1
|
Zhang HY, Shu YQ, Li Y, Hu YL, Wu ZH, Li ZP, Deng Y, Zheng ZJ, Zhang XJ, Gong LF, Luo Y, Wang XY, Li HP, Liao XP, Li G, Ren H, Qiu W, Sun J. Metabolic disruption exacerbates intestinal damage during sleep deprivation by abolishing HIF1α-mediated repair. Cell Rep 2024; 43:114915. [PMID: 39527478 DOI: 10.1016/j.celrep.2024.114915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/22/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Sleep deprivation (SD) has been reported to induce intestinal damage by several mechanisms, yet its role in modulating epithelial repair remains unclear. In this study, we find that chronic SD leads to colonic damage through continuous hypoxia. However, HIF1α, which generally responds to hypoxia to modulate barrier integrity, was paradoxically dysregulated in the colon. Further investigation revealed that a metabolic disruption during SD causes accumulation of α-ketoglutarate in the colon. The excessive α-ketoglutarate degrades HIF1α protein through PHD2 (prolyl hydroxylase 2) to abolish the intestinal repair functions of HIF1α. Collectively, these findings provide insights into how SD can exacerbate intestinal damage by fine-tuning metabolism to abolish HIF1α-mediated repair.
Collapse
Affiliation(s)
- Hai-Yi Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ya-Qing Shu
- The Third Affiliated Hospital of Sun Yat-sen University, Department of Neurology, Guangzhou, China
| | - Yan Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ya-Lin Hu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhi-Hong Wu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhi-Peng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yao Deng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zi-Jian Zheng
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Liu-Fei Gong
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yang Luo
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiao-Yu Wang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | | | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gong Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wei Qiu
- The Third Affiliated Hospital of Sun Yat-sen University, Department of Neurology, Guangzhou, China.
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China; National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
2
|
Jeong J, Kang BH, Ju S, Park NY, Kim D, Dinh NTB, Lee J, Rhee CY, Cho DH, Kim H, Chung DK, Bunch H. Lactiplantibacillus plantarum K8 lysates regulate hypoxia-induced gene expression. Sci Rep 2024; 14:6275. [PMID: 38491188 PMCID: PMC10943017 DOI: 10.1038/s41598-024-56958-7] [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: 07/19/2023] [Accepted: 03/13/2024] [Indexed: 03/18/2024] Open
Abstract
Hypoxic responses have been implicated in critical pathologies, including inflammation, immunity, and tumorigenesis. Recently, efforts to identify effective natural remedies and health supplements are increasing. Previous studies have reported that the cell lysates and the cell wall-bound lipoteichoic acids of Lactiplantibacillus plantarum K8 (K8) exert anti-inflammatory and immunomodulative effects. However, the effect of K8 on cellular hypoxic responses remains unknown. In this study, we found that K8 lysates had a potent suppressive effect on gene expression under hypoxia. K8 lysates markedly downregulated hypoxia-induced HIF1α accumulation in the human bone marrow and lung cancer cell lines, SH-SY5Y and H460. Consequently, the transcription of known HIF1α target genes, such as p21, GLUT1, and ALDOC, was notably suppressed in the K8 lysate supplement and purified lipoteichoic acids of K8, upon hypoxic induction. Intriguingly, K8 lysates decreased the expression of PHD2 and VHL proteins, which are responsible for HIF1α destabilization under normoxic conditions, suggesting that K8 may regulate HIF1α stability in a non-canonical pathway. Overall, our results suggest that K8 lysates desensitize the cells to hypoxic stresses and suppress HIF1α-mediated hypoxic gene activation.
Collapse
Affiliation(s)
- Jaehyeon Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Byeong-Hee Kang
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sangmin Ju
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Na Yeon Park
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Deukyeong Kim
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ngoc Thi Bao Dinh
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeongho Lee
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Chang Yun Rhee
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioRearch Group, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Co. Ltd., Yongin, 17104, Republic of Korea
| | - Dae Kyun Chung
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Republic of Korea
| | - Heeyoun Bunch
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
3
|
Johnston SN, Tsingas M, Ain R, Barve RA, Risbud MV. Increased HIF-2α activity in the nucleus pulposus causes intervertebral disc degeneration in the aging mouse spine. Front Cell Dev Biol 2024; 12:1360376. [PMID: 38510179 PMCID: PMC10950937 DOI: 10.3389/fcell.2024.1360376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Hypoxia-inducible factors (HIFs) are essential to the homeostasis of hypoxic tissues. Although HIF-2α, is expressed in nucleus pulposus (NP) cells, consequences of elevated HIF-2 activity on disc health remains unknown. We expressed HIF-2α with proline to alanine substitutions (P405A; P531A) in the Oxygen-dependent degradation domain (HIF-2αdPA) in the NP tissue using an inducible, nucleus pulposus-specific K19CreERT allele to study HIF-2α function in the adult intervertebral disc. Expression of HIF-2α in NP impacted disc morphology, as evident from small but significantly higher scores of degeneration in NP of 24-month-old K19CreERT; HIF-2αdPA (K19-dPA) mice. Noteworthy, comparisons of grades within each genotype between 14 months and 24 months indicated that HIF-2α overexpression contributed to more pronounced changes than aging alone. The annulus fibrosus (AF) compartment in the 14-month-old K19-dPA mice exhibited lower collagen turnover and Fourier transform-infrared (FTIR) spectroscopic imaging analyses showed changes in the biochemical composition of the 14- and 24-month-old K19-dPA mice. Moreover, there were changes in aggrecan, chondroitin sulfate, and COMP abundance without alterations in NP phenotypic marker CA3, suggesting the overexpression of HIF-2α had some impact on matrix composition but not the cell phenotype. Mechanistically, the global transcriptomic analysis showed enrichment of differentially expressed genes in themes closely related to NP cell function such as cilia, SLIT/ROBO pathway, and HIF/Hypoxia signaling at both 14- and 24-month. Together, these findings underscore the role of HIF-2α in the pathogenesis of disc degeneration in the aged spine.
Collapse
Affiliation(s)
- Shira N. Johnston
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Maria Tsingas
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Rahatul Ain
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
- Graduate Program in Pharmacology, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - Ruteja A. Barve
- Department of Genetics, Genome Technology Access Centre at the McDonnell Genome Institute, Washington University, School of Medicine, St. Louis, MO, United States
| | - Makarand V. Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Johnston SN, Tsingas M, Ain R, Barve RA, Risbud MV. Increased HIF-2α Activity in the Nucleus Pulposus Causes Intervertebral Disc Degeneration in the Aging Mouse Spine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573086. [PMID: 38187709 PMCID: PMC10769411 DOI: 10.1101/2023.12.22.573086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Hypoxia-inducible factors (HIFs) are essential to the homeostasis of hypoxic tissues. Although HIF-2α, is expressed in nucleus pulposus (NP) cells, consequences of elevated HIF-2 activity on disc health remains unknown. We expressed HIF-2α with proline to alanine substitutions (P405A;P531A) in the Oxygen-dependent degradation domain (HIF-2αdPA) in the NP tissue using an inducible, nucleus pulposus-specific K19 CreERT allele to study HIF-2α function in the adult intervertebral disc. Expression of HIF-2α in NP impacted disc morphology, as evident from small but significantly higher scores of degeneration in NP of 24-month-old K19 CreERT ; HIF-2α dPA (K19-dPA) mice. Noteworthy, comparisons of grades within each genotype between 14 months and 24 months indicated that HIF-2α overexpression contributed to more pronounced changes than aging alone. The annulus fibrosus (AF) compartment in the 14-month-old K19-dPA mice exhibited lower collagen turnover and Fourier transform-infrared (FTIR) spectroscopic imaging analyses showed changes in the biochemical composition of the 14-and 24-month-old K19-dPA mice. Moreover, there were changes in aggrecan, chondroitin sulfate, and COMP abundance without alterations in NP phenotypic marker CA3, suggesting the overexpression of HIF-2α had some impact on matrix composition but not the cell phenotype. Mechanistically, the global transcriptomic analysis showed enrichment of differentially expressed genes in themes closely related to NP cell function such as cilia, SLIT/ROBO pathway, and HIF/Hypoxia signaling at both 14- and 24-months. Together, these findings underscore the role of HIF-2α in the pathogenesis of disc degeneration in the aged spine.
Collapse
|
5
|
Liao Q, Deng H, Wang Z, Yu G, Zhu C, Jia S, Liu W, Bai Y, Sun X, Chen X, Xiao W, Liu X. Deletion of prolyl hydroxylase domain-containing enzyme 3 (phd3) in zebrafish facilitates hypoxia tolerance. J Biol Chem 2023; 299:105420. [PMID: 37923141 PMCID: PMC10724695 DOI: 10.1016/j.jbc.2023.105420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Prolyl hydroxylase domain (PHD)-containing enzyme 3 (PHD3) belongs to the Caenorhabditis elegans gene egl-9 family of prolyl hydroxylases. PHD3 catalyzes proline hydroxylation of hypoxia-inducible factor α (HIF-α) and promotes HIF-α proteasomal degradation through coordination with the pVHL complex under normoxic conditions. However, the relationship between PHD3 and the hypoxic response is not well understood. In this study, we used quantitative real-time PCR assay and O-dianisidine staining to characterize the hypoxic response in zebrafish deficient in phd3. We found that the hypoxia-responsive genes are upregulated and the number of erythrocytes was increased in phd3-null zebrafish compared with their wild-type siblings. On the other hand, we show overexpression of phd3 suppresses HIF-transcriptional activation. In addition, we demonstrate phd3 promotes polyubiquitination of zebrafish hif-1/2α proteins, leading to their proteasomal degradation. Finally, we found that compared with wild-type zebrafish, phd3-null zebrafish are more resistant to hypoxia treatment. Therefore, we conclude phd3 has a role in hypoxia tolerance. These results highlight the importance of modulation of the hypoxia signaling pathway by phd3 in hypoxia adaptation.
Collapse
Affiliation(s)
- Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Hongyan Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; College of Life Science, Wuhan University, Wuhan, P. R.China
| | - Zixuan Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Guangqing Yu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China
| | - Chunchun Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China
| | - Shuke Jia
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Wen Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Yao Bai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Xueyi Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Xiaoyun Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; Hubei Hongshan Laboratory, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, P. R.China.
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R.China; University of Chinese Academy of Sciences, Beijing, P. R.China; The Innovation of Seed Design, Chinese Academy of Sciences, Wuhan, P. R.China.
| |
Collapse
|
6
|
Shan C, Xia Y, Wu Z, Zhao J. HIF-1α and periodontitis: Novel insights linking host-environment interplay to periodontal phenotypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 184:50-78. [PMID: 37769974 DOI: 10.1016/j.pbiomolbio.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
Periodontitis, the sixth most prevalent epidemic disease globally, profoundly impacts oral aesthetics and masticatory functionality. Hypoxia-inducible factor-1α (HIF-1α), an oxygen-dependent transcriptional activator, has emerged as a pivotal regulator in periodontal tissue and alveolar bone metabolism, exerts critical functions in angiogenesis, erythropoiesis, energy metabolism, and cell fate determination. Numerous essential phenotypes regulated by HIF are intricately associated with bone metabolism in periodontal tissues. Extensive investigations have highlighted the central role of HIF and its downstream target genes and pathways in the coupling of angiogenesis and osteogenesis. Within this concise perspective, we comprehensively review the cellular phenotypic alterations and microenvironmental dynamics linking HIF to periodontitis. We analyze current research on the HIF pathway, elucidating its impact on bone repair and regeneration, while unraveling the involved cellular and molecular mechanisms. Furthermore, we briefly discuss the potential application of targeted interventions aimed at HIF in the field of bone tissue regeneration engineering. This review expands our biological understanding of the intricate relationship between the HIF gene and bone angiogenesis in periodontitis and offers valuable insights for the development of innovative therapies to expedite bone repair and regeneration.
Collapse
Affiliation(s)
- Chao Shan
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - YuNing Xia
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Zeyu Wu
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China
| | - Jin Zhao
- Department of Dentistry, Xinjiang Medical University, Ürümqi, China; The First Affiliated Hospital of Xinjiang Medical University (Affiliated Stomatology Hospital), Ürümqi, China; Xinjiang Uygur Autonomous Region Institute of Stomatology, Ürümqi, China.
| |
Collapse
|
7
|
Wu J, Yu L, Liu Y, Xiao B, Ye X, Zhao H, Xi Y, Shi Z, Wang W. Hypoxia regulates adipose mesenchymal stem cells proliferation, migration, and nucleus pulposus-like differentiation by regulating endoplasmic reticulum stress via the HIF-1α pathway. J Orthop Surg Res 2023; 18:339. [PMID: 37158945 PMCID: PMC10169485 DOI: 10.1186/s13018-023-03818-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023] Open
Abstract
OBJECTIVE Hypoxia can promote stem cell proliferation and migration through HIF-1α. Hypoxia can regulate cellular endoplasmic reticulum (ER) stress. Some studies have reported the relationship among hypoxia, HIF-α, and ER stress, however, while little is known about HIF-α and ER stress in ADSCs under hypoxic conditions. The purpose of the study was to investigate the role and relationship of hypoxic conditions, HIF-1α and ER stress in regulating adipose mesenchymal stem cells (ADSCs) proliferation, migration, and NPC-like differentiation. METHOD ADSCs were pretreated with hypoxia, HIF-1α gene transfection, and HIF-1α gene silence. The ADSCs proliferation, migration, and NPC-like differentiation were assessed. The expression of HIF-1α in ADSCs was regulated; then, the changes of ER stress level in ADSCs were observed to investigate the relationship between ER stress and HIF-1α in ADSCs under hypoxic conditions. RESULT The cell proliferation and migration assay results show that hypoxia and HIF-1α overexpression can significantly increase the ADSCs proliferation and migration, while HIF-1α inhibition can significantly decrease the ADSCs proliferation and migration. The HIF-1α and co-cultured with NPCs played an important role in the directional differentiation of ADSCs into NPCs. The hypoxia-regulated ER stress in ADSCs through the HIF-1α pathway, thereby regulating the cellular state of ADSCs, was also observed. CONCLUSION Hypoxia and HIF-1α play important roles in proliferation, migration, and NPC-like differentiation of ADSCs. This study provides preliminary evidence that HIF-1α-regulated ER stress thus affects ADSCs proliferation, migration, and differentiation. Therefore, HIF-1α and ER may serve as key points to improve the efficacy of ADSCs in treating disc degeneration.
Collapse
Affiliation(s)
- Jianxin Wu
- Department of Orthopaedics, First Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai, People’s Republic of China
| | - Lei Yu
- Department of Orthopedic Surgery and Neurosurgery, No. 906 Hospital of the People’s Liberation Army, Ningbo, Zhejiang People’s Republic of China
| | - Yi Liu
- Department of Orthopedics, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Kangfu Road, Tianjin, People’s Republic of China
| | - Bing Xiao
- Department of Orthopaedics, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, People’s Republic of China
| | - Xiaojian Ye
- Department of Orthopaedics, Tongren Hospital of Shanghai Jiaotong University, No. 1111, Xianxia Road, Shanghai, People’s Republic of China
| | - Hong Zhao
- Department of Orthopedics, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Kangfu Road, Tianjin, People’s Republic of China
| | - Yanhai Xi
- Department of Orthopaedics, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, People’s Republic of China
| | - Zhicai Shi
- Department of Orthopaedics, First Affiliated Hospital of Naval Medical University, No. 168 Changhai Road, Shanghai, People’s Republic of China
| | - Weiheng Wang
- Department of Orthopaedics, Second Affiliated Hospital of Naval Medical University, No. 415 Fengyang Road, Shanghai, People’s Republic of China
| |
Collapse
|
8
|
Xiao C, Liu S, Ge G, Jiang H, Wang L, Chen Q, Jin C, Mo J, Li J, Wang K, Zhang Q, Zhou J. Roles of hypoxia-inducible factor in hepatocellular carcinoma under local ablation therapies. Front Pharmacol 2023; 14:1086813. [PMID: 36814489 PMCID: PMC9939531 DOI: 10.3389/fphar.2023.1086813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common digestive malignancies. HCC It ranges as the fifth most common cause of cancer mortality worldwide. While The prognosis of metastatic or advanced HCC is still quite poor. Recently, locoregional treatment, especially local ablation therapies, plays an important role in the treatment of HCC. Radiofrequency ablation (RFA) and high-intensity focused ultrasound (HIFU) ablation are the most common-used methods effective and feasible for treating HCC. However, the molecular mechanisms underlying the actions of ablation in the treatments for HCC and the HCC recurrence after ablation still are poorly understood. Hypoxia-inducible factor (HIF), the key gene switch for adaptive responses to hypoxia, has been found to play an essential role in the rapid aggressive recurrence of HCC after ablation treatment. In this review, we summarized the current evidence of the roles of HIF in the treatment of HCC with ablation. Fifteen relevant studies were included and further analyzed. Among them, three clinical studies suggested that HIF-1α might serve as a crucial role in the RAF treatment of HCC or the local recurrence of HCC after RFA. The remainder included experimental studies demonstrated that HIF-1, 2α might target the different molecules (e.g., BNIP3, CA-IX, and arginase-1) and signaling cascades (e.g., VEGFA/EphA2 pathway), constituting a complex network that promoted HCC invasion and metastasis after ablation. Currently, the inhibitors of HIF have been developed, providing important proof of targeting HIF for the prevention of HCC recurrence after IRFA and HIFU ablation. Further confirmation by prospective clinical and in-depth experimental studies is still warranted to illustrate the effects of HIF in HCC recurrence followed ablation treatment in the future.
Collapse
Affiliation(s)
- Chunying Xiao
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Sheng Liu
- Department of Hepatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ge Ge
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Jin Li
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Kunpeng Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| | - Qianqian Zhang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianyu Zhou
- Department of Ultrasound, Taizhou Central Hospital (Taizhou University, Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
9
|
Hu L, Hu J, Huang Y, Zheng S, Yin J, Li X, Li D, Lv C, Li S, Hu W. Hypoxia-mediated activation of hypoxia-inducible factor-1α in head and neck squamous cell carcinoma: A review. Medicine (Baltimore) 2023; 102:e32533. [PMID: 36607847 PMCID: PMC9829281 DOI: 10.1097/md.0000000000032533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Since the 1950s, hypoxia has been recognized as a crucial characteristic of cancer cells and their microenvironment. Indeed, hypoxia promotes the growth, survival, and metastasis of cancer cells. In the early 1990s, we found that as many phenomena in hypoxia can occur through hypoxia-inducible factor-1α (HIF1α). HIF1α is known as an angiogenesis converter in hypoxia, which promotes tumorigenesis, development, immune escape, recurrence, etc; This page goes into great detail on how HIF1α is activated during hypoxia and how the 2 signaling channels interact. It specifically emphasizes the significance of reactive oxygen species, the function of the PI3K/the serine/threonine kinase Akt/mammalian target of rapamycin cascade, and outlines the similarities between the 2 important factors (reactive oxygen species and PI3K/the serine/threonine kinase Akt/mammalian target of rapamycin cascade), nuclear factor κB, for HIF1α Important implications, in an effort to offer fresh views for the treatment of head and neck squamous cell carcinoma and HIF1α research.
Collapse
Affiliation(s)
- Lanxin Hu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jinwei Hu
- Clinical Medicine, Nanchang University Queen Mary School, Nanchang, China
| | - Yanlin Huang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sihan Zheng
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ji Yin
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xiaohui Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Daiying Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Caifeng Lv
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Sen Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Wenjian Hu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- * Correspondence: Wenjian Hu, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, O.182 Chunhui Road Longmatan District Luzhou Sichuan 646000, China (e-mail: )
| |
Collapse
|
10
|
Salvagno M, Coppalini G, Taccone FS, Strapazzon G, Mrakic-Sposta S, Rocco M, Khalife M, Balestra C. The Normobaric Oxygen Paradox-Hyperoxic Hypoxic Paradox: A Novel Expedient Strategy in Hematopoiesis Clinical Issues. Int J Mol Sci 2022; 24:ijms24010082. [PMID: 36613522 PMCID: PMC9820104 DOI: 10.3390/ijms24010082] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Hypoxia, even at non-lethal levels, is one of the most stressful events for all aerobic organisms as it significantly affects a wide spectrum of physiological functions and energy production. Aerobic organisms activate countless molecular responses directed to respond at cellular, tissue, organ, and whole-body levels to cope with oxygen shortage allowing survival, including enhanced neo-angiogenesis and systemic oxygen delivery. The benefits of hypoxia may be evoked without its detrimental consequences by exploiting the so-called normobaric oxygen paradox. The intermittent shift between hyperoxic-normoxic exposure, in addition to being safe and feasible, has been shown to enhance erythropoietin production and raise hemoglobin levels with numerous different potential applications in many fields of therapy as a new strategy for surgical preconditioning aimed at frail patients and prevention of postoperative anemia. This narrative review summarizes the physiological processes behind the proposed normobaric oxygen paradox, focusing on the latest scientific evidence and the potential applications for this strategy. Future possibilities for hyperoxic-normoxic exposure therapy include implementation as a synergistic strategy to improve a patient's pre-surgical condition, a stimulating treatment in critically ill patients, preconditioning of athletes during physical preparation, and, in combination with surgery and conventional chemotherapy, to improve patients' outcomes and quality of life.
Collapse
Affiliation(s)
- Michele Salvagno
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Giacomo Coppalini
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Giacomo Strapazzon
- Institute of Mountain Emergency Medicine, Eurac Research, 39100 Bolzano, Italy
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology—National Research Council (CNR-IFC), 20162 Milano, Italy
| | - Monica Rocco
- Dipartimento di Scienze Medico Chirurgiche e Medicina Traslazionale, Sapienza University of Rome, 00189 Rome, Italy
| | - Maher Khalife
- Department of Anesthesiology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Costantino Balestra
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1050 Brussels, Belgium
- Anatomical Research and Clinical Studies, Vrije Universiteit Brussels (VUB), 1090 Brussels, Belgium
- DAN Europe Research Division (Roseto-Brussels), 1020 Brussels, Belgium
- Physical Activity Teaching Unit, Motor Sciences Department, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
- Correspondence:
| |
Collapse
|
11
|
Johnston SN, Madhu V, Shapiro IM, Risbud MV. Conditional Deletion of HIF-2α in Mouse Nucleus Pulposus Reduces Fibrosis and Provides Mild and Transient Protection From Age-Dependent Structural Changes in Intervertebral Disc. J Bone Miner Res 2022; 37:2512-2530. [PMID: 36117450 PMCID: PMC9772060 DOI: 10.1002/jbmr.4707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/05/2022] [Accepted: 07/29/2022] [Indexed: 01/19/2023]
Abstract
Hypoxia-inducible factors (HIFs) are critical to the development and homeostasis of hypoxic tissues. Although HIF-2α, one of the main HIF-α isoforms, is expressed in nucleus pulposus (NP) cells, its functions remain unknown. We deleted HIF-2α in the NP tissue using a notochord-specific FoxA2Cre allele to study HIF-2α function in the adult intervertebral disc. Unlike observations in HIF-1αcKO mice, fate mapping studies using Rosa26-mTmG reporter showed that HIF-2α loss in NP did not negatively impact cell survival or affect compartment development. Rather, loss of HIF-2α resulted in slightly better attributes of NP morphology in 14-month-old HIF-2αcKO mice as evident from lower scores of degeneration. These 14-month-old HIF-2αcKO mice also exhibited significant reduction in NP tissue fibrosis and lower collagen turnover in the annulus fibrosis (AF) compartment. Imaging-Fourier transform-infrared (FTIR) analyses showed decreased collagen and protein content in the NP and maintained chondroitin sulfate levels in 14-month-old HIF-2αcKO . Mechanistically, global transcriptomic analysis showed enrichment of differentially expressed genes with Gene Ontology (GO) terms related to metabolic processes and cell development, molecular functions concerned with histone and protein binding, and associated pathways, including oxidative stress. Noteworthy, these morphological differences were not apparent in 24-month-old HIF-2αcKO , indicating that aging is the dominant factor in governing disc health. Together these data suggest that loss of HIF-2α in the NP compartment is not detrimental to the intervertebral disc development but rather mitigates NP tissue fibrosis and offers mild but transient protection from age-dependent early degenerative changes. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shira N. Johnston
- Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Vedavathi Madhu
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Irving M. Shapiro
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Makarand V. Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
12
|
Jones NM, Nathanson AD, Chell S, DeAngelis E, Whelan G, Willé D, Cheng K. The prolyl hydroxylase inhibitor GSK1120360A reduces early brain injury, but protection is not maintained in a neonatal rat model of hypoxic ischaemic encephalopathy. Int J Dev Neurosci 2022; 82:423-435. [PMID: 35662244 DOI: 10.1002/jdn.10199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 05/31/2022] [Indexed: 11/06/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) in newborns is associated with high morbidity and mortality, with many babies suffering long-term neurological deficits. Currently, treatment options are limited to therapeutic hypothermia, which is not appropriate for use in all babies. Previous studies have shown protective effects of increasing the transcription factor-hypoxia-inducible factor-1 (HIF-1) in animal models, by using mild hypoxia or compounds that act as prolyl hydroxylase inhibitors (PHIs). Here, we aimed to examine the neuroprotective actions of an orally active, small molecule PHI, GSK1120360A in a neonatal rat model of hypoxia-ischemia (HI) compared to another PHI, desferrioxamine (DFX). Sprague-Dawley rats underwent HI surgery on postnatal day 7 (P7), where unilateral carotid artery occlusion was performed followed by hypoxia (8% oxygen, 3 h). Initial testing showed that GSK1120360A and erythropoietin levels were detectable in plasma at 6 h following oral exposure to GSK1120360A. For the short-term neuroprotection study, pups were assigned to receive either saline (s.c), desferrioxamine (DFX-200 mg/kg, s.c), methylcellulose (1%, oral) or GSK1120360A (30 mg/kg, oral) immediately after HI. Histological analysis showed that GSK1120360A in this setting reduced brain injury size 7 days after HI, compared to the methylcellulose vehicle control group. DFX had no significant effect on injury size compared to saline group at the same 7 day timepoint. In the long-term neuroprotection study, pups were randomly assigned to be administered methylcellulose (1%, oral) or GSK1120360A (30 mg/kg, oral) immediately after HI. On P42, rats underwent behavioural testing using the forelimb grip strength, grid walking and novel object recognition tasks, and brains were collected for histological analysis. Long-term behavioural deficits were observed in grid walking, grip strength and novel object recognition tests after HI which were not improved in the GSK1120360A treatment group compared to the methylcellulose group. Similarly, there was no improvement in injury size on P42 in the GSK1120360A study group compared to the methylcellulose group. Here, we have shown that GSK1120360A can reduce brain injury at 7 days but that this neuroprotective benefit is not maintained when examined at 5 weeks after HI.
Collapse
Affiliation(s)
- Nicole M Jones
- Department of Pharmacology, School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Anton D Nathanson
- Department of Pharmacology, School of Medical Sciences, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Simon Chell
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | | | - Greg Whelan
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | - David Willé
- Medicines Research Centre, GlaxoSmithKline, Stevenage, UK
| | | |
Collapse
|
13
|
Bora-Singhal N, Saha B, Mohankumar D, Padmanabhan J, Coppola D, Chellappan S. A Novel PHD2/VHL-mediated Regulation of YAP1 Contributes to VEGF Expression and Angiogenesis. CANCER RESEARCH COMMUNICATIONS 2022; 2:624-638. [PMID: 35937460 PMCID: PMC9351435 DOI: 10.1158/2767-9764.crc-21-0084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/10/2022] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
The transcriptional co-activator YAP1 is the major oncogenic component of the Hippo signaling pathway and contributes to the genesis and progression of various tumors, including non-small cell lung cancer (NSCLC). YAP1 levels are regulated by the canonical Hippo kinases, MST1/2 and LATS1/2, which modulate its cytoplasmic retention and proteasomal degradation. While non-canonical regulation of YAP1 has been reported, its role in hypoxic response is not fully elucidated. The studies presented here show that YAP1 levels and function are modulated by VHL and PHD2. YAP1 could regulate multiple genes involved in angiogenesis through E2F1; it also associates with HIF1α in cancer cells under hypoxic conditions, inducing the VEGF-A promoter. Under normoxic conditions, PHD2 associates with and hydroxylates specific proline residues on YAP1, facilitating its interaction with VHL and promoting ubiquitination and subsequent proteasomal degradation. Exposure to hypoxia dissociates YAP1 from PHD2 and VHL, elevating YAP1 levels and enhancing its association with HIF1α. YAP1-HIF1α interaction was higher in NSCLC and RCC samples, indicating a role for this interaction in the genesis of these cancers. Our results thus reveal a novel mode of regulation of YAP1 by PHD2 and VHL in normoxic cells, suggesting that YAP1-mediated induction of VEGF and other genes contributes to hypoxic response in tumors.
Collapse
Affiliation(s)
| | - Biswarup Saha
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida
| | | | - Jaya Padmanabhan
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, Florida
| | - Domenico Coppola
- Department of Anatomic pathology, Moffitt Cancer Center, Tampa, Florida
| | | |
Collapse
|
14
|
Swamy G, Salo P, Duncan N, Jirik F, Matyas J. IL-1Ra deficiency accelerates intervertebral disc degeneration in C57BL6J mice. JOR Spine 2022; 5:e1201. [PMID: 35783913 PMCID: PMC9238285 DOI: 10.1002/jsp2.1201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/24/2022] [Accepted: 03/29/2022] [Indexed: 11/26/2022] Open
Abstract
The expression of Interleukin-1ß (IL-1ß) and its antagonist and Interleukin-1 receptor antagonist (IL-1Ra) are correlated with greater human intervertebral disc (IVD) degeneration, suggesting that elevated IL-1β activity promotes disc degeneration. Many in vitro studies support such a mechanistic relationship, whereas few in vivo investigations have been reported. The present study tests the effect of increased IL-1β activity on intervertebral disc in mice with an IL-1Ra gene deletion. IL-1Ra-/- mice and wild-type (WT) C57Bl6J mice were examined at 3 and 12 months of age. Caudal IVD segments were evaluated for disc degeneration by histopathology, functional testing, and inflammatory gene expression relevant to IL-1β pathways. To test differences in injury response, pinprick annular puncture was performed on IL-1Ra-/- and WT mice and evaluated similarly. IL-1Ra-/- IVDs had significantly worse histopathology at 3 months compared to WT controls, but not at 12 months. IL-1Ra-/- IVDs exhibited significantly more viscous mechanical properties than WT IVDs. qPCR revealed downregulation of inflammatory genes at 3 and 12 months in IL-1Ra-/- IVDs, with concomitant downregulation of anabolic and catabolic genes. Annular puncture yielded no appreciable differences between 2-week and 6-week post-injured WT and IL1-Ra-/- IVDs in histopathology or biomechanics, but inflammatory gene expression was sharply downregulated in IL-1Ra-/- mice at 2 weeks, returning by 6 weeks post injury. In the present study, IL-1Ra deletion resulted in increased IVD histopathology, inferior biomechanics, and transiently decreased pro-inflammatory cytokine gene expression. The histopathology of IL-1Ra-/- IVDs on a C57BL/6J background is less severe than a previous report of IL1Ra-/- on a BALB/c background, yet both strains exhibit IVD degeneration, reinforcing a mechanistic role of IL-1β signaling in IVD pathobiology. Despite a pro-inflammatory environment, the annular puncture was no worse in IL-1Ra-/- mice, suggesting that response to injury involves pathways other than inflammation. Overall, this study supports the hypothesis that IL-1β-driven inflammation is important in IVD degeneration.
Collapse
Affiliation(s)
- Ganesh Swamy
- Cumming School of MedicineMcCaig Institute of Bone and Joint Health University of CalgaryCalgaryAlbertaCanada
| | - Paul Salo
- Department of SurgeryCumming School of MedicineCalgaryAlbertaCanada
| | - Neil Duncan
- Department of Civil EngineeringSchulich School of EngineeringCalgaryAlbertaCanada
| | - Frank Jirik
- Department of MedicineHealth Research Innovation CentreCalgaryAlbertaCanada
| | - John Matyas
- Department of Comparative Biology & Experimental MedicineFaculty of Veterinary Medicine University of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
15
|
Liu SR, Ren D, Wu HT, Yao SQ, Song ZH, Geng LD, Wang PC. Reparative effects of chronic intermittent hypobaric hypoxia pre‑treatment on intervertebral disc degeneration in rats. Mol Med Rep 2022; 25:173. [PMID: 35315494 PMCID: PMC8971903 DOI: 10.3892/mmr.2022.12689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/25/2022] [Indexed: 11/24/2022] Open
Abstract
Previous studies have indicated that chronic intermittent hypobaric hypoxia (CIHH) preconditioning can inhibit TNF-α and other related inflammatory cytokines and exerts protective effect on intervertebral disc degeneration disease (IDD) in rats; however, the mechanism is still unclear. The present study aimed to explore the repair mechanisms of CIHH on IDD in rats. In the experiment, 48 adult Sprague-Dawley rats were selected and randomly divided into an experimental group (CIHH-IDD), a degenerative group (IDD) and a control group (CON). The CIHH-IDD group of rats (n=16) were treated with CIHH (simulated 3000 m altitude, 5 h per day, 28 days; PO2=108.8 mmHg) before disc degeneration surgery. The IDD group of rats (n=16) underwent tail-vertebral intervertebral disc surgery to establish a model of intervertebral disc degeneration. The CON group of rats (n=16) did not receive any treatments. After surgery, the disc height index was calculated using X-ray analysis of rat tail vertebrae, the degeneration process was observed and repair was evaluated by chemically staining degenerative intervertebral disc tissue slices. The expression levels of basic fibroblast growth factor (bFGF), TGFβ1, Collagen I and Collagen II were measured in the intervertebral disc tissue using western blotting; while the expression levels of bFGF, TGFβ1 and hypoxia-inducible factor 1-α (HIF-1α) were measured in rat serum using ELISA. The results demonstrated that: i) The degree of intervertebral disc height degeneration in CIHH-IDD rats was significantly lower compared with that in IDD rats (P<0.05); ii) the expression levels of bFGF, TGFβ1 and HIF-1α were higher in CIHH-IDD rat serum compared with those in IDD rat serum (P<0.05); iii) optical microscopy revealed that the degree of disc degeneration was relatively mild in CIHH-IDD rats; and iv) the protein expression levels of bFGF, TGFβ1 and collagen II were increased in CIHH-IDD rat intervertebral disc tissues compared with those of IDD rats, while the overexpression of collagen I protein was inhibited. Overall, after CIHH pre-treatment, the expression levels of bFGF and TGFβ1 were up-regulated, which play notable roles in repairing degenerative intervertebral discs in rats.
Collapse
Affiliation(s)
- Shu-Ren Liu
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Dong Ren
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Hao-Tan Wu
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Shuang-Quan Yao
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Zhao-Hui Song
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Lin-Dan Geng
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| | - Peng-Cheng Wang
- Major Laboratory of Orthopaedic Biomechanics in Hebei Province, Department of Orthopaedic Trauma Service Centre, The Third Hospital of Hebei Medical University, Hebei, Shijiazhuang 050051, P.R. China
| |
Collapse
|
16
|
Smith MM, Hayes AJ, Melrose J. Pentosan Polysulphate (PPS), a Semi-Synthetic Heparinoid DMOAD With Roles in Intervertebral Disc Repair Biology emulating The Stem Cell Instructive and Tissue Reparative Properties of Heparan Sulphate. Stem Cells Dev 2022; 31:406-430. [PMID: 35102748 DOI: 10.1089/scd.2022.0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review highlights the attributes of pentosan polysulphate (PPS) in the promotion of intervertebral disc (IVD) repair processes. PPS has been classified as a disease modifying osteoarthritic drug (DMOAD) and many studies have demonstrated its positive attributes in the countering of degenerative changes occurring in cartilaginous tissues during the development of osteoarthritis (OA). Degenerative changes in the IVD also involve inflammatory cytokines, degradative proteases and cell signalling pathways similar to those operative in the development of OA in articular cartilage. PPS acts as a heparan sulphate (HS) mimetic to effect its beneficial effects in cartilage. The IVD contains small cell membrane HS-proteoglycans (HSPGs) such as syndecan, and glypican and a large multifunctional HS/chondroitin sulphate (CS) hybrid proteoglycan (HSPG2/perlecan) that have important matrix stabilising properties and sequester, control and present growth factors from the FGF, VEGF, PDGF and BMP families to cellular receptors to promote cell proliferation, differentiation and matrix synthesis. HSPG2 also has chondrogenic properties and stimulates the synthesis of extracellular matrix (ECM) components, expansion of cartilaginous rudiments and has roles in matrix stabilisation and repair. Perlecan is a perinuclear and nuclear proteoglycan in IVD cells with roles in chromatin organisation and control of transcription factor activity, immunolocalises to stem cell niches in cartilage, promotes escape of stem cells from quiescent recycling, differentiation and attainment of pluripotency and migratory properties. These participate in tissue development and morphogenesis, ECM remodelling and repair. PPS also localises in the nucleus of stromal stem cells, promotes development of chondroprogenitor cell lineages, ECM synthesis and repair and discal repair by resident disc cells. The availability of recombinant perlecan and PPS offer new opportunities in repair biology. These multifunctional agents offer welcome new developments in repair strategies for the IVD.
Collapse
Affiliation(s)
- Margaret M Smith
- The University of Sydney Raymond Purves Bone and Joint Research Laboratories, 247198, St Leonards, New South Wales, Australia;
| | - Anthony J Hayes
- Cardiff School of Biosciences, University of Cardiff, UK, Bioimaging Unit, Cardiff, Wales, United Kingdom of Great Britain and Northern Ireland;
| | - James Melrose
- Kolling Institute, University of Sydney, Royal North Shore Hospital, Raymond Purves Lab, Sydney Medical School Northern, Level 10, Kolling Institute B6, Royal North Shore Hospital, St. Leonards, New South Wales, Australia, 2065.,University of New South Wales, 7800, Graduate School of Biomedical Engineering, University of NSW, Sydney, New South Wales, Australia, 2052;
| |
Collapse
|
17
|
Jiang Y, Duan LJ, Fong GH. Oxygen-sensing mechanisms in development and tissue repair. Development 2021; 148:273632. [PMID: 34874450 DOI: 10.1242/dev.200030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Under normoxia, hypoxia inducible factor (HIF) α subunits are hydroxylated by PHDs (prolyl hydroxylase domain proteins) and subsequently undergo polyubiquitylation and degradation. Normal embryogenesis occurs under hypoxia, which suppresses PHD activities and allows HIFα to stabilize and regulate development. In this Primer, we explain molecular mechanisms of the oxygen-sensing pathway, summarize HIF-regulated downstream events, discuss loss-of-function phenotypes primarily in mouse development, and highlight clinical relevance to angiogenesis and tissue repair.
Collapse
Affiliation(s)
- Yida Jiang
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Li-Juan Duan
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.,Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
18
|
Korbecki J, Simińska D, Gąssowska-Dobrowolska M, Listos J, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Chronic and Cycling Hypoxia: Drivers of Cancer Chronic Inflammation through HIF-1 and NF-κB Activation: A Review of the Molecular Mechanisms. Int J Mol Sci 2021; 22:ijms221910701. [PMID: 34639040 PMCID: PMC8509318 DOI: 10.3390/ijms221910701] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic (continuous, non-interrupted) hypoxia and cycling (intermittent, transient) hypoxia are two types of hypoxia occurring in malignant tumors. They are both associated with the activation of hypoxia-inducible factor-1 (HIF-1) and nuclear factor κB (NF-κB), which induce changes in gene expression. This paper discusses in detail the mechanisms of activation of these two transcription factors in chronic and cycling hypoxia and the crosstalk between both signaling pathways. In particular, it focuses on the importance of reactive oxygen species (ROS), reactive nitrogen species (RNS) together with nitric oxide synthase, acetylation of HIF-1, and the action of MAPK cascades. The paper also discusses the importance of hypoxia in the formation of chronic low-grade inflammation in cancerous tumors. Finally, we discuss the effects of cycling hypoxia on the tumor microenvironment, in particular on the expression of VEGF-A, CCL2/MCP-1, CXCL1/GRO-α, CXCL8/IL-8, and COX-2 together with PGE2. These factors induce angiogenesis and recruit various cells into the tumor niche, including neutrophils and monocytes which, in the tumor, are transformed into tumor-associated neutrophils (TAN) and tumor-associated macrophages (TAM) that participate in tumorigenesis.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland;
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St., 20-093 Lublin, Poland;
| | - Izabela Gutowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (D.S.); (I.G.); (D.C.)
- Correspondence: ; Tel.: +48-(91)-466-1515
| |
Collapse
|
19
|
Rajasekaran S, Thangavel C, Djuric N, Raveendran M, Soundararajan DCR, Nayagam SM, Matchado MS, Sri Vijay Anand KS, Venkateshwaran K. Profiling extra cellular matrix associated proteome of human fetal nucleus pulposus in search for regenerative targets. Sci Rep 2021; 11:19013. [PMID: 34561485 PMCID: PMC8463528 DOI: 10.1038/s41598-021-97620-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 08/24/2021] [Indexed: 01/07/2023] Open
Abstract
Degeneration of the intervertebral disc is associated with a decrease in extra-cellular matrix (ECM) content due to an imbalance in anabolic and catabolic signaling. Our previous study profiled the core matrisome of fetal NP’s and identified various proteins with anabolic potential for regenerative therapies. This study aims to complement those results by exploring ECM regulators, associated proteins and secreted factors of the fetal nucleus pulposus (NP). Proteomic data of 9 fetal, 7 healthy adults (age 22–79), and 11 degenerated NP’s was analyzed. Based on the selection criteria, a total of 45 proteins were identified, of which 14 were uniquely expressed or upregulated in fetus compared to adult NP’s. Pathway analysis with these proteins revealed a significant upregulation of one pathway and two biological processes, in which 12 proteins were involved. Prolyl 4 hydroxylase (P4HA) 1 and 2, Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD) 1, and Heat shock protein 47 (SERPINH1) were involved in ‘collagen biosynthesis’ pathway. In addition, PLOD 1, SERPINH1, Annexin A1 and A4, CD109 and Galectin 3 (LGALS3) were all involved in biological process of ‘tissue development’. Furthermore Annexin A1, A4 and A5, LGALS-3 and SERPINF1 were featured in ‘negative regulation of cell death’. In conclusion, additionally to core ECM proteome, this study reveals ECM regulators and ECM affiliated proteins of interest to study for regenerative therapies, and their potential should be validated in future mechanistic experiments.
Collapse
Affiliation(s)
| | - Chitra Thangavel
- Ganga Research Centre, No 91, Mettuppalayam Road, Coimbatore, 641030, India
| | - Niek Djuric
- Ganga Research Centre, No 91, Mettuppalayam Road, Coimbatore, 641030, India.,Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Muthurajan Raveendran
- Department of Plant Biotechnology, Tamil Nadu Agricultural University, Coimbatore, 641003, India
| | | | | | | | - K S Sri Vijay Anand
- Department of Spine Surgery, Ganga Hospital, 313, Mettuppalayam Road, Coimbatore, 641043, India
| | | |
Collapse
|
20
|
The role of HIF proteins in maintaining the metabolic health of the intervertebral disc. Nat Rev Rheumatol 2021; 17:426-439. [PMID: 34083809 PMCID: PMC10019070 DOI: 10.1038/s41584-021-00621-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 01/18/2023]
Abstract
The physiologically hypoxic intervertebral disc and cartilage rely on the hypoxia-inducible factor (HIF) family of transcription factors to mediate cellular responses to changes in oxygen tension. During homeostatic development, oxygen-dependent prolyl hydroxylases, circadian clock proteins and metabolic intermediates control the activities of HIF1 and HIF2 in these tissues. Mechanistically, HIF1 is the master regulator of glycolytic metabolism and cytosolic lactate levels. In addition, HIF1 regulates mitochondrial metabolism by promoting flux through the tricarboxylic acid cycle, inhibiting downsteam oxidative phosphorylation and controlling mitochondrial health through modulation of the mitophagic pathway. Accumulation of metabolic intermediates from HIF-dependent processes contribute to intracellular pH regulation in the disc and cartilage. Namely, to prevent changes in intracellular pH that could lead to cell death, HIF1 orchestrates a bicarbonate buffering system in the disc, controlled by carbonic anhydrase 9 (CA9) and CA12, sodium bicarbonate cotransporters and an intracellular H+/lactate efflux mechanism. In contrast to HIF1, the role of HIF2 remains elusive; in disorders of the disc and cartilage, its function has been linked to both anabolic and catabolic pathways. The current knowledge of hypoxic cell metabolism and regulation of HIF1 activity provides a strong basis for the development of future therapies designed to repair the degenerative disc.
Collapse
|
21
|
Wang D, Hartman R, Han C, Zhou CM, Couch B, Malkamaki M, Roginskaya V, Van Houten B, Mullett SJ, Wendell SG, Jurczak MJ, Kang J, Lee J, Sowa G, Vo N. Lactate oxidative phosphorylation by annulus fibrosus cells: evidence for lactate-dependent metabolic symbiosis in intervertebral discs. Arthritis Res Ther 2021; 23:145. [PMID: 34020698 PMCID: PMC8139157 DOI: 10.1186/s13075-021-02501-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 04/03/2021] [Indexed: 01/03/2023] Open
Abstract
Background Intervertebral disc degeneration contributes to low back pain. The avascular intervertebral disc consists of a central hypoxic nucleus pulpous (NP) surrounded by the more oxygenated annulus fibrosus (AF). Lactic acid, an abundant end-product of NP glycolysis, has long been viewed as a harmful waste that acidifies disc tissue and decreases cell viability and function. As lactic acid is readily converted into lactate in disc tissue, the objective of this study was to determine whether lactate could be used by AF cells as a carbon source rather than being removed from disc tissue as a waste byproduct. Methods Import and conversion of lactate to tricarboxylic acid (TCA) cycle intermediates and amino acids in rabbit AF cells were measured by heavy-isotope (13C-lactate) tracing experiments using mass spectrometry. Levels of protein expression of lactate converting enzymes, lactate importer and exporter in NP and AF tissues were quantified by Western blots. Effects of lactate on proteoglycan (35S-sulfate) and collagen (3H-proline) matrix protein synthesis and oxidative phosphorylation (Seahorse XFe96 Extracellular Flux Analyzer) in AF cells were assessed. Results Heavy-isotope tracing experiments revealed that AF cells imported and converted lactate into TCA cycle intermediates and amino acids using in vitro cell culture and in vivo models. Addition of exogenous lactate (4mM) in culture media induced expression of the lactate importer MCT1 and increased oxygen consumption rate by 50%, mitochondrial ATP-linked respiration by 30%, and collagen synthesis by 50% in AF cell cultures grown under physiologic oxygen (2-5% O2) and glucose concentration (1-5mM). AF tissue highly expresses MCT1, LDH-H, an enzyme that preferentially converts lactate to pyruvate, and PDH, an enzyme that converts pyruvate to acetyl-coA. In contrast, NP tissue highly expresses MCT4, a lactate exporter, and LDH-M, an enzyme that preferentially converts pyruvate to lactate. Conclusions These findings support disc lactate-dependent metabolic symbiosis in which lactate produced by the hypoxic, glycolytic NP cells is utilized by the more oxygenated AF cells via oxidative phosphorylation for energy and matrix production, thus shifting the current research paradigm of viewing disc lactate as a waste product to considering it as an important biofuel. These scientifically impactful results suggest novel therapeutic targets in disc metabolism and degeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02501-2.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA
| | - Robert Hartman
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA.,University of Pittsburgh Medical Center Enterprises, Pittsburgh, PA, 15213, USA
| | - Chao Han
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA.,Tianjin Hospital, 406 Jiefang South Road Hexi District, Tianjin, PR China
| | - Chao-Ming Zhou
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA
| | - Brandon Couch
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA
| | - Matias Malkamaki
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA
| | - Vera Roginskaya
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Bennett Van Houten
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.,Health Sciences Metabolomics and Lipidomics Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stacy G Wendell
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.,Health Sciences Metabolomics and Lipidomics Core, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J Jurczak
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.,Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - James Kang
- Department of Orthopedics, Brigham and Women's Hospital, School of Medicine, Harvard University, 75 Francis Street, Boston, MA, 02115, USA
| | - Joon Lee
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA
| | - Gwendolyn Sowa
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA. .,Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| | - Nam Vo
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopedic and Spine Research, University of Pittsburgh, 200 Lothrop Street, E1644 Biomedical Science Tower, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
22
|
Kim JW, Jeon N, Shin DE, Lee SY, Kim M, Han DH, Shin JY, Lee S. Regeneration in Spinal Disease: Therapeutic Role of Hypoxia-Inducible Factor-1 Alpha in Regeneration of Degenerative Intervertebral Disc. Int J Mol Sci 2021; 22:ijms22105281. [PMID: 34067899 PMCID: PMC8155933 DOI: 10.3390/ijms22105281] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/05/2023] Open
Abstract
The intervertebral disc (IVD) is a complex joint structure comprising three primary components—namely, nucleus pulposus (NP), annulus fibrosus (AF), and cartilaginous endplate (CEP). The IVD retrieves oxygen from the surrounding vertebral body through CEP by diffusion and likely generates ATP via anaerobic glycolysis. IVD degeneration is characterized by a cascade of cellular, compositional, structural changes. With advanced age, pronounced changes occur in the composition of the disc extracellular matrix (ECM). NP and AF cells in the IVD possess poor regenerative capacity compared with that of other tissues. Hypoxia-inducible factor (HIF) is a master transcription factor that initiates a coordinated cellular cascade in response to a low oxygen tension environment, including the regulation of numerous enzymes in response to hypoxia. HIF-1α is essential for NP development and homeostasis and is involved in various processes of IVD degeneration process, promotes ECM in NP, maintains the metabolic activities of NP, and regulates dystrophic mineralization of NP, as well as angiogenesis, autophagy, and apoptosis during IVD degeneration. HIF-1α may, therefore, represent a diagnostic tool for early IVD degeneration and a therapeutic target for inhibiting IVD degeneration
Collapse
Affiliation(s)
- Jin-Woo Kim
- Department of Orthopaedic Surgery, Nowon Eulji Medical Center, Eulji University, Seoul 01830, Korea; (J.-W.K.); (N.J.); (M.K.)
| | - Neunghan Jeon
- Department of Orthopaedic Surgery, Nowon Eulji Medical Center, Eulji University, Seoul 01830, Korea; (J.-W.K.); (N.J.); (M.K.)
| | - Dong-Eun Shin
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13488, Korea; (D.-E.S.); (D.H.H.)
| | - So-Young Lee
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si 13488, Korea;
| | - Myongwhan Kim
- Department of Orthopaedic Surgery, Nowon Eulji Medical Center, Eulji University, Seoul 01830, Korea; (J.-W.K.); (N.J.); (M.K.)
| | - Dong Hun Han
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13488, Korea; (D.-E.S.); (D.H.H.)
| | - Jae Yeon Shin
- Department of Computer Science, College of IT Engineering, SeMyung University, Jechun 27136, Korea;
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13488, Korea; (D.-E.S.); (D.H.H.)
- Correspondence: ; Tel.: +82-31-780-5289; Fax: +82-31-708-3578
| |
Collapse
|
23
|
Hirota K. HIF-α Prolyl Hydroxylase Inhibitors and Their Implications for Biomedicine: A Comprehensive Review. Biomedicines 2021; 9:biomedicines9050468. [PMID: 33923349 PMCID: PMC8146675 DOI: 10.3390/biomedicines9050468] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Oxygen is essential for the maintenance of the body. Living organisms have evolved systems to secure an oxygen environment to be proper. Hypoxia-inducible factor (HIF) plays an essential role in this process; it is a transcription factor that mediates erythropoietin (EPO) induction at the transcriptional level under hypoxic environment. After successful cDNA cloning in 1995, a line of studies were conducted for elucidating the molecular mechanism of HIF activation in response to hypoxia. In 2001, cDNA cloning of dioxygenases acting on prolines and asparagine residues, which play essential roles in this process, was reported. HIF-prolyl hydroxylases (PHs) are molecules that constitute the core molecular mechanism of detecting a decrease in the partial pressure of oxygen, or hypoxia, in the cells; they can be called oxygen sensors. In this review, I discuss the process of molecular cloning of HIF and HIF-PH, which explains hypoxia-induced EPO expression; the development of HIF-PH inhibitors that artificially or exogenously activate HIF by inhibiting HIF-PH; and the significance and implications of medical intervention using HIF-PH inhibitors.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
24
|
What Are the Potential Roles of Nuclear Perlecan and Other Heparan Sulphate Proteoglycans in the Normal and Malignant Phenotype. Int J Mol Sci 2021; 22:ijms22094415. [PMID: 33922532 PMCID: PMC8122901 DOI: 10.3390/ijms22094415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/27/2022] Open
Abstract
The recent discovery of nuclear and perinuclear perlecan in annulus fibrosus and nucleus pulposus cells and its known matrix stabilizing properties in tissues introduces the possibility that perlecan may also have intracellular stabilizing or regulatory roles through interactions with nuclear envelope or cytoskeletal proteins or roles in nucleosomal-chromatin organization that may regulate transcriptional factors and modulate gene expression. The nucleus is a mechano-sensor organelle, and sophisticated dynamic mechanoresponsive cytoskeletal and nuclear envelope components support and protect the nucleus, allowing it to perceive and respond to mechano-stimulation. This review speculates on the potential roles of perlecan in the nucleus based on what is already known about nuclear heparan sulphate proteoglycans. Perlecan is frequently found in the nuclei of tumour cells; however, its specific role in these diseased tissues is largely unknown. The aim of this review is to highlight probable roles for this intriguing interactive regulatory proteoglycan in the nucleus of normal and malignant cell types.
Collapse
|
25
|
Xu YC, Gu Y, Yang JY, Xi K, Tang JC, Bian J, Cai F, Chen L. RACK1 mediates the advanced glycation end product-induced degradation of HIF-1α in nucleus pulposus cells via competing with HSP90 for HIF-1α binding. Cell Biol Int 2021; 45:1316-1326. [PMID: 33620117 DOI: 10.1002/cbin.11574] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 11/09/2022]
Abstract
Hyperglycemia can drive advanced glycation end product (AGE) accumulation and associated nucleus pulposus cell (NPC) dysfunction, but the basis for this activity has not been elucidated. Hypoxia-inducible factor-1α (HIF-1α) is subject to cell-type-specific AGE-mediated regulation. In the current study, we assessed the mechanistic relationship between AGE accumulation and HIF-1α degradation in NPCs. Immunohistochemical staining of degenerated nucleus pulposus (NP) samples was used to assess AGE levels. AGE impact on NPC survival and glycolysis-related gene expression was assessed via 3-(4,5)-dimethylthiazol(-z-y1)-3,5-di-phenyltetrazolium bromide assay and quantitative reverse-transcription polymerase chain reaction (qRT-PCR), while HIF-1α expression in NPCs following AGE treatment was monitored via Western blot analysis and qRT-PCR. Additionally, a luciferase reporter assay was used to monitor HIF-1α transcriptional activity. The importance of the receptor for activated C-kinase 1 (RACK1) as a mediator of HIF-1α degradation was evaluated through gain- and loss-of-function experiments. Competitive binding of RACK1 and HSP90 to HIF-1α was evaluated via immunoprecipitation. Increased AGE accumulation was evident in NP samples from diabetic patients, and AGE treatment resulted in reduced HIF-1α protein levels in NPCs that coincided with reduced HIF-1α transcriptional activity. AGE treatment impaired the stability of HIF-1α, leading to its RACK1-mediated proteasomal degradation in a manner independent of the canonical PHD-mediated degradation pathway. Additionally, RACK1 competed with HSP90 for HIF-1α binding following AGE treatment. AGE treatment of NPCs leads to HIF-1α protein degradation. RACK1 competes with HSP90 for HIF-1α binding following AGE treatment, resulting in posttranslational HIF-1α degradation. These results suggest that AGE is an intervertebral disc degeneration risk factor, and highlight potential avenues for the treatment or prevention of this disease.
Collapse
Affiliation(s)
- Yi-Chang Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Gu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jia-Ying Yang
- Department of Endocrinology and Metabolism, Shanghai Genenal Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Xi
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Cheng Tang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang Bian
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Cai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
26
|
Zheng F, Chen J, Zhang X, Wang Z, Chen J, Lin X, Huang H, Fu W, Liang J, Wu W, Li B, Yao H, Hu H, Song E. The HIF-1α antisense long non-coding RNA drives a positive feedback loop of HIF-1α mediated transactivation and glycolysis. Nat Commun 2021; 12:1341. [PMID: 33637716 PMCID: PMC7910558 DOI: 10.1038/s41467-021-21535-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 01/25/2021] [Indexed: 12/22/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a master driver of glucose metabolism in cancer cells. Here, we demonstrate that a HIF-1α anti-sense lncRNA, HIFAL, is essential for maintaining and enhancing HIF-1α-mediated transactivation and glycolysis. Mechanistically, HIFAL recruits prolyl hydroxylase 3 (PHD3) to pyruvate kinase 2 (PKM2) to induce its prolyl hydroxylation and introduces the PKM2/PHD3 complex into the nucleus via binding with heterogeneous nuclear ribonucleoprotein F (hnRNPF) to enhance HIF-1α transactivation. Reciprocally, HIF-1α induces HIFAL transcription, which forms a positive feed-forward loop to maintain the transactivation activity of HIF-1α. Clinically, high HIFAL expression is associated with aggressive breast cancer phenotype and poor patient outcome. Furthermore, HIFAL overexpression promotes tumor growth in vivo, while targeting both HIFAL and HIF-1α significantly reduces their effect on cancer growth. Overall, our results indicate a critical regulatory role of HIFAL in HIF-1α-driven transactivation and glycolysis, identifying HIFAL as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Fang Zheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaoqian Zhang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Jiewen Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaorong Lin
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hongyan Huang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wenkui Fu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jing Liang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
| | - Wei Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Bo Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hai Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Bioland Laboratory, Guangzhou, 510005, China.
- Fountain-Valley Institute for Life Sciences, 4th Floor, Building D, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Huangpu District, Guangzhou, 510535, China.
| |
Collapse
|
27
|
Snuggs JW, Tessier S, Bunning RAB, Shapiro IM, Risbud MV, Le Maitre CL. TonEBP regulates the hyperosmotic expression of aquaporin 1 and 5 in the intervertebral disc. Sci Rep 2021; 11:3164. [PMID: 33542263 PMCID: PMC7862284 DOI: 10.1038/s41598-021-81838-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/22/2020] [Indexed: 01/30/2023] Open
Abstract
The central region of the intervertebral disc (IVD) is rich in proteoglycans, leading to a hyperosmotic environment, which fluctuates with daily loading. The cells of the nucleus pulposus (NP cells) have adapted to this environment via the function of tonicity enhancer binding protein (TonEBP), and NP cells have been shown to express several water channels known as aquaporins (AQP). We have previously shown that AQP1 and 5 decrease during IVD degeneration. Here, the regulation of AQP1 and 5 by hyperosmotic conditions and the role of TonEBP in this regulation was investigated. AQP1 and 5 gene expression was upregulated by hyperosmotic conditions mimicking the osmolality of the healthy IVD, which was abrogated by TonEBP knockdown. Furthermore, AQP1 and 5 immunopositivity was significantly reduced in TonEBPΔ/Δ E17.5 mice when compared with wildtype controls, indicating in vivo expression of AQP1 and 5 is controlled at least in part by TonEBP. This hyperosmotic regulation of AQP1 and 5 could help to explain the decreased AQP1 and 5 expression during degeneration, when the osmolality of the NP decreases. Together this data suggests that TonEBP-regulated osmo-adaptation may be disrupted during IVD degeneration when the expression of both AQPs is reduced.
Collapse
Affiliation(s)
- J. W. Snuggs
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| | - S. Tessier
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA
| | - R. A. B. Bunning
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| | - I. M. Shapiro
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
| | - M. V. Risbud
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
| | - C. L. Le Maitre
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| |
Collapse
|
28
|
Li Y, Liu S, Pan D, Xu B, Xing X, Zhou H, Zhang B, Zhou S, Ning G, Feng S. The potential role and trend of HIF‑1α in intervertebral disc degeneration: Friend or foe? (Review). Mol Med Rep 2021; 23:239. [PMID: 33537810 PMCID: PMC7893690 DOI: 10.3892/mmr.2021.11878] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Lower back pain (LBP) is one of the most common reasons for seeking medical advice in orthopedic clinics. Increasingly, research has shown that symptomatic intervertebral disc degeneration (IDD) is mostly related to LBP. This review first outlines the research and findings of studies into IDD, from the physiological structure of the intervertebral disc (IVD) to various pathological cascades. The vicious cycles of IDD are re-described in relation to the analysis of the relationship among the pathological mechanisms involved in IDD. Interestingly, a ‘chief molecule’ was found, hypoxia-inducible factor-1α (HIF-1α), that may regulate all other mechanisms involved in IDD. When the vicious cycle is established, the low oxygen tension activates the expression of HIF-1α, which subsequently enters into the hypoxia-induced HIF pathways. The HIF pathways are dichotomized as friend and foe pathways according to the oxygen tension of the IVD microenvironment. Combined with clinical outcomes and previous research, the trend of IDD development has been predicted in this paper. Lastly, an early precautionary diagnosis and treatment method is proposed whereby nucleus pulposus tissue for biopsy can be obtained through IVD puncture guided by B-ultrasound when the patient is showing symptoms but MRI imaging shows negative results. The assessment criteria for biopsy and the feasibility, superiority and challenges of this approach have been discussed. Overall, it is clear that HIF-1α is an indispensable reference indicator for the accurate diagnosis and treatment of IDD.
Collapse
Affiliation(s)
- Yongjin Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shen Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Dayu Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Baoshan Xu
- Department of Spine Surgery, Tianjin Hospital, Tianjin 300000, P.R. China
| | - Xuewu Xing
- Department of Orthopedic Surgery, First Central Clinical of Tianjin Medical University, Tianjin 300052, P.R. China
| | - Hengxing Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Bin Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Suzhe Zhou
- Department of Orthopedics, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200034, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
29
|
Korbecki J, Kojder K, Kapczuk P, Kupnicka P, Gawrońska-Szklarz B, Gutowska I, Chlubek D, Baranowska-Bosiacka I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors-A Review of Literature. Int J Mol Sci 2021; 22:ijms22020843. [PMID: 33467722 PMCID: PMC7830156 DOI: 10.3390/ijms22020843] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an integral component of the tumor microenvironment. Either as chronic or cycling hypoxia, it exerts a similar effect on cancer processes by activating hypoxia-inducible factor-1 (HIF-1) and nuclear factor (NF-κB), with cycling hypoxia showing a stronger proinflammatory influence. One of the systems affected by hypoxia is the CXC chemokine system. This paper reviews all available information on hypoxia-induced changes in the expression of all CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8 (IL-8), CXCL9, CXCL10, CXCL11, CXCL12 (SDF-1), CXCL13, CXCL14, CXCL15, CXCL16, CXCL17) as well as CXC chemokine receptors—CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7 and CXCR8. First, we present basic information on the effect of these chemoattractant cytokines on cancer processes. We then discuss the effect of hypoxia-induced changes on CXC chemokine expression on the angiogenesis, lymphangiogenesis and recruitment of various cells to the tumor niche, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T cells (Tregs) and tumor-infiltrating lymphocytes (TILs). Finally, the review summarizes data on the use of drugs targeting the CXC chemokine system in cancer therapies.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Barbara Gawrońska-Szklarz
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
30
|
Kling L, Schreiber A, Eckardt KU, Kettritz R. Hypoxia-inducible factors not only regulate but also are myeloid-cell treatment targets. J Leukoc Biol 2020; 110:61-75. [PMID: 33070368 DOI: 10.1002/jlb.4ri0820-535r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Hypoxia describes limited oxygen availability at the cellular level. Myeloid cells are exposed to hypoxia at various bodily sites and even contribute to hypoxia by consuming large amounts of oxygen during respiratory burst. Hypoxia-inducible factors (HIFs) are ubiquitously expressed heterodimeric transcription factors, composed of an oxygen-dependent α and a constitutive β subunit. The stability of HIF-1α and HIF-2α is regulated by oxygen-sensing prolyl-hydroxylases (PHD). HIF-1α and HIF-2α modify the innate immune response and are context dependent. We provide a historic perspective of HIF discovery, discuss the molecular components of the HIF pathway, and how HIF-dependent mechanisms modify myeloid cell functions. HIFs enable myeloid-cell adaptation to hypoxia by up-regulating anaerobic glycolysis. In addition to effects on metabolism, HIFs control chemotaxis, phagocytosis, degranulation, oxidative burst, and apoptosis. HIF-1α enables efficient infection defense by myeloid cells. HIF-2α delays inflammation resolution and decreases antitumor effects by promoting tumor-associated myeloid-cell hibernation. PHDs not only control HIF degradation, but also regulate the crosstalk between innate and adaptive immune cells thereby suppressing autoimmunity. HIF-modifying pharmacologic compounds are entering clinical practice. Current indications include renal anemia and certain cancers. Beneficial and adverse effects on myeloid cells should be considered and could possibly lead to drug repurposing for inflammatory disorders.
Collapse
Affiliation(s)
- Lovis Kling
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Schreiber
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
31
|
Prolyl-hydroxylase inhibitors for the treatment of anemia in chronic kidney disease. Curr Opin Nephrol Hypertens 2020; 28:600-606. [PMID: 31567284 DOI: 10.1097/mnh.0000000000000554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Prolyl-hydroxylase inhibitors are a novel class of orally administered drugs that are under development for the treatment of anemia in patients with chronic kidney disease. This review discusses the biology of these drugs and their target - hypoxia-inducible factor and potential advantages and disadvantages of these therapies. Finally, we will discuss current trials in patients with both chronic kidney disease and end-stage renal disease. RECENT FINDINGS Recent smaller studies have found that prolyl-hydroxylase are as effective as erythropoietin in treating anemia of chronic kidney disease. We do not yet know if they have the same cardiovascular and cancer-related risk profile and these questions will be answered by large phase III trials that are ongoing. SUMMARY Although prolyl hydroxylase inhibitors have much potential, questions remain regarding their efficacy and safety. Should these concerns prove to be unfounded, the treatment of anemia in chronic kidney disease will likely be transformed over the next decade.
Collapse
|
32
|
Chen Y, Koike Y, Chi L, Ahmed A, Miyake H, Li B, Lee C, Delgado-Olguín P, Pierro A. Formula feeding and immature gut microcirculation promote intestinal hypoxia, leading to necrotizing enterocolitis. Dis Model Mech 2019; 12:dmm040998. [PMID: 31704804 PMCID: PMC6918740 DOI: 10.1242/dmm.040998] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/28/2019] [Indexed: 01/09/2023] Open
Abstract
Major risk factors for necrotizing enterocolitis (NEC) are formula feeding and prematurity; however, their pathogenic mechanisms are unknown. Here, we found that insufficient arginine/nitric oxide synthesis limits blood flow in the intestinal microvasculature, leading to hypoxia, mucosal damage and NEC in the premature intestine after formula feeding. Formula feeding led to increased intestinal hypoxia in pups at postnatal day (P)1 and P5, but not in more mature pups at P9. Accordingly, blood flow in the intestinal microvasculature increased after formula feeding in P9 pups only. mRNA profiling revealed that regulators of arginine/nitric oxide synthesis are at higher levels in endothelial cells of the intestine in P9 than in P1 pups. Importantly, arginine supplementation increased intestinal microvasculature blood flow and prevented NEC, whereas an arginine antagonist exacerbated NEC. Our results suggest that balancing intestinal oxygen demand and supply in the premature intestine by modulating arginine/nitric oxide could be used to prevent NEC.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yong Chen
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
- Department of Pediatric Surgery, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899 Singapore
| | - Yuhki Koike
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Lijun Chi
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Abdalla Ahmed
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Hiromu Miyake
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| | - Bo Li
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Carol Lee
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
| | - Paul Delgado-Olguín
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S1A8, Canada
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON M5S3H2, Canada
| | - Agostino Pierro
- Translational Medicine, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON M5G1X8, Canada
| |
Collapse
|
33
|
Elevated expression of hypoxia-inducible factor-2alpha regulated catabolic factors during intervertebral disc degeneration. Life Sci 2019; 232:116565. [PMID: 31251999 DOI: 10.1016/j.lfs.2019.116565] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
HEADINGS AIMS The present study determined whether nucleus pulposus (NP) cells express hypoxia-inducible factor-2alpha (HIF-2α) and assessed its role in regulating the expression of catabolic factors during intervertebral disc degeneration. MATERIALS AND METHODS Human degenerated NP tissues were acquired to examine the HIF-2α expression levels using immunohistochemistry, western blotting, and Real-time PCR. Human NP cells were cultivated under normoxic or hypoxic conditions, and the HIF-2α expression was determined. Then, human NP cells were treated with HIF-2α plasmids, HIF-2α siRNA, and tumor necrosis factor-alpha (TNF-α) to evaluate the role of HIF-2α in regulating matrix metalloproteinase (MMP) and aggrecanase expression. An in vivo rabbit disc degeneration model was established to demonstrate that HIF-2α plays a critical role in disc degeneration. KEY FINDINGS We found that HIF-2α had a markedly elevated expression in human degenerated discs in the Grade III stage. HIF-2α protein and gene transcript levels in vitro were relatively higher under hypoxic conditions. The expression of MMP-13, ADAMTS-4 was decreased significantly in HIF-2α silencing condition, while the over-expression resulted in significantly increased levels of MMP-13 and ADAMTS-4. When cytokine TNF-α was added, HIF-2α was induced by nuclear factor-κB (NF-κB). The in vivo experiments showed that the HIF-2α controlled the catabolic factors MMP-13 and ADAMTS-4 that regulated the collagen II and aggrecan metabolism in disc degeneration. SIGNIFICANCE HIF-2α is a catabolic regulator in disc degeneration and directly controls the catabolic genes. The suppression of HIF-2α expression leads to decelerates extracellular matrix degradation that might represent a therapeutic target for the degenerative disc.
Collapse
|
34
|
Pan H, Strickland A, Madhu V, Johnson ZI, Chand SN, Brody JR, Fertala A, Zheng Z, Shapiro IM, Risbud MV. RNA binding protein HuR regulates extracellular matrix gene expression and pH homeostasis independent of controlling HIF-1α signaling in nucleus pulposus cells. Matrix Biol 2019; 77:23-40. [PMID: 30092282 PMCID: PMC6367062 DOI: 10.1016/j.matbio.2018.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/02/2018] [Accepted: 08/05/2018] [Indexed: 12/19/2022]
Abstract
Nucleus pulposus (NP) cells reside in the hypoxic niche of the intervertebral disc. Studies have demonstrated that RNA-binding protein HuR modulates hypoxic signaling in several cancers, however, its function in the disc is unknown. HuR did not show cytoplasmic translocation in hypoxia and its silencing did not alter levels of Hif-1α or HIF-targets in NP cells. RNA-Sequencing data revealed that important extracellular matrix-related genes including several collagens, MMPs, aggrecan, Tgf-β3 and Sdc4 were regulated by HuR. Further analysis of HuR-silenced NP cells confirmed that HuR maintained expression of these matrix genes. We confirmed decreased levels of secreted collagen I and Sdc4 and increased pro-MMP13 in HuR-knockdown cells. In addition, messenger ribonucleoprotein immunoprecipitation demonstrated HuR binding to Tgf-β3 and Sdc4 mRNAs. Interestingly, while HuR bound to Hif-1α and Vegf mRNAs, it was clear that compensatory mechanisms sustained their expression when HuR was silenced. Noteworthy, despite the presence of multiple HuR-binding sites and reported interaction in other cell types, HuR showed no binding to Pgk1, Eno1, Pdk1 and Pfkfb3 in NP cells. Metabolic studies showed a significant decrease in the extracellular acidification rate (ECAR) and mitochondrial oxygen consumption rate (OCR) and acidic pH in HuR-silenced NP cells, without appreciable change in total OCR. These changes were likely due to decreased Ca12 expression in HuR silenced cells. Taken together, our study demonstrates for the first time that HuR regulates extracellular matrix (ECM) and pH homeostasis of NP cells and has important implications in the maintenance of intervertebral disc health.
Collapse
Affiliation(s)
- Hehai Pan
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Adam Strickland
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vedavathi Madhu
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zariel I Johnson
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Saswati N Chand
- Department of Surgery, The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan R Brody
- Department of Surgery, The Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Aker L, Ghannam M, Alzuabi MA, Jumah F, Alkhdour SM, Mansour S, Samara A, Cronk K, Massengale J, Holsapple J, Adeeb N, Oskouian RJ, Tubbs RS. Molecular Biology and Interactions in Intervertebral Disc Development, Homeostasis, and Degeneration, with Emphasis on Future Therapies: A Systematic Review. ACTA ACUST UNITED AC 2017. [DOI: 10.26632/ss.3.2017.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Schoepflin ZR, Silagi ES, Shapiro IM, Risbud MV. PHD3 is a transcriptional coactivator of HIF-1α in nucleus pulposus cells independent of the PKM2-JMJD5 axis. FASEB J 2017; 31:3831-3847. [PMID: 28495754 PMCID: PMC5572688 DOI: 10.1096/fj.201601291r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/24/2017] [Indexed: 12/17/2022]
Abstract
The role of prolyl hydroxylase (PHD)-3 as a hypoxia inducible factor (HIF)-1α cofactor is controversial and remains unknown in skeletal tissues. We investigated whether PHD3 controls HIF-1 transcriptional activity in nucleus pulposus (NP) cells through the pyruvate kinase muscle (PKM)-2-Jumonji domain--containing protein (JMJD5) axis. PHD3-/- mice (12.5 mo old) showed increased incidence of intervertebral disc degeneration with a concomitant decrease in expression of the HIF-1α targets VEGF-A, glucose transporter-1, and lactate dehydrogenase A. PHD3 silencing decreased hypoxic activation of HIF-1α C-terminal transactivation domain (C-TAD), but not HIF-1α-N-terminal-(N)-TAD or HIF-2α-TAD. Moreover, PHD3 suppression in NP cells resulted in decreased HIF-1α enrichment on target promoters and lower expression of select HIF-1 targets. Contrary to other cell types, manipulation of PKM2 and JMJD5 levels had no effect on HIF-1 activity in NP cells. Likewise, stabilization of tetrameric PKM2 by a chemical approach had no effect on PHD3-dependent HIF-1 activity. Coimmunoprecipitation assays showed lack of association between HIF-1α and PKM2 in NP cells. Results support the role of the PHD3 as a cofactor for HIF-1, independent of PKM2-JMJD5.-Schoepflin, Z. R., Silagi, E. S., Shapiro, I. M., Risbud, M. V. PHD3 is a transcriptional coactivator of HIF-1α in nucleus pulposus cells independent of the PKM2-JMJD5 axis.
Collapse
Affiliation(s)
- Zachary R Schoepflin
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Elizabeth S Silagi
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
37
|
Wang J, Pan H, Li X, Zhang K, Li Z, Wang H, Zheng Z, Liu H. Hypoxia suppresses serum deprivation-induced degradation of the nucleus pulposus cell extracellular matrix through the JNK and NF-κB pathways. J Orthop Res 2017; 35:2059-2066. [PMID: 27869311 DOI: 10.1002/jor.23486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/16/2016] [Indexed: 02/04/2023]
Abstract
Intervertebral disc (IVD) degeneration is associated with the imbalance between anabolism and catabolism of the nucleus pulposus (NP) extracellular matrix (ECM). Serum deprivation (SD) has been reported to exacerbate IVD degeneration; however, the effect of SD on ECM metabolism is not fully understood. Hypoxia plays important roles in maintaining the physiological functions of IVD cells; however, whether hypoxia has any effect on NP ECM production under conditions of SD is still unclear. In the current study, we established an in vitro SD model by exposing NP cells to serum-free medium. SD decreased the expression of aggrecan and collagen II, as well as the production of sulfated glycosaminoglycan (sGAG) in a time-dependent manner. However, hypoxia abolished SD-mediated down-regulation of aggrecan and collagen II expression via JNK1/2 activation. Moreover, hypoxia abolished SD-induced MMP-3 and MMP-13 expression by inhibiting NF-κB activation, p65 translocation, and MMP-3 and MMP-13 promoter activity. These results indicated that, hypoxia maintained ECM production under conditions of SD. This effect was elicited in part through JNK1/2-mediated up-regulation of matrix gene expression and down-regulation of MMP expression, through the inhibition of NF-κB. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2059-2066, 2017.
Collapse
Affiliation(s)
- Jianru Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Hehai Pan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Kuibo Zhang
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, Guangdong, China
| | - Zemin Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Hua Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Hui Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| |
Collapse
|
38
|
Johnson ZI, Doolittle AC, Snuggs JW, Shapiro IM, Le Maitre CL, Risbud MV. TNF-α promotes nuclear enrichment of the transcription factor TonEBP/NFAT5 to selectively control inflammatory but not osmoregulatory responses in nucleus pulposus cells. J Biol Chem 2017; 292:17561-17575. [PMID: 28842479 DOI: 10.1074/jbc.m117.790378] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/03/2017] [Indexed: 01/07/2023] Open
Abstract
Intervertebral disc degeneration (IDD) causes chronic back pain and is linked to production of proinflammatory molecules by nucleus pulposus (NP) and other disc cells. Activation of tonicity-responsive enhancer-binding protein (TonEBP)/NFAT5 by non-osmotic stimuli, including proinflammatory molecules, occurs in cells involved in immune response. However, whether inflammatory stimuli activate TonEBP in NP cells and whether TonEBP controls inflammation during IDD is unknown. We show that TNF-α, but not IL-1β or LPS, promoted nuclear enrichment of TonEBP protein. However, TNF-α-mediated activation of TonEBP did not cause induction of osmoregulatory genes. RNA sequencing showed that 8.5% of TNF-α transcriptional responses were TonEBP-dependent and identified genes regulated by both TNF-α and TonEBP. These genes were over-enriched in pathways and diseases related to inflammatory response and inhibition of matrix metalloproteases. Based on RNA-sequencing results, we further investigated regulation of novel TonEBP targets CXCL1, CXCL2, and CXCL3 TonEBP acted synergistically with TNF-α and LPS to induce CXCL1-proximal promoter activity. Interestingly, this regulation required a highly conserved NF-κB-binding site but not a predicted TonE, suggesting cross-talk between these two members of the Rel family. Finally, analysis of human NP tissue showed that TonEBP expression correlated with canonical osmoregulatory targets TauT/SLC6A6, SMIT/SLC5A3, and AR/AKR1B1, supporting in vitro findings that the inflammatory milieu during IDD does not interfere with TonEBP osmoregulation. In summary, whereas TonEBP participates in the proinflammatory response to TNF-α, therapeutic strategies targeting this transcription factor for treatment of disc disease must spare osmoprotective, prosurvival, and matrix homeostatic activities.
Collapse
Affiliation(s)
- Zariel I Johnson
- From the Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Alexandra C Doolittle
- From the Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Joseph W Snuggs
- the Biomolecular Sciences Research Centre, Sheffield Hallam University, S1 1WB Sheffield, United Kingdom
| | - Irving M Shapiro
- From the Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| | - Christine L Le Maitre
- the Biomolecular Sciences Research Centre, Sheffield Hallam University, S1 1WB Sheffield, United Kingdom
| | - Makarand V Risbud
- From the Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107 and
| |
Collapse
|
39
|
Liu C, Choi H, Johnson ZI, Tian J, Shapiro IM, Risbud MV. Lack of evidence for involvement of TonEBP and hyperosmotic stimulus in induction of autophagy in the nucleus pulposus. Sci Rep 2017; 7:4543. [PMID: 28674405 PMCID: PMC5495809 DOI: 10.1038/s41598-017-04876-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 05/16/2017] [Indexed: 12/04/2022] Open
Abstract
Nucleus pulposus (NP) cells reside in a physiologically hyperosmotic environment within the intervertebral disc. TonEBP/NFAT5 is an osmo-sensitive transcription factor that controls expression of genes critical for cell survival under hyperosmotic conditions. A recent report on NP and studies of other cell types have shown that hyperosmolarity triggers autophagy. However, little is known whether such autophagy induction occurs through TonEBP. The goal of this study was to investigate the role of TonEBP in hyperosmolarity-dependent autophagy in NP. Loss-of-function studies showed that autophagy in NP cells was not TonEBP-dependent; hyperosmolarity did not upregulate autophagy as previously reported. NP tissue of haploinsufficient TonEBP mice showed normal pattern of LC3 staining. NP cells did not increase LC3-II or LC3-positive puncta under hyperosmotic conditions. Bafilomycin-A1 treatment and tandem mCherry-EGFP-LC3B reporter transfection demonstrated that the autophagic flux was unaffected by hyperosmolarity. Even under serum-free conditions, NP cells did not induce autophagy with increasing osmolarity. Hyperosmolarity did not change the phosphorylation of ULK1 by mTOR and AMPK. An ex vivo disc organ culture study supported that extracellular hyperosmolarity plays no role in promoting autophagy in the NP. We conclude that hyperosmolarity does not play a role in autophagy induction in NP cells.
Collapse
Affiliation(s)
- Chao Liu
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Orthopaedics, The Central Hospital of Songjiang District, Shanghai, China
| | - Hyowon Choi
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zariel I Johnson
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jiwei Tian
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Fuhrmann DC, Brüne B. Mitochondrial composition and function under the control of hypoxia. Redox Biol 2017; 12:208-215. [PMID: 28259101 PMCID: PMC5333533 DOI: 10.1016/j.redox.2017.02.012] [Citation(s) in RCA: 434] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/13/2022] Open
Abstract
Hypoxia triggers several mechanisms to adapt cells to a low oxygen environment. Mitochondria are major consumers of oxygen and a potential source of reactive oxygen species (ROS). In response to hypoxia they exchange or modify distinct subunits of the respiratory chain and adjust their metabolism, especially lowering the citric acid cycle. Intermediates of the citric acid cycle participate in regulating hypoxia inducible factors (HIF), the key mediators of adaptation to hypoxia. Here we summarize how hypoxia conditions mitochondria with consequences for ROS-production and the HIF-pathway. Hypoxia provokes changes in mitochondrial morphology, metabolism, and respiration. Hypoxia calls forth changes in redox signaling. HIF-signaling is linked to mitochondrial metabolism and ROS formation. Hypoxia adjusts ETC complex formation, activity, respiration, and ROS formation.
Collapse
Affiliation(s)
- Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| |
Collapse
|
41
|
Vo NV, Hartman RA, Patil PR, Risbud MV, Kletsas D, Iatridis JC, Hoyland JA, Le Maitre CL, Sowa GA, Kang JD. Molecular mechanisms of biological aging in intervertebral discs. J Orthop Res 2016; 34:1289-306. [PMID: 26890203 PMCID: PMC4988945 DOI: 10.1002/jor.23195] [Citation(s) in RCA: 280] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/03/2016] [Indexed: 02/04/2023]
Abstract
Advanced age is the greatest risk factor for the majority of human ailments, including spine-related chronic disability and back pain, which stem from age-associated intervertebral disc degeneration (IDD). Given the rapid global rise in the aging population, understanding the biology of intervertebral disc aging in order to develop effective therapeutic interventions to combat the adverse effects of aging on disc health is now imperative. Fortunately, recent advances in aging research have begun to shed light on the basic biological process of aging. Here we review some of these insights and organize the complex process of disc aging into three different phases to guide research efforts to understand the biology of disc aging. The objective of this review is to provide an overview of the current knowledge and the recent progress made to elucidate specific molecular mechanisms underlying disc aging. In particular, studies over the last few years have uncovered cellular senescence and genomic instability as important drivers of disc aging. Supporting evidence comes from DNA repair-deficient animal models that show increased disc cellular senescence and accelerated disc aging. Additionally, stress-induced senescent cells have now been well documented to secrete catabolic factors, which can negatively impact the physiology of neighboring cells and ECM. These along with other molecular drivers of aging are reviewed in depth to shed crucial insights into the underlying mechanisms of age-related disc degeneration. We also highlight molecular targets for novel therapies and emerging candidate therapeutics that may mitigate age-associated IDD. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1289-1306, 2016.
Collapse
Affiliation(s)
- Nam V. Vo
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert A. Hartman
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prashanti R. Patil
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Makarand V. Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - James C. Iatridis
- Leni & Peter W May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Judith A. Hoyland
- Centre for Tissue Injury and Repair, Faculty of Medical and Human Sciences, University of Manchester M13 9PT and NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester Academic Health Science Centre, Manchester, UK
| | - Christine L. Le Maitre
- Musculoskeletal and Regenerative Medicine Research Group, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK
| | - Gwendolyn A. Sowa
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James D. Kang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
42
|
Singh Y, Shi X, Zhang S, Umbach AT, Chen H, Salker MS, Lang F. Prolyl hydroxylase 3 (PHD3) expression augments the development of regulatory T cells. Mol Immunol 2016; 76:7-12. [DOI: 10.1016/j.molimm.2016.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/28/2016] [Accepted: 06/02/2016] [Indexed: 01/04/2023]
|
43
|
Wang F, Cai F, Shi R, Wei JN, Wu XT. Hypoxia regulates sumoylation pathways in intervertebral disc cells: implications for hypoxic adaptations. Osteoarthritis Cartilage 2016; 24:1113-24. [PMID: 26826302 DOI: 10.1016/j.joca.2016.01.134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the hypoxic regulation of sumoylation pathways and cell viability in nucleus pulposus (NP) and annulus fibrosus (AF) cells. DESIGN Expression of small ubiquitin-like modifier (SUMO) molecules, SUMO E1 activating enzymes SAE1 and SAE2, SUMO E2 conjugating enzyme UBC9, and de-sumoylation enzyme sentrin/SUMO-specific proteases (SENP)1 was immunolocalized in rat intervertebral disc (IVD) cells. NP and AF cells were cultured in hypoxia and cell viability was evaluated by quantifying cell proliferation, cellular senescence, apoptosis, and cell cycle distribution. Hypoxic regulation of sumoylation pathways was studied by analyzing the transcription and expression of SUMO molecules and sumoylation enzymes. Loss of function study using SENP1 siRNA was performed to investigate the regulatory role of sumoylation on the function of hypoxia inducible factor 1α (HIF-1α) and the hypoxic tolerance of IVD cells. RESULTS Sumoylation pathways were expressed in IVD cells and localized predominantly in nuclei. Both NP and AF cells maintained viability under hypoxia and upregulated the expression of SENP1. In NP cells hypoxia transiently increased the expression of SUMO-1, SUMO-2/3, SAE2, and UBC9, whereas SUMO-1 was elevated while SUMO-2/3, SAE1, SAE2, and UBC9 were reduced by low oxygen tensions in AF cells. Although downregulation of SENP1 decreased the transcriptional activity of HIF-1α, the viability of disc cells showed no significant loss under hypoxia. CONCLUSIONS NP and AF cells equally tolerate oxygen deficiency, but differently regulate the sumoylation pathways under hypoxia. The distinct sumoylation dynamics may help extend our understanding of the cell-specific regulation of the molecular basis that promotes cell survival in the hypoxic IVD.
Collapse
Affiliation(s)
- F Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - F Cai
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - R Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - J-N Wei
- Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - X-T Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| |
Collapse
|
44
|
Schoepflin ZR, Shapiro IM, Risbud MV. Class I and IIa HDACs Mediate HIF-1α Stability Through PHD2-Dependent Mechanism, While HDAC6, a Class IIb Member, Promotes HIF-1α Transcriptional Activity in Nucleus Pulposus Cells of the Intervertebral Disc. J Bone Miner Res 2016; 31:1287-99. [PMID: 26765925 PMCID: PMC4891304 DOI: 10.1002/jbmr.2787] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/29/2015] [Accepted: 01/08/2016] [Indexed: 12/17/2022]
Abstract
The objective of this study was to determine the role of histone deacetylases (HDACs) in regulating HIF-1α protein stability and activity in nucleus pulposus (NP) cells. Treatment of NP cells with pan-HDAC inhibitor TSA resulted in decreased HIF-1α levels under both normoxia and hypoxia in a dose-dependent fashion. TSA-mediated HIF-1α degradation was rescued by concomitant inhibition of not only the 26S proteasome but also PHD2 function. Moreover, TSA treatment of PHD2(-/-) cells had little effect on HIF-1α levels, supporting the notion that inhibition of PHD2 function by HDACs contributed to HIF-1α stabilization. Surprisingly, class-specific HDAC inhibitors did not affect HIF-1α protein stability, indicating that multiple HDACs controlled HIF-1α stability by regulating HIF-1α-PHD2 interaction in NP cells. Interestingly, lower-dose TSA that did not affect HIF-1α stability decreased its activity and target gene expression. Likewise, rescue of TSA-mediated HIF-1α protein degradation by blocking proteasomal or PHD activity did not restore HIF-1 activity, suggesting that HDACs independently regulate HIF-1α stability and activity. Noteworthy, selective inhibition of HDAC6 and not of class I and IIa HDACs decreased HIF-1-mediated transcription under hypoxia to a similar extent as lower-dose TSA, contrasting the reported role of HDAC6 as a transcriptional repressor in other cell types. Moreover, HDAC6 inhibition completely blocked TSA effects on HIF-1 activity. HDAC6 associated with and deacetylated HSP90, an important cofactor for HIF-1 function in NP cells, and HDAC6 inhibition decreased p300 transactivation in NP cells. Taken together, these results suggest that although multiple class I and class IIa HDACs control HIF-1 stability, HDAC6, a class IIb HDAC, is a novel mediator of HIF-1 activity in NP cells possibly through promoting action of critical HIF-1 cofactors. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Zachary R Schoepflin
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
45
|
Heyman SN, Leibowitz D, Mor-Yosef Levi I, Liberman A, Eisenkraft A, Alcalai R, Khamaisi M, Rosenberger C. Adaptive response to hypoxia and remote ischaemia pre-conditioning: a new hypoxia-inducible factors era in clinical medicine. Acta Physiol (Oxf) 2016; 216:395-406. [PMID: 26449466 DOI: 10.1111/apha.12613] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/01/2015] [Accepted: 10/02/2015] [Indexed: 01/30/2023]
Abstract
Transient ischaemia leads to tolerance to subsequent protracted ischaemia. This 'ischaemia pre-conditioning' results from the induction of numerous protective genes, involved in cell metabolism, proliferation and survival, in antioxidant capacity, angiogenesis, vascular tone and erythropoiesis. Hypoxia-inducible factors (HIF) play a pivotal role in this transcriptional adaptive response. HIF prolyl hydroxylases (PHDs), serving as oxygen sensors, control HIFα degradation. HIF-mediated ischaemic pre-conditioning can be achieved with the administration of PHD inhibitors, with the attenuation of organ injury under various hypoxic and toxic insults. Clinical trials are currently under way, evaluating PHD inhibitors as inducers of erythropoietin. Once their safety is established, their potential use might be further tested in clinical trials in various forms of acute ischaemic and toxic organ damage. Repeated transient limb ischaemia was also found to attenuate ischaemic injury in remote organs. This 'remote ischaemic pre-conditioning' phenomenon (RIP) has been extensively studied recently in small clinical trials, preceding, or in parallel with an abrupt insult, such as myocardial infarction, cardiac surgery or radiocontrast administration. Initial results are promising, suggesting organ protection. Large-scale multi-centre studies are currently under way, evaluating the protective potential of RIP in cardiac surgery, in the management of myocardial infarction and in organ transplantation. The mechanisms of organ protection provided by RIP are poorly understood, but HIF seemingly play a role as well. Thus, Inhibition of HIF degradation with PHD inhibitors, as well as RIP (in part through HIF), might develop into novel clinical interventions in organ protection in the near future.
Collapse
Affiliation(s)
- S. N. Heyman
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - D. Leibowitz
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - I. Mor-Yosef Levi
- Department of Nephrology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Liberman
- Department of Neurology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Eisenkraft
- The Research Institute for Military Medicine; The Hebrew University Medical School and the Israeli Defense Force Medical Corps; Jerusalem Israel
| | - R. Alcalai
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | | | | |
Collapse
|
46
|
Johnson ZI, Gogate SS, Day R, Binch A, Markova DZ, Chiverton N, Cole A, Conner M, Shapiro IM, Le Maitre CL, Risbud MV. Aquaporin 1 and 5 expression decreases during human intervertebral disc degeneration: Novel HIF-1-mediated regulation of aquaporins in NP cells. Oncotarget 2016; 6:11945-58. [PMID: 25844601 PMCID: PMC4494915 DOI: 10.18632/oncotarget.3631] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/05/2015] [Indexed: 01/07/2023] Open
Abstract
Objectives of this study were to investigate whether AQP1 and AQP5 expression is altered during intervertebral disc degeneration and if hypoxia and HIF-1 regulate their expression in NP cells. AQP expression was measured in human tissues from different degenerative grades; regulation by hypoxia and HIF-1 was studied using promoter analysis and gain- and loss-of-function experiments. We show that both AQPs are expressed in the disc and that mRNA and protein levels decline with human disease severity. Bioinformatic analyses of AQP promoters showed multiple evolutionarily conserved HREs. Surprisingly, hypoxia failed to induce promoter activity or expression of either AQP. While genomic chromatin immunoprecipitation showed limited binding of HIF-1α to conserved HREs, their mutation did not suppress promoter activities. Stable HIF-1α suppression significantly decreased mRNA and protein levels of both AQPs, but HIF-1α failed to induce AQP levels following accumulation. Together, our results demonstrate that AQP1 and AQP5 expression is sensitive to human disc degeneration and that HIF-1α uniquely maintains basal expression of both AQPs in NP cells, independent of oxemic tension and HIF-1 binding to promoter HREs. Diminished HIF-1 activity during degeneration may suppress AQP levels in NP cells, compromising their ability to respond to extracellular osmolarity changes.
Collapse
Affiliation(s)
- Zariel I Johnson
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shilpa S Gogate
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rebecca Day
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Abbie Binch
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Dessislava Z Markova
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Neil Chiverton
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ashley Cole
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Matt Conner
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
47
|
Reciprocal regulation by hypoxia-inducible factor-2α and the NAMPT-NAD(+)-SIRT axis in articular chondrocytes is involved in osteoarthritis. Osteoarthritis Cartilage 2015. [PMID: 26209889 DOI: 10.1016/j.joca.2015.07.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Hypoxia-inducible factor-2α (HIF-2α) transcriptionally upregulates Nampt in articular chondrocytes. NAMPT, which exhibits nicotinamide phosphoribosyltransferase activity, in turn causes osteoarthritis (OA) in mice by stimulating the expression of matrix-degrading enzymes. Here, we sought to elucidate whether HIF-2α activates the NAMPT-NAD(+)-SIRT axis in chondrocytes and thereby contributes to the pathogenesis of OA. METHODS Assays of NAD levels, SIRT activity, reporter gene activity, mRNA, and protein levels were conducted in primary cultured mouse articular chondrocytes. Experimental OA in mice was induced by intra-articular (IA) injection of adenovirus expressing HIF-2α (Ad-Epas1) or NAMPT (Ad-Nampt). The functions of SIRT in OA were examined by IA co-injection of SIRT inhibitors or adenovirus expressing individual SIRT isoforms or shRNA targeting specific SIRT isoforms. RESULTS HIF-2α activated the NAMPT-NAD(+)-SIRT axis in chondrocytes by upregulating NAMPT, which stimulated NAD(+) synthesis and thereby activated SIRT family members. The activated NAMPT-SIRT pathway, in turn, promoted HIF-2α protein stability by negatively regulating its hydroxylation and 26S proteasome-mediated degradation, resulting in increased HIF-2α transcriptional activity. Among SIRT family members (SIRT1-7), SIRT2 and SIRT4 were positively associated with HIF-2α stability and transcriptional activity in chondrocytes. This reciprocal regulation was required for the expression of catabolic matrix metalloproteinases (MMP3, MMP12, and MMP13) and OA cartilage destruction caused by IA injection of Ad-Epas1 Ad-Nampt. CONCLUSION The reciprocal regulation of HIF-2α and the NAMPT-NAD(+)-SIRT axis in articular chondrocytes is involved in OA cartilage destruction caused by HIF-2α or NAMPT.
Collapse
|
48
|
Zhang J, Wang C, Chen X, Takada M, Fan C, Zheng X, Wen H, Liu Y, Wang C, Pestell RG, Aird KM, Kaelin WG, Liu XS, Zhang Q. EglN2 associates with the NRF1-PGC1α complex and controls mitochondrial function in breast cancer. EMBO J 2015; 34:2953-70. [PMID: 26492917 DOI: 10.15252/embj.201591437] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022] Open
Abstract
The EglN2/PHD1 prolyl hydroxylase is an important oxygen sensor contributing to breast tumorigenesis. Emerging studies suggest that there is functional cross talk between oxygen sensing and mitochondrial function, both of which play an essential role for sustained tumor growth. However, the potential link between EglN2 and mitochondrial function remains largely undefined. Here, we show that EglN2 depletion decreases mitochondrial respiration in breast cancer under normoxia and hypoxia, which correlates with decreased mitochondrial DNA in a HIF1/2α-independent manner. Integrative analyses of gene expression profile and genomewide binding of EglN2 under hypoxic conditions reveal nuclear respiratory factor 1 (NRF1) motif enrichment in EglN2-activated genes, suggesting NRF1 as an EglN2 binding partner. Mechanistically, by forming an activator complex with PGC1α and NRF1 on chromatin, EglN2 promotes the transcription of ferridoxin reductase (FDXR) and maintains mitochondrial function. In addition, FDXR, as one of effectors for EglN2, contributes to breast tumorigenesis in vitro and in vivo. Our findings suggest that EglN2 regulates mitochondrial function in ERα-positive breast cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chengyang Wang
- Department of Bioinformatics, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xi Chen
- Department of Molecular and Cellular Biology, The Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mamoru Takada
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Xingnan Zheng
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Haitao Wen
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Yong Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chenguang Wang
- Program of Radiation Protection and Drug Discovery, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin, China
| | - Richard G Pestell
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Katherine M Aird
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Xiaole Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
49
|
Inducible glomerular erythropoietin production in the adult kidney. Kidney Int 2015; 88:1345-1355. [PMID: 26398496 DOI: 10.1038/ki.2015.274] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/26/2015] [Accepted: 07/02/2015] [Indexed: 02/07/2023]
Abstract
Hypoxia-inducible factor (HIF)-2-triggered erythropoietin production in renal interstitial fibroblast-like cells is the physiologically relevant source of erythropoietin for regulating erythropoiesis. During renal fibrosis, these cells transform into myofibroblasts and lose their ability to produce sufficient erythropoietin leading to anemia. To find if other cells for erythropoietin production might exist in the kidney we tested for the capability of nonepithelial glomerular cells to elaborate erythropoietin. Therefore, HIF transcription factors were stabilized by cell-specific deletion of the von Hippel-Lindau (VHL) gene. Inducible deletion of VHL in glomerular connexin40-expressing cells (endothelial, renin-expressing, and mesangial cells) markedly increased glomerular erythropoietin mRNA expression levels, plasma erythropoietin concentrations, and hematocrit values. These changes were mimicked by inducible cell-specific VHL deletion in renin-expressing and in mesangial cells but not in endothelial cells. The increases of erythropoietin production were absent, when VHL was co-deleted with HIF-2. The induction of glomerular erythropoietin expression was associated with the downregulation of juxtaglomerular renin expression, again in a HIF-2-dependent manner. Thus, VHL deletion in renin-expressing and in mesangial cells induces the capability to produce relevant amounts of erythropoietin and to suppress renin expression in the adult kidney if HIF-2 is stabilized.
Collapse
|
50
|
Esfahani M, Karimi F, Afshar S, Niknazar S, Sohrabi S, Najafi R. Prolyl hydroxylase inhibitors act as agents to enhance the efficiency of cell therapy. Expert Opin Biol Ther 2015; 15:1739-55. [PMID: 26325448 DOI: 10.1517/14712598.2015.1084281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION In stem cell-based therapy as a subtype of regenerative medicine, stem cells can be used to replace or repair injured tissue and cells in order to treat disease. Stem cells have the ability to integrate into injured areas and produce new cells via processes of proliferation and differentiation. Several studies have demonstrated that hypoxia increases self-renewal, proliferation and post-homing differentiation of stem cells through the regulation of hypoxia-inducible factor-1 (HIF-1)-mediated gene expression. Thus, pharmacological interventions including prolyl hydroxylase (PHD) inhibitors are considered as promising solutions for stem cell-based therapy. PHD inhibitors stabilize the HIF-1 and activate its pathway through preventing proteasomal degradation of HIF-1. AREAS COVERED This review focuses on the role of hypoxia, HIF-1 and especially PHD inhibitors on cell therapy. PHD structure and function are discussed as well as their inhibitors. In addition, we have investigated several preclinical studies in which PHD inhibitors improved the efficiency of cell-based therapies. EXPERT OPINION The data reviewed here suggest that PHD inhibitors are effective operators in improving stem cell therapy. However, because of some limitations, these compounds should be properly examined before clinical application.
Collapse
Affiliation(s)
- Maryam Esfahani
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Fatemeh Karimi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Saeid Afshar
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Somayeh Niknazar
- b 2 Shahid Beheshti University of Medical Science, Hearing Disorders Research Center , Tehran, the Islamic Republic of Iran
| | - Sareh Sohrabi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Rezvan Najafi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| |
Collapse
|