmonary aging and age-related lung diseases such as chronic obstructive pulmonary disease idiopathic pulmonary fibrosis and lung cancer. HIF plays a pivotal role in cellular adaptation to hypoxia, regulating processes like angiogenesis, metabolism, and inflammation. Meanwhile, cellular senescence leads to irreversible cell cycle arrest, triggering the senescence-associated secretory phenotype which contributes to chronic inflammation, tissue remodeling, and fibrosis. Dysregulation of these pathways accelerates lung aging and disease progression by promoting oxidative stress, mitochondrial dysfunction, and epigenetic alterations. Recent studies indicate that HIF and senescence interact at multiple levels, where HIF can both induce and suppress senescence, depending on cellular conditions. While transient HIF activation supports tissue repair and stress resistance, chronic dysregulation exacerbates pulmonary pathologies. Furthermore, emerging evidence suggests that targeting HIF and senescence pathways could offer new therapeutic strategies to mitigate age-related lung diseases. This review explores the intricate crosstalk between these mechanisms, shedding light on how their interplay influences pulmonary aging and disease progression. Additionally, we discuss potential interventions, including senolytic therapies and HIF modulators, that could enhance lung health and longevity.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - A Rekha
- Dr D Y Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mukesh Kumari
- NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Ronan MacLoughlin
- Aerogen, IDA Business Park, Dangan, Galway, H91 HE94, Ireland
- School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- School of Pharmacy & Pharmaceutical Sciences, Trinity College, Dublin, D02 PN40, Ireland
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo, NSW, 2007, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo, NSW, 2007, Australia.
- Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| |
Collapse
|
32
|
Andryszkiewicz W, Gąsiorowska J, Kübler M, Kublińska K, Pałkiewicz A, Wiatkowski A, Szwedowicz U, Choromańska A. Glucose Metabolism and Tumor Microenvironment: Mechanistic Insights and Therapeutic Implications. Int J Mol Sci 2025; 26:1879. [PMID: 40076506 PMCID: PMC11900028 DOI: 10.3390/ijms26051879] [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: 01/22/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic reprogramming in cancer cells involves changes in glucose metabolism, glutamine utilization, and lipid production, as well as promoting increased cell proliferation, survival, and immune resistance by altering the tumor microenvironment. Our study analyzes metabolic reprogramming in neoplastically transformed cells, focusing on changes in glucose metabolism, glutaminolysis, and lipid synthesis. Moreover, we discuss the therapeutic potential of targeting cancer metabolism, focusing on key enzymes involved in glycolysis, the pentose phosphate pathway, and amino acid metabolism, including lactate dehydrogenase A, hexokinase, phosphofructokinase and others. The review also highlights challenges such as metabolic heterogeneity, adaptability, and the need for personalized therapies to overcome resistance and minimize adverse effects in cancer treatment. This review underscores the significance of comprehending metabolic reprogramming in cancer cells to engineer targeted therapies, personalize treatment methodologies, and surmount challenges, including metabolic plasticity and therapeutic resistance.
Collapse
Affiliation(s)
- Wiktoria Andryszkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Julia Gąsiorowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Maja Kübler
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Karolina Kublińska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Agata Pałkiewicz
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Adam Wiatkowski
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.A.); (J.G.); (M.K.); (K.K.); (A.P.); (A.W.)
| | - Urszula Szwedowicz
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
33
|
Yang L, Wen Y, Yuan Z, Zhao D, Weng P, Li Y, Chen Q, Zhang W, Hu H, Yu C. Hypoxia-mediated SUMOylation of FADD exacerbates endothelial cell injury via the RIPK1-RIPK3-MLKL signaling axis. Cell Death Dis 2025; 16:121. [PMID: 39984463 PMCID: PMC11845712 DOI: 10.1038/s41419-025-07441-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 01/13/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Vascular endothelial cells are the predominant cell type in the cardiovascular system, and their dysfunction and death following hypoxic injury contribute to vascular lesions, playing an essential role in cardiovascular disease. Despite its importance, the mechanisms underlying vascular endothelial cell injury under hypoxia and potential therapeutic interventions remain poorly understood. Here, we constructed both an in vivo hypoxia model in C57BL/6 mice and an in vitro hypoxia model in HUVEC cells. Our findings demonstrated that hypoxia induces necroptosis in vascular endothelial cells and exacerbates inflammatory injury in vivo and in vitro, as evidenced by immunofluorescence and western blot. We identified FADD as a critical regulator of hypoxia-mediated necroptosis, with FADD knockdown significantly reversing hypoxia-induced necroptosis. Mechanistically, hypoxia affected protein conformation through SUMOylation of FADD and competitively inhibited its ubiquitination, leading to an increase in protein half-life and protein level of FADD. Furthermore, SUMOylation increased the interaction between FADD and RIPK1 and induced the formation of the FADD-RIPK1-RIPK3 complex, thereby promoting necroptosis in vascular endothelial cells. The SUMOylation inhibitor ginkgolic acid (GA) notably reduced hypoxia-induced vascular endothelial injury and inflammatory responses in male mice. Taken together, our research has uncovered a new process by which SUMOylation of FADD regulates hypoxia-induced necroptosis in endothelial cells, providing potential therapeutic targets for hypoxia-related cardiovascular diseases.
Collapse
Affiliation(s)
- Liming Yang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yilin Wen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Dezhang Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Ping Weng
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Yueyue Li
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Qingyang Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Wanping Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China
| | - Hui Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, Chongqing, China.
| |
Collapse
|
34
|
Singh V, Singh R, Goswami P, Manna PP, Basu Baul TS, Mandal A, Singh AK, Koch B. Influences of aqua-(2-formylbenzoato) triphenyltin(IV) on regression of hypoxic solid tumor through mitochondrial mediated pathway by inhibiting Hif-1 alpha. Sci Rep 2025; 15:5302. [PMID: 39939702 PMCID: PMC11821849 DOI: 10.1038/s41598-025-89761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/07/2025] [Indexed: 02/14/2025] Open
Abstract
Tumor hypoxia is the major hindrance behind cancer chemotherapy and the foremost reason for the less effectiveness of most anticancer drugs. We herein inquire into the mechanistic part and therapeutic efficacy of our previously reported compound, aqua-(2-formylbenzoato) triphenyltin (IV) (abbreviated as OTC), in a hypoxic solid tumor-bearing mouse model (BALB/c). In addition to solid tumors, we investigated the therapeutic potential of OTC in intraperitoneal tumor and in in vitro system. Following treatment, mitochondrial dynamics, tumor load regression, survival analysis and histopathological parameters were analyzed. Furthermore, the differential expression levels of cleaved PARP-1, Hif-1α, VEGF and apoptotic genes such as Bax, Bcl-2, p53, and caspase 3 at the mRNA and protein levels were assessed. Our findings demonstrate that OTC significantly induces tumor regression and increased survivability by down regulating the expression of the hypoxia-associated genes Hif-1α and VEGF while elevating the levels of cleaved PARP-1 and p53. In contrast, the commercially available drug doxorubicin was found less effective and failed to respond in the tumor microenvironment. Furthermore, increased mitochondrial aggregation and membrane permeability and activation of Bax, caspase 3 and caspase-9 and release of Cytochrome-c from the mitochondrial membrane at RNA level confirms the mitochondrial pathway of apoptosis. Therefore, our present findings reveal that newly synthesized OTC potentially induces apoptosis and could be a promising compound against the tumor microenvironment.
Collapse
Affiliation(s)
- Virendra Singh
- Cancer Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Pooja Goswami
- Cancer Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Tushar S Basu Baul
- Centre for Advanced Studies in Chemistry, North Eastern Hill University, NEHU Permanent Campus, Umshing, Shillong, 793 022, India
| | - Abhijit Mandal
- Department of Radiotherapy and Radiation Medicine, Banaras Hindu University, Varanasi, 221005, India
| | - Arvind Kumar Singh
- Genetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Biplob Koch
- Cancer Biology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
35
|
Dickerson AG, Joseph CA, Kashfi K. Current Approaches and Innovations in Managing Preeclampsia: Highlighting Maternal Health Disparities. J Clin Med 2025; 14:1190. [PMID: 40004721 PMCID: PMC11856135 DOI: 10.3390/jcm14041190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Preeclampsia (PE) is a major cause of maternal mortality and morbidity, affecting 3-6% of pregnancies worldwide and ranking among the top six causes of maternal deaths in the U.S. PE typically develops after 20 weeks of gestation and is characterized by new-onset hypertension and/or end-organ dysfunction, with or without proteinuria. Current management strategies for PE emphasize early diagnosis, blood pressure control, and timely delivery. For prevention, low-dose aspirin (81 mg/day) is recommended for high-risk women between 12 and 28 weeks of gestation. Magnesium sulfate is also advised to prevent seizures in preeclamptic women at risk of eclampsia. Emerging management approaches include antiangiogenic therapies, hypoxia-inducible factor suppression, statins, and supplementation with CoQ10, nitric oxide, and hydrogen sulfide donors. Black women are at particularly high risk for PE, potentially due to higher rates of hypertension and cholesterol, compounded by healthcare disparities and possible genetic factors, such as the APOL1 gene. This review explores current and emerging strategies for managing PE and addresses the underlying causes of health disparities, offering potential solutions to improve outcomes.
Collapse
Affiliation(s)
- Alexis G. Dickerson
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
| | - Christiana A. Joseph
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
- Department of Chemistry and Physics, State University of New York at Old Westbury, Old Westbury, NY 11568, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
- Department of Chemistry and Physics, State University of New York at Old Westbury, Old Westbury, NY 11568, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10091, USA
| |
Collapse
|
36
|
Dwyer KD, Snyder CA, Coulombe KLK. Cardiomyocytes in Hypoxia: Cellular Responses and Implications for Cell-Based Cardiac Regenerative Therapies. Bioengineering (Basel) 2025; 12:154. [PMID: 40001674 PMCID: PMC11851968 DOI: 10.3390/bioengineering12020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Myocardial infarction (MI) is a severe hypoxic event, resulting in the loss of up to one billion cardiomyocytes (CMs). Due to the limited intrinsic regenerative capacity of the heart, cell-based regenerative therapies, which feature the implantation of stem cell-derived cardiomyocytes (SC-CMs) into the infarcted myocardium, are being developed with the goal of restoring lost muscle mass, re-engineering cardiac contractility, and preventing the progression of MI into heart failure (HF). However, such cell-based therapies are challenged by their susceptibility to oxidative stress in the ischemic environment of the infarcted heart. To maximize the therapeutic benefits of cell-based approaches, a better understanding of the heart environment at the cellular, tissue, and organ level throughout MI is imperative. This review provides a comprehensive summary of the cardiac pathophysiology occurring during and after MI, as well as how these changes define the cardiac environment to which cell-based cardiac regenerative therapies are delivered. This understanding is then leveraged to frame how cell culture treatments may be employed to enhance SC-CMs' hypoxia resistance. In this way, we synthesize both the complex experience of SC-CMs upon implantation and the engineering techniques that can be utilized to develop robust SC-CMs for the clinical translation of cell-based cardiac therapies.
Collapse
Affiliation(s)
| | | | - Kareen L. K. Coulombe
- Institute for Biology, Engineering, and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA; (K.D.D.); (C.A.S.)
| |
Collapse
|
37
|
Lee PWT, Kobayashi M, Dohkai T, Takahashi I, Yoshida T, Harada H. 2-Oxoglutarate-dependent dioxygenases as oxygen sensors: their importance in health and disease. J Biochem 2025; 177:79-104. [PMID: 39679914 DOI: 10.1093/jb/mvae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024] Open
Abstract
Since low oxygen conditions below physiological levels, hypoxia, are associated with various diseases, it is crucial to understand the molecular basis behind cellular response to hypoxia. Hypoxia-inducible factors (HIFs) have been revealed to primarily orchestrate the hypoxic response at the transcription level and have continuously attracted great attention over the past three decades. In addition to these hypoxia-responsive effector proteins, 2-oxoglutarate-dependent dioxygenase (2-OGDD) superfamily including prolyl-4-hydroxylase domain-containing proteins (PHDs) and factor inhibiting HIF-1 (FIH-1) has attracted even greater attention in recent years as factors that act as direct oxygen sensors due to their necessity of oxygen for the regulation of the expression and activity of the regulatory subunit of HIFs. Herein, we present a detailed classification of 2-OGDD superfamily proteins, such as Jumonji C-domain-containing histone demethylases, ten-eleven translocation enzymes, AlkB family of DNA/RNA demethylases and lysyl hydroxylases, and discuss their specific functions and associations with various diseases. By introducing the multifaceted roles of 2-OGDD superfamily proteins in the hypoxic response, this review aims to summarize the accumulated knowledge about the complex mechanisms governing cellular adaptation to hypoxia in various physiological and pathophysiological contexts.
Collapse
Affiliation(s)
- Peter W T Lee
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takakuni Dohkai
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Itsuki Takahashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takumi Yoshida
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
38
|
Forkan CP, Shrestha A, Yu A, Chuang C, Pociot F, Yarani R. Could hypoxic conditioning augment the potential of mesenchymal stromal cell-derived extracellular vesicles as a treatment for type 1 diabetes? Stem Cell Res Ther 2025; 16:37. [PMID: 39901225 PMCID: PMC11792614 DOI: 10.1186/s13287-025-04153-4] [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: 11/04/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
Type1 Diabetes (T1D) is an autoimmune disorder characterised by the loss of pancreatic β-cells. This β cell loss occurs primarily through inflammatory pathways culminating in apoptosis. Mesenchymal stromal cells (MSCs) have been heavily studied for therapeutic applications due to their regenerative, anti-apoptotic, immunomodulatory, and anti-inflammatory properties. The therapeutic effects of MSCs are mediated through cell-to-cell contact, differentiation, and the release of paracrine factors, which include the release of extracellular vesicles (EVs). Culturing MSCs in hypoxia, a low oxygen tension state more analogous to their physiological environment, seems to increase the therapeutic efficacy of MSC cell therapy, enhancing their immunomodulatory, anti-inflammatory, and anti-fibrotic properties. This is also the case with MSC-derived EVs, which show altered properties based on the parent cell preconditioning. In this review, we examine the evidence supporting the potential application of hypoxic preconditioning in strengthening MSC-EVs for treating the inflammatory and apoptotic causes of β cell loss in T1D.
Collapse
Affiliation(s)
- Cathal Patrick Forkan
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Aruna Shrestha
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Alfred Yu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Christine Chuang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark.
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA.
| |
Collapse
|
39
|
Yuan F, Liu J, Zhong L, Liu P, Li T, Yang K, Gao W, Zhang G, Sun J, Zou X. Enhanced therapeutic effects of hypoxia-preconditioned mesenchymal stromal cell-derived extracellular vesicles in renal ischemic injury. Stem Cell Res Ther 2025; 16:39. [PMID: 39901252 PMCID: PMC11792194 DOI: 10.1186/s13287-025-04166-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been shown to provide significant protection against renal ischemia-reperfusion injury (IRI). Hypoxia has emerged as a promising strategy to enhance the tissue repair capabilities of MSCs. However, the specific effects of hypoxia on MSCs and MSC-EVs, as well as their therapeutic potential in renal IRI, remain unclear. In this study, we investigated the alterations occurring in MSCs and the production of MSC-EVs following hypoxia pre-treatment, and further explored the key intrinsic mechanisms underlying the therapeutic effects of hypoxic MSC-EVs in the treatment of renal IRI. METHODS Human umbilical cord MSCs were cultured under normoxic and hypoxic conditions. Proliferation and related pathways were measured, and RNA sequencing was used to detect changes in the transcriptional profile. MSC-EVs from both normoxic and hypoxic conditions were isolated and characterized. In vivo, the localization and therapeutic effects of MSC-EVs were assessed in a rat renal IRI model. Histological examinations were conducted to evaluate the structure, proliferation, and apoptosis of IRI kidney tissue respectively. Renal function was assessed by measuring serum creatinine and blood urea nitrogen levels. In vitro, the therapeutic potential of MSC-EVs were measured in renal tubular epithelial cells injured by antimycin A. Protein sequencing analysis of hypoxic MSC-EVs was performed, and the depletion of Glutathione S-Transferase Omega 1 (GSTO1) in hypoxic MSC-EVs was carried out to verify its key role in alleviating renal injury. RESULTS Hypoxia alters MSCs transcriptional profile, promotes their proliferation, and increases the production of EVs. Hypoxia-pretreated MSC-EVs demonstrated a superior ability to mitigate renal IRI, enhancing proliferation and reducing apoptosis of renal tubular epithelial cells both in vivo and in vitro. Protein profiling of the EVs revealed an accumulation of numerous anti-oxidative stress proteins, with GSTO1 being particularly prominent. Knockdown of GSTO1 significantly reduced the antioxidant and therapeutic effects on renal IRI of hypoxic MSC-EVs. CONCLUSIONS Hypoxia significantly promotes the generation of MSC-EVs and enhances their therapeutic effects on renal IRI. The antioxidant stress effect induced by GSTO1 is identified as one of the most critical underlying mechanisms. Our findings highlight that hypoxia-pretreated MSC-EVs represent a novel and promising therapeutic strategy for renal IRI.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jie Liu
- Department of Neurology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhong
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Pengtao Liu
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Li
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Kexin Yang
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Guangyuan Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu Province, China.
| | - Jie Sun
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Xiangyu Zou
- Department of Urology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
40
|
Li X, Liu B, Li G, Wang H, Yang J, Wen H, He F. tgfβ1a/vegfa gene expression and methylation in response to acute hypoxia in Japanese flounder (Paralichthys olivaceus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 163:105319. [PMID: 39826665 DOI: 10.1016/j.dci.2025.105319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/12/2025] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
The physiological response and molecular mechanism of the immune response of Japanese flounder under hypoxia stress remain unclear. In this study, we examined the immune-related physiological indexes and the molecular mechanisms of Japanese flounders under acute hypoxia stress. The results showed that the levels of serum ALT, ALP, AST and LDH in hypoxia stress group were significantly increased (P < 0.01). Through quantitative real-time PCR and double in situ hybridization, we found acute hypoxia stress induced immune response of skeletal muscle and gill filaments. The transcriptional regulation mechanism of this immune signaling pathway was investigated by dual luciferase assay. In addition, DNA methylation levels of genes were detected to explore epigenetic modifications of this pathway. As a transcription factor, Foxo1a can interact tgfβ1a(AGATGTTTTTT) and vegfa(TTCTTTTTATA, TACTGTTGCTA) sequences of the promoter regions. The DNA methylation levels of tgfβ1a and vegfa genes were significantly affected by hypoxia and negatively correlated with their expression. These experiments showed that the expression of immune related genes tgfβ1a and vegfa were regulated by transcription factor Foxo1a and DNA methylation. Our study provides theoretical foundations for acute hypoxia stress induced immune response of Japanese flounder.
Collapse
Affiliation(s)
- Xiaohui Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Binghua Liu
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Guangling Li
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Hao Wang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Jun Yang
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China
| | - Feng He
- Key Laboratory of Mariculture, Ocean University of China, Ministry of Education, Qingdao, 266003, PR China.
| |
Collapse
|
41
|
Ma N, Liu P, Li N, Hu Y, Kang L. Exploring the pharmacological mechanisms for alleviating OSA: Adenosine A2A receptor downregulation of the PI3K/Akt/HIF‑1 pathway (Review). Biomed Rep 2025; 22:21. [PMID: 39720297 PMCID: PMC11668141 DOI: 10.3892/br.2024.1899] [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: 08/29/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Obstructive sleep apnea (OSA) is the most common type of sleep apnea, which leads to episodes of intermittent hypoxia due to obstruction of the upper airway. A key feature of OSA is the upregulation and stabilization of hypoxia-inducible factor 1 (HIF-1), a crucial metabolic regulator that facilitates rapid adaptation to changes in oxygen availability. Adenosine A2A receptor (A2AR), a major adenosine receptor, regulates HIF-1 under hypoxic conditions, exerting anti-inflammatory properties and affecting lipid metabolism. The present study explored the roles of A2AR in OSA regulation, specifically focusing on its effects via the PI3K/Akt/HIF-1 pathway. The findings enhance our understanding the pharmacological potential of A2AR in OSA management and suggest future research directions in exploring its clinical applications.
Collapse
Affiliation(s)
- Nini Ma
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Peijie Liu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Ning Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Yushi Hu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| | - Liang Kang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 641418, P.R. China
| |
Collapse
|
42
|
Zhou M, Dong S, Wang J, Luo X, Li R, Zhang Y, Ding H, Tan X, Qiao Z, Yang K, Chen W. Differential expression of HIF-1α and its hypoxia-related inducers in the spleens of plateau yaks and plain yellow cattle. Histol Histopathol 2025; 40:225-235. [PMID: 38864176 DOI: 10.14670/hh-18-768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
The present study aims to investigate the distribution and expression characteristics of HIF-1α, VEGF, VEGFR-2, VCAM-1, and IL-4 in the spleen of plateau yaks and plain yellow cattle and to speculate the possible regulatory role of HIF-1α and its related hypoxia-inducible factors in the adaptation of the yak spleen to the plateau hypoxic environment. Histological features were observed using H&E and PAS stains. Immunohistochemical staining and optical density analysis were applied to investigate the distribution and differences in the expression of HIF-1α, VEGF, VEGFR-2, VCAM-1, and IL-4 in the spleen of yaks and cattle. The results showed that the area of splenic trabeculae and splenic nodules was significantly larger in the yak than in yellow cattle (P<0.05). Glycogen was mainly distributed in splenic arterial endothelial cells, vascular smooth muscle cells, splenic blood sinusoidal endothelial cells, and fibroblasts, and the distribution was significantly higher in the spleen of yaks than in cattle (P<0.05). HIF-1α, VEGF, VEGFR-2, VCAM-1, and IL-4 were mainly expressed in lymphocytes, arterial endothelial cells, vascular smooth muscle cells, splenic blood sinusoidal endothelial cells, and fibroblast cytoplasm, with higher expression in yak spleen (P<0.05). In conclusion, combining the differences in spleen tissue structure, glycogen distribution, and expression distribution of several hypoxia-related factors between yaks and cattle, we suggest that HIF-1α, VEGF, VEGFR-2, VCAM-1, and IL-4 may be important factors in the adaptation of yak spleen to the plateau environment, which provides a theoretical basis for further exploring the adaptation mechanism of plateau hypoxia in yaks.
Collapse
Affiliation(s)
- Manlin Zhou
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lan Zhou, Gansu, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Shihui Dong
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Jun Wang
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Xuehui Luo
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Rui Li
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lan Zhou, Gansu, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Yiyang Zhang
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lan Zhou, Gansu, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Haie Ding
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Xiao Tan
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Zilin Qiao
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lan Zhou, Gansu, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
| | - Kun Yang
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Lan Zhou, Gansu, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lan Zhou, Gansu, China.
| | - Weiji Chen
- College of Life Science and Engineering, Northwest Minzu University, Lan Zhou, Gansu, China
| |
Collapse
|
43
|
Wang Z, Zhu C, Sun X, Deng H, Liu W, Jia S, Bai Y, Xiao W, Liu X. Spring viremia of carp virus infection induces hypoxia response in zebrafish by stabilizing hif1α. J Virol 2025; 99:e0149124. [PMID: 39601573 PMCID: PMC11784138 DOI: 10.1128/jvi.01491-24] [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: 08/27/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
The hypoxia signaling pathway controls hypoxia adaptation and tolerance of organisms, which is regulated by multiple mechanisms. Viral infection elicits various pathophysiological responses in the host. However, whether viral infection can affect the hypoxia response is not yet fully understood. In this study, we found that Spring viremia of carp virus (SVCV) infection in zebrafish caused symptoms similar to those in zebrafish under hypoxic conditions. Further assays indicated that SVCV infection activated the hypoxia signaling pathway in zebrafish. In addition, SVCV infection caused increased glycolysis and reactive oxygen species (ROS) levels in cells. Mechanistically, SVCV-G protein interacted with hif1α-a/b and attenuated their K48-linked polyubiquitination, leading to their stabilization and subsequent enhancement of target gene expression. Moreover, treatment with the HIF1α-specific inhibitor PX478 enhanced the antiviral ability against SVCV infection in zebrafish and zebrafish cells. This study reveals a relationship between SVCV infection and the hypoxia signaling pathway in fish and provides a strategy for reducing the damage of viral disease in the aquaculture industry. IMPORTANCE Viral infection triggers various pathophysiological responses in the host. The hypoxia signaling pathway controls hypoxia adaptation and tolerance of organisms. However, whether viral infection can affect the hypoxia response is not yet fully understood. This study showed that Spring viremia of carp virus (SVCV) infection activated the hypoxia signaling pathway and induced a hypoxia response. The SVCV-G protein interacted with hif1α-a/b and reduced their K48-linked polyubiquitination, leading to their stabilization and subsequent enhancement of target gene expression. Additionally, treatment with the HIF1α-specific inhibitor PX478 enhanced the antiviral ability against SVCV infection in zebrafish and zebrafish cells. Our findings not only reveal a relationship between SVCV infection and the hypoxia signaling pathway in fish but also provide a strategy for reducing the damage of viral disease in the aquaculture industry.
Collapse
Affiliation(s)
- Zixuan Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chunchun Zhu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xueyi Sun
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Hongyan Deng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Life Science, Wuhan University, Wuhan, China
| | - Wen Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Shuke Jia
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Yao Bai
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Wuhan Xiao
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
- College of Life Science, Wuhan University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Xing Liu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
- College of Life Science, Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Nishigaki A, Ishida M, Tsubokura H, Hisamatsu Y, Hirose Y, Okada H. HIF-1α Promotes Luteinization via NDRG1 Induction in the Human Ovary. Biomedicines 2025; 13:328. [PMID: 40002742 PMCID: PMC11852844 DOI: 10.3390/biomedicines13020328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a crucial role in various physiological and pathological processes of the ovary. However, the timing of HIF-1α expression and its specific biological function in the follicular development of the human ovary remain unclear. Therefore, in this study, we aimed to examine whether HIF-1α and its downstream gene, N-myc downstream-regulated gene 1 (NDRG1), exhibit stage-specific expression during the follicular development process in the human ovary. Methods: We used ovarian tissues from eight women with regular menstrual cycles who were not undergoing hormonal treatment. We investigated HIF-1α and NDRG1 expression and localization using immunohistochemistry. Further, we transfected human ovarian granulosa (KGN) cells with HIF-1α small interfering RNA (siRNA) to investigate the influence of HIF-1α on NDRG1 expression and progesterone synthesis. Results: The immunohistochemical analysis of human ovarian tissues revealed that HIF-1α was localized in the cytoplasm of granulosa cells (GCs) at both the primary and secondary follicular stages. Conversely, in tertiary and later developmental stages, HIF-1α was observed exclusively in the nucleus of GCs. Furthermore, while NDRG1 was not detected in primary follicles, it was present in all GCs beyond the tertiary stage. Notably, transfection of KGN cells with HIF-1α siRNA significantly decreased NDRG1 expression, at both the mRNA and protein levels, and in progesterone synthesis. Conclusion: Our results indicate that HIF-1α and NDRG1 are integral to follicular development and the early luteinization of pre-ovulatory follicles.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoji Hisamatsu
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| |
Collapse
|
45
|
García-Pérez-de-Sevilla G, González-de-la-Flor Á. Impact of Fatty Acid Supplementation on Migraine Outcomes: A Systematic Review and Meta-analysis. Nutr Rev 2025:nuae219. [PMID: 39823374 DOI: 10.1093/nutrit/nuae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
CONTEXT Migraines are a prevalent neurological condition that significantly impacts the quality of life. Although narrative reviews and clinical trials suggest the potential effects of fatty acid supplementation as a promising approach for migraine prophylaxis, the findings remain inconsistent. OBJECTIVE The aim was to evaluate the efficacy of fatty acid supplementation on migraine clinical outcomes through a systematic review and meta-analysis. DATA SOURCES This systematic review and meta-analysis was conducted using electronic databases including Medline, Scopus, Web of Science, and CINAHL from their inception up to October 7, 2024. DATA EXTRACTION Studies meeting the PICOS criteria were included: adults diagnosed with migraine, dietary supplementation with fatty acids, assessing migraine clinical outcomes, and only randomized controlled trials. Methodological quality and risk of bias were assessed independently by 2 reviewers using the Cochrane Risk of Bias 2 tool. DATA ANALYSIS Meta-analyses were conducted using Review Manager software. A random-effects model was applied to account for heterogeneity among studies. A standardized mean difference (SMD) of 0.2, 0.5, and 0.8 was interpreted as small, moderate, and large effects, respectively. Six randomized controlled trials, including 407 participants with chronic migraines, were analyzed. Interventions varied, including omega-3 supplementation with or without omega-6 restriction, lasting from 8 to 16 weeks, and 1 trial assessing alpha-lipoic acid. The overall risk of bias was assessed as low. The pooled analysis demonstrated significant reductions in headache intensity (SMD = -1.77; 95% CI: -3.32 to -0.21; P = .03), headache duration (SMD = -0.77; 95% CI: -1.05 to -0.50; P < .00001), headache frequency (SMD = -1.91; 95% CI: -2.61 to -1.21; P < .00001), and HIT-6 score (SMD = -2.44; 95% CI: -4.13 to -0.76; P = .004). CONCLUSION This meta-analysis provides moderate evidence that fatty acid supplementation, particularly omega-3, improves migraine clinical outcomes. Additional high-quality, randomized controlled trials are needed to confirm these findings, particularly for the promising effects of alpha-lipoic acid. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42024592084.
Collapse
Affiliation(s)
- Guillermo García-Pérez-de-Sevilla
- Universidad Europea de Madrid, Department of Physiotherapy, Faculty of Medicine, Health and Sports, 28670 Villaviciosa de odón, Madrid, Spain
| | - Ángel González-de-la-Flor
- Universidad Europea de Madrid, Department of Physiotherapy, Faculty of Medicine, Health and Sports, 28670 Villaviciosa de odón, Madrid, Spain
| |
Collapse
|
46
|
Koloi S, Ganguly I, Singh S, Dixit S. Whole genome re-sequencing reveals high altitude adaptation signatures and admixture in Ladakhi cattle. Gene 2025; 933:148957. [PMID: 39306203 DOI: 10.1016/j.gene.2024.148957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 08/19/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Ladakhi cattle, native to the high-altitude region of Ladakh in northern India (ranging from 3,000 to 5,000 m above sea level), have evolved unique genetic adaptations to thrive in harsh environmental conditions, such as hypoxia, extreme cold, and low humidity. This study explored the genome of Ladakhi cattle to investigate genetic structure, selection signatures, and adaptive mechanisms. Whole genome sequencing reads, generated on Illumina NovaSeq 6000 platform, were aligned to the Bos taurus reference genome with BWA-MEM. SNPs were identified and filtered using GATK and bcftools, and functionally annotated with SnpEff. For population genomic analysis, PCA and admixture modeling assessed genetic structure, while Neighbor-Joining trees, LD decay, nucleotide diversity (π), and FST evaluated phylogenetic relationships and genetic variation. Selective sweeps were detected using RAiSD, and gene-set enrichment and protein-protein interaction analyses were conducted to explore functional pathways related to adaptation. The study revealed 3,759,279 unique SNPs and demonstrated that Ladakhi cattle form a distinct genetic cluster with an estimated admixture of 68 % Bos indicus and 32 % Bos taurus ancestry. Key findings include rapid linkage disequilibrium decay, low inbreeding level, and the identification of selection signatures and genes associated with hypoxia response, energy metabolism, and cold adaptation. Mean nucleotide diversity (π, 0.0037) and FST values indicated moderate genetic differentiation from other breeds. The analysis highlighted selection signatures for genes like HIF1A, ENO4, ANGPT1, EPO, NOS3, MAPK3, HMOX1, BCL2,CAMK2D, MTOR, AKT2,PIK3CB, and MAP2K1, among others, including various keratin and heat shock proteins. The interaction between genes associated with hypoxia signaling (HIF-1) and other enriched pathways such as PI3K, mTOR, NFκB, ERK, and ER stress, reveals a complex mechanism for managing hypoxic stress in Ladakhi cattle. These findings offer valuable insights for breeding programs aimed at enhancing livestock resilience in extreme environments and enhance understanding of mammalian adaptation to high-altitude conditions.
Collapse
Affiliation(s)
- Subrata Koloi
- Division of Animal Genetics, ICAR-National Bureau of Animal Genetic Resources, Karnal 132001, India; Division of Animal Genetics and Breeding, ICAR-National Dairy Research Institute, Karnal 132001, India
| | - Indrajit Ganguly
- Division of Animal Genetics, ICAR-National Bureau of Animal Genetic Resources, Karnal 132001, India.
| | - Sanjeev Singh
- Division of Animal Genetics, ICAR-National Bureau of Animal Genetic Resources, Karnal 132001, India
| | - Satpal Dixit
- Division of Animal Genetics, ICAR-National Bureau of Animal Genetic Resources, Karnal 132001, India.
| |
Collapse
|
47
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
48
|
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
|
49
|
Patel JIA, Poyya J, Padakannaya A, Kurdekar NM, Khandagale AS, Joshi CG, Kanade SR, Satyamoorthy K. Mechanistic insights into gut microbe derived siderophores and PHD2 interactions with implications for HIF-1α stabilization. Sci Rep 2025; 15:1113. [PMID: 39774022 PMCID: PMC11707245 DOI: 10.1038/s41598-024-83730-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
In oxygen-deprived conditions, cells respond by activating adaptive mechanisms to bolster their survival and protect tissue integrity. A key player in this process is the HIF-1α signaling cascade, meticulously regulated by Prolyl Hydroxylase Domain 2 (PHD2), which orchestrates cellular responses to varying oxygen levels. The primary aim of this investigation is to utilize gut siderophores as inhibitors of PHD2 in ischemic conditions. This study also helps in understanding the structural mechanisms by which gut microbiota regulate HIF-1α via PHD2 inhibition through the secretion of siderophores. We explore potential PHD2 inhibitors through in-silico approaches, specifically molecular docking, binding pose metadynamics, molecular dynamics simulations, and free energy calculations. We evaluated siderophores secreted by gut microbiota as candidate inhibitors for PHD2. Docking studies revealed that Salmochelin SX exhibits the highest binding affinity to PHD2 (- 9.527 kcal/mol), interacting with key residues such as ASP254, TYR310, ASP315, and ARG322. Despite its high affinity, binding pose metadynamics indicated instability for Salmochelin SX, whereas Staphyloferrin A demonstrated superior stability. Molecular dynamics simulations confirmed stable ligand interactions with PHD2, highlighting HIS313 and ASP315 as critical for inhibition. Principal Component Analysis (PCA) and Free Energy Landscape (FEL) analyses underscored conformational changes and binding stability, suggesting that these interactions may stabilize PHD2's active site and have potential therapeutic implications. Additionally, the study reveals how gut microbiota prevent gut dysbiosis through the stabilization of HIF-1α signaling by secreting siderophores.
Collapse
Affiliation(s)
- Jainabbi Irshad Ahmed Patel
- SDM Research Institute for Biomedical Sciences, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, 580009, India
| | - Jagadeesha Poyya
- SDM Research Institute for Biomedical Sciences, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, 580009, India.
| | - Apeksha Padakannaya
- SDM Research Institute for Biomedical Sciences, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, 580009, India
| | - Namrata Manjunath Kurdekar
- SDM Research Institute for Biomedical Sciences, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, 580009, India
| | - Ajay Sathayanarayan Khandagale
- SDM Research Institute for Biomedical Sciences, Shri Dharmasthala Manjunatheshwara University, Dharwad, Karnataka, 580009, India
| | | | - Santosh R Kanade
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India
| | - Kapaettu Satyamoorthy
- Shri Dharmasthala Manjunatheshwara (SDM) University, Manjushree Nagar, Sattur, Dharwad, Karnataka, 580009, India
| |
Collapse
|
50
|
Yang X, Wang X, Yang Z, Lu H. Iron-Mediated Regulation in Adipose Tissue: A Comprehensive Review of Metabolism and Physiological Effects. Curr Obes Rep 2025; 14:4. [PMID: 39753935 DOI: 10.1007/s13679-024-00600-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/14/2025]
Abstract
PURPOSE OF REVIEW Review the latest data regarding the intersection of adipose tissue (AT) and iron to meet the needs of AT metabolism and the progression of related diseases. RECENT FINDINGS Iron is involved in fundamental biological metabolic processes and is precisely fine-tuned within the body to maintain cellular, tissue and even systemic iron homeostasis. AT not only serves as an energy storage depot but also represents the largest endocrine organ in the human body, maintaining systemic metabolic homeostasis. It is involved in physiological processes such as energy storage, insulin sensitivity regulation and lipid metabolism. As a unique iron-sensing tissue, AT expresses related regulatory factors, including the classic hepcidin, ferroportin (FPN), iron regulatory protein/iron responsive element (IRP/IRE) and ferritin. Consequently, the interaction between AT and iron is intricately intertwined. Imbalance of iron homeostasis produces the potential risks of steatosis, impaired glucose tolerance and insulin resistance, leading to AT dysfunction diseases, including obesity, type 2 diabetes and metabolic dysfunction-associated steatotic liver disease (MASLD). Despite the role of AT iron has garnered increasing attention in recent years, a comprehensive review that systematically organizes the connection between iron and AT remains lacking. Given the necessity of iron homeostasis, emphasizing its potential impact on AT function and metabolism regulation provides valuable insights into physiological effects such as adipocyte differentiation and thermogenesis. Futhermore, regulators including adipokines, mitochondria and macrophages have been mentioned, along with analyzing the novel perspective of iron as a key mediator influencing the fat-gut crosstalk.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Zhe Yang
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Hongyun Lu
- Department of Endocrinology and Metabolism, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China.
| |
Collapse
|