51
|
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
|
52
|
Jiang WI, Cao Y, Xue Y, Ji Y, Winer BY, Chandra R, Zhang XF, Zhang M, Singhal NS, Pierce JT, Chen S, Ma DK. Suppressing APOE4-induced neural pathologies by targeting the VHL-HIF axis. Proc Natl Acad Sci U S A 2025; 122:e2417515122. [PMID: 39874294 PMCID: PMC11804744 DOI: 10.1073/pnas.2417515122] [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: 08/29/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
The ε4 variant of human apolipoprotein E (APOE4) is a key genetic risk factor for neurodegeneration in Alzheimer's disease and elevated all-cause mortality in humans. Understanding the factors and mechanisms that can mitigate the harmful effects of APOE4 has significant implications. In this study, we find that inactivating the VHL-1 (Von Hippel-Lindau) protein can suppress mortality, neural and behavioral pathologies caused by transgenic human APOE4 in Caenorhabditis elegans. The protective effects of VHL-1 deletion are recapitulated by stabilized HIF-1 (hypoxia-inducible factor), a transcription factor degraded by VHL-1. HIF-1 activates a genetic program that safeguards against mitochondrial dysfunction, oxidative stress, proteostasis imbalance, and endolysosomal rupture-critical cellular events linked to neural pathologies and mortality. Furthermore, genetic inhibition of Vhl reduces cerebral vascular injury and synaptic lesions in APOE4 mice, suggesting an evolutionarily conserved mechanism. Thus, we identify the VHL-HIF axis as a potent modulator of APOE4-induced neural pathologies and propose that targeting this pathway in nonproliferative tissues may curb cellular damage, protect against neurodegeneration, and reduce tissue injuries and mortality.
Collapse
Affiliation(s)
- Wei I. Jiang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Yiming Cao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yue Xue
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yichun Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Benjamin Y. Winer
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- HHMI, Chevy Chase, MD20815
| | - Rashmi Chandra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Xingyuan Fischer Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Mengqi Zhang
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Neel S. Singhal
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Jonathan T. Pierce
- Department of Neuroscience, The Center for Learning and Memory, Waggoner Center for Alcohol and Addiction Research, Institute of Neuroscience, University of Texas at Austin, Austin, TX78712
| | - Song Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Department of Physiology, University of California San Francisco, San Francisco, CA94158
- Innovative Genomics Institute, University of California, Berkeley, CA94720
| |
Collapse
|
53
|
Gheitasi I, Akbari G, Savari F. Physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated in underlying of ischemia/reperfusion injury in different organs. Mol Cell Biochem 2025; 480:855-868. [PMID: 39001984 DOI: 10.1007/s11010-024-05052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/15/2024]
Abstract
Ischemia-reperfusion (I/R) injury, as a pathological phenomenon, takes place when blood supply to an organ is disrupted and then aggravated during restoration of blood flow. Ischemic preconditioning (IPC) is a potent method for attenuating subsequent events of IR damage in numerous organs. IPC protocol is determined by a brief and sequential time periods of I/R before the main ischemia. MicroRNAs are endogenous non-coding RNAs that regulate post-transcriptionally target mRNA translation via degrading it and/or suppressing protein synthesis. This review introduces the physiological and cellular mechanisms of ischemic preconditioning microRNAs-mediated after I/R insult in different organs such as the liver, kidney, heart, brain, and intestine. Data of this review have been collected from the scientific articles published in databases such as Science Direct, Scopus, PubMed, Web of Science, and Scientific Information Database from 2000 to 2023. Based on these literature studies, IPC/IR intervention can affect cellular mechanisms including oxidative stress, apoptosis, angiogenesis, and inflammation through up-regulation or down-regulation of multiple microRNAs and their target genes.
Collapse
Affiliation(s)
- Izadpanah Gheitasi
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Ghaidafeh Akbari
- Department of Physiology, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Feryal Savari
- Department of Medical Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| |
Collapse
|
54
|
Burtscher J, Denti V, Gostner JM, Weiss AK, Strasser B, Hüfner K, Burtscher M, Paglia G, Kopp M, Dünnwald T. The interplay of NAD and hypoxic stress and its relevance for ageing. Ageing Res Rev 2025; 104:102646. [PMID: 39710071 DOI: 10.1016/j.arr.2024.102646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD) is an essential regulator of cellular metabolism and redox processes. NAD levels and the dynamics of NAD metabolism change with increasing age but can be modulated via the diet or medication. Because NAD metabolism is complex and its regulation still insufficiently understood, achieving specific outcomes without perturbing delicate balances through targeted pharmacological interventions remains challenging. NAD metabolism is also highly sensitive to environmental conditions and can be influenced behaviorally, e.g., by exercise. Changes in oxygen availability directly and indirectly affect NAD levels and may result from exposure to ambient hypoxia, increased oxygen demand during exercise, ageing or disease. Cellular responses to hypoxic stress involve rapid alterations in NAD metabolism and depend on many factors, including age, glucose status, the dose of the hypoxic stress and occurrence of reoxygenation phases, and exhibit complex time-courses. Here we summarize the known determinants of NAD-regulation by hypoxia and evaluate the role of NAD in hypoxic stress. We define the specific NAD responses to hypoxia and identify a great potential of the modulation of NAD metabolism regarding hypoxic injuries. In conclusion, NAD metabolism and cellular hypoxia responses are strongly intertwined and together mediate protective processes against hypoxic insults. Their interactions likely contribute to age-related changes and vulnerabilities. Targeting NAD homeostasis presents a promising avenue to prevent/treat hypoxic insults and - conversely - controlled hypoxia is a potential tool to regulate NAD homeostasis.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria.
| | - Vanna Denti
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Johanna M Gostner
- Medical University of Innsbruck, Biocenter, Institute of Medical Biochemistry, Innsbruck, Austria
| | - Alexander Kh Weiss
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Barbara Strasser
- Ludwig Boltzmann Institute for Rehabilitation Research, Vienna, Austria; Faculty of Medicine, Sigmund Freud Private University, Vienna, Austria
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Hospital for Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Paglia
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, MB, Italy
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Tobias Dünnwald
- Institute for Sports Medicine, Alpine Medicine and Health Tourism (ISAG), UMIT TIROL - Private University for Health Sciences and Health Technology, Hall in Tirol, Austria
| |
Collapse
|
55
|
Rani R, Kutum R, Punera DS, Yadav AP, Bansal V, Prasher B. Physiological, biochemical, and genome-wide expression patterns during graded normobaric hypoxia in healthy individuals. Physiol Genomics 2025; 57:49-64. [PMID: 39716895 DOI: 10.1152/physiolgenomics.00056.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 12/25/2024] Open
Abstract
The regulation of oxygen homeostasis is critical in physiology and disease pathogenesis. High-altitude environment or hypoxia (lack of oxygen) can lead to adverse health conditions such as high-altitude pulmonary edema (HAPE) despite initial adaptive physiological responses. Studying genetic, hematological and biochemical, and the physiological outcomes of hypoxia together could yield a comprehensive understanding and potentially uncover valuable biomarkers for predicting responses. To this end, healthy individuals (n = 51) were recruited and exposed to graded normobaric hypoxia. Physiological parameters such as heart rate (HR), heart rate variability (HRV), oxygen saturation (Spo2), and blood pressure (BP) were constantly monitored, and a blood sample was collected before and after the hypoxia exposure for the hematological and gene-expression profiles. HR was elevated, and Spo2 and HRV were significantly reduced in a fraction of inspired oxygen ([Formula: see text])-dependent manner. After exposure to hypoxia, there was a minimal decrease in HCT, red blood cell distribution width (RDW)-coefficient of variation (CV), mean platelet volume (MPV), platelet distribution width, plateletcrit, eosinophils, lymphocytes, and HDL cholesterol. Additionally, there was a marginal increase observed in neutrophils. The effect of hypoxia was further assessed at the genome-wide expression level in a subset of individuals. Eighty-two genes significantly differed after hypoxia exposure, with 46 upregulated genes and 36 downregulated genes (P ≤ 0.05 and log2-fold change greater than ±0.5). We also conducted an integrative analysis of global gene expression profiles linked with physiological parameters, and we uncovered numerous reliable gene signatures associated with BP, Spo2, HR, and HRV in response to graded normobaric hypoxia.NEW & NOTEWORTHY Our study delves into the multifaceted response to hypoxia, integrating gene expression and hematological, biochemical, and physiological assessments. Hypoxia, crucial in both physiology and pathology, prompts varied responses, necessitating a thorough systemic understanding. Examining healthy subjects exposed to graded normobaric hypoxia, we observed significant shifts in heart rate, oxygen saturation, and heart rate variability. Moreover, genomic analysis unveiled distinct gene signatures associated with physiological parameters, offering insights into molecular perturbations and adaptations to oxygen deprivation.
Collapse
Affiliation(s)
- Ritu Rani
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi, India
- CSIR's Ayurgenomics Unit-TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rintu Kutum
- CSIR's Ayurgenomics Unit-TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Deep Shikha Punera
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi, India
- CSIR's Ayurgenomics Unit-TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anand Prakash Yadav
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi, India
- Vallabhbhai Patel Chest Institute, Delhi, India
| | | | - Bhavana Prasher
- Centre of Excellence for Applied Development of Ayurveda Prakriti and Genomics, Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi, India
- CSIR's Ayurgenomics Unit-TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
56
|
Bozyel B, Doğan Ö, Elgün S, Özdemir B. Hypoxic Responses in Periodontal Tissues: Influence of Smoking and Periodontitis. J Clin Periodontol 2025; 52:249-257. [PMID: 39491490 PMCID: PMC11743020 DOI: 10.1111/jcpe.14087] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
AIM This study aimed to investigate the hypoxic changes in periodontal tissues resulting from smoking and periodontitis by assessing levels of hypoxia-inducible factors (HIF-1α, HIF-2α, HIF-3α) and vascular endothelial growth factor (VEGF) in gingival crevicular fluid (GCF). MATERIALS AND METHODS The study comprised 22 periodontally healthy non-smokers (Group H), 22 periodontally healthy smokers (Group HS), 22 non-smokers with periodontitis (Group P) and 22 smokers with periodontitis (Group PS). Clinical periodontal parameters were documented, and GCF samples were collected and analysed using enzyme-linked immunosorbent assay (ELISA). RESULTS Significantly elevated levels of HIF-1α, HIF-3α and VEGF were observed in Groups HS, P and PS compared to Group H (p < 0.05). Moreover, higher HIF-2α levels were detected in the Groups HS and P compared to Group H (p < 0.05). Significant correlations were detected between all evaluated hypoxia biomarkers in the Group P (p < 0.05) except HIF-2α and HIF-3α. However, in the PS group, significant correlation appeared only between HIF-1α and HIF-2α (p < 0.05). CONCLUSION Our findings indicate that smoking and periodontitis induce comparable hypoxic effects in periodontal tissues, as evidenced by the evaluated biomarkers. Further research is warranted to gain a deeper understanding of the mechanisms underlying hypoxia in periodontal tissues.
Collapse
Affiliation(s)
- Bejna Bozyel
- Department of PeriodontologyFaculty of Dentistry, Gazi UniversityAnkaraTurkey
| | - Özlem Doğan
- Department of Medical BiochemistryFaculty of Medicine, Ankara UniversityAnkaraTurkey
| | - Serenay Elgün
- Department of Medical BiochemistryFaculty of Medicine, Ankara UniversityAnkaraTurkey
| | - Burcu Özdemir
- Department of PeriodontologyFaculty of Dentistry, Gazi UniversityAnkaraTurkey
| |
Collapse
|
57
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
58
|
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
|
59
|
Gong L, Zou C, Zhang H, Yang F, Qi G, Ma Z. Landscape of Noncoding RNA in the Hypoxic Tumor Microenvironment. Genes (Basel) 2025; 16:140. [PMID: 40004471 PMCID: PMC11855738 DOI: 10.3390/genes16020140] [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: 01/06/2025] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Amidst the prevalent and notable characteristic of a hypoxic microenvironment present in the majority of solid tumors, a burgeoning number of studies have revealed the significance of noncoding RNAs (ncRNAs) in hypoxic tumor regions. The transcriptome of cancers is highly heterogeneous, with noncoding transcripts playing crucial roles. Long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs) are two distinctive classes of ncRNA that are garnering increasing attention. Biologically, they possess intriguing properties and possess significant regulatory functions. Clinically, they present as promising biomarkers and therapeutic targets. Additionally, recent research has evaluated the clinical applications of these ncRNAs in RNA-based treatments and noninvasive liquid biopsies. This review provides a comprehensive summary of recent studies on lncRNAs and circRNAs within the hypoxic tumor microenvironment. Furthermore, the clinical significance of lncRNAs and circRNAs in cancer diagnosis and treatment is emphasized, which could pave the way for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (L.G.); (C.Z.); (H.Z.); (F.Y.); (G.Q.)
| |
Collapse
|
60
|
Rogers RS, Mootha VK. Hypoxia as a medicine. Sci Transl Med 2025; 17:eadr4049. [PMID: 39841808 DOI: 10.1126/scitranslmed.adr4049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025]
Abstract
Oxygen is essential for human life, yet a growing body of preclinical research is demonstrating that chronic continuous hypoxia can be beneficial in models of mitochondrial disease, autoimmunity, ischemia, and aging. This research is revealing exciting new and unexpected facets of oxygen biology, but translating these findings to patients poses major challenges, because hypoxia can be dangerous. Overcoming these barriers will require integrating insights from basic science, high-altitude physiology, clinical medicine, and sports technology. Here, we explore the foundations of this nascent field and outline a path to determine how chronic continuous hypoxia can be safely, effectively, and practically delivered to patients.
Collapse
Affiliation(s)
- Robert S Rogers
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02114, USA
| |
Collapse
|
61
|
Shi Y, Gilkes DM. HIF-1 and HIF-2 in cancer: structure, regulation, and therapeutic prospects. Cell Mol Life Sci 2025; 82:44. [PMID: 39825916 PMCID: PMC11741981 DOI: 10.1007/s00018-024-05537-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/27/2024] [Accepted: 12/01/2024] [Indexed: 01/20/2025]
Abstract
Hypoxia, or a state of low tissue oxygenation, has been characterized as an important feature of solid tumors that is related to aggressive phenotypes. The cellular response to hypoxia is controlled by Hypoxia-inducible factors (HIFs), a family of transcription factors. HIFs promote the transcription of gene products that play a role in tumor progression including proliferation, angiogenesis, metastasis, and drug resistance. HIF-1 and HIF-2 are well known and widely described. Although these proteins share a high degree of homology, HIF-1 and HIF-2 have non-redundant roles in cancer. In this review, we summarize the similarities and differences between HIF-1α and HIF-2α in their structure, expression, and DNA binding. We also discuss the canonical and non-canonical regulation of HIF-1α and HIF-2α under hypoxic and normal conditions. Finally, we outline recent strategies aimed at targeting HIF-1α and/or HIF-2α.
Collapse
Affiliation(s)
- Yi Shi
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele M Gilkes
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
62
|
Wang S, Xu Q, Liu W, Zhang N, Qi Y, Tang F, Ge R. Regulation of PHD2 by HIF-1α in Erythroid Cells: Insights into Erythropoiesis Under Hypoxia. Int J Mol Sci 2025; 26:762. [PMID: 39859474 PMCID: PMC11765976 DOI: 10.3390/ijms26020762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
The hypoxia-inducible factor (HIF) pathway has been demonstrated to play a pivotal role in the process of high-altitude adaptation. PHD2, a key regulator of the HIF pathway, has been found to be associated with erythropoiesis. However, the relationship between changes in Phd2 abundance and erythroid differentiation under hypoxic conditions remains to be elucidated. A hemin-induced K562 erythroid differentiation model was used to explore the effects of PHD2 knockdown under hypoxia. Erythroid differentiation was assessed by flow cytometry and immunofluorescence. HIF-1α's regulation of PHD2 was examined using luciferase assays and ChIP-seq. CRISPR/Cas9 was applied to knock out EGLN1 and HIF1A, and a fluorescent reporter system was developed to track PHD2 expression. PHD2 knockdown enhanced erythroid differentiation, evident by increased CD71 and CD235a expression. Reporter assays and ChIP-seq identified an HIF-1α binding site in the EGLN1 5' UTR, confirming HIF-1α as a regulator of PHD2 expression. The fluorescent reporter system provided real-time monitoring of endogenous PHD2 expression, showing that HIF-1α significantly modulates PHD2 levels under hypoxic conditions. PHD2 influences erythropoiesis under hypoxia, with HIF-1α regulating its expression. This feedback loop between HIF-1α and PHD2 sheds light on mechanisms driving erythroid differentiation under low-oxygen conditions.
Collapse
Affiliation(s)
- Shunjuan Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Qiying Xu
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Wenjing Liu
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Na Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Yuelin Qi
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Feng Tang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810016, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining 810016, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810016, China
| |
Collapse
|
63
|
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
|
64
|
Liu NN, Pan LJ, Xiao ZJ, Xu ZF. [Familial erythrocytosis type 2 due to VHL germline mutations: a case report and literature review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2025; 46:75-80. [PMID: 40059686 PMCID: PMC11886440 DOI: 10.3760/cma.j.cn121090-20241011-00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Indexed: 03/14/2025]
Abstract
To enhance the understanding of familial erythrocytosis type 2 (ECYT2) resulting from compound heterozygous mutations in the VHL gene. Methods: We conducted a retrospective analysis of the case data from a patient with ECYT2 to investigate its pathogenesis, clinical features, diagnosis and treatment options, as well as prognosis, while also reviewing the relevant literature. Results: A 31-year-old man was admitted to the hospital due to facial and hand flushing that had persisted for 29 years. Whole exome sequencing revealed compound heterozygous mutations in VHL p.P81L and p.N90T. Both of his parents were found to carry only one of these heterozygous mutations, yet they exhibited normal phenotypes. Based on the patient's hematological tests, a clear diagnosis of ECYT2 was established. Following treatment with erythrocytapheresis and daily administration of aspirin at a dosage of 100 mg, the patient experienced relief from dizziness and headaches associated with blood hyperviscosity, without any thrombotic or bleeding complications during this period. Conclusions: ECYT2 is a rare group of autosomal recessive genetic disorders. This case of ECYT2, resulting from compound heterozygous mutations in the VHL gene, represents the first report in China. Clinically, it is characterized by elevated red cell mass, normal or increased serum erythropoietin levels, and normal hemoglobin oxygen affinity levels. These factors contribute to thrombotic and bleeding complications that can lead to early mortality.
Collapse
Affiliation(s)
- N N Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China Tianjin Institutes of Health Science, Tianjin 301600, China
| | - L J Pan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Z J Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Z F Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
65
|
Wu Y, Zhang Z, Cai H, Zhang W, Zhang L, Li Z, Yang L, Chen Y, Corner TP, Song Z, Yue J, Yang F, Li X, Schofield CJ, Zhang X. Discovery of ZG-2305, an Orally Bioavailable Factor Inhibiting HIF Inhibitor for the Treatment of Obesity and Fatty Liver Disease. J Med Chem 2025; 68:212-235. [PMID: 39432709 DOI: 10.1021/acs.jmedchem.4c01698] [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] [Indexed: 10/23/2024]
Abstract
Genetic loss of the 2-oxoglutarate oxygenase factor inhibiting hypoxia-inducible factor (FIH) enhances both glycolysis and aerobic metabolism. FIH is thus a potential target for adiposity control and improving hepatic steatosis. We describe development of a series of novel, potent, and selective FIH inhibitors that occupy both the FIH catalytic site and a recently defined tyrosine conformational-flip pocket. ZG-2305, with a Ki of 79.6 nM for FIH, manifests 38-fold selectivity over the hypoxia-inducible factor (HIF) prolyl hydroxylase PHD2. Oral administration of ZG-2305 in the western-diet induced obesity mouse model results in improved lipid accumulation and recovery from abnormal body weight/hepatic steatosis. Amelioration of nonalcoholic steatohepatitis (NASH) related pathological phenotypes in the HF-CDAA-diet induced NASH mouse model was observed. Preliminary preclinical studies indicate ZG-2305 has good pharmacokinetic properties and an acceptable safety profile. The results imply ZG-2305 is a promising candidate for treatment of obesity and fatty liver disease.
Collapse
Affiliation(s)
- Yue Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Zewei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Haiping Cai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Weiqing Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Linjian Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Zhihong Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Le Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yafen Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Thomas P Corner
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Zhe Song
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Yue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Fulai Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Xiaojin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
66
|
Heidarian Y, Fasteen TD, Mungcal L, Buddika K, Mahmoudzadeh NH, Nemkov T, D'Alessandro A, Tennessen JM. Hypoxia-inducible factor 1α is required to establish the larval glycolytic program in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631819. [PMID: 39829828 PMCID: PMC11741260 DOI: 10.1101/2025.01.07.631819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The rapid growth that occurs during Drosophila larval development requires a dramatic rewiring of central carbon metabolism to support biosynthesis. Larvae achieve this metabolic state, in part, by coordinately up-regulating the expression of genes involved in carbohydrate metabolism. The resulting metabolic program exhibits hallmark characteristics of aerobic glycolysis and establishes a physiological state that supports growth. To date, the only factor known to activate the larval glycolytic program is the Drosophila Estrogen-Related Receptor (dERR). However, dERR is dynamically regulated during the onset of this metabolic switch, indicating that other factors must be involved. Here we discover that Sima, the Drosophila ortholog of Hif1α, is also essential for establishing the larval glycolytic program. Using a multi-omics approach, we demonstrate that sima mutants fail to properly activate aerobic glycolysis and die during larval development with metabolic defects that phenocopy dERR mutants. Moreover, we demonstrate that dERR and Sima/Hif1α protein accumulation is mutually dependent, as loss of either transcription factor results in decreased abundance of the other protein. Considering that the mammalian homologs of ERR and Hif1α also cooperatively regulate aerobic glycolysis in cancer cells, our findings establish the fly as a powerful genetic model for studying the interaction between these two key metabolic regulators.
Collapse
Affiliation(s)
- Yasaman Heidarian
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Tess D Fasteen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Liam Mungcal
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
- Affiliate Member, Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA
| |
Collapse
|
67
|
Ahn JH, da Silva Pedrosa M, Lopez LR, Tibbs TN, Jeyachandran JN, Vignieri EE, Rothemich A, Cumming I, Irmscher AD, Haswell CJ, Zamboni WC, Yu YRA, Ellermann M, Denson LA, Arthur JC. Intestinal E. coli-produced yersiniabactin promotes profibrotic macrophages in Crohn's disease. Cell Host Microbe 2025; 33:71-88.e9. [PMID: 39701098 DOI: 10.1016/j.chom.2024.11.012] [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: 05/09/2024] [Revised: 11/11/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
Inflammatory bowel disease (IBD)-associated fibrosis causes significant morbidity. Mechanisms are poorly understood but implicate the microbiota, especially adherent-invasive Escherichia coli (AIEC). We previously demonstrated that AIEC producing the metallophore yersiniabactin (Ybt) promotes intestinal fibrosis in an IBD mouse model. Since macrophages interpret microbial signals and influence inflammation/tissue remodeling, we hypothesized that Ybt metal sequestration disrupts this process. Here, we show that macrophages are abundant in human IBD-fibrosis tissue and mouse fibrotic lesions, where they co-localize with AIEC. Ybt induces profibrotic gene expression in macrophages via stabilization and nuclear translocation of hypoxia-inducible factor 1-alpha (HIF-1α), a metal-dependent immune regulator. Importantly, Ybt-producing AIEC deplete macrophage intracellular zinc and stabilize HIF-1α through inhibition of zinc-dependent HIF-1α hydroxylation. HIF-1α+ macrophages localize to sites of disease activity in human IBD-fibrosis strictures and mouse fibrotic lesions, highlighting their physiological relevance. Our findings reveal microbiota-mediated metal sequestration as a profibrotic trigger targeting macrophages in the inflamed intestine.
Collapse
Affiliation(s)
- Ju-Hyun Ahn
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marlus da Silva Pedrosa
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lacey R Lopez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Taylor N Tibbs
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joanna N Jeyachandran
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emily E Vignieri
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Aaron Rothemich
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ian Cumming
- Department of Pulmonary and Critical Care Medicine, Duke University, Durham, NC 27710, USA
| | - Alexander D Irmscher
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Corey J Haswell
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C Zamboni
- UNC Advanced Translational Pharmacology and Analytical Chemistry Lab, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yen-Rei A Yu
- Department of Pulmonary and Critical Care Medicine, Duke University, Durham, NC 27710, USA; Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Melissa Ellermann
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Lee A Denson
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Janelle C Arthur
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
68
|
Zhang J, Yao M, Xia S, Zeng F, Liu Q. Systematic and comprehensive insights into HIF-1 stabilization under normoxic conditions: implications for cellular adaptation and therapeutic strategies in cancer. Cell Mol Biol Lett 2025; 30:2. [PMID: 39757165 DOI: 10.1186/s11658-024-00682-7] [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: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are essential transcription factors that orchestrate cellular responses to oxygen deprivation. HIF-1α, as an unstable subunit of HIF-1, is usually hydroxylated by prolyl hydroxylase domain enzymes under normoxic conditions, leading to ubiquitination and proteasomal degradation, thereby keeping low levels. Instead of hypoxia, sometimes even in normoxia, HIF-1α translocates into the nucleus, dimerizes with HIF-1β to generate HIF-1, and then activates genes involved in adaptive responses such as angiogenesis, metabolic reprogramming, and cellular survival, which presents new challenges and insights into its role in cellular processes. Thus, the review delves into the mechanisms by which HIF-1 maintains its stability under normoxia including but not limited to giving insights into transcriptional, translational, as well as posttranslational regulation to underscore the pivotal role of HIF-1 in cellular adaptation and malignancy. Moreover, HIF-1 is extensively involved in cancer and cardiovascular diseases and potentially serves as a bridge between them. An overview of HIF-1-related drugs that are approved or in clinical trials is summarized, highlighting their potential capacity for targeting HIF-1 in cancer and cardiovascular toxicity related to cancer treatment. The review provides a comprehensive insight into HIF-1's regulatory mechanism and paves the way for future research and therapeutic development.
Collapse
Affiliation(s)
- Jiayi Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Mingxuan Yao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shiting Xia
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
69
|
Rosenblum JS, Cole Y, Dang D, Lookian PP, Alkaissi H, Patel M, Cappadona AJ, Jha A, Edwards N, Donahue DR, Munasinghe J, Wang H, Knutsen RH, Pappo AS, Lechan RM, Kozel BA, Smirniotopoulos JG, Kim HJ, Vortmeyer A, Miettinen M, Heiss JD, Zhuang Z, Pacak K. Head and neck paraganglioma in Pacak-Zhuang syndrome. JNCI Cancer Spectr 2025; 9:pkaf001. [PMID: 39821441 PMCID: PMC11790058 DOI: 10.1093/jncics/pkaf001] [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: 06/02/2024] [Revised: 11/15/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Head and neck paragangliomas (HNPGLs) are typically slow-growing, hormonally inactive tumors of parasympathetic paraganglia. Inactivation of prolyl-hydroxylase domain-containing 2 protein causing indirect gain-of-function of hypoxia-inducible factor-2α (HIF-2α), encoded by EPAS1, was recently shown to cause carotid body hyperplasia. We previously described a syndrome with multiple sympathetic paragangliomas caused by direct gain-of-function variants in EPAS1 (Pacak-Zhuang syndrome, PZS) and developed a corresponding mouse model. METHODS We evaluated a cohort of patients with PZS (n = 9) for HNPGL by positron emission tomography, magnetic resonance imaging, and computed tomography and measured carotid body size compared to literature reference values. Resected tumors were evaluated by histologic sectioning and staining. We evaluated the corresponding mouse model at multiple developmental stages (P8 and adult) for lesions of the head and neck by high resolution ex vivo imaging and performed immunohistochemical staining on histologic sections of the identified lesions. RESULTS hree patients had imaging consistent with HNPGL, one of which warranted resection and was confirmed on histology. Three additional patients had carotid body enlargement (Z-score > 2.0), and 3 had carotid artery malformations. We found that 9 of 10 adult variant mice had carotid body tumors and 6 of 8 had a paraganglioma on the cranio-caval vein, the murine homologue of the superior vena cava; these were also found in 4 of 5 variant mice at post-natal day 8. These tumors and the one resected from a patient were positive for tyrosine hydroxylase, synaptophysin, and chromogranin A. Brown fat in a resected patient tumor carried the EPAS1 pathogenic variant. CONCLUSIONS These findings (1) suggest HNPGL as a feature of PZS and (2) show that these pathogenic variants are sufficient to cause the development of these tumors, which we believe represents a continuous spectrum of disease starting from hyperplasia.
Collapse
Affiliation(s)
- Jared S Rosenblum
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yasemin Cole
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Danielle Dang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Pashayar P Lookian
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hussam Alkaissi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mayank Patel
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- General Surgical Pathology Section, National Institutes of Health, Bethesda, MD, United States
| | - Anthony J Cappadona
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Nancy Edwards
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Danielle R Donahue
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jeeva Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Herui Wang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Russell H Knutsen
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alberto S Pappo
- Division of Solid Tumor, St Jude Children’s Research Hospital, Memphis, TN, United States
| | - Ronald M Lechan
- Division of Endocrinology, Diabetes & Metabolism, Tufts Medical Center, Boston, MA, United States
| | - Beth A Kozel
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - James G Smirniotopoulos
- Department of Radiology, George Washington University, Washington, DC, United States
- MedPix® National Library of Medicine, Bethesda, MD, United States
| | - H Jeffrey Kim
- Department of Otolaryngology, Georgetown University School of Medicine, Washington, DC, United States
- Office of Clinical Director, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, United States
| | - Alexander Vortmeyer
- Clinical Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Markku Miettinen
- General Surgical Pathology Section, National Institutes of Health, Bethesda, MD, United States
| | - John D Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- Center for Adrenal Endocrine Tumors, AKESO, Prague 5, Czech Republic
| |
Collapse
|
70
|
Zhang Q, Gu R, Dai Y, Chen J, Ye P, Zhu H, He W, Nie X. Molecular mechanisms of ubiquitination in wound healing. Biochem Pharmacol 2025; 231:116670. [PMID: 39613112 DOI: 10.1016/j.bcp.2024.116670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Wound healing is a complex biological process involving multiple cellular and molecular mechanisms. Ubiquitination, a crucial post-translational modification, plays a vital role in regulating various aspects of wound healing through protein modification and degradation. This review comprehensively examines the molecular mechanisms of ubiquitination in wound healing, focusing on its regulation of inflammatory responses, macrophage polarization, angiogenesis, and the activities of fibroblasts and keratinocytes. We discuss how ubiquitination modifies key signaling pathways, including TGF-β/Smad3, NF-κB, and HIF-α, which are essential for proper wound healing. Understanding these mechanisms provides insights into potential therapeutic strategies for treating impaired wound healing, particularly in conditions such as diabetes. The review highlights recent advances in understanding ubiquitination's role in wound healing and discusses future research directions for developing targeted therapeutic approaches.
Collapse
Affiliation(s)
- Qianbo Zhang
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Rifang Gu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; School Medical Office, Zunyi Medical University, Zunyi 563006, PR China.
| | - Yuhe Dai
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Jitao Chen
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Penghui Ye
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Huan Zhu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Wenping He
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, PR China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, PR China.
| |
Collapse
|
71
|
Amin N, Wu F, Zhao BX, Shi Z, Abdelsadik A, Elshazly Younis A, Naz Abbasi I, Sundus J, Badry Hussein A, Geng Y, Fang M. Hif-1α ablation reduces the efficiency of NeuroD1 gene-based therapy and aggravates the brain damage following ischemic stroke. Expert Opin Drug Deliv 2025; 22:121-138. [PMID: 39632618 DOI: 10.1080/17425247.2024.2435458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Hypoxia-inducible factor 1α [HIF1α] regulates gene expression, allowing the organism to respond to low oxygen levels. Meanwhile, astrocytes participate in inflammatory processes and are associated with neurotoxic chemicals that can increase stroke volume, contributing considerably to the devastating effects of a stroke. OBJECTIVE To evaluate whether Hif-1α ablation from the central nervous system is implicated in motor dysfunction and ischemic brain damage following stroke. Furthermore, to explore if Hif-1α ablation affects the therapeutic impact of NeuroD1 gene-based therapy. METHODS Endothelin-1 [ET-1] was injected to induce ischemic stroke in mice. Both wild-type and Hypoxia-inducible factor 1α conditional knockout [Hif-1α CKO] mice were used. The effect of Hif-1α ablation was assessed by the neuron numbers, astrocyte activity, vascular endothelial growth factor [VEGF] expression, and behavioral tests. Moreover, western blot, ELISA, and RNA sequencing were used. Then, we used pAAV2/9-GfaABC1D-NeuroD1-P2A-EGFP-WPRE injection to examine the impact of NeuroD1 in Hif-1α CKO mice following ischemic stroke. RESULTS We found that following stroke, motor dysfunction significantly increased in Hif-1α CKO mice. Furthermore, elevation of apoptosis and activation in both microglia and astrocytes were observed, consequently up-regulating neuroinflammation. Meanwhile, Hif-1α ablation significantly decreased the efficiency of NeuroD1 gene-based therapy. CONCLUSION Our findings demonstrate that Hif-1α ablation from the nervous system is implicated in ischemic stroke pathogenesis mainly by increasing neuron cell death and inducing astrocytes as well as decreasing the efficiency of NeuroD1. These data support the idea that manipulating HIF-1α is a viable therapeutic for ischemic stroke.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Department of Neurology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Fei Wu
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ahmed Abdelsadik
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
- Laboratory of Immunometabolism, Aswan University, Aswan, Egypt
| | | | - Irum Naz Abbasi
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Javaria Sundus
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry Hussein
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Marong Fang
- Department of Neurology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou, China
- Institute of System Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
72
|
Archontakis-Barakakis P, Mavridis T, Chlorogiannis DD, Barakakis G, Laou E, Sessler DI, Gkiokas G, Chalkias A. Intestinal oxygen utilisation and cellular adaptation during intestinal ischaemia-reperfusion injury. Clin Transl Med 2025; 15:e70136. [PMID: 39724463 DOI: 10.1002/ctm2.70136] [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: 08/28/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The gastrointestinal tract can be deranged by ailments including sepsis, trauma and haemorrhage. Ischaemic injury provokes a common constellation of microscopic and macroscopic changes that, together with the paradoxical exacerbation of cellular dysfunction and death following restoration of blood flow, are collectively known as ischaemia-reperfusion injury (IRI). Although much of the gastrointestinal tract is normally hypoxemic, intestinal IRI results when there is inadequate oxygen availability due to poor supply (pathological hypoxia) or abnormal tissue oxygen use and metabolism (dysoxia). Intestinal oxygen uptake usually remains constant over a wide range of blood flows and pressures, with cellular function being substantively compromised when ischaemia leads to a >50% decline in intestinal oxygen consumption. Restoration of perfusion and oxygenation provokes additional injury, resulting in mucosal damage and disruption of intestinal barrier function. The primary cellular mechanism for sensing hypoxia and for activating a cascade of cellular responses to mitigate the injury is a family of heterodimer proteins called hypoxia-inducible factors (HIFs). The HIF system is connected to numerous biochemical and immunologic pathways induced by IRI and the concentration of those proteins increases during hypoxia and dysoxia. Activation of the HIF system leads to augmented transcription of specific genes in various types of affected cells, but may also augment apoptotic and inflammatory processes, thus aggravating gut injury. KEY POINTS: During intestinal ischaemia, mitochondrial oxygen uptake is reduced when cellular oxygen partial pressure decreases to below the threshold required to maintain normal oxidative metabolism. Upon reperfusion, intestinal hypoxia may persist because microcirculatory flow remains impaired and/or because available oxygen is consumed by enzymes, intestinal cells and neutrophils.
Collapse
Affiliation(s)
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital incorporating the National Children's Hospital (AMNCH), Dublin, Ireland
| | | | - Georgios Barakakis
- Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, Athens, Greece
| | - Daniel I Sessler
- Center for Outcomes Research and Department of Anesthesiology, UTHealth, Houston, Texas, USA
- Outcomes Research Consortium®, Houston, Texas, USA
| | - George Gkiokas
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Chalkias
- Outcomes Research Consortium®, Houston, Texas, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Critical Care Medicine, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
73
|
Nishizawa H, Funasaki S, Ma W, Kubota Y, Watanabe K, Arima Y, Kuroda S, Ito T, Furuya M, Motoshima T, Nishiyama A, Mehanna S, Satou Y, Hasumi H, Jikuya R, Makiyama K, Tamura T, Oike Y, Tanaka Y, Suda T, Schmidt LS, Linehan WM, Baba M, Kamba T. HIF1α Plays a Crucial Role in the Development of TFE3-Rearranged Renal Cell Carcinoma by Orchestrating a Metabolic Shift Toward Fatty Acid Synthesis. Genes Cells 2025; 30:e13195. [PMID: 39807625 PMCID: PMC11729263 DOI: 10.1111/gtc.13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Tumor development often requires cellular adaptation to a unique, high metabolic state; however, the molecular mechanisms that drive such metabolic changes in TFE3-rearranged renal cell carcinoma (TFE3-RCC) remain poorly understood. TFE3-RCC, a rare subtype of RCC, is defined by the formation of chimeric proteins involving the transcription factor TFE3. In this study, we analyzed cell lines and genetically engineered mice, demonstrating that the expression of the chimeric protein PRCC-TFE3 induced a hypoxia-related signature by transcriptionally upregulating HIF1α and HIF2α. The upregulation of HIF1α by PRCC-TFE3 led to increased cellular ATP production by enhancing glycolysis, which also supplied substrates for the TCA cycle while maintaining mitochondrial oxidative phosphorylation. We crossed TFE3-RCC mouse models with Hif1α and/or Hif2α knockout mice and found that Hif1α, rather than Hif2α, is essential for tumor development in vivo. RNA-seq and metabolomic analyses of the kidney tissues from these mice revealed that ketone body production is inversely correlated with tumor development, whereas de novo lipid synthesis is upregulated through the HIF1α/SREBP1-dependent mechanism in TFE3-RCC. Our data suggest that the coordinated metabolic shift via the PRCC-TFE3/HIF1α/SREBP1 axis is a key mechanism by which PRCC-TFE3 enhances cancer cell metabolism, promoting tumor development in TFE3-RCC.
Collapse
Grants
- JP21K19721 Japan Society for the Promotion of Science
- HHSN261201500003C NCI NIH HHS
- JP24K09315 Japan Society for the Promotion of Science
- JP 24K02578 Japan Society for the Promotion of Science
- JPMXP0618217493 Ministry of Education, Culture, Sports, Science and Technology
- JP20K09560 Japan Society for the Promotion of Science
- JPMXP0622717006 Ministry of Education, Culture, Sports, Science and Technology
- JP21K09374 Japan Society for the Promotion of Science
- JP23K24474 Japan Society for the Promotion of Science
- JP21K06000 Japan Society for the Promotion of Science
- HHSN261201500003I NCI NIH HHS
- JP23K27589 Japan Society for the Promotion of Science
- JPMXP0723833149 Ministry of Education, Culture, Sports, Science and Technology
- Japan Society for the Promotion of Science
- Ministry of Education, Culture, Sports, Science and Technology
- National Cancer Institute
Collapse
Affiliation(s)
- Hidekazu Nishizawa
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Shintaro Funasaki
- Divison of Molecular and Vascular Biology, IRDAKumamoto UniversityKumamotoJapan
| | - Wenjuan Ma
- Cambridge Stem Cell Institute, University of CambridgeCambridgeUK
| | - Yoshiaki Kubota
- Department of AnatomyInstitute for Advanced Medical Research and Keio University School of MedicineTokyoJapan
| | | | - Yuichiro Arima
- Developmental Cardiology Laboratory, International Research Center for Medical Science (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Shoichiro Kuroda
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Takaaki Ito
- Department of Medical TechnologyKumamoto Health Science University Faculty of Health SciencesKumamotoJapan
| | - Mitsuko Furuya
- Department of Surgical PathologyHokkaido University HospitalSapporoJapan
| | - Takanobu Motoshima
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Akira Nishiyama
- Department of ImmunologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Sally Mehanna
- Biotechnology Department, Faculty of Nanotechnology for Postgraduate Studies, Cairo UniversityAd DoqiEgypt
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus InfectionKumamoto UniversityKumamotoJapan
| | - Hisashi Hasumi
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Ryosuke Jikuya
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Kazuhide Makiyama
- Department of UrologyYokohama City University Graduate School of MedicineKanagawaJapan
| | - Tomohiko Tamura
- Department of ImmunologyYokohama City University Graduate School of MedicineKanagawaJapan
- Advanced Medical Research CenterYokohama City UniversityKanagawaJapan
| | - Yuichi Oike
- Department of Molecular Genetics, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Toshio Suda
- Laboratory of Stem Cell Regulation, International Research Center for Medical Science (IRCMS)Kumamoto UniversityKumamotoJapan
| | - Laura S. Schmidt
- Urologic Oncology BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
- Basic Science Program, Frederick National Laboratory for Cancer ResearchNational Cancer InstituteFrederickMarylandUSA
| | - W. Marston Linehan
- Urologic Oncology BranchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Masaya Baba
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tomomi Kamba
- Department of Urology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
74
|
Seo SH, Lee JH, Choi EK, Rho SB, Yoon K. C/EBPβ Regulates HIF-1α-Driven Invasion of Non-Small-Cell Lung Cancer Cells. Biomolecules 2024; 15:36. [PMID: 39858431 PMCID: PMC11764306 DOI: 10.3390/biom15010036] [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: 11/22/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Metastatic cancer accounts for most cancer-related deaths, and identifying specific molecular targets that contribute to metastatic progression is crucial for the development of effective treatments. Hypoxia, a feature of solid tumors, plays a role in cancer progression by inducing resistance to therapy and accelerating metastasis. Here, we report that CCAAT/enhancer-binding protein beta (C/EBPβ) transcriptionally regulates hypoxia-inducible factor 1 subunit alpha (HIF1A) and thus promotes migration and invasion of non-small-cell lung cancer (NSCLC) cells under hypoxic conditions. Our results show that knockdown or forced expression of C/EBPβ was correlated with HIF-1α expression and that C/EBPβ directly bound to the promoter region of HIF1A. Silencing HIF1A inhibited the enhanced migration and invasion induced by C/EBPβ overexpression in NSCLC cells under hypoxia. Expression of the HIF-1α target gene, SLC2A1, was also altered in a C/EBPβ-dependent manner, and knockdown of SLC2A1 reduced migration and invasion enhanced by C/EBPβ overexpression. These results indicate that C/EBPβ is a critical regulator for the invasion of NSCLC cells in the hypoxic tumor microenvironment. Collectively, the C/EBPβ-HIF-1α-GLUT1 axis represents a potential therapeutic target for preventing metastatic progression of NSCLC and improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Kyungsil Yoon
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.H.S.); (J.H.L.); (S.B.R.)
| |
Collapse
|
75
|
Caca J, Bartelt A, Egea V. Hypoxia Regulates Brown Adipocyte Differentiation and Stimulates miR-210 by HIF-1α. Int J Mol Sci 2024; 26:117. [PMID: 39795975 PMCID: PMC11720532 DOI: 10.3390/ijms26010117] [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: 12/06/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
MicroRNAs (miRNAs) are short sequences of single-stranded non-coding RNAs that target messenger RNAs, leading to their repression or decay. Interestingly, miRNAs play a role in the cellular response to low oxygen levels, known as hypoxia, which is associated with reactive oxygen species and oxidative stress. However, the physiological implications of hypoxia-induced miRNAs ("hypoxamiRs") remain largely unclear. Here, we investigate the role of miR-210 in brown adipocyte differentiation and thermogenesis. We treated the cells under sympathetic stimulation with hypoxia, CoCl2, or IOX2. To manipulate miR-210, we performed reverse transfection with antagomiRs. Adipocyte markers expression, lipid accumulation, lipolysis, and oxygen consumption were measured. Hypoxia hindered BAT differentiation and suppressed sympathetic stimulation. Hypoxia-induced HIF-1α stabilization increased miR-210 in brown adipocytes. Interestingly, miR-210-5p enhanced differentiation under normoxic conditions but was insufficient to rescue the inhibition of brown adipocyte differentiation under hypoxic conditions. Although adrenergic stimulation activated HIF-1α signaling and upregulated miR-210 expression, inhibition of miR-210-5p did not significantly influence UCP1 expression or oxygen consumption. In summary, hypoxia and adrenergic stimulation upregulated miR-210, which impacted brown adipocyte differentiation and thermogenesis. These findings offer new insights for the physiological role of hypoxamiRs in brown adipose tissue, which could aid in understanding oxidative stress and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jan Caca
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research, 85764 Neuherberg, Germany
- Chair of Translational Nutritional Medicine, Department of Molecular Life Sciences, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, 85354 Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
- German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| |
Collapse
|
76
|
Lavilla-Puerta M, Giuntoli B. Designed to breathe: synthetic biology applications in plant hypoxia. PLANT PHYSIOLOGY 2024; 197:kiae623. [PMID: 39673416 DOI: 10.1093/plphys/kiae623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 12/16/2024]
Abstract
Over the past years, plant hypoxia research has produced a considerable number of new resources to monitor low oxygen responses in model species, mainly Arabidopsis thaliana. Climate change urges the development of effective genetic strategies aimed at improving plant resilience during flooding events. This need pushes forward the search for optimized tools that can reveal the actual oxygen available to plant cells, in different organs or under various conditions, and elucidate the mechanisms underlying plant hypoxic responses, complementing the existing transcriptomics, proteomics, and metabolic analysis methods. Oxygen-responsive reporters, dyes, and nanoprobes are under continuous development, as well as novel synthetic strategies that make precision control of plant hypoxic responses realistic. In this review, we summarize the recent progress made in the definition of tools for oxygen response monitoring in plants, either adapted from bacterial and animal research or peculiar to plants. Moreover, we highlight how adoption of a synthetic biology perspective has enabled the design of novel genetic circuits for the control of oxygen-dependent responses in plants. Finally, we discuss the current limitations and challenges toward the implementation of synbio solutions in the plant low-oxygen biology field.
Collapse
Affiliation(s)
- Mikel Lavilla-Puerta
- Plant Molecular Biology Section, Department of Biology, University of Oxford, OX1 3RB Oxford, UK
| | | |
Collapse
|
77
|
Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, El-kott AF, AlShehri MA, Morsy K, Negm S, Kira AY. Nanoscale Systems for Local Activation of Hypoxia-Inducible Factor-1 Alpha: A New Approach in Diabetic Wound Management. Int J Nanomedicine 2024; 19:13735-13762. [PMID: 39723173 PMCID: PMC11669355 DOI: 10.2147/ijn.s497041] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/03/2024] [Indexed: 12/28/2024] Open
Abstract
Chronic wounds in diabetic patients experience significant clinical challenges due to compromised healing processes. Hypoxia-inducible factor-1 alpha (HIF-1α) is a critical regulator in the cellular response to hypoxia, enhancing angiogenesis and tissue restoration. Nevertheless, the cellular response to the developed chronic hypoxia within diabetes is impaired, likely due to the destabilization of HIF-1α via degradation by prolyl hydroxylase domain (PHD) enzymes. Researchers have extensively explored HIF-1α activation as a potential pathway for diabetic wound management, focusing mainly on deferoxamine (DFO) as a potent agent to stabilize HIF-1α. This review provides an update of the other recent pharmacological agents managing HIF-1α activation, including novel PHD inhibitors (roxadustat and daprodustat) and Von Hippel-Lindau protein (VHL) antagonists, which could be potential alternatives for the local treatment of diabetic wounds. Furthermore, it highlights how localized delivery via advanced nanostructures can enhance the efficacy of these novel therapies. Importantly, by addressing these points, the current review can offer a promising area for research. Given that, these novel drugs have minimal applications in diabetic wound healing, particularly in the context of local application through nanomaterials. This gap presents an exciting opportunity for further investigation, as combining these drugs with localized nanotechnology could avoid undesired systemic side effects and sustain drug release within wound site, offering a transformative platform for diabetes wound treatment.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, 63514, Egypt
| | - Mai A Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M20 4BX, UK
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Attalla F El-kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, Abha, 62529, Saudi Arabia
| | - Ahmed Y Kira
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| |
Collapse
|
78
|
Pauzaite T, Nathan JA. A closer look at the role of deubiquitinating enzymes in the Hypoxia Inducible Factor pathway. Biochem Soc Trans 2024; 52:2253-2265. [PMID: 39584532 PMCID: PMC11668284 DOI: 10.1042/bst20230861] [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: 08/27/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024]
Abstract
Hypoxia Inducible transcription Factors (HIFs) are central to the metazoan oxygen-sensing response. Under low oxygen conditions (hypoxia), HIFs are stabilised and govern an adaptive transcriptional programme to cope with prolonged oxygen starvation. However, when oxygen is present, HIFs are continuously degraded by the proteasome in a process involving prolyl hydroxylation and subsequent ubiquitination by the Von Hippel Lindau (VHL) E3 ligase. The essential nature of VHL in the HIF response is well established but the role of other enzymes involved in ubiquitination is less clear. Deubiquitinating enzymes (DUBs) counteract ubiquitination and provide an important regulatory aspect to many signalling pathways involving ubiquitination. In this review, we look at the complex network of ubiquitination and deubiquitination in controlling HIF signalling in normal and low oxygen tensions. We discuss the relative importance of DUBs in opposing VHL, and explore roles of DUBs more broadly in hypoxia, in both VHL and HIF independent contexts. We also consider the catalytic and non-catalytic roles of DUBs, and elaborate on the potential benefits and challenges of inhibiting these enzymes for therapeutic use.
Collapse
Affiliation(s)
- Tekle Pauzaite
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah, Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, U.K
| | - James A. Nathan
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah, Biomedical Centre, Department of Medicine, University of Cambridge, Cambridge CB2 0AW, U.K
| |
Collapse
|
79
|
Abdallah M, Voland R, Decamp M, Flickinger J, Pacioles T, Jamil M, Silbermins D, Shenouda M, Valsecchi M, Bir A, Shweihat Y, Bastidas J, Chowdhury N, Kachynski Y, Eldib H, Wright T, Mahdi A, Al-Nusair J, Nwanwene K, Varlotto J. Evaluation of Anti-Angiogenic Therapy Combined with Immunotherapy and Chemotherapy as a Strategy to Treat Locally Advanced and Metastatic Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:4207. [PMID: 39766108 PMCID: PMC11674749 DOI: 10.3390/cancers16244207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/23/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has made recent improvements in disease-free survival (DFS) and/or overall survival (OS) in all stages of non-small-cell lung cancer (NSCLC). Here, we review the tumor microenvironment and its immunosuppressive effects and discuss how anti-angiogenic therapies may potentiate the anti-carcinogenic effects of immunotherapy. We also review all the past literature and discuss strategies of combining anti-angiogenic therapy and immunotherapy +/- chemotherapy and hypothesize how we can use this strategy for non-small-cell lung cancer in metastatic previously untreated/previously treated settings in previously treated EGFR-mutated NSCLC for the upfront treatment of brain metastases prior to radiation therapy and for the incorporation of this strategy into stage III unresectable disease. We assert the use of anti-angiogenic therapy and immunotherapy when combined appropriately with chemotherapy and radiotherapy has the potential to increase the long-term survivals in both the stage III and metastatic setting so that we can now consider more patients to experience curative treatment.
Collapse
Affiliation(s)
- Mahmoud Abdallah
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Rick Voland
- Department of Ophthalmology, University of Wisconsin, Madison, WI 53705, USA;
| | - Malcolm Decamp
- Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
| | - John Flickinger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA;
| | - Toni Pacioles
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Muhammad Jamil
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Damian Silbermins
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Mina Shenouda
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Matias Valsecchi
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Arvinder Bir
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Yousef Shweihat
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Juan Bastidas
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Nepal Chowdhury
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Yury Kachynski
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Howide Eldib
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Thomas Wright
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Ahmad Mahdi
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Jowan Al-Nusair
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Kemnasom Nwanwene
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - John Varlotto
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| |
Collapse
|
80
|
Zhang X, Meng Z, Yang C, Wang C, Zhang K, Shi A, Guo J, Feng Y, Zeng Y. miR-210 loss leads to widespread phenotypic and gene expression changes in human 293T cells. Front Genet 2024; 15:1486252. [PMID: 39737000 PMCID: PMC11683127 DOI: 10.3389/fgene.2024.1486252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Hypoxia responses are critical for myriad physiological and pathological processes, such as development, tissue repair, would healing, and tumorigenesis. microRNAs (miRNAs) are a class of small non-coding RNAs that exert their functions by inhibiting the expression of their target genes, and miR-210 is the miRNA universally and most conspicuously upregulated by hypoxia in mammalian systems. For its relationship to hypoxia, miR-210 has been studied extensively, yet no consensus exists on the roles and mechanisms of miR-210 in human physiological processes or diseases, and we know little about genuine miR-210 target genes in humans. Methods To better investigate the functions and mechanisms of human miR-210, therefore, we derived the human miR-210 gene knockout (KO) 293T cell lines using the CRISPR/Cas9 technology. We then examined the cellular phenotypes and gene expression profiles of 293T cells under normoxia and hypoxia conditions. Results and Discussion We found that the loss of miR-210 altered a variety of cellular phenotypes including proliferation and apoptosis. Subsequent global gene expression analyses identified plausible mechanisms underlying these phenotypic changes in 293T cells. In particular, we showed that miR-210 might target the expression of BNIP3L as a potential mechanism to suppress apoptosis. Surprisingly, the mRNA levels of most previously reported miR-210 target genes were not induced upon miR-210 KO, suggesting a need to reexamining and studying human miR-210 functions directly and comprehensively. Thus, our work established a human cellular system and opportunity to unravel the complexity of the regulatory networks by miR-210.
Collapse
Affiliation(s)
- Xiaoxiao Zhang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Zhen Meng
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Chengyong Yang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Chenghao Wang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Kexin Zhang
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Anxin Shi
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Yong Feng
- Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yan Zeng
- Department of Zoology, College of Life Sciences, Nanjing Agricultural University, Nanjing, Jiangsu, China
| |
Collapse
|
81
|
Yang J, Li Y, Huang Y, Chen H, Sui P. Unlocking lung regeneration: insights into progenitor cell dynamics and metabolic control. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:31. [PMID: 39676102 PMCID: PMC11646969 DOI: 10.1186/s13619-024-00212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Regenerative responses are particularly important in the lungs, which are critical for gas exchange and frequently challenged by environmental insults. The lung progenitor cells play a central role in the lung regeneration response, and their dysfunction is associated with various lung diseases. Understanding the mechanisms regulating lung progenitor cell function is essential for developing new therapeutic approaches to promote lung regeneration. This review summarizes recent advancements in the field of lung regeneration, focusing on the metabolic control of lung progenitor cell function. We discuss cell lineage plasticity and cell-cell signaling under different physiological conditions. Additionally, we highlight the connection between progenitor cell dysfunction and lung diseases, emphasizing the need to develop new therapeutic strategies in regenerative medicine to improve lung regenerative capacity.
Collapse
Affiliation(s)
- Jiaying Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yawen Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ying Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huaiyong Chen
- Department of Basic Medicine, Tianjin University Haihe Hospital, Tianjin, 300350, China.
- Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin, China.
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin, China.
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin, China.
| | - Pengfei Sui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
82
|
Moreira ET, Lourenço MP, Cunha-Fernandes T, Silva TI, Siqueira LD, Castro-Faria-Neto HC, Reis PA. Minocycline inhibits microglial activation in the CA1 hippocampal region and prevents long-term cognitive sequel after experimental cerebral malaria. J Neuroimmunol 2024; 397:578480. [PMID: 39504755 DOI: 10.1016/j.jneuroim.2024.578480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
Cerebral malaria is the worst complication of malaria infection, has a high mortality rate, and may cause different neurodysfunctions, including cognitive decline. Neuroinflammation is an important cause of cognitive damage in neurodegenerative diseases, and microglial cells can be activated in a disease-associated profile leading to tissue damage and neuronal death. Here, we demonstrated that treatment with minocycline reduced blood-brain barrier breakdown and modulated ICAM1 mRNA expression; reduced proinflammatory cytokines, such as TNF-α, IL-1β, IFN-γ, and IL-6; and prevented long-term cognitive decline in contextual and aversive memory tasks. Taken together, our data suggest that microglial cells are activated during experimental cerebral malaria, leading to neuroinflammatory events that end up in cognitive damage. In addition, pharmacological modulation of microglial activation, by drugs such as minocycline may be an important therapeutic strategy in the prevention of long-term memory impairment.
Collapse
Affiliation(s)
- E T Moreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Universidade Cruzeiro do Sul, Brazil; Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - M P Lourenço
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - T Cunha-Fernandes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - T I Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - L D Siqueira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - H C Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - P A Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
83
|
Zhang Z, Wang D, Xu R, Li X, Wang Z, Zhang Y. The Physiological Functions and Therapeutic Potential of Hypoxia-Inducible Factor-1α in Vascular Calcification. Biomolecules 2024; 14:1592. [PMID: 39766299 PMCID: PMC11674127 DOI: 10.3390/biom14121592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
HIF-1α plays a crucial regulatory role in vascular calcification (VC), primarily influencing the osteogenic differentiation of VSMCs through oxygen-sensing mechanisms. Under hypoxic conditions, the stability of HIF-1α increases, avoiding PHD and VHL protein-mediated degradation, which promotes its accumulation in cells and then activates gene expressions related to calcification. Additionally, HIF-1α modulates the metabolic state of VSMCs by regulating the pathways that govern the switch between glycolysis and oxidative phosphorylation, thereby further advancing the calcification process. The interaction between HIF-1α and other signaling pathways, such as nuclear factor-κB, Notch, and Wnt/β-catenin, creates a complex regulatory network that serves as a critical driving force in VC. Therefore, a deeper understanding of the role and regulatory mechanism of the HIF-1α signaling during the development and progression of VC is of great significance, as it is not only a key molecular marker for understanding the pathological mechanisms of VC but also represents a promising target for future anti-calcification therapies.
Collapse
Affiliation(s)
- Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China; (Z.Z.); (R.X.)
| | - Defan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen 361102, China;
| | - Renfeng Xu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China; (Z.Z.); (R.X.)
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA;
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China; (Z.Z.); (R.X.)
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA;
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA;
| |
Collapse
|
84
|
Zhang X, Zhang H, Dong Q, Qin Y, Cao Y, Zhu H, Ma Z, Li Z, Rao Z, Ning P, Tian Z, Xia Y, Yang P, Wang Z. Bypassing Ca 2+ Influx for Antimetastasis Photodynamic Therapy via Robust Nucleus-Targeted Near-Infrared Cyanines. NANO LETTERS 2024; 24:15817-15826. [PMID: 39584561 DOI: 10.1021/acs.nanolett.4c04789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Hypoxia-induced tumor metastasis severely hinders the efficacy of photodynamic therapy (PDT) in cancer treatment. Current strategies predominantly offer palliative suppression of the HIF-1α pathway, emphasizing the urgent need for innovative PDT approaches to prevent metastasis from the outset. Our study revealed that typical PDT triggers an increase in cytoplasmic Ca2+ levels, activating HIF-1α, and that reducing Ca2+ levels can, in turn, mitigate metastasis. Considering cytoplasm's role in Ca2+ storage and regulation, we propose that PDT-induced metastasis can be addressed at its source by precise intracellular localization of photosensitizers (PSs). We developed near-infrared (NIR) cyanine PSs with inherent nucleus targeting capabilities. These PSs effectively inhibit cytoplasmic Ca2+ elevation and reduce HIF-1α activity upon irradiation, achieving remarkable antimetastatic effects in 4T1 tumors. Consequently, our findings highlight the pivotal role of Ca2+ in PDT-induced metastasis and provide a robust approach for circumventing metastasis from the outset using new nucleus-targeting organic PSs.
Collapse
Affiliation(s)
- Xianghan Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Huaicong Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Qunyan Dong
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yuan Qin
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yutian Cao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Haixing Zhu
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zimeng Ma
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zehua Li
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhiping Rao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Pengbo Ning
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zuhong Tian
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yuqiong Xia
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Peng Yang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
85
|
Huo G, Lin Y, Liu L, He Y, Qu Y, Liu Y, Zhu R, Wang B, Gong Q, Han Z, Yin H. Decoding ferroptosis: transforming orthopedic disease management. Front Pharmacol 2024; 15:1509172. [PMID: 39712490 PMCID: PMC11659002 DOI: 10.3389/fphar.2024.1509172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
As a mechanism of cell death, ferroptosis has gained popularity since 2012. The process is distinguished by iron toxicity and phospholipid accumulation, in contrast to autophagy, apoptosis, and other cell death mechanisms. It is implicated in the advancement of multiple diseases across the body. Researchers currently know that osteosarcoma, osteoporosis, and other orthopedic disorders are caused by NRF2, GPX4, and other ferroptosis star proteins. The effective relief of osteoarthritis symptoms from deterioration has been confirmed by clinical treatment with multiple ferroptosis inhibitors. At the same time, it should be reminded that the mechanisms involved in ferroptosis that regulate orthopedic diseases are not currently understood. In this manuscript, we present the discovery process of ferroptosis, the mechanisms involved in ferroptosis, and the role of ferroptosis in a variety of orthopedic diseases. We expect that this manuscript can provide a new perspective on clinical diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Guanlin Huo
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Yi Qu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, The Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Bo Wang
- Department of Orthopaedics, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Qing Gong
- Orthopaedic Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhongyu Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongbing Yin
- Orthopedic Center, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
86
|
Ide H, Miike K, Ohmori T, Maruyama K, Izumi Y, Tanigawa S, Nishinakamura R. Mouse embryonic kidney transplantation identifies maturation defects in the medulla. Sci Rep 2024; 14:30293. [PMID: 39639083 PMCID: PMC11621804 DOI: 10.1038/s41598-024-81984-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Kidney organoids are connected to the host circulation and mature after transplantation. However, they are still immature compared to the adult kidneys, and their precise maturation stages remain unclear. By transplanting the mouse embryonic kidney as a model system for organoid transplantation, we report here the maturation defects of the graft, especially in the medulla. Single cell profiling of the developing kidneys in vivo identified gene sets associated with the maturation of the collecting duct epithelium and medullary stroma. These data revealed an upregulation of genes associated with channel/transporter functions and immune defense, as well as a downregulation of neuronal genes. Using these marker genes, we found that the maturation of the collecting duct and medullary stroma in the grafts barely corresponds to the perinatal stage, which was confirmed histologically by using representative genes. Thus, the gene sets obtained serve as maturation coordinates for the renal medulla and will be helpful in analyzing its maturation defects after transplantation. They will also provide a useful basis for further maturation of transplanted kidney organoids.
Collapse
Affiliation(s)
- Hiroshi Ide
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Koichiro Miike
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Tomoko Ohmori
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Kosuke Maruyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Shunsuke Tanigawa
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| |
Collapse
|
87
|
Alshehri B. Cytochrome c and cancer cell metabolism: A new perspective. Saudi Pharm J 2024; 32:102194. [PMID: 39564377 PMCID: PMC11570848 DOI: 10.1016/j.jsps.2024.102194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024] Open
Abstract
Cytochrome c is a vital electron carrier in the mitochondrial respiratory chain. When the outer membrane of mitochondria becomes permeable, cytochrome c is discharged into the cytoplasm, where it initiates the intrinsic apoptosis pathway. The complex interaction between cytochrome c and apoptosis protease-activating factor-1 (Apaf-1) leads to the formation of the apoptosome and activation of a cascade of caspases, highlighting the critical role of cytochrome c in controlling cell death mechanisms. Additionally, cytochrome c undergoes post-translational modifications, especially phosphorylation, which intricately regulate its roles in both respiration and apoptosis. These modifications add layers of complexity to how cytochrome c effectively controls cellular functions. cytochrome c becomes a lighthouse in the intricate web of cancer, its expression patterns providing hints about prognosis and paths toward treatment. Reduced levels of cytochrome c have been observed in cancer tissues, indicating a potential inhibition of apoptosis. For instance, in glioma tissues, cytochrome c levels were lower compared to healthy tissues, and this reduction became more pronounced in advanced stages of the disease. However, the role of cytochrome c in cancer becomes more intricate as it becomes intertwined with the metabolic reprogramming of cancer cells. This suggests that cytochrome c plays a crucial role in tumor progression and resistance to treatment. Viewing cytochrome c as a molecular mosaic reveals that it is not merely a protein, but also a central player in determining cellular fate. This realization opens up exciting avenues for potential advancements in cancer diagnosis and treatment strategies. Despite the advancements made, the narrative surrounding cytochrome c remains incomplete, urging further exploration into its complexities and the biological implications linked to cancer. cytochrome c stands as a beacon of hope and a promising target for therapy in the battle against cancer, particularly due to its significant involvement in tumor metabolism. It holds the potential for a future where innovative solutions can be developed to address the intricate challenges of cellular fate. In this review, we have endeavored to illuminate the multifaceted domain of cytochrome c drawing connections among apoptosis, metabolic reprogramming, and the Warburg effect in the context of cancer.
Collapse
Affiliation(s)
- Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah-11952, Saudi Arabia
| |
Collapse
|
88
|
Otsuka E, Kitamura M, Funakoshi S, Mukae H, Nishino T. Roxadustat has risks of reversible central hypothyroidism in patients undergoing hemodialysis: a single-center retrospective cohort study. Ren Fail 2024; 46:2410375. [PMID: 39378117 PMCID: PMC11463015 DOI: 10.1080/0886022x.2024.2410375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
Roxadustat, a hypoxia-inducible factor-prolyl hydroxylase inhibitor, has proven efficacy in the treatment of renal anemia; however, evidence indicates that it may cause central hypothyroidism. The prevalence and reversibility of roxadustat-induced central hypothyroidism in patients undergoing hemodialysis remain unclear. Here, we retrospectively analyzed thyroid-stimulating hormone (TSH), free thyroxine (FT4), and free triiodothyronine (FT3) levels in 51 patients (mean age: 72.3 ± 10.7 years; 58.8% male) undergoing hemodialysis before, during, and after halting roxadustat treatment. TSH levels were significantly decreased from a median of 2.46 (interquartile range:1.60-4.51) mU/L before roxadustat treatment to 1.36 (0.72-2.41) mU/L during treatment (p < 0.001), and improved to 2.56 (1.78-4.63) mU/L after halting roxadustat (p < 0.001). Similarly, FT4 levels decreased from 1.11 (0.97-1.24) ng/dL before roxadustat treatment to 0.92 (0.71-1.03) ng/dL during treatment (p < 0.001) and improved to 1.05 (0.93-1.17) ng/dL after halting roxadustat (p < 0.001). FT3 levels were 2.04 (1.78-2.31) pg/mL before starting roxadustat, 1.97 (1.69-2.27) pg/mL during treatment, and 1.90 (1.63-2.18) pg/mL after halting roxadustat, with no significant difference between each group. Moreover, 2.0% of patients exhibited extremely low TSH levels (≤0.1 mU/L) and low TSH levels (>0.1 mU/L to <0.4 mU/L) before starting roxadustat and that percentage increased to 5.9% and 7.8%, respectively, during treatment. After roxadustat cessation, extremely low or low TSH levels recovered in all patients. Taken together, the results indicate that roxadustat can cause reversible central hypothyroidism in patients undergoing hemodialysis.
Collapse
Affiliation(s)
- Emiko Otsuka
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Nagasaki Renal Center, Nagasaki, Japan
| | - Mineaki Kitamura
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Nagasaki Renal Center, Nagasaki, Japan
| | | | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
89
|
Pathak S, Singh V, Kumar G N, Jayandharan GR. AAV-mediated combination gene therapy of inducible Caspase 9 and miR-199a-5p is therapeutic in hepatocellular carcinoma. Cancer Gene Ther 2024; 31:1796-1803. [PMID: 39385010 DOI: 10.1038/s41417-024-00844-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Advanced-stage hepatocellular carcinoma (HCC) remains an untreatable disease with an overall survival of less than one year. One of the critical molecular mediators contributing to increased resistance to therapy and relapse, is increased hypoxia-inducible factor 1α (HIF-1α) levels, leading to metastasis of tumor cells. Several microRNAs are known to be dysregulated and impact HIF-1α expression in HCC. An in silico analysis demonstrated that hsa-miR-199a-5p is downregulated at various stages of HCC and is known to repress HIF-1α expression. Based on this analysis, we developed a combinatorial suicide gene therapy by employing hepatotropic Adeno-associated virus-based vectors encoding an inducible caspase 9 (iCasp9) and miR-199a. The overexpression of miR-199a-5p alone significantly decreased ( ~ 2-fold vs. Mock treated cells, p < 0.05) HIF-1α mRNA levels, with a concomitant increase in cancer cell cytotoxicity in Huh7 cells in vitro and in xenograft models in vivo. To further enhance the efficacy of gene therapy, we evaluated the synergistic therapeutic effect of AAV8-miR-199a and AAV6-iCasp9 in a xenograft model of HCC. Our data revealed that mice receiving combination suicide gene therapy exhibited reduced expression of HIF-1α ( ~ 4-fold vs. Mock, p < 0.001), with a significant reduction in tumor growth when compared to mock-treated animals. These findings underscore the therapeutic potential of downregulating HIF-1α during suicide gene therapy for HCC.
Collapse
Affiliation(s)
- Subhajit Pathak
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Vijayata Singh
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Narendra Kumar G
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Giridhara R Jayandharan
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India.
| |
Collapse
|
90
|
Mei T, Ye T, Huang D, Xie Y, Xue Y, Zhou D, Wang W, Chen J. Triggering immunogenic death of cancer cells by nanoparticles overcomes immunotherapy resistance. Cell Oncol (Dordr) 2024; 47:2049-2071. [PMID: 39565509 DOI: 10.1007/s13402-024-01009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
Immunotherapy resistance poses a significant challenge in oncology, necessitating novel strategies to enhance the therapeutic efficacy. Immunogenic cell death (ICD), including necroptosis, pyroptosis and ferroptosis, triggers the release of tumor-associated antigens and numerous bioactive molecules. This release can potentiate a host immune response, thereby overcoming resistance to immunotherapy. Nanoparticles (NPs) with their biocompatible and immunomodulatory properties, are emerging as promising vehicles for the delivery of ICD-inducing agents and immune-stimulatory adjuvants to enhance immune cells tumoral infiltration and augment immunotherapy efficacy. This review explores the mechanisms underlying immunotherapy resistance, and offers an in-depth examination of ICD, including its principles and diverse modalities of cell death that contribute to it. We also provide a thorough overview of how NPs are being utilized to trigger ICD and bolster antitumor immunity. Lastly, we highlight the potential of NPs in combination with immunotherapy to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Ting Mei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Ye
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dingkun Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Yuxiu Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Ying Xue
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, 430022, China.
- Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
91
|
Woubshete M, Chan LI, Diallinas G, Byrne B. The dimer of human SVCT1 is key for transport function. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184390. [PMID: 39369805 DOI: 10.1016/j.bbamem.2024.184390] [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: 05/07/2024] [Revised: 08/06/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Humans and other primates lack the ability to synthesize the essential nutrient, Vitamin C, which is derived exclusively from the diet. Crucial for effective vitamin C uptake are the Na+ dependent Vitamin C transporters, SVCT1 and SVCT2, members of the nucleobase ascorbate transporter (NAT) family. SVCT1 and 2 actively transport the reduced form of Vitamin C, ascorbic acid, into key tissues. The recent structure of the mouse SVCT1 revealed the molecular basis of substrate binding and that, like the other structurally characterised members of the NAT family, it exists as a closely associated dimer. SVCT1 is likely to function via the elevator mechanism with the core domain of each protomer able to bind substrate and move through the membrane carrying the substrate across the membrane. Here we explored the function of a range of variants of the human SVCT1, revealing a range of residues involved in substrate selection and binding, and confirming the importance of the C-terminus in membrane localisation. Furthermore, using a dominant negative mutant we show that the dimer is essential for transport function, as previously seen in the fungal homologue, UapA. In addition, we show that a localisation deficient C-terminal truncation of SVCT1 blocks correct localisation of co-expressed, associated wildtype SVCT1. These results clearly show the importance of the dimer in both correct SVCT1 trafficking and transport activity.
Collapse
Affiliation(s)
- Menebere Woubshete
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Lok I Chan
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - George Diallinas
- Department of Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15784 Athens, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 70013 Heraklion, Greece
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
92
|
He J, Yu Y, Liu W, Li Z, Qi Z, Weng S, Guo C, He J. Molecular mechanism of infectious spleen and kidney necrosis virus in manipulating the hypoxia-inducible factor pathway to augment virus replication. Virulence 2024; 15:2349027. [PMID: 38680083 PMCID: PMC11085990 DOI: 10.1080/21505594.2024.2349027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
Infectious spleen and kidney necrosis virus (ISKNV), a member of the genus Megalocytivirus in the family Iridoviridae, can infect over 50 fish species and cause significant economic losses in Asia. Our previous study showed that hypoxia triggers the hypoxia-inducible factor pathway (HIF-pathway), leading to increased replication of ISKNV through promoting the upregulation of viral hypoxic response genes like orf077r. This study delved into the molecular mechanism of how ISKNV manipulates the HIF-pathway to enhance its replication. In vitro and in vivo experiments confirmed that ISKNV infection activated the HIF-pathway, which in turn promoted ISKNV replication. These findings suggest that ISKNV actively manipulates the HIF-pathway. Co-immunoprecipitation experiments revealed that the ISKNV-encoded protein VP077R interacts with the Von Hippel-Lindau (VHL) protein at the HIF-binding region, competitively inhibiting the interaction of HIF-1α with VHL. This prevents HIF degradation and activates the HIF-pathway. Furthermore, VP077R interacts with factor-inhibiting HIF (FIH), recruiting FIH and S-phase kinase-associated protein 1 (Skp1) to form an FIH - VP077R - Skp1 complex. This complex promotes FIH protein degradation via ubiquitination, further activating the HIF-pathway. These findings indicated that ISKNV takes over the HIF-pathway by releasing two "brakes" on this pathway (VHL and FIH) via VP077R, facilitating virus replication. We speculate that hypoxia initiates a positive feedback loop between ISKNV VP077R and the HIF pathway, leading to the outbreak of ISKNV disease. This work offers valuable insights into the complex interactions between the environment, host, and virus.
Collapse
Affiliation(s)
- Jian He
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yang Yu
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Wenhui Liu
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Zhimin Li
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Zhang Qi
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Changjun Guo
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- State Key Laboratory for Biocontrol/Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Guangdong Provincial Observation and Research Station for Marine Ranching of the Lingdingyang Bay, School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| |
Collapse
|
93
|
Frazer R, Arranz JÁ, Estévez SV, Parikh O, Krabbe LM, Vasudev NS, Doehn C, Marschner N, Waddell T, Ince W, Goebell PJ. Tivozanib Monotherapy in the Frontline Setting for Patients with Metastatic Renal Cell Carcinoma and Favorable Prognosis. Curr Oncol Rep 2024; 26:1639-1650. [PMID: 39565522 PMCID: PMC11646210 DOI: 10.1007/s11912-024-01613-7] [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] [Accepted: 10/05/2024] [Indexed: 11/21/2024]
Abstract
PURPOSE OF REVIEW In this review, we discuss which patients with metastatic clear cell renal cell carcinoma (mRCC) may be most suitable for frontline tyrosine kinase inhibitor (TKI) monotherapy, a treatment option supported by emerging long-term efficacy data including overall survival and quality of life. We specifically focus on tivozanib, a potent and selective inhibitor of vascular endothelial growth factor receptor, which has comparable efficacy to other single-agent TKIs in frontline treatment for mRCC while exhibiting fewer off-target side effects. RECENT FINDINGS Combination therapy with TKIs and checkpoint inhibitors (CPIs) and CPI/CPI combination therapies, as well as TKI monotherapy are recommended frontline treatment options for mRCC. Treatment decisions are complex and based on several factors, including the patient's International Metastatic RCC Database Consortium risk status, age, comorbidities, and personal preferences related to response, tolerability, and quality of life. TKIs not only serve as backbone of most combination therapies for mRCC, but also remain a viable monotherapy option in the first-line setting for patients in favorable risk groups and those with contraindications to CPI combination therapies. Given that overall survival benefits have not yet been confirmed for CPI-containing combination regimens in favorable risk patients, we argue that frontline single-agent TKI treatment remains a standard of care option for these patients. This is supported by treatment guidelines, even in the era of TKI/CPI combination therapies.
Collapse
Affiliation(s)
| | | | | | - Omi Parikh
- Lancashire Teaching Hospitals NHS Foundation Trust, Chorley, UK
| | | | | | | | | | - Tom Waddell
- The Christie NHS Foundation Trust, Manchester, UK
| | - Will Ince
- Department of Oncology, Cambridge University NHS Foundation Trust, Cambridge, UK
| | - Peter J Goebell
- Uniklinik Erlangen, Urologische und Kinderurologische Klinik, Erlangen, Germany
| |
Collapse
|
94
|
Bechmann N, Rosenblum JS, Alzahrani AS. Current views on the role of HIF-2α in the pathogenesis and syndromic presentation of pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab 2024; 38:101955. [PMID: 39426935 DOI: 10.1016/j.beem.2024.101955] [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] [Indexed: 10/21/2024]
Abstract
Pathogenic variants (PVs) in EPAS1, which encodes hypoxia-inducible factor-2α (HIF-2α), could be the underlying genetic cause of about 3%-6% of pheochromocytoma and paragangliomas (PPGLs). EPAS1-related PPGLs may occur as isolated tumors or as part of Pacak-Zhuang Syndrome (PZS) with two or more of a triad of PPGL, polycythemia, and somatostatinoma. HIF-2α plays a critical role in the regulation of the cellular hypoxia pathway. When a gain-of-function PV is acquired, HIF-2α evades steady-state hydroxylation by the prolyl hydroxylase type 2 (PHD2), which accelerates von Hippel-Lindau (VHL)-mediated proteasomal degradation. In this situation, HIF-2α is stabilized and can translocate to the nucleus, inducing the expression of several genes involved in tumorigenesis. This leads to the development of PPGL and other manifestations of PZS. EPAS1-related PPGLs usually occur in the second or third decade of life, more frequently in females, and are usually multiple, adrenal and extra-adrenal, and norepinephrine-secreting. In addition, these tumors carry an increased metastatic potential and have been reported with metastatic disease in up to 30% of cases. While polycythemia is fairly common in PZS, somatostatinomas are rare. It has been suggested that the character of the acquired PV in EPAS1, which affects its binding to PHD2, correlates with certain phenotypes in PZS. PVs in EPAS1 that have been found in related sporadic PPGLs have also been associated with hypoxic conditions including cyanotic congenital heart disease, hemoglobinopathies and high altitude. Understanding the hypoxia pathway and its role in the pathogenesis of PPGL may open a new avenue for developing effective therapies for these tumors. Indeed, one of these therapies is Belzutifan, a HIF-2α inhibitor that is being tested in the treatment of metastatic PPGLs.
Collapse
Affiliation(s)
- Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Eunice Kennedy Shriver National Institute of Child Health and Development, Bethesda, MD 20892, United States; Department of Medicine and Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia.
| | - Jared S Rosenblum
- Eunice Kennedy Shriver National Institute of Child Health and Development, Bethesda, MD 20892, United States.
| | - Ali S Alzahrani
- Department of Medicine and Department of Molecular Oncology, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia.
| |
Collapse
|
95
|
Schreiber T, Scharner B, Thévenod F. Insoluble HIFa protein aggregates by cadmium disrupt hypoxia-prolyl hydroxylase (PHD)-hypoxia inducible factor (HIFa) signaling in renal epithelial (NRK-52E) and interstitial (FAIK3-5) cells. Biometals 2024; 37:1629-1642. [PMID: 39256317 PMCID: PMC11618182 DOI: 10.1007/s10534-024-00631-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: 06/15/2024] [Accepted: 08/24/2024] [Indexed: 09/12/2024]
Abstract
The kidney is the main organ that senses changes in systemic O2 pressure by hypoxia-PHD-HIFa (HPH) signaling, resulting in adaptive target gene activation, including erythropoietin (EPO). The non-essential transition metal cadmium (Cd) is nephrotoxic and disrupts the renal HPH pathway, which may promote Cd-associated chronic renal disease (CKD). A deeper molecular understanding of Cd interference with renal HPH signaling is missing, and no data with renal cell lines are available. In rat kidney NRK-52E cells, which model the proximal tubule, and murine fibroblastoid atypical interstitial kidney (FAIK3-5) cells, which mimic renal EPO-producing cells, the chemical hypoxia mimetic dimethyloxalylglycine (DMOG; 1 mmol/l) or hypoxia (1% O2) activated HPH signaling. Cd2+ (2.5-20 µmol/l for ≤ 24 h) preferentially induced necrosis (trypan blue uptake) of FAIK3-5 cells at high Cd whereas NRK-52E cells specially developed apoptosis (PARP-1 cleavage) at all Cd concentrations. Cd (12.5 µmol/l) abolished HIFa stabilization and prevented upregulation of target genes (quantitative real-time polymerase chain reaction and immunoblotting) induced by DMOG or hypoxia in both cell lines, which was caused by the formation of insoluble HIFa aggregates. Strikingly, hypoxic preconditioning (1% O2 for 18 h) reduced apoptosis of FAIK3-5 and NRK-52E cells at low Cd concentrations and decreased insoluble HIFa proteins. Hence, drugs mimicking hypoxic preconditioning could reduce CKD induced by chronic low Cd exposure.
Collapse
Affiliation(s)
- Timm Schreiber
- Institute of Physiology and Pathophysiology and ZBAF, Faculty of Health, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany.
| | - Bettina Scharner
- Institute of Physiology and Pathophysiology and ZBAF, Faculty of Health, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
| | - Frank Thévenod
- Institute of Physiology and Pathophysiology and ZBAF, Faculty of Health, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany.
- Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany.
| |
Collapse
|
96
|
Carullo N, Sorbo D, Faga T, Pugliese S, Zicarelli MT, Costa D, Ielapi N, Battaglia Y, Pisani A, Coppolino G, Bolignano D, Michael A, Serra R, Andreucci M. Anemia and Mineral Bone Disorder in Kidney Disease Patients: The Role of FGF-23 and Other Related Factors. Int J Mol Sci 2024; 25:12838. [PMID: 39684548 DOI: 10.3390/ijms252312838] [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: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Anemia and mineral and bone disorder (MBD) are significant complications of chronic kidney disease (CKD). The erythropoietin (Epo) pathway plays a key role in both of these processes in CKD. Another molecule that plays an important role in CKD-MBD is fibroblast growth factor (FGF)-23, whose main role is to maintain serum phosphate levels in the normal range, acting via its co-receptor Klotho; however, its activity may also be related to anemia and inflammation. In this review, the regulation of Epo and FGF-23 and the molecular mechanisms of their action are outlined. Furthermore, the complex interaction between EPO and FGF-23 is discussed, as well as their association with other anemia-related factors and processes such as Klotho, vitamin D, and iron deficiency. Together, these may be part of a "kidney-bone marrow-bone axis" that promotes CKD-MBD.
Collapse
Affiliation(s)
- Nazareno Carullo
- "G. Jazzolino" Hospital, A.S.P. Vibo Valentia, I89900 Vibo Valentia, Italy
| | - David Sorbo
- San Bortolo Hospital, ULSS 8 Berica, I36100 Vicenza, Italy
| | - Teresa Faga
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Sara Pugliese
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Maria Teresa Zicarelli
- Amantea Outpatient Clinic, A.S.P. Cosenza, I87032 Amantea, Italy
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Costa
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, I00185 Rome, Italy
| | - Yuri Battaglia
- Department of Medicine, University of Verona, I37129 Verona, Italy
| | - Antonio Pisani
- Department of Public Health, University of Naples Federico II, I80131 Naples, Italy
| | - Giuseppe Coppolino
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Ashour Michael
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, I88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences, "Magna Graecia" University, I88100 Catanzaro, Italy
| |
Collapse
|
97
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
98
|
Wang X, Lu Y, Zhao R, Zhu B, Liu J, Yue Q, Wu R, Han S, Gao Y, Chen J, Gong J, He D, Xu T, Ying J. Global surveillance of circulating microRNA for diagnostic and prognostic assessment of acute myocardial infarction based on the plasma small RNA sequencing. Biomark Res 2024; 12:143. [PMID: 39563415 DOI: 10.1186/s40364-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Circulating microRNAs (miRNAs) are recently a rapidly increasing of interest as non-invasive biomarkers for diagnosis and prognosis of acute myocardial infarction (AMI). Previous studies revealed that several miRNAs exhibited the capacity for diagnosis and prognosis of AMI, the reasons why these circulating miRNAs are concerned as targets for investigation are quite cryptogenic, presumably due to the lack of clues provided by global surveillance at the transcriptome level, and the current data for some miRNAs are controversial and inconsistent among independent studies. METHODS To comprehensively profiling the potential miRNAs for diagnosis and prognosis of AMI, we reported transcriptomes of circulating miRNAs in the plasma of 27 healthy controls, 64 AMI patients (37 STEMI and 27 NSTEMI) and 20 AMI patients who were subjected to reperfusion therapy. Meanwhile, the cTnI of AMI patients was parallel determined. Differentially-circulated miRNAs were analyzed between each group. All detected circulating miRNAs were examined by ROC analysis and then LASSO dimension reduction to obtain an optimal panel for diagnosis of AMI. A five-year period follow-up towards the AMI and reperfusion patients was performed, and the prognostic value of circulating miRNAs in these patients was estimated by using the Cox regression model, ROC and Kaplan-Meier curves. RESULTS Comprehensive global differences of miRNAs transcriptome among AMI, reperfusion patients and healthy controls were identified. A total of 40 miRNAs, called high diagnostic performance miRNAs, including several previous well-studied miRNAs with AUC greater than 0.85 were shown to discriminate AMI with healthy controls. In addition, 29 miRNAs were analyzed to be strongly correlated with the plasma cTnI level, of which 20 overlapped with high diagnostic performance miRNAs. These overlapped miRNAs are over-represented in the pathways which actually reflect the pathological cause of myocardial infarction, as well as the regulation of gene expression and energetic pathway of cellular response to hypoxia. Finally, two miRNAs were analyzed to be significantly correlated to all-cause mortality. CONCLUSION This is the first time to survey plasma miRNAs for the development of AMI diagnostic and prognostic biomarkers at the transcriptome level. A subset of miRNAs exhibited potential diagnostic and prognostic merits for AMI.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
- Institute of Translational Medicine, Baotou Central Hospital, Baotou, China
| | - Yaojun Lu
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Ruiping Zhao
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
- Institute of Translational Medicine, Baotou Central Hospital, Baotou, China
| | - Bing Zhu
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
- Institute of Translational Medicine, Baotou Central Hospital, Baotou, China
| | - Jian Liu
- Dian Diagnostics Group Co., Ltd, Hangzhou, China
| | - Qiang Yue
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Rina Wu
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Shuwen Han
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Yuanyuan Gao
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Juan Chen
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Jie Gong
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Danna He
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China
| | - Teng Xu
- Department of Cardiology/Chest Pain Center, Baotou Central Hospital, Baotou, China.
- Institute of Translational Medicine, Baotou Central Hospital, Baotou, China.
| | - Jianchao Ying
- Wenzhou Key Laboratory of Emergency, Critical Care, and Disaster Medicine/Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
99
|
Soroush A, Dunn JF. A Hypoxia-Inflammation Cycle and Multiple Sclerosis: Mechanisms and Therapeutic Implications. Curr Treat Options Neurol 2024; 27:6. [PMID: 39569339 PMCID: PMC11573864 DOI: 10.1007/s11940-024-00816-4] [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] [Accepted: 10/24/2024] [Indexed: 11/22/2024]
Abstract
Purpose of Review Multiple sclerosis (MS) is a complex neurodegenerative disease characterized by inflammation, demyelination, and neurodegeneration. Significant hypoxia exists in brain of people with MS (pwMS), likely contributing to inflammatory, neurodegenerative, and vascular impairments. In this review, we explore the concept of a negative feedback loop between hypoxia and inflammation, discussing its potential role in disease progression based on evidence of hypoxia, and its implications for therapeutic targets. Recent Findings In the experimental autoimmune encephalomyelitis (EAE) model, hypoxia has been detected in gray matter (GM) using histological stains, susceptibility MRI and implanted oxygen sensitive probes. In pwMS, hypoxia has been quantified using near-infrared spectroscopy (NIRS) to measure cortical tissue oxygen saturation (StO2), as well as through blood-based biomarkers such as Glucose Transporter-1 (GLUT-1). We outline the potential for the hypoxia-inflammation cycle to drive tissue damage even in the absence of plaques. Inflammation can drive hypoxia through blood-brain barrier (BBB) disruption and edema, mitochondrial dysfunction, oxidative stress, vessel blockage and vascular abnormalities. The hypoxia can, in turn, drive more inflammation. Summary The hypoxia-inflammation cycle could exacerbate neuroinflammation and disease progression. We explore therapeutic approaches that target this cycle, providing information about potential treatments in MS. There are many therapeutic approaches that could block this cycle, including inhibiting hypoxia-inducible factor 1-α (HIF-1α), blocking cell adhesion or using vasodilators or oxygen, which could reduce either inflammation or hypoxia. This review highlights the potential significance of the hypoxia-inflammation pathway in MS and suggests strategies to break the cycle. Such treatments could improve quality of life or reduce rates of progression.
Collapse
Affiliation(s)
- Ateyeh Soroush
- Department of Neuroscience, University of Calgary, Calgary, Alberta Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta Canada
- Experimental Imaging Center (EIC), Cal Wenzel Precision Health Building (CWPH Building) University of Calgary, 2500 University Dr NW, Calgary, AB T2N 1N4 Canada
| | - Jeff F Dunn
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, Alberta Canada
- Department of Radiology, University of Calgary, Calgary, Alberta Canada
- Experimental Imaging Center (EIC), Cal Wenzel Precision Health Building (CWPH Building) University of Calgary, 2500 University Dr NW, Calgary, AB T2N 1N4 Canada
| |
Collapse
|
100
|
Gong X, Yang SY, Wang ZY, Tang M. The role of hypoxic microenvironment in autoimmune diseases. Front Immunol 2024; 15:1435306. [PMID: 39575238 PMCID: PMC11578973 DOI: 10.3389/fimmu.2024.1435306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
The hypoxic microenvironment, characterized by significantly reduced oxygen levels within tissues, has emerged as a critical factor in the pathogenesis and progression of various autoimmune diseases (AIDs). Central to this process is the hypoxia-inducible factor-1 (HIF-1), which orchestrates a wide array of cellular responses under low oxygen conditions. This review delves into the multifaceted roles of the hypoxic microenvironment in modulating immune cell function, particularly highlighting its impact on immune activation, metabolic reprogramming, and angiogenesis. Specific focus is given to the mechanisms by which hypoxia contributes to the development and exacerbation of diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), and dermatomyositis (DM). In these conditions, the hypoxic microenvironment not only disrupts immune tolerance but also enhances inflammatory responses and promotes tissue damage. The review also discusses emerging therapeutic strategies aimed at targeting the hypoxic pathways, including the application of HIF-1α inhibitors, mTOR inhibitors, and other modulators of the hypoxic response. By providing a comprehensive overview of the interplay between hypoxia and immune dysfunction in AIDs, this review offers new perspectives on the underlying mechanisms of these diseases and highlights potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xun Gong
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Su-Yin Yang
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen-Yu Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|