1
|
Luo Y, Li WX, Zheng QS, Yan JQ, Yang YD, Shen SR, Zhang QH, Liang G, Wang Y, Chen DD, Hu X, Luo W. OTUD1 deficiency attenuates myocardial ischemia/reperfusion induced cardiomyocyte apoptosis by regulating RACK1 phosphorylation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01567-x. [PMID: 40394237 DOI: 10.1038/s41401-025-01567-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/14/2025] [Indexed: 05/22/2025]
Abstract
Myocardial infarction (MI) is an important risk factor of cardiovascular disease (CVD) and its incidence has been on the rise globally. Myocardial ischemia/reperfusion (I/R) injury is frequently detected in the ischemic myocardium. Recent studies have shown that ubiquitination plays an important role in the cardiac pathophysiological processes. Herein, we investigated the role and molecular mechanism of Ovarian tumor deubiquitinase 1 (OTUD1) in I/R induced myocardial injury. It was observed that the myocardial OTUD1 was upregulated in I/R-induced heart tissues and global deletion of OTUD1 significantly ameliorated I/R induced myocardial injury and dysfunction. Similarly, silencing or overexpression OTUD1 affected the hypoxia/reoxygenation (H/R) induced cell apoptosis in cultured cardiomyocytes. Mechanistically, immunoprecipitation-mass spectrometry revealed that OTUD1 directly bound to receptor for activated C-kinase 1 (RACK1) which has been identified as a scaffold protein for multiple kinases including mitogen-activated protein kinase (MAPKs) and Inhibitor of nuclear factor kappa B kinase (IKK). OTUD1 could cleave K63-linked polyubiquitin chains to enhance RACK1 phosphorylation, thus modulating MAPKs and nuclear factor kappa B (NF-κB) signaling. Finally, silencing of RACK1 reverses OTUD1-promoted H/R induced myocardial apoptosis. In conclusion, our findings suggest that OTUD1 promotes I/R-induced heart injury by deubiquitinating RACK1, suggesting that OTUD1 is a potential therapeutic target for myocardial I/R.
Collapse
Affiliation(s)
- Yue Luo
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou, 325800, China
| | - Wei-Xin Li
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qing-Song Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jue-Qian Yan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yu-Die Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Si-Rui Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qian-Hui Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Ding-Dao Chen
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou, 325800, China.
| | - Xiang Hu
- Department of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Wu Luo
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou, 325800, China.
- Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
Adlakha A, Williams TJ, Shou X, Reed AK, Lenhard B, Armstrong-James D. Interferon-gamma rescues dendritic cell calcineurin-dependent responses to Aspergillus fumigatus via Stat3 to Stat1 switching. iScience 2025; 28:111535. [PMID: 39898039 PMCID: PMC11787545 DOI: 10.1016/j.isci.2024.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/11/2024] [Accepted: 12/03/2024] [Indexed: 02/04/2025] Open
Abstract
Invasive pulmonary aspergillosis is a lethal opportunistic fungal infection in transplant recipients receiving calcineurin inhibitors. We previously identified a role for the calcineurin pathway in innate immune responses to A. fumigatus and have used exogenous interferon-gamma successfully to treat aspergillosis in this setting. Here we show that calcineurin inhibitors block dendritic cell maturation in response to A. fumigatus, impairing the Th1 polarization of CD4 cells. Interferon gamma, an immunotherapeutic option for invasive aspergillosis, restored maturation and promoted Th1 polarization via a dendritic cell dependent effect that was co-dependent on T cell interaction. We find that interferon gamma activates alternative transcriptional pathways to calcineurin-NFAT for the augmentation of pathogen handling. Histone modification ChIP-Seq analysis revealed dominant control by an interferon gamma induced regulatory switch from STAT3 to STAT1 transcription factor binding underpinning these observations. These findings provide key insight into the mechanisms of immunotherapy in organ transplant recipients with invasive fungal diseases.
Collapse
Affiliation(s)
- Amit Adlakha
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- MRC London Institute of Medical Sciences, Imperial College, London W12 0NN, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Thomas J. Williams
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Xinxin Shou
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
| | - Anna K. Reed
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Boris Lenhard
- MRC London Institute of Medical Sciences, Imperial College, London W12 0NN, UK
| | - Darius Armstrong-James
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| |
Collapse
|
3
|
Zhang J, Yao M, Xia S, Zeng F, Liu Q. Systematic and comprehensive insights into HIF-1 stabilization under normoxic conditions: implications for cellular adaptation and therapeutic strategies in cancer. Cell Mol Biol Lett 2025; 30:2. [PMID: 39757165 DOI: 10.1186/s11658-024-00682-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
Hypoxia-inducible factors (HIFs) are essential transcription factors that orchestrate cellular responses to oxygen deprivation. HIF-1α, as an unstable subunit of HIF-1, is usually hydroxylated by prolyl hydroxylase domain enzymes under normoxic conditions, leading to ubiquitination and proteasomal degradation, thereby keeping low levels. Instead of hypoxia, sometimes even in normoxia, HIF-1α translocates into the nucleus, dimerizes with HIF-1β to generate HIF-1, and then activates genes involved in adaptive responses such as angiogenesis, metabolic reprogramming, and cellular survival, which presents new challenges and insights into its role in cellular processes. Thus, the review delves into the mechanisms by which HIF-1 maintains its stability under normoxia including but not limited to giving insights into transcriptional, translational, as well as posttranslational regulation to underscore the pivotal role of HIF-1 in cellular adaptation and malignancy. Moreover, HIF-1 is extensively involved in cancer and cardiovascular diseases and potentially serves as a bridge between them. An overview of HIF-1-related drugs that are approved or in clinical trials is summarized, highlighting their potential capacity for targeting HIF-1 in cancer and cardiovascular toxicity related to cancer treatment. The review provides a comprehensive insight into HIF-1's regulatory mechanism and paves the way for future research and therapeutic development.
Collapse
Affiliation(s)
- Jiayi Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Mingxuan Yao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shiting Xia
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
4
|
Yao H, Tian J, Cheng S, Dou H, Zhu Y. The mechanism of hypoxia-inducible factor-1α enhancing the transcriptional activity of transferrin ferroportin 1 and regulating the Nrf2/HO-1 pathway in ferroptosis after cerebral ischemic injury. Neuroscience 2024; 559:26-38. [PMID: 39168172 DOI: 10.1016/j.neuroscience.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Cerebral ischemic/reperfusion (I/R) injury has high disability and morbidity. Hypoxia-inducible factor-1α (HIF-1α) may enhance the transcriptional activity of transferrin ferroportin 1 (FPN1) in regulating ferroptosis after cerebral ischemia injury (CII). In this study, cerebral I/R injury rat models were established and treated with pcDNA3.1-HIF-1α, pcDNA3.1-NC lentiviral plasmid, or ML385 (a specific Nrf2 inhibitor). Additionally, oxygen-glucose deprivation/reoxygenation (OGD/R) exposed PC12 cells were used as an in vitro model of cerebral ischemia and treated with pcDNA3.1-HIF-1α, si-FPN1, or ML385. The results elicited that cerebral I/R injury rats exhibited increased Longa scores, TUNEL and NeuN co-positive cells, Fe2+ concentration, ROS and HIF-1α levels, and MDA content, while reduced cell density and number, GSH content, and GPX4 protein level. Morphologically abnormal and disordered hippocampal neurons were also observed in CII rats. HIF-1α inhibited brain neuron ferroptosis and ameliorated I/R injury. HIF-1α alleviated OGD-induced PC12 cell ferroptosis. OGD/R decreased FPN1 protein level in PC12 cells, and HIF-1α enhanced FPN1 transcriptional activity. FPN1 knockdown reversed HIF-1α-mediated alleviation of OGD/R-induced ferroptosis. HIF-1α activated the Nrf2/HO-1 pathway by enhancing FPN1 expression and alleviating OGD/R-induced ferroptosis. Conjointly, HIF-1α enhanced the transcriptional activity of FPN1, activated the Nrf2/HO-1 pathway, and inhibited ferroptosis of brain neurons, thereby improving I/R injury in CII rats.
Collapse
Affiliation(s)
- Haiqian Yao
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Jianan Tian
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Shi Cheng
- Department of Orthopaedics, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Haitong Dou
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Yulan Zhu
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
5
|
Otsuka S, Dutta D, Wu CJ, Alam MS, Ashwell JD. Calcineurin is an adaptor required for assembly of the TCR signaling complex. Cell Rep 2024; 43:114568. [PMID: 39088318 PMCID: PMC11407306 DOI: 10.1016/j.celrep.2024.114568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024] Open
Abstract
The serine/threonine phosphatase calcineurin is a component of the T cell receptor (TCR) signalosome, where it promotes T cell activation by dephosphorylating LckS59. Using small interfering RNA (siRNA)-mediated knockdown and CRISPR-Cas9-targeted genetic disruption of the calcineurin A chain α and β isoforms, we find that calcineurin also functions as an adaptor in TCR-signaled human T cells. Unlike inhibition of its phosphatase activity, in the absence of calcineurin A, TCR signaling results in attenuated actin rearrangement, markedly reduced TCR-Lck microcluster formation and recruitment of the adaptor RhoH, and diminished phosphorylation of critical targets downstream of Lck such as TCRζ and ZAP-70. Reconstitution of deficient T cells with either calcineurin Aα or Aβ restores TCR microcluster formation and signaling, as does reconstitution with a phosphatase-inactive Aα chain. These results assign a non-enzymatic adaptor function to calcineurin in the formation and stabilization of a functional TCR signaling complex.
Collapse
Affiliation(s)
- Shizuka Otsuka
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Debjani Dutta
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan-Jin Wu
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Muhammad S Alam
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Roy S, Roy S, Halder S, Jana K, Ukil A. Leishmania exploits host cAMP/EPAC/calcineurin signaling to induce an IL-33-mediated anti-inflammatory environment for the establishment of infection. J Biol Chem 2024; 300:107366. [PMID: 38750790 PMCID: PMC11208913 DOI: 10.1016/j.jbc.2024.107366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 06/10/2024] Open
Abstract
Host anti-inflammatory responses are critical for the progression of visceral leishmaniasis, and the pleiotropic cytokine interleukin (IL)-33 was found to be upregulated in infection. Here, we documented that IL-33 induction is a consequence of elevated cAMP-mediated exchange protein activated by cAMP (EPAC)/calcineurin-dependent signaling and essential for the sustenance of infection. Leishmania donovani-infected macrophages showed upregulation of IL-33 and its neutralization resulted in decreased parasite survival and increased inflammatory responses. Infection-induced cAMP was involved in IL-33 production and of its downstream effectors PKA and EPAC, only the latter was responsible for elevated IL-33 level. EPAC initiated Rap-dependent phospholipase C activation, which triggered the release of intracellular calcium followed by calcium/calmodulin complex formation. Screening of calmodulin-dependent enzymes affirmed involvement of the phosphatase calcineurin in cAMP/EPAC/calcium/calmodulin signaling-induced IL-33 production and parasite survival. Activated calcineurin ensured nuclear localization of the transcription factors, nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha required for IL-33 transcription, and we further confirmed this by chromatin immunoprecipitation assay. Administering specific inhibitors of nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha in BALB/c mouse model of visceral leishmaniasis decreased liver and spleen parasite burden along with reduction in IL-33 level. Splenocyte supernatants of inhibitor-treated infected mice further documented an increase in tumor necrosis factor alpha and IL-12 level with simultaneous decrease of IL-10, thereby indicating an overall disease-escalating effect of IL-33. Thus, this study demonstrates that cAMP/EPAC/calcineurin signaling is crucial for the activation of IL-33 and in effect creates anti-inflammatory responses, essential for infection.
Collapse
Affiliation(s)
- Souravi Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Shalini Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Satyajit Halder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata, India.
| |
Collapse
|
7
|
Mukhopadhyay A, Tsukasaki Y, Chan WC, Le JP, Kwok ML, Zhou J, Natarajan V, Mostafazadeh N, Maienschein-Cline M, Papautsky I, Tiruppathi C, Peng Z, Rehman J, Ganesh B, Komarova Y, Malik AB. trans-Endothelial neutrophil migration activates bactericidal function via Piezo1 mechanosensing. Immunity 2024; 57:52-67.e10. [PMID: 38091995 PMCID: PMC10872880 DOI: 10.1016/j.immuni.2023.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The regulation of polymorphonuclear leukocyte (PMN) function by mechanical forces encountered during their migration across restrictive endothelial cell junctions is not well understood. Using genetic, imaging, microfluidic, and in vivo approaches, we demonstrated that the mechanosensor Piezo1 in PMN plasmalemma induced spike-like Ca2+ signals during trans-endothelial migration. Mechanosensing increased the bactericidal function of PMN entering tissue. Mice in which Piezo1 in PMNs was genetically deleted were defective in clearing bacteria, and their lungs were predisposed to severe infection. Adoptive transfer of Piezo1-activated PMNs into the lungs of Pseudomonas aeruginosa-infected mice or exposing PMNs to defined mechanical forces in microfluidic systems improved bacterial clearance phenotype of PMNs. Piezo1 transduced the mechanical signals activated during transmigration to upregulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4, crucial for the increased PMN bactericidal activity. Thus, Piezo1 mechanosensing of increased PMN tension, while traversing the narrow endothelial adherens junctions, is a central mechanism activating the host-defense function of transmigrating PMNs.
Collapse
Affiliation(s)
- Amitabha Mukhopadhyay
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jonathan P Le
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Man Long Kwok
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Jian Zhou
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Viswanathan Natarajan
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA; Department of Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Nima Mostafazadeh
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ian Papautsky
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Zhangli Peng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois, Chicago, IL 60612, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Balaji Ganesh
- Flow Cytometry Core, Research Resources Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia Komarova
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
8
|
Powell JT, Kayesh R, Ballesteros-Perez A, Alam K, Niyonshuti P, Soderblom EJ, Ding K, Xu C, Yue W. Assessing Trans-Inhibition of OATP1B1 and OATP1B3 by Calcineurin and/or PPIase Inhibitors and Global Identification of OATP1B1/3-Associated Proteins. Pharmaceutics 2023; 16:63. [PMID: 38258074 PMCID: PMC10818623 DOI: 10.3390/pharmaceutics16010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Organic anion transporting polypeptide (OATP) 1B1 and OATP1B3 are key determinants of drug-drug interactions (DDIs). Various drugs including the calcineurin inhibitor (CNI) cyclosporine A (CsA) exert preincubation-induced trans-inhibitory effects upon OATP1B1 and/or OATP1B3 (abbreviated as OATP1B1/3) by unknown mechanism(s). OATP1B1/3 are phosphoproteins; calcineurin, which dephosphorylates and regulates numerous phosphoproteins, has not previously been investigated in the context of preincubation-induced trans-inhibition of OATP1B1/3. Herein, we compare the trans-inhibitory effects exerted on OATP1B1 and OATP1B3 by CsA, the non-analogous CNI tacrolimus, and the non-CNI CsA analogue SCY-635 in transporter-overexpressing human embryonic kidney (HEK) 293 stable cell lines. Preincubation (10-60 min) with tacrolimus (1-10 µM) rapidly and significantly reduces OATP1B1- and OATP1B3-mediated transport up to 0.18 ± 0.03- and 0.20 ± 0.02-fold compared to the control, respectively. Both CsA and SCY-635 can trans-inhibit OATP1B1, with the inhibitory effects progressively increasing over a 60 min preincubation time. At each equivalent preincubation time, CsA has greater trans-inhibitory effects toward OATP1B1 than SCY-635. Preincubation with SCY-635 for 60 min yielded IC50 of 2.2 ± 1.4 µM against OATP1B1, which is ~18 fold greater than that of CsA (0.12 ± 0.04 µM). Furthermore, a proteomics-based screening for protein interactors was used to examine possible proteins and processes contributing to OATP1B1/3 regulation and preincubation-induced inhibition by CNIs and other drugs. A total of 861 and 357 proteins were identified as specifically associated with OATP1B1 and OATP1B3, respectively, including various protein kinases, ubiquitin-related enzymes, the tacrolimus (FK506)-binding proteins FKBP5 and FKBP8, and several known regulatory targets of calcineurin. The current study reports several novel findings that expand our understanding of impaired OATP1B1/3 function; these include preincubation-induced trans-inhibition of OATP1B1/3 by the CNI tacrolimus, greater preincubation-induced inhibition by CsA compared to its non-CNI analogue SCY-635, and association of OATP1B1/3 with various proteins relevant to established and candidate OATP1B1/3 regulatory processes.
Collapse
Affiliation(s)
- John T. Powell
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Ruhul Kayesh
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Alexandra Ballesteros-Perez
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Khondoker Alam
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Pascaline Niyonshuti
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| | - Erik J. Soderblom
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC 27708, USA
| | - Kai Ding
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Chao Xu
- Department of Biostatistics & Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (K.D.); (C.X.)
| | - Wei Yue
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (J.T.P.)
| |
Collapse
|
9
|
Lee Q, Chan WC, Qu X, Sun Y, Abdelkarim H, Le J, Saqib U, Sun MY, Kruse K, Banerjee A, Hitchinson B, Geyer M, Huang F, Guaiquil V, Mutso AA, Sanders M, Rosenblatt MI, Maienschein-Cline M, Lawrence MS, Gaponenko V, Malik AB, Komarova YA. End binding-3 inhibitor activates regenerative program in age-related macular degeneration. Cell Rep Med 2023; 4:101223. [PMID: 37794584 PMCID: PMC10591057 DOI: 10.1016/j.xcrm.2023.101223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 07/19/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023]
Abstract
Wet age-related macular degeneration (AMD), characterized by leaky neovessels emanating from the choroid, is a main cause of blindness. As current treatments for wet AMD require regular intravitreal injections of anti-vascular endothelial growth factor (VEGF) biologics, there is a need for the development of less invasive treatments. Here, we designed an allosteric inhibitor of end binding-3 (EB3) protein, termed EBIN, which reduces the effects of environmental stresses on endothelial cells by limiting pathological calcium signaling. Delivery of EBIN via eye drops in mouse and non-human primate (NHP) models of wet AMD prevents both neovascular leakage and choroidal neovascularization. EBIN reverses the epigenetic changes induced by environmental stresses, allowing an activation of a regenerative program within metabolic-active endothelial cells comprising choroidal neovascularization (CNV) lesions. These results suggest the therapeutic potential of EBIN in preventing the degenerative processes underlying wet AMD.
Collapse
Affiliation(s)
- Quinn Lee
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Wan Ching Chan
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Xinyan Qu
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Ying Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Jonathan Le
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Uzma Saqib
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Mitchell Y Sun
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Kevin Kruse
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Avik Banerjee
- Department of Chemistry, The University of Illinois, Chicago, IL 60612, USA
| | - Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Melissa Geyer
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Fei Huang
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Victor Guaiquil
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Amelia A Mutso
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | - Mark I Rosenblatt
- Department of Ophthalmology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Asrar B Malik
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Yulia A Komarova
- Department of Pharmacology and The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612, USA.
| |
Collapse
|
10
|
Wang Y, Liu X, Huang W, Liang J, Chen Y. The intricate interplay between HIFs, ROS, and the ubiquitin system in the tumor hypoxic microenvironment. Pharmacol Ther 2022; 240:108303. [PMID: 36328089 DOI: 10.1016/j.pharmthera.2022.108303] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/16/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alterations in protein ubiquitination and hypoxia-inducible factor (HIF) signaling both contribute to tumorigenesis and tumor progression. Ubiquitination is a dynamic process that is coordinately regulated by E3 ligases and deubiquitinases (DUBs), which have emerged as attractive therapeutic targets. HIF expression and transcriptional activity are usually increased in tumors, leading to poor clinical outcomes. Reactive oxygen species (ROS) are upregulated in tumors and have multiple effects on HIF signaling and the ubiquitin system. A growing body of evidence has shown that multiple E3 ligases and UBDs function synergistically to control the expression and activity of HIF, thereby allowing cancer cells to cope with the hypoxic microenvironment. Conversely, several E3 ligases and DUBs are regulated by hypoxia and/or HIF signaling. Hypoxia also induces ROS production, which in turn modulates the stability or activity of HIF, E3 ligases, and DUBs. Understanding the complex networks between E3 ligase, DUBs, ROS, and HIF will provide insights into the fundamental mechanism of the cellular response to hypoxia and help identify novel molecular targets for cancer treatment. We review the current knowledge on the comprehensive relationship between E3 ligase, DUBs, ROS, and HIF signaling, with a particular focus on the use of E3 ligase or DUB inhibitors in cancer.
Collapse
Affiliation(s)
- Yijie Wang
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China
| | - Xiong Liu
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Weixiao Huang
- School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Junjie Liang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China.
| | - Yan Chen
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China; School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
11
|
Li D, Wang T, Lai J, Zeng D, Chen W, Zhang X, Zhu X, Zhang G, Hu Z. Silencing TRPM2 enhanced erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Cytotechnology 2022; 74:559-577. [PMID: 36238268 PMCID: PMC9525503 DOI: 10.1007/s10616-022-00545-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/25/2022] [Indexed: 11/03/2022] Open
Abstract
Ferroptosis is a regulated form of cell death driven by small molecules or conditions that induce lipid-based reactive oxygen species (ROS) accumulation. Cation channel transient receptor potential melastatin-2 (TRPM2) is crucial for cancer cell survival. Our bioinformatic analysis revealed that TRPM2 is associated with cellular responses to chemical stimulus and oxidative stress, implying the potential role of TRPM2 in ferroptosis. Gastric cancer cells were treated with the ferroptosis-inducer, Erastin and RSL3. siRNA transfection was used to silence TRPM2. The levels of GSH, Fe2+, ROS and lipid peroxidation, and the activity of GPx activity were evaluated by flow cytometry and spectrophotometer. The effect of TRPM2 on ubiquitination of HIF-1α and Nrf2 were evaluated by co-immunoprecipitation. Erastin and RSL3 induced the up-regulation of TRPM2 in gastric cancer cell lines, especially in SGC7901 and MGC803. These two cells also showed stronger resistance to Erastin and RSL3 than the other cell lines. TRPM2 knockdown reduced the concentration of GSH and GPx activity, but enhanced the concentration of Fe2+, ROS and lipid peroxidation, which are significant indicators of ferroptosis. Importantly, silencing TRPM2 enhanced the inhibitory effects of Erastin and RSL3 on gastric cancer cell viability, migration, and invasion. TRPM2 stabilized and finally elevated the abundance of HIF-1α and Nrf2 in SGC7901 and MGC803 cells upon Erastin and RSL3. Activation of HIF-1α impaired Erastin- and RSL3-induced ferroptosis after TRPM2 knockdown. Collectively, silencing TRPM2 enhanced Erastin- and RSL3-induced ferroptosis in gastric cancer cells through destabilizing HIF-1α and Nrf2 proteins. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00545-z.
Collapse
Affiliation(s)
- Dingyun Li
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Ting Wang
- Department of Physical Diagnosis, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Deqiang Zeng
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Weijuan Chen
- Clinical Laboratory, Yue Bei People’s Hospital, No. 133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaochong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Xiaofeng Zhu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Guoxiong Zhang
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| | - Zhiwei Hu
- Department of Gastrointestinal Surgery, Yue Bei People’s Hospital, No.133 Huimin South Road, Wujiang District, Shaoguan, 512026 Guangdong China
| |
Collapse
|
12
|
Li X, Bu F, Ma S, Cananzi F, Zhao Y, Xiao M, Min L, Luo C. The Janus-faced role of TRPM2-S in retroperitoneal liposarcoma via increasing ROS levels. Cell Commun Signal 2022; 20:128. [PMID: 36008839 PMCID: PMC9404563 DOI: 10.1186/s12964-022-00873-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Retroperitoneal liposarcoma (RPLS) is a specific soft tissue sarcoma with a high recurrence rate. The short isoform of transient receptor potential cation channel subfamily M member 2 (TRPM2-S) plays an important role in the regulation of reactive oxygen species (ROS). However, the association between TRPM2-S and RPLS and its underlying mechanisms remains unclear. METHODS The expression of both TRPM2-S and TRPM2-L in RPLS tissues was verified by kimmunohistochemistry (IHC). The regulation on Ca2+ influx by TRPM2-S was evaluated by Fluo-4 AM staining. The effect of TRPM2-S on cell proliferation and apoptosis was tested by 5-Ethynyl-2'-deoxyuridine (EdU) staining and Flow cytometry respectively. The level of cellular ROS was assessed by the DCFH-DA probe. Different concentrations of H2O2 were used to provide oxidative stress on RPLS cells. The underlying mechanisms were further explored by Western blotting. RESULTS The IHC assays showed that TRPM2-S, but not TRPM2-L, was prognostic in RPLS. Low TRPM2-S level was associated with poor disease-free survival (DFS). Calcium influx signal intensity was significantly decreased under TRPM2-S overexpression, which resulted in a decrease in the levels of FOXO3a and PTEN. Correspondingly, the levels of pERK, pAKT, pP65, pGSK-3β, Bcl-2, and β-catenin were upregulated, and cellular ROS was gently increased under TRPM2-S overexpression. Moreover, TRPM2-S slightly promoted cell proliferation and inhibited apoptosis of RPLS cell lines under normoxia, but largely increased apoptosis rates under oxidative stress. The cleaved caspase3 was significantly upregulated by TRPM2-S overexpression under oxidative stress. N-Acetyl-L-cysteine (NAC), a small molecule antioxidant, could largely rescue RPLS cells from the apoptosis induced by H2O2. CONCLUSION TRPM2-S exerts Janus-faced effects in RPLS by increasing the ROS levels via inhibition on FOXO3a, which promotes cell proliferation under normoxia but induces apoptosis under oxidative stress. Video abstract.
Collapse
Affiliation(s)
- Xiangji Li
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Peking University Eighth School of Clinical Medicine, Beijing, 102206, People's Republic of China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Fanqin Bu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Shixiang Ma
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Peking University Eighth School of Clinical Medicine, Beijing, 102206, People's Republic of China
| | - Ferdinando Cananzi
- Department of Biomedical Sciences, Humanitas University, 20089, Milan, Italy
| | - Yu Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Mengmeng Xiao
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Peking University Eighth School of Clinical Medicine, Beijing, 102206, People's Republic of China.
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China.
| | - Chenghua Luo
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Peking University Eighth School of Clinical Medicine, Beijing, 102206, People's Republic of China.
| |
Collapse
|
13
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
14
|
Singh V, Singh R, Kushwaha R, Verma SP, Tripathi AK, Mahdi AA. The Molecular Role of HIF1α Is Elucidated in Chronic Myeloid Leukemia. Front Oncol 2022; 12:912942. [PMID: 35847841 PMCID: PMC9279726 DOI: 10.3389/fonc.2022.912942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic myeloid leukemia (CML) is potentially fatal blood cancer, but there is an unmet need to discover novel molecular biomarkers. The hypothesis of this study aimed to elucidate the relationship of HIF1α with the redox system, Krebs cycles, notch1, and other regulatory proteins to better understand the pathophysiology and clinical relevance in chronic myeloid leukemia (CML) patients, as the molecular mechanism of this axis is still not clear. This study included CML patient samples (n = 60; 60: blood; 10: bone marrow tissues) and compared them with healthy controls (n = 20; blood). Clinical diagnosis confirmed on bone marrow aspiration, marrow trephine biopsy, and BCR/ABL1 translocation. Cases were subclassified into chronic, accelerated, and blast crises as per WHO guidelines. Molecular experiments included redox parameters, DNA fragmentation, Krebs cycle metabolites, and gene expression by RT-PCR/Western blot/LC-MS, PPI (STRING), Pearson correlation, and ROC curve analysis. Here, our findings show that p210/p190BCR/ABL1 translocation is common in all blast crisis phases of CML. Redox factor/Krebs oncometabolite concentrations were high, leading to upregulation and stabilization of HIF1α. HIF1α leads to the pathogenesis in CML cells by upregulating their downstream genes (Notch 2/4/Ikaros/SIRT1/Foxo-3a/p53, etc.). Whereas, downregulated ubiquitin proteasomal and apoptotic factors in CML pateints, can trigger degradation of HIF1α through proline hydroxylation. However, HIF1α showed a negative corelation with the notch1 pathway. Notch1 plays a tumor-suppressive role in CML and might have the potential to be used as a diagnostic marker along with other factors in CML patients. The outcome also revealed that oxidant treatment could not be effective in augmentation with conventional therapy because CML cells can enhance the levels of antioxidants for their survival. HIF1α might be a novel therapeutic target other than BCR/ABL1 translocation.
Collapse
Affiliation(s)
- Vivek Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, India
| | - Ranjana Singh
- Department of Biochemistry, King George’s Medical University, Lucknow, India
- *Correspondence: Ranjana Singh, ;
| | - Rashmi Kushwaha
- Department of Pathology, King George’s Medical University, Lucknow, India
| | | | - Anil Kumar Tripathi
- Department of Clinical Hematology, King George’s Medical University, Lucknow, India
| | - Abbas Ali Mahdi
- Department of Biochemistry, King George’s Medical University, Lucknow, India
| |
Collapse
|
15
|
Characterization of the Impacts of Living at High Altitude in Taif: Oxidative Stress Biomarker Alterations and Immunohistochemical Changes. Curr Issues Mol Biol 2022; 44:1610-1625. [PMID: 35723368 PMCID: PMC9164078 DOI: 10.3390/cimb44040110] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
At high elevations, the human body experiences a number of pathological, physiological, and biochemical changes, all of which have adverse impacts on human health and organ vitality. This study aimed to investigate the alterations in the liver and kidney biomarkers, oxidative stress markers, gene expression, and cellular histology of rats maintained at high altitudes and normal sea level. A total of twenty male Wistar rats at 2 months of age were randomly assigned to two groups. The rats in group A were maintained at normal sea level in Jeddah, whereas rats in group B were maintained in an area in Taif 2600 m above sea level. After 2 months of housing, orbital blood samples were collected for the analysis of significant biochemical indicators of oxidative stress biomarkers of the liver and kidneys. Liver and kidney tissues from both groups were taken to examine the hepatorenal changes occurring at the biochemical, histological, immunohistochemical, and genetic levels. The results revealed substantial increases in the serum levels of liver and kidney biomarkers (GPT, GOT, urea, and creatinine) and decreases in the serum levels of antioxidant biomarkers (SOD, catalase, GSH, and NO). In parallel, the levels of the malondialdehyde (MDA) tissue damage marker and inflammatory cytokines (IL-1β, TNF-α, and IFN-γ) were increased in the high-altitude group compared to the normal sea level group. In addition, there were significant alterations in the oxidative and inflammatory status of rats that lived at high altitude, with considerable upregulation in the expression of hepatic VEGF, type 1 collagen, Cox-2, TNF-α, and iNOS as well as renal EPASI, CMYC, HIF-α, and EGLN-2 genes in the high-altitude group compared with controls housed at normal sea level. In conclusion, living at high altitude induces hepatorenal damage and biochemical and molecular alterations, all of which may serve as critical factors that must be taken into account for organisms living at high altitudes.
Collapse
|
16
|
Peng SS, Li Y, Chen Q, Hu Q, He Y, Che L, Jiang PP. Intestinal and Mucosal Microbiome Response to Oral Challenge of Enterotoxigenic Escherichia coli in Weaned Pigs. Pathogens 2022; 11:pathogens11020160. [PMID: 35215105 PMCID: PMC8879466 DOI: 10.3390/pathogens11020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/10/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is closely associated with diarrhoea in children in resource-limited countries. This study aims to investigate the change of the mucosal microbiome and protein expression in the ileum induced by E. coli K88 (ETEC) using pigs as a model. Seven weaned male pigs were orally given ETEC (1 × 109 CFU, n = 7), and the other seven received saline (CON, n = 7). Ileal tissues were obtained 48 hours after the ETEC challenge for both proteomic and mucosal microbiome analyses. Nine proteins were found with altered abundance between the two groups, including a decrease in FABP1 and FABP6, involved in bile acid circulation. The TLR-9 mediated pathway was also affected showing increased transcription of genes SIGIRR and MyD88. Correlations between the ileal proteins and mucosal bacterial taxa found included a positive correlation between Lactobacilllus and PPP3CA (r = 0.9, p < 0.001) and a negative correlation between Prevotella with CTNND1 (r = −0.7, p < 0.01). In conclusion, ETEC infection caused inflammation and impaired the circulation of bile acids and the mucosal microbiome may affect the expression of intestinal proteins. Further studies are needed to explain the exact roles of these affected processes in the pathogenesis of ETEC-triggered diarrhoea.
Collapse
Affiliation(s)
- Shan-Shan Peng
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
| | - Yingjie Li
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Qiuhong Chen
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Qi Hu
- The Neomics Institute, Shenzhen 518122, China;
| | - Ying He
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
| | - Lianqiang Che
- Key Laboratory for Animal Disease Resistance and Nutrition of the Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.L.); (Q.C.); (Y.H.)
- Correspondence: (L.C.); (P.-P.J.)
| | - Ping-Ping Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: (L.C.); (P.-P.J.)
| |
Collapse
|
17
|
Wang M, Liu Y, Liang Y, Naruse K, Takahashi K. Systematic Understanding of Pathophysiological Mechanisms of Oxidative Stress-Related Conditions-Diabetes Mellitus, Cardiovascular Diseases, and Ischemia-Reperfusion Injury. Front Cardiovasc Med 2021; 8:649785. [PMID: 33928135 PMCID: PMC8076504 DOI: 10.3389/fcvm.2021.649785] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) plays a role in intracellular signal transduction under physiological conditions while also playing an essential role in diseases such as hypertension, ischemic heart disease, and diabetes, as well as in the process of aging. The influence of ROS has some influence on the frequent occurrence of cardiovascular diseases (CVD) in diabetic patients. In this review, we considered the pathophysiological relationship between diabetes and CVD from the perspective of ROS. In addition, considering organ damage due to ROS elevation during ischemia-reperfusion, we discussed heart and lung injuries. Furthermore, we have focused on the transient receptor potential (TRP) channels and L-type calcium channels as molecular targets for ROS in ROS-induced tissue damages and have discussed about the pathophysiological mechanism of the injury.
Collapse
Affiliation(s)
| | | | | | | | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
18
|
Harb IA, Ashour H, Sabry D, El-Yasergy DF, Hamza WM, Mostafa A. Nicorandil prevents the nephrotoxic effect of cyclosporine-A in albino rats through modulation of HIF-1α/VEGF/eNOS signaling. Can J Physiol Pharmacol 2021; 99:411-417. [PMID: 32822562 DOI: 10.1139/cjpp-2020-0012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Despite that cyclosporine-A (CsA) is a widely used immunosuppressive drug, its nephrotoxic effect limits its long-term administration. Herein we tried to investigate its renal effect on endothelial dysfunction targeting the hypoxia-inducible factor (HIF-1α) / vascular endothelial growth factor (VEGF) / endothelial nitric oxide synthase (eNOS) pathway and the possible modulation by nicorandil. Eight groups of adult male Wistar rats were included: (1) control; (2) vehicle group (received oil); (3) glibenclamide 5 mg·kg-1·day-1 administered orally; (4) nicorandil 10 mg·kg-1·day-1 administered orally; (5) CsA 25 mg·kg-1·day-1 administered orally; (6) combined administration of CsA and nicorandil; (7) glibenclamide was added to CsA; and (8) both CsA and nicorandil were combined with glibenclamide. The treatment continued for six weeks. Combined nicorandil with CsA improved renal function deterioration initiated by CsA. CsA decreased the renal expression levels (P < 0.001) of HIF-1α, eNOS, and VEGF, inducing endothelial dysfunction and triggering inflammation, and upregulated the profibrotic marker transforming growth factor (TGF-β). Nicorandil fixed the disturbed HIF-1α/VEGF/eNOS signaling. Nicorandil corrected the renal functions, confirmed by the improved histological glomerular tuft retraction that was obvious in the CsA group, without significant influence by glibenclamide. Proper protection from CsA-induced nephrotoxicity was achieved by nicorandil. Nicorandil reversed the disturbed HIF-1α/VEGF/eNOS pathway created by CsA.
Collapse
Affiliation(s)
- Inas A Harb
- Department of Pharmacology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hend Ashour
- Department of Physiology, Faculty of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
- Department of Physiology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina Fawzy El-Yasergy
- Department of Pathology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Wael Mostafa Hamza
- Department of Pathology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abeer Mostafa
- Department of Medical Biochemistry and Molecular Biology, Kasr Alainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Wang J, Chen X, Hu H, Yao M, Song Y, Yang A, Xu X, Zhang N, Gao J, Liu B. PCAT-1 facilitates breast cancer progression via binding to RACK1 and enhancing oxygen-independent stability of HIF-1α. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:310-324. [PMID: 33850635 PMCID: PMC8020346 DOI: 10.1016/j.omtn.2021.02.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/25/2021] [Indexed: 12/20/2022]
Abstract
Hypoxia induces a series of cellular adaptive responses that enable promotion of inflammation and cancer development. Hypoxia-inducible factor-1α (HIF-1α) is involved in the hypoxia response and cancer promotion, and it accumulates in hypoxia and is degraded under normoxic conditions. Here we identify prostate cancer associated transcript-1 (PCAT-1) as a hypoxia-inducible long non-coding RNA (lncRNA) that regulates HIF-1α stability, crucial for cancer progression. Extensive analyses of clinical data indicate that PCAT-1 is elevated in breast cancer patients and is associated with pathological grade, tumor size, and poor clinical outcomes. Through gain- and loss-of-function experiments, we find that PCAT-1 promotes hypoxia-associated breast cancer progression including growth, migration, invasion, colony formation, and metabolic regulation. Mechanistically, PCAT-1 directly interacts with the receptor of activated protein C kinase-1 (RACK1) protein and prevents RACK1 from binding to HIF-1α, thus protecting HIF-1α from RACK1-induced oxygen-independent degradation. These findings provide new insight into lncRNA-mediated mechanisms for HIF-1α stability and suggest a novel role of PCAT-1 as a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Jianlong Wang
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xuyi Chen
- Department of Neurosurgery, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin 300162, China
| | - Haijuan Hu
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengting Yao
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin 300071, China
| | - Yanbiao Song
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Aimin Yang
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xiuhua Xu
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Ning Zhang
- Department of Biomedical Engineering, Tianjin Key Lab of BME Measurement, Tianjin University, Tianjin 300072, China
| | - Jianzhao Gao
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin 300071, China
| | - Bin Liu
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
20
|
Samec M, Liskova A, Koklesova L, Mersakova S, Strnadel J, Kajo K, Pec M, Zhai K, Smejkal K, Mirzaei S, Hushmandi K, Ashrafizadeh M, Saso L, Brockmueller A, Shakibaei M, Büsselberg D, Kubatka P. Flavonoids Targeting HIF-1: Implications on Cancer Metabolism. Cancers (Basel) 2021; 13:E130. [PMID: 33401572 PMCID: PMC7794792 DOI: 10.3390/cancers13010130] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia is described as an oxygen deprivation in malignant tissue. The hypoxic condition is a consequence of an imbalance between rapidly proliferating cells and a vascularization that leads to lower oxygen levels in tumors. Hypoxia-inducible factor 1 (HIF-1) is an essential transcription factor contributing to the regulation of hypoxia-associated genes. Some of these genes modulate molecular cascades associated with the Warburg effect and its accompanying pathways and, therefore, represent promising targets for cancer treatment. Current progress in the development of therapeutic approaches brings several promising inhibitors of HIF-1. Flavonoids, widely occurring in various plants, exert a broad spectrum of beneficial effects on human health, and are potentially powerful therapeutic tools against cancer. Recent evidences identified numerous natural flavonoids and their derivatives as inhibitors of HIF-1, associated with the regulation of critical glycolytic components in cancer cells, including pyruvate kinase M2(PKM2), lactate dehydrogenase (LDHA), glucose transporters (GLUTs), hexokinase II (HKII), phosphofructokinase-1 (PFK-1), and pyruvate dehydrogenase kinase (PDK). Here, we discuss the results of most recent studies evaluating the impact of flavonoids on HIF-1 accompanied by the regulation of critical enzymes contributing to the Warburg phenotype. Besides, flavonoid effects on glucose metabolism via regulation of HIF-1 activity represent a promising avenue in cancer-related research. At the same time, only more-in depth investigations can further elucidate the mechanistic and clinical connections between HIF-1 and cancer metabolism.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (M.S.); (A.L.); (L.K.)
| | - Sandra Mersakova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 03601 Martin, Slovakia; (S.M.); (J.S.)
| | - Jan Strnadel
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 03601 Martin, Slovakia; (S.M.); (J.S.)
| | - Karol Kajo
- Department of Pathology, St. Elizabeth Cancer Institute Hospital, 81250 Bratislava, Slovakia;
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Karel Smejkal
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého třída 1946/1, 61200 Brno, Czech Republic;
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, 1477893855 Tehran, Iran;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, 1419963114 Tehran, Iran;
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, 00185 Rome, Italy;
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany; (A.B.); (M.S.)
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| |
Collapse
|
21
|
Zhang S, Zhang M, Chen J, Zhao J, Su J, Zhang X. Ginsenoside Compound K Regulates HIF-1α-Mediated Glycolysis Through Bclaf1 to Inhibit the Proliferation of Human Liver Cancer Cells. Front Pharmacol 2020; 11:583334. [PMID: 33363466 PMCID: PMC7753211 DOI: 10.3389/fphar.2020.583334] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
This study aimed to demonstrate that ginsenoside compound K (20 (S)-ginsenoside CK; CK) downregulates Bcl-2-associated transcription factor 1 (Bclaf1), which inhibits the hypoxia-inducible factor-1α (HIF-1α)-mediated glycolysis pathway to inhibit the proliferation of liver cancer cells. Treatment of hepatoma cells (Bel-7404 and Huh7) under hypoxic conditions with different concentrations of CK showed that CK inhibited the proliferation of hepatoma cells in a time- and concentration-dependent manner; furthermore, the ability of the cells to form colonies was reduced, and cell growth was blocked in the G0/G1 phase. CK promoted the degradation of HIF-1α ubiquitination in liver cancer cells by regulating the expression of HIF-1α and related ubiquitination proteins; moreover, it reduced the activity of key enzymes involved in glycolysis, the pressure of cellular glycolysis, and the rate of real-time ATP production, thereby inhibiting the glycolysis pathway. It also decreased the expression of Bclaf1 in hypoxic liver cancer cells and thus reduced the ability of Bclaf1 to bind to HIF-1α. CK treatment of Bel-7404 and Huh7 cells with CRISPR/Cas9-engineered knock out of Bclaf1 gene under hypoxic conditions further suppressed the expression of HIF-1α, promoted HIF-1α ubiquitination, and inhibited the glycolysis pathway. In a rat model of primary liver cancer induced by diethylnitrosamine, positron emission tomography and computed tomography scans showed that after CK administration, tumor tissue volumes were reduced and glucose uptake capacity decreased. Increased Bclaf1 and HIF-1α expression promoted the ubiquitination of HIF-1α and inhibited the glycolysis pathway, thereby inhibiting the proliferation of liver cancer cells. In summary, this study confirmed by in vitro and in vivo experiments that in hypoxic liver cancer cells CK downregulates the expression of Bclaf1, inhibits the HIF-1α-mediated glycolysis pathway, and inhibits cell proliferation, suggesting that the CK-mediated effects on Bclaf1 may represent a novel therapeutic approach for the treatment of liver cancer patients.
Collapse
Affiliation(s)
- Silin Zhang
- College of Medicine, Yanbian University, Yanji, China
| | | | - Jiaxin Chen
- College of Medicine, Yanbian University, Yanji, China
| | - Jiaqi Zhao
- College of Medicine, Yanbian University, Yanji, China
| | - Jielin Su
- College of Medicine, Yanbian University, Yanji, China
| | - Xuewu Zhang
- College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
22
|
Wagner N, Wagner KD. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020; 9:cells9051133. [PMID: 32375405 PMCID: PMC7291220 DOI: 10.3390/cells9051133] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family. Three different isoforms, PPAR alpha, PPAR beta/delta and PPAR gamma have been identified. They all form heterodimers with retinoic X receptors to activate or repress downstream target genes dependent on the presence/absence of ligands and coactivators or corepressors. PPARs differ in their tissue expression profile, ligands and specific agonists and antagonists. PPARs attract attention as potential therapeutic targets for a variety of diseases. PPAR alpha and gamma agonists are in clinical use for the treatment of dyslipidemias and diabetes. For both receptors, several clinical trials as potential therapeutic targets for cancer are ongoing. In contrast, PPAR beta/delta has been suggested as a therapeutic target for metabolic syndrome. However, potential risks in the settings of cancer are less clear. A variety of studies have investigated PPAR beta/delta expression or activation/inhibition in different cancer cell models in vitro, but the relevance for cancer growth in vivo is less well documented and controversial. In this review, we summarize critically the knowledge of PPAR beta/delta functions for the different hallmarks of cancer biological capabilities, which interplay to determine cancer growth.
Collapse
|
23
|
Taheem DK, Jell G, Gentleman E. Hypoxia Inducible Factor-1α in Osteochondral Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:105-115. [PMID: 31774026 PMCID: PMC7166133 DOI: 10.1089/ten.teb.2019.0283] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022]
Abstract
Damage to osteochondral (OC) tissues can lead to pain, loss of motility, and progress to osteoarthritis. Tissue engineering approaches offer the possibility of replacing damaged tissues and restoring joint function; however, replicating the spatial and functional heterogeneity of native OC tissue remains a pressing challenge. Chondrocytes in healthy cartilage exist in relatively low-oxygen conditions, while osteoblasts in the underlying bone experience higher oxygen pressures. Such oxygen gradients also exist in the limb bud, where they influence OC tissue development. The cellular response to these spatial variations in oxygen pressure, which is mediated by the hypoxia inducible factor (HIF) pathway, plays a central role in regulating osteo- and chondrogenesis by directing progenitor cell differentiation and promoting and maintaining appropriate extracellular matrix production. Understanding the role of the HIF pathway in OC tissue development may enable new approaches to engineer OC tissue. In this review, we discuss strategies to spatially and temporarily regulate the HIF pathway in progenitor cells to create functional OC tissue for regenerative therapies. Impact statement Strategies to engineer osteochondral (OC) tissue are limited by the complex and varying microenvironmental conditions in native bone and cartilage. Indeed, native cartilage experiences low-oxygen conditions, while the underlying bone is relatively normoxic. The cellular response to these low-oxygen conditions, which is mediated through the hypoxia inducible factor (HIF) pathway, is known to promote and maintain the chondrocyte phenotype. By using tissue engineering scaffolds to spatially and temporally harness the HIF pathway, it may be possible to improve OC tissue engineering strategies for the regeneration of damaged cartilage and its underlying subchondral bone.
Collapse
Affiliation(s)
- Dheraj K. Taheem
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Gavin Jell
- Division of Surgery and Interventional Sciences, University College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
24
|
Suppression of TRPM7 Inhibited Hypoxia-Induced Migration and Invasion of Androgen-Independent Prostate Cancer Cells by Enhancing RACK1-Mediated Degradation of HIF-1 α. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6724810. [PMID: 32215176 PMCID: PMC7079255 DOI: 10.1155/2020/6724810] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/22/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Transient receptor potential melastatin subfamily member 7 (TRPM7) was essential in the growth and metastatic ability of prostate cancer cells. However, the effects and the relevant molecular mechanisms of TRPM7 on metastasis of prostate cancer under hypoxic atmosphere remain unclear. This study investigated the role of TRPM7 in the metastatic ability of androgen-independent prostate cancer cells under hypoxia. First, data mining was carried out to disclose the relationship between the TRPM7 gene level and the survival of prostate cancer patients. Specific siRNAs were used to knockdown target genes. Western blotting and qPCR were employed to determine protein and gene expression, respectively. The gene transcription activity was evaluated by luciferase activity assay of promoter gene. The protein interaction was determined by coimmunoprecipitation. Wound healing and transwell assays were employed to evaluated cell migration and invasion, respectively. Open access database results showed that high expression of TRPM7 was closely related to the poor survival of prostate cancer patients. Hypoxia simultaneously increased TRPM7 expression and induced HIF-1α accumulation in androgen-independent prostate cancer cells. Knockdown of TRPM7 significantly promoted HIF-1α degradation through the proteasome and inhibited EMT changes in androgen-independent prostate cancer cells under hypoxic condition. Moreover, TRPM7 knockdown increased the phosphorylation of RACK1 and strengthened the interaction between RACK1 and HIF-1α but attenuated the binding of HSP90 to HIF-1α. Whereas knockdown of RACK1 increased the binding of HSP90 to HIF-1α. Furthermore, both TRPM7 and HIF-1α knockdown significantly suppressed hypoxia-induced Annexin A1 protein expression, and suppression of HIF-1α/Annexin A1 signaling significantly inhibited hypoxia-induced cell migration and invasion of androgen-independent prostate cancer cells. Our findings demonstrate that TRPM7 knockdown promotes HIF-1α degradation via an oxygen-independent mechanism involving increased binding of RAKC1 to HIF-1α, and TRPM7-HIF-1α-Annexin A1 signaling axis plays a crucial role in the EMT, cell migration, and invasion of androgen-independent prostate cancer cells under hypoxic conditions.
Collapse
|
25
|
|
26
|
Fitzpatrick SF. Immunometabolism and Sepsis: A Role for HIF? Front Mol Biosci 2019; 6:85. [PMID: 31555665 PMCID: PMC6742688 DOI: 10.3389/fmolb.2019.00085] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming of innate immune cells occurs during both the hyperinflammatory and immunotolerant phases of sepsis. The hypoxia inducible factor (HIF) signaling pathway plays a vital role in regulating these metabolic changes. This review initially summarizes the HIF-driven changes in metabolic dynamics of innate immune cells in response to sepsis. The hyperinflammatory phase of sepsis is accompanied by a metabolic switch from oxidative phosphorylation to HIF-1α mediated glycolysis. Furthermore, HIF driven alterations in arginine metabolism also occur during this phase. This promotes sepsis pathophysiology and the development of clinical symptoms. These early metabolic changes are followed by a late immunotolerant phase, in which suppressed HIF signaling promotes a switch from aerobic glycolysis to fatty acid oxidation, with a subsequent anti-inflammatory response developing. Recently the molecular mechanisms controlling HIF activation during these early and late phases have begun to be elucidated. In the final part of this review the contribution of toll-like receptors, transcription factors, metabolic intermediates, kinases and reactive oxygen species, in governing the HIF-induced metabolic reprogramming of innate immune cells will be discussed. Importantly, understanding these regulatory mechanisms can lead to the development of novel diagnostic and therapeutic strategies targeting the HIF-dependent metabolic state of innate immune cells.
Collapse
Affiliation(s)
- Susan F Fitzpatrick
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Solis AG, Bielecki P, Steach HR, Sharma L, Harman CCD, Yun S, de Zoete MR, Warnock JN, To SDF, York AG, Mack M, Schwartz MA, Dela Cruz CS, Palm NW, Jackson R, Flavell RA. Mechanosensation of cyclical force by PIEZO1 is essential for innate immunity. Nature 2019; 573:69-74. [PMID: 31435009 DOI: 10.1038/s41586-019-1485-8] [Citation(s) in RCA: 406] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 07/16/2019] [Indexed: 12/21/2022]
Abstract
Direct recognition of invading pathogens by innate immune cells is a critical driver of the inflammatory response. However, cells of the innate immune system can also sense their local microenvironment and respond to physiological fluctuations in temperature, pH, oxygen and nutrient availability, which are altered during inflammation. Although cells of the immune system experience force and pressure throughout their life cycle, little is known about how these mechanical processes regulate the immune response. Here we show that cyclical hydrostatic pressure, similar to that experienced by immune cells in the lung, initiates an inflammatory response via the mechanically activated ion channel PIEZO1. Mice lacking PIEZO1 in innate immune cells showed ablated pulmonary inflammation in the context of bacterial infection or fibrotic autoinflammation. Our results reveal an environmental sensory axis that stimulates innate immune cells to mount an inflammatory response, and demonstrate a physiological role for PIEZO1 and mechanosensation in immunity.
Collapse
Affiliation(s)
- Angel G Solis
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Piotr Bielecki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Holly R Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lokesh Sharma
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Sanguk Yun
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, CT, USA.,Department of Cell Biology, Yale Cardiovascular Research Center, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale Cardiovascular Research Center, Yale University, Yale University, New Haven, CT, USA
| | - Marcel R de Zoete
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - James N Warnock
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - S D Filip To
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS, USA
| | - Autumn G York
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Matthias Mack
- Department of Internal Medicine II-Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Martin A Schwartz
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, CT, USA.,Department of Cell Biology, Yale Cardiovascular Research Center, Yale University, New Haven, CT, USA.,Department of Biomedical Engineering, Yale Cardiovascular Research Center, Yale University, Yale University, New Haven, CT, USA
| | - Charles S Dela Cruz
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruaidhrí Jackson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA. .,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
28
|
Abstract
Eukaryotic cells require sufficient oxygen (O2) for biological activity and survival. When the oxygen demand exceeds its supply, the oxygen levels in local tissues or the whole body decrease (termed hypoxia), leading to a metabolic crisis, threatening physiological functions and viability. Therefore, eukaryotes have developed an efficient and rapid oxygen sensing system: hypoxia-inducible factors (HIFs). The hypoxic responses are controlled by HIFs, which induce the expression of several adaptive genes to increase the oxygen supply and support anaerobic ATP generation in eukaryotic cells. Hypoxia also contributes to a functional decline during the aging process. In this review, we focus on the molecular mechanisms regulating HIF-1α and aging-associated signaling proteins, such as sirtuins, AMP-activated protein kinase, mechanistic target of rapamycin complex 1, UNC-51-like kinase 1, and nuclear factor κB, and their roles in aging and aging-related diseases. In addition, the effects of prenatal hypoxia and obstructive sleep apnea (OSA)-induced intermittent hypoxia have been reviewed due to their involvement in the progression and severity of many diseases, including cancer and other aging-related diseases. The pathophysiological consequences and clinical manifestations of prenatal hypoxia and OSA-induced chronic intermittent hypoxia are discussed in detail.
Collapse
|
29
|
Involvement of E3 Ligases and Deubiquitinases in the Control of HIF-α Subunit Abundance. Cells 2019; 8:cells8060598. [PMID: 31208103 PMCID: PMC6627837 DOI: 10.3390/cells8060598] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
The ubiquitin and hypoxia-inducible factor (HIF) pathways are cellular processes involved in the regulation of a variety of cellular functions. Enzymes called ubiquitin E3 ligases perform protein ubiquitylation. The action of these enzymes can be counteracted by another group of enzymes called deubiquitinases (DUBs), which remove ubiquitin from target proteins. The balanced action of these enzymes allows cells to adapt their protein content to a variety of cellular and environmental stress factors, including hypoxia. While hypoxia appears to be a powerful regulator of the ubiquitylation process, much less is known about the impact of DUBs on the HIF system and hypoxia-regulated DUBs. Moreover, hypoxia and DUBs play crucial roles in many diseases, such as cancer. Hence, DUBs are considered to be promising targets for cancer cell-specific treatment. Here, we review the current knowledge about the role DUBs play in the control of HIFs, the regulation of DUBs by hypoxia, and their implication in cancer progression.
Collapse
|
30
|
Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Han HJ. Role of HIF1 α Regulatory Factors in Stem Cells. Int J Stem Cells 2019; 12:8-20. [PMID: 30836734 PMCID: PMC6457711 DOI: 10.15283/ijsc18109] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF1) is a master transcription factor that induces the transcription of genes involved in the metabolism and behavior of stem cells. HIF1-mediated adaptation to hypoxia is required to maintain the pluripotency and survival of stem cells under hypoxic conditions. HIF1 activity is well known to be tightly controlled by the alpha subunit of HIF1 (HIF1α). Understanding the regulatory mechanisms that control HIF1 activity in stem cells will provide novel insights into stem cell biology under hypoxia. Recent research has unraveled the mechanistic details of HIF1α regulating processes, suggesting new strategies for regulating stem cells. This review summarizes recent experimental studies on the role of several regulatory factors (including calcium, 2-oxoglutarate-dependent dioxygenase, microtubule network, importin, and coactivators) in regulating HIF1α activity in stem cells.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National Universit
| |
Collapse
|
31
|
Hayashi Y, Yokota A, Harada H, Huang G. Hypoxia/pseudohypoxia-mediated activation of hypoxia-inducible factor-1α in cancer. Cancer Sci 2019; 110:1510-1517. [PMID: 30844107 PMCID: PMC6501028 DOI: 10.1111/cas.13990] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Since the first identification of hypoxic cells in sections of carcinomas in the 1950s, hypoxia has been known as a central hallmark of cancer cells and their microenvironment. Indeed, hypoxia benefits cancer cells in their growth, survival, and metastasis. The historical discovery of hypoxia‐inducible factor‐1α (HIF1A) in the early 1990s had a great influence on the field as many phenomena in hypoxia could be explained by HIF1A. However, not all regions or types of tumors are necessarily hypoxic. Thus, it is difficult to explain whole cancer pathobiology by hypoxia, especially in the early stage of cancer. Upregulation of glucose metabolism in cancer cells has been well known. Oxygen‐independent glycolysis is activated in cancer cells even in the normoxia condition, which is known as the Warburg effect. Accumulating evidence and recent advances in cancer metabolism research suggest that hypoxia‐independent mechanisms for HIF signaling activation is a hallmark for cancer. There are various mechanisms that generate pseudohypoxic conditions, even in normoxia. Given the importance of HIF1A for cancer pathobiology, the pseudohypoxia concept could shed light on the longstanding mystery of the Warburg effect and accelerate better understanding of the diverse phenomena seen in a variety of cancers.
Collapse
Affiliation(s)
- Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Asumi Yokota
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
32
|
Abstract
The TRP ion channel TRPM2 has an essential function in cell survival and protects the viability of a number of cell types after oxidative stress. It is highly expressed in many cancers including breast, prostate, and pancreatic cancer, melanoma, leukemia, and neuroblastoma, suggesting it promotes cancer cell survival. TRPM2 is activated by production of ADP-ribose (ADPR) following oxidative stress, which binds to the C-terminus of TRPM2, resulting in channel opening. In a number of cancers including neuroblastoma, TRPM2 has been shown to preserve viability and mechanisms have been identified. Activation of TRPM2 results in expression of transcription factors and kinases important in cell proliferation and survival including HIF-1/2α, CREB, nuclear factor (erythroid-derived 2)-related factor-2 (Nrf2), and Pyk2, and Src phosphorylation. Together, HIF-1/2α and CREB regulate expression of genes encoding proteins with roles in mitochondrial function including members of the electron transport complex involved in ATP production. These contribute to lower mitochondrial ROS production while expression of antioxidants regulated by HIF-1/2α, FOXO3a, CREB, and Nrf2 is maintained. CREB is also important in control of expression of key proteins involved in autophagy. When TRPM2-mediated calcium influx is inhibited, mitochondria are dysfunctional, cellular bioenergetics are reduced, production of ROS is increased, and autophagy and DNA repair are impaired, decreasing tumor growth and increasing chemotherapy sensitivity. Inhibition of TRPM2 expression or function results in decreased tumor proliferation and/or viability in many malignancies including breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, and T-cell and acute myelogenous leukemia. However, in a small number of malignancies, activation of TRPM2 rather than inhibition has been reported to reduce tumor cell survival. Here, TRPM2-mediated Ca2+ signaling and mechanisms of regulation of cancer cell growth and survival are reviewed and controversies discussed. Evidence suggests that targeting TRPM2 may be a novel therapeutic approach in many cancers.
Collapse
Affiliation(s)
- Barbara A Miller
- Departments of Pediatrics, and Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA.
| |
Collapse
|
33
|
Karagiota A, Mylonis I, Simos G, Chachami G. Protein phosphatase PPP3CA (calcineurin A) down-regulates hypoxia-inducible factor transcriptional activity. Arch Biochem Biophys 2019; 664:174-182. [PMID: 30776328 DOI: 10.1016/j.abb.2019.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 01/07/2023]
Abstract
Hypoxia-inducible factors (HIF) are master regulators of the response to hypoxia. Although several kinases are known to modify their oxygen sensitive HIF-α subunits or affect indirectly their function, little is known about the role of phosphatases in HIF control. To address this issue, a library containing siRNAs for the 25 known catalytic subunits of human phosphatases was used to screen for their effect on HIF transcriptional activity in HeLa cells. Serine-threonine phosphatase PPP3CA (calcineurin A, isoform a) was identified as the strongest candidate for a negative regulator of HIF activity. Indeed, independent silencing of PPP3CA expression stimulated HIF transcriptional activity under hypoxia, without increasing the protein levels of HIF-1α or HIF-2α. Overexpression of a constitutively active PPP3CA form, but not its catalytically inactive counterpart, inhibited HIF activity and expression of HIF target genes but did not affect HIF-1α or HIF-2α expression. These results were phenocopied by treatment with the ionophore ionomycin, that activates endogenous PPP3CA. The effect of ionomycin was mediated by PPP3CA as it was largely abolished by PPP3CA silencing. Furthermore, ionomycin enhanced the down-regulation of HIF activity by wild-type PPP3CA overexpression. Overall, our results suggest the involvement of PPP3CA in fine-tuning the HIF-dependent transcriptional response to hypoxia.
Collapse
Affiliation(s)
- Angeliki Karagiota
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ilias Mylonis
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - George Simos
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Larissa, Greece; Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada.
| | - Georgia Chachami
- Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
34
|
Liang ZL, Zhang XY, Wang F, Zhang K, Liu HF, Liu HL. Understanding molecular mechanisms of Rhodiola rosea for the treatment of acute mountain sickness through computational approaches (a STROBE-compliant article). Medicine (Baltimore) 2018; 97:e11886. [PMID: 30278484 PMCID: PMC6181534 DOI: 10.1097/md.0000000000011886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rhodiola rosea has been used in the treatment of acute mountain sickness (AMS) for a long time, but the mechanism of its action is not still completely clear. In this paper, the therapeutic mechanism of R rosea for AMS was investigated by analysis of the relationship between R rosea compositions and hypoxia-inducible factor 1 (HIF-1) degradation pathway.System biology and network biology, computational approaches were used to explore the molecular mechanisms of traditional Chinese medicine (TCM).Our results showed that chemical compositions of R rosea could inhibit the targets of HIF-1 degradation pathway in multi-composition/multi-target ways.We conclude that the 18 components with more than 2 targets and 5 targets (arrest-defective-1 [ARD1], forkhead transcription factor [FOXO4], osteosarcoma-9 [OS-9], prolyl hydroxylase 2 [PHD2], human double minute 2 [Hdm2]) deserve to be noticed, and PHD2, receptor for activated C-kinase1 (RACK1) and spermidine/spermine-N1-acetyltransferase-1 (SSAT1) may be the targets of active ingredients of rhodionin, rhodiosin, and rhodiolatuntoside, respectively.
Collapse
|
35
|
Glinton K, DeBerge M, Yeap XY, Zhang J, Forbess J, Luo X, Thorp EB. Acute and chronic phagocyte determinants of cardiac allograft vasculopathy. Semin Immunopathol 2018; 40:593-603. [PMID: 30141073 DOI: 10.1007/s00281-018-0699-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/03/2018] [Indexed: 01/09/2023]
Abstract
Post-transplant immunosuppression has reduced the incidence of T cell-mediated acute rejection, yet long-term cardiac graft survival rates remain a challenge. An important determinant of chronic solid organ allograft complication is accelerated vascular disease of the transplanted graft. In the case of cardiac allograft vasculopathy (CAV), the precise cellular etiology remains inadequately understood; however, histologic evidence hints at the accumulation and activation of innate phagocytes as a causal contributing factor. This includes monocytes, macrophages, and immature dendritic cell subsets. In addition to crosstalk with adaptive T and B immune cells, myeloid phagocytes secrete paracrine signals that directly activate fibroblasts and vascular smooth muscle cells, both of which contribute to fibrous intimal thickening. Though maladaptive phagocyte functions may promote CAV, directed modulation of myeloid cell function, at the molecular level, holds promise for tolerance and prolonged cardiac graft function.
Collapse
Affiliation(s)
- Kristofor Glinton
- Department of Pathology, The Feinberg School of Medicine, Northwestern University, 300 East Superior St, Chicago, IL, 60611, USA.,Feinberg Cardiovascular and Renal Research Institute, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Matthew DeBerge
- Department of Pathology, The Feinberg School of Medicine, Northwestern University, 300 East Superior St, Chicago, IL, 60611, USA.,Feinberg Cardiovascular and Renal Research Institute, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Xin-Yi Yeap
- Department of Pathology, The Feinberg School of Medicine, Northwestern University, 300 East Superior St, Chicago, IL, 60611, USA.,Feinberg Cardiovascular and Renal Research Institute, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA
| | - Jenny Zhang
- Department of Surgery, The Feinberg School of Medicine, Northwestern University, 251 East Huron St, Chicago, IL, 60611, USA
| | - Joseph Forbess
- Ann and Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Xunrong Luo
- Feinberg Cardiovascular and Renal Research Institute, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA.,Department of Surgery, The Feinberg School of Medicine, Northwestern University, 251 East Huron St, Chicago, IL, 60611, USA.,Department of Medicine, The Feinberg School of Medicine, Northwestern University, 251 East Huron St, Chicago, IL, 60611, USA
| | - Edward B Thorp
- Department of Pathology, The Feinberg School of Medicine, Northwestern University, 300 East Superior St, Chicago, IL, 60611, USA. .,Feinberg Cardiovascular and Renal Research Institute, The Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL, 60611, USA.
| |
Collapse
|
36
|
Azimi I. The interplay between HIF-1 and calcium signalling in cancer. Int J Biochem Cell Biol 2018; 97:73-77. [DOI: 10.1016/j.biocel.2018.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/18/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022]
|
37
|
Wilson RB. Hypoxia, cytokines and stromal recruitment: parallels between pathophysiology of encapsulating peritoneal sclerosis, endometriosis and peritoneal metastasis. Pleura Peritoneum 2018; 3:20180103. [PMID: 30911653 PMCID: PMC6405013 DOI: 10.1515/pp-2018-0103] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
Peritoneal response to various kinds of injury involves loss of peritoneal mesothelial cells (PMC), danger signalling, epithelial-mesenchymal transition and mesothelial-mesenchymal transition (MMT). Encapsulating peritoneal sclerosis (EPS), endometriosis (EM) and peritoneal metastasis (PM) are all characterized by hypoxia and formation of a vascularized connective tissue stroma mediated by vascular endothelial growth factor (VEGF). Transforming growth factor-β1 (TGF-β1) is constitutively expressed by the PMC and plays a major role in the maintenance of a transformed, inflammatory micro-environment in PM, but also in EPS and EM. Persistently high levels of TGF-β1 or stimulation by inflammatory cytokines (interleukin-6 (IL-6)) induce peritoneal MMT, adhesion formation and fibrosis. TGF-β1 enhances hypoxia inducible factor-1α expression, which drives cell growth, extracellular matrix production and cell migration. Disruption of the peritoneal glycocalyx and exposure of the basement membrane release low molecular weight hyaluronan, which initiates a cascade of pro-inflammatory mediators, including peritoneal cytokines (TNF-α, IL-1, IL-6, prostaglandins), growth factors (TGF-α, TGF-β, platelet-derived growth factor, VEGF, epidermal growth factor) and the fibrin/coagulation cascade (thrombin, Tissue factor, plasminogen activator inhibitor [PAI]-1/2). Chronic inflammation and cellular transformation are mediated by damage-associated molecular patterns, pattern recognition receptors, AGE-RAGE, extracellular lactate, pro-inflammatory cytokines, reactive oxygen species, increased glycolysis, metabolomic reprogramming and cancer-associated fibroblasts. The pathogenesis of EPS, EM and PM shows similarities to the cellular transformation and stromal recruitment of wound healing.
Collapse
Affiliation(s)
- Robert Beaumont Wilson
- Upper GI Surgery Department, Liverpool Hospital, Elizabeth St, Liverpool, 2170, NSW, Australia
| |
Collapse
|
38
|
Pallet N, Fernández-Ramos AA, Loriot MA. Impact of Immunosuppressive Drugs on the Metabolism of T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:169-200. [DOI: 10.1016/bs.ircmb.2018.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Iommarini L, Porcelli AM, Gasparre G, Kurelac I. Non-Canonical Mechanisms Regulating Hypoxia-Inducible Factor 1 Alpha in Cancer. Front Oncol 2017; 7:286. [PMID: 29230384 PMCID: PMC5711814 DOI: 10.3389/fonc.2017.00286] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/13/2017] [Indexed: 12/21/2022] Open
Abstract
Hypoxia-inducible factor 1 alpha (HIF-1α) orchestrates cellular adaptation to low oxygen and nutrient-deprived environment and drives progression to malignancy in human solid cancers. Its canonical regulation involves prolyl hydroxylases (PHDs), which in normoxia induce degradation, whereas in hypoxia allow stabilization of HIF-1α. However, in certain circumstances, HIF-1α regulation goes beyond the actual external oxygen levels and involves PHD-independent mechanisms. Here, we gather and discuss the evidence on the non-canonical HIF-1α regulation, focusing in particular on the consequences of mitochondrial respiratory complexes damage on stabilization of this pleiotropic transcription factor.
Collapse
Affiliation(s)
- Luisa Iommarini
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| |
Collapse
|
40
|
Vignali PDA, Barbi J, Pan F. Metabolic Regulation of T Cell Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1011:87-130. [DOI: 10.1007/978-94-024-1170-6_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Herawati M, Wardaya, Mulyawan W, Farhan FS, Ferdinal F, Jusman SWA, Sadikin M. Expression of Hypoxia-Inducible Factor-1α and Myoglobin in Rat Heart as Adaptive Response to Intermittent Hypobaric Hypoxia Exposure. HAYATI JOURNAL OF BIOSCIENCES 2017. [DOI: 10.1016/j.hjb.2017.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
42
|
Okuyama T, Shirakawa J, Yanagisawa H, Kyohara M, Yamazaki S, Tajima K, Togashi Y, Terauchi Y. Identification of the matricellular protein Fibulin-5 as a target molecule of glucokinase-mediated calcineurin/NFAT signaling in pancreatic islets. Sci Rep 2017; 7:2364. [PMID: 28539593 PMCID: PMC5443834 DOI: 10.1038/s41598-017-02535-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 04/12/2017] [Indexed: 12/12/2022] Open
Abstract
Glucokinase-mediated glucose signaling induces insulin secretion, proliferation, and apoptosis in pancreatic β-cells. However, the precise molecular mechanisms underlying these processes are not clearly understood. Here, we demonstrated that glucokinase activation using a glucokinase activator (GKA) significantly upregulated the expression of Fibulin-5 (Fbln5), a matricellular protein involved in matrix-cell signaling, in isolated mouse islets. The islet Fbln5 expression was induced by ambient glucose in a time- and dose-dependent manner and further enhanced by high-fat diet or the deletion of insulin receptor substrate 2 (IRS-2), whereas the GKA-induced increase in Fbln5 expression was diminished in Irs-2-deficient islets. GKA-induced Fbln5 upregulation in the islets was blunted by a glucokinase inhibitor, KATP channel opener, Ca2+ channel blocker and calcineurin inhibitor, while it was augmented by harmine, a dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) 1 A inhibitor. Although deletion of Fbln5 in mice had no significant effects on the glucose tolerance or β-cell functions, adenovirus-mediated Fbln5 overexpression increased glucose-stimulated insulin secretion in INS-1 rat insulinoma cells. Since the islet Fbln5 expression is regulated through a glucokinase/KATP channel/calcineurin/nuclear factor of activated T cells (NFAT) pathway crucial for the maintenance of β-cell functions, further investigation of Fbln5 functions in the islets is warranted.
Collapse
Affiliation(s)
- Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Jun Shirakawa
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan.
| | - Hiromi Yanagisawa
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Shunsuke Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Kazuki Tajima
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan.
| |
Collapse
|
43
|
Pan S, Zhang T, Rong Z, Hu L, Gu Z, Wu Q, Dong S, Liu Q, Lin Z, Deutschova L, Li X, Dixon A, Bruford MW, Zhan X. Population transcriptomes reveal synergistic responses of DNA polymorphism and RNA expression to extreme environments on the Qinghai-Tibetan Plateau in a predatory bird. Mol Ecol 2017; 26:2993-3010. [PMID: 28277617 DOI: 10.1111/mec.14090] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 02/10/2017] [Accepted: 02/28/2017] [Indexed: 01/04/2023]
Abstract
Low oxygen and temperature pose key physiological challenges for endotherms living on the Qinghai-Tibetan Plateau (QTP). Molecular adaptations to high-altitude living have been detected in the genomes of Tibetans, their domesticated animals and a few wild species, but the contribution of transcriptional variation to altitudinal adaptation remains to be determined. Here we studied a top QTP predator, the saker falcon, and analysed how the transcriptome has become modified to cope with the stresses of hypoxia and hypothermia. Using a hierarchical design to study saker populations inhabiting grassland, steppe/desert and highland across Eurasia, we found that the QTP population is already distinct despite having colonized the Plateau <2000 years ago. Selection signals are limited at the cDNA level, but of only seventeen genes identified, three function in hypoxia and four in immune response. Our results show a significant role for RNA transcription: 50% of upregulated transcription factors were related to hypoxia responses, differentiated modules were significantly enriched for oxygen transport, and importantly, divergent EPAS1 functional variants with a refined co-expression network were identified. Conservative gene expression and relaxed immune gene variation may further reflect adaptation to hypothermia. Our results exemplify synergistic responses between DNA polymorphism and RNA expression diversity in coping with common stresses, underpinning the successful rapid colonization of a top predator onto the QTP. Importantly, molecular mechanisms underpinning highland adaptation involve relatively few genes, but are nonetheless more complex than previously thought and involve fine-tuned transcriptional responses and genomic adaptation.
Collapse
Affiliation(s)
- Shengkai Pan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China.,Institute of Zoology Joint Laboratory for Biocomplexity Research, Cardiff University, Beichen West Road, Beijing, 100101, China.,University of Chinese Academy of Sciences, Yuquan Road, Beijing, 100049, China
| | - Tongzuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China
| | | | - Li Hu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China.,University of Chinese Academy of Sciences, Yuquan Road, Beijing, 100049, China.,BGI-Shenzhen, Shenzhen, 518083, China
| | - Zhongru Gu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China.,University of Chinese Academy of Sciences, Yuquan Road, Beijing, 100049, China
| | - Qi Wu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China
| | - Shanshan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qiong Liu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China.,State Key Laboratory of Earth Surface Processes and Resource Ecology & MOE Key Laboratory for Biodiversity Science and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Zhenzhen Lin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China
| | - Lucia Deutschova
- Raptor Protection of Slovakia, Kuklovská 5, SK-841 04, Bratislava 4, Slovakia
| | - Xinhai Li
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China
| | - Andrew Dixon
- Institute of Zoology Joint Laboratory for Biocomplexity Research, Cardiff University, Beichen West Road, Beijing, 100101, China.,International Wildlife Consultants Ltd., PO Box 19, Carmarthen, SA33 5YL, UK.,Environment Agency-Abu Dhabi, PO Box 45553, Al Mamoura Building (A), Muroor Road, Abu Dhabi, United Arab Emirates
| | - Michael W Bruford
- Institute of Zoology Joint Laboratory for Biocomplexity Research, Cardiff University, Beichen West Road, Beijing, 100101, China.,Organisms and Environment Division, Cardiff School of Bioscience, Cardiff University, Cardiff, CF10 3AX, UK
| | - Xiangjiang Zhan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road, Beijing, 100101, China.,Institute of Zoology Joint Laboratory for Biocomplexity Research, Cardiff University, Beichen West Road, Beijing, 100101, China
| |
Collapse
|
44
|
Bolger GB. The RNA-binding protein SERBP1 interacts selectively with the signaling protein RACK1. Cell Signal 2017; 35:256-263. [PMID: 28267599 DOI: 10.1016/j.cellsig.2017.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022]
Abstract
The RACK1 protein interacts with numerous proteins involved in signal transduction, the cytoskeleton, and mRNA splicing and translation. We used the 2-hybrid system to identify additional proteins interacting with RACK1 and isolated the RNA-binding protein SERBP1. SERPB1 shares amino acid sequence homology with HABP4 (also known as Ki-1/57), a component of the RNA spicing machinery that has been shown previously to interact with RACK1. Several different isoforms of SERBP1, generated by alternative mRNA splicing, interacted with RACK1 with indistinguishable interaction strength, as determined by a 2-hybrid beta-galactosidase assay. Analysis of deletion constructs of SERBP1 showed that the C-terminal third of the SERBP1 protein, which contains one of its two substrate sites for protein arginine N-methyltransferase 1 (PRMT1), is necessary and sufficient for it to interact with RACK1. Analysis of single amino acid substitutions in RACK1, identified in a reverse 2-hybrid screen, showed very substantial overlap with those implicated in the interaction of RACK1 with the cAMP-selective phosphodiesterase PDE4D5. These data are consistent with SERBP1 interacting selectively with RACK1, mediated by an extensive interaction surface on both proteins.
Collapse
Affiliation(s)
- Graeme B Bolger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-3300, USA; Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL 35294-3300, USA.
| |
Collapse
|
45
|
Nielsen MH, Flygaard RK, Jenner LB. Structural analysis of ribosomal RACK1 and its role in translational control. Cell Signal 2017; 35:272-281. [PMID: 28161490 DOI: 10.1016/j.cellsig.2017.01.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 12/28/2022]
Abstract
Receptor for Activated C-Kinase 1 (RACK1) belongs to the WD40 family of proteins, known to act as scaffolding proteins in interaction networks. Accordingly, RACK1 is found to have numerous interacting partners ranging from kinases and signaling proteins to membrane bound receptors and ion channels. Interestingly, RACK1 has also been identified as a ribosomal protein present in all eukaryotic ribosomes. Structures of eukaryotic ribosomes have shown RACK1 to be located at the back of the head of the small ribosomal subunit. This suggests that RACK1 could act as a ribosomal scaffolding protein recruiting regulators of translation to the ribosome, and several studies have in fact found RACK1 to play a role in regulation of translation. To fully understand the role of RACK1 we need to understand whether the many reported interaction partners of RACK1 bind to free or ribosomal RACK1. In this review we provide a structural analysis of ribosome-bound RACK1 to provide a basis for answering this fundamental question. Our analysis shows that RACK1 is tightly bound to the ribosome through highly conserved and specific interactions confirming RACK1 as an integral ribosomal protein. Furthermore, we have analyzed whether reported binding sites for RACK1 interacting partners with a proposed role in translational control are accessible on ribosomal RACK1. Our analysis shows that most of the interaction partners with putative regulatory functions have binding sites that are available on ribosomal RACK1, supporting the role of RACK1 as a ribosomal signaling hub. We also discuss the possible role for RACK1 in recruitment of ribosomes to focal adhesion sites and regulation of local translation during cell spreading and migration.
Collapse
Affiliation(s)
- Maja Holch Nielsen
- Department of Molecular Biology and Genetics, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Aarhus University, Denmark
| | - Rasmus Kock Flygaard
- Department of Molecular Biology and Genetics, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Aarhus University, Denmark
| | - Lasse Bohl Jenner
- Department of Molecular Biology and Genetics, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Aarhus University, Denmark
| |
Collapse
|
46
|
Asc1p/RACK1 Connects Ribosomes to Eukaryotic Phosphosignaling. Mol Cell Biol 2017; 37:MCB.00279-16. [PMID: 27821475 DOI: 10.1128/mcb.00279-16] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/24/2016] [Indexed: 02/07/2023] Open
Abstract
WD40 repeat proteins fold into characteristic β-propeller structures and control signaling circuits during cellular adaptation processes within eukaryotes. The RACK1 protein of Saccharomyces cerevisiae, Asc1p, consists exclusively of a single seven-bladed β-propeller that operates from the ribosomal base at the head region of the 40S subunit. Here we show that the R38D K40E ribosomal binding-compromised variant (Asc1DEp) is severely destabilized through mutation of phosphosite T143 to a dephosphorylation-mimicking alanine, probably through proteasomal degradation, leading to asc1- phenotypes. Phosphosite Y250 contributes to resistance to translational inhibitors but does not influence Asc1DEp stability. Beyond its own phosphorylation at T143, Y250, and other sites, Asc1p heavily influences the phosphorylation of as many as 90 proteins at 120 sites. Many of these proteins are regulators of fundamental processes ranging from mRNA translation to protein transport and turnover, cytoskeleton organization, and cellular signaling. Our data expose Asc1p/RACK1 as a key factor in phosphosignaling and manifest it as a control point at the head of the ribosomal 40S subunit itself regulated through posttranslational modification.
Collapse
|
47
|
CHEUNG JOSEPHY, MILLER BARBARAA. Transient Receptor Potential-Melastatin Channel Family Member 2: Friend or Foe. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2017; 128:308-329. [PMID: 28790515 PMCID: PMC5525431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Transient receptor potential melastatin 2 (Trpm2) channels are nonvoltage-activated channels permeable to monovalent and divalent cations, and are expressed in heart, brain, kidney, vasculature, and hematopoietic cells. Trpm2 is overexpressed in bladder, lung, breast, liver, head, and neck cancers. Classically, Trpm2 activation induces cell injury and death by Ca2+ overload or enhanced inflammatory response. Recent studies show that Trpm2 protects lungs from endotoxin-induced injury by reducing reactive oxygen species production in phagocytes; and improves cardiac function after ischemia-reperfusion injury by preserving mitochondrial respiration and cellular adenosine triphosphate levels while decreasing reactive oxygen species levels. In neuroblastoma xenografts, Trpm2 overexpression promotes tumor growth through modulation of hypoxia-inducible transcription factor expression and cellular bioenergetics; whereas Trpm2 inhibition results in enhanced sensitivity to doxorubicin. The robust expression in cancer cells and its pro-survival and proliferative properties make Trpm2 a rational target for cancer therapy. Indiscriminate Trpm2 inhibition, however, may engender serious untoward side effects in other vital organs.
Collapse
Affiliation(s)
- JOSEPH Y. CHEUNG
- Correspondence and reprint requests: Joseph Y. Cheung, MD, PhD,
Department of Medicine, Lewis Katz School of Medicine of Temple University, 3401 N. Broad Street, Suite 807, Philadelphia, Pennsylvania 19140
| | | |
Collapse
|
48
|
Bao L, Chen SJ, Conrad K, Keefer K, Abraham T, Lee JP, Wang J, Zhang XQ, Hirschler-Laszkiewicz I, Wang HG, Dovat S, Gans B, Madesh M, Cheung JY, Miller BA. Depletion of the Human Ion Channel TRPM2 in Neuroblastoma Demonstrates Its Key Role in Cell Survival through Modulation of Mitochondrial Reactive Oxygen Species and Bioenergetics. J Biol Chem 2016; 291:24449-24464. [PMID: 27694440 DOI: 10.1074/jbc.m116.747147] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/29/2016] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential melastatin 2 (TRPM2) ion channel has an essential function in modulating cell survival following oxidant injury and is highly expressed in many cancers including neuroblastoma. Here, in xenografts generated from neuroblastoma cells in which TRPM2 was depleted with CRISPR/Cas9 technology and in in vitro experiments, tumor growth was significantly inhibited and doxorubicin sensitivity increased. The hypoxia-inducible transcription factor 1/2α (HIF-1/2α) signaling cascade including proteins involved in oxidant stress, glycolysis, and mitochondrial function was suppressed by TRPM2 depletion. TRPM2-depleted SH-SY5Y neuroblastoma cells demonstrated reduced oxygen consumption and ATP production after doxorubicin, confirming impaired cellular bioenergetics. In cells in which TRPM2 was depleted, mitochondrial superoxide production was significantly increased, particularly following doxorubicin. Ectopic expression of superoxide dismutase 2 (SOD2) reduced ROS and preserved viability of TRPM2-depleted cells, however, failed to restore ATP levels. Mitochondrial reactive oxygen species (ROS) were also significantly increased in cells in which TRPM2 function was inhibited by TRPM2-S, and pretreatment of these cells with the antioxidant MitoTEMPO significantly reduced ROS levels in response to doxorubicin and protected cell viability. Expression of the TRPM2 pore mutant E960D, in which calcium entry through TRPM2 is abolished, also resulted in significantly increased mitochondrial ROS following doxorubicin treatment, showing the critical role of TRPM2-mediated calcium entry. These findings demonstrate the important function of TRPM2 in modulation of cell survival through mitochondrial ROS, and the potential of targeted inhibition of TRPM2 as a therapeutic approach to reduce cellular bioenergetics, tumor growth, and enhance susceptibility to chemotherapeutic agents.
Collapse
Affiliation(s)
- Lei Bao
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Shu-Jen Chen
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Kathleen Conrad
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Kerry Keefer
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Thomas Abraham
- Neural and Behavioral Sciences and Microscopy Imaging Facility
| | - John P Lee
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - JuFang Wang
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Xue-Qian Zhang
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Iwona Hirschler-Laszkiewicz
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Hong-Gang Wang
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Sinisa Dovat
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and
| | - Brian Gans
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Muniswamy Madesh
- The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Molecular Genetics and Medical Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Joseph Y Cheung
- the Departments of Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; The Center of Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Barbara A Miller
- Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033; Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033 and.
| |
Collapse
|
49
|
The effect of immunosuppressive molecules on T-cell metabolic reprogramming. Biochimie 2016; 127:23-36. [DOI: 10.1016/j.biochi.2016.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/22/2016] [Indexed: 12/22/2022]
|
50
|
Heaven MR, Flint D, Randall SM, Sosunov AA, Wilson L, Barnes S, Goldman JE, Muddiman DC, Brenner M. Composition of Rosenthal Fibers, the Protein Aggregate Hallmark of Alexander Disease. J Proteome Res 2016; 15:2265-82. [PMID: 27193225 PMCID: PMC5036859 DOI: 10.1021/acs.jproteome.6b00316] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alexander disease (AxD) is a neurodegenerative disorder characterized by astrocytic protein aggregates called Rosenthal fibers (RFs). We used mouse models of AxD to determine the protein composition of RFs to obtain information about disease mechanisms including the hypothesis that sequestration of proteins in RFs contributes to disease. A method was developed for RF enrichment, and analysis of the resulting fraction using isobaric tags for relative and absolute quantitation mass spectrometry identified 77 proteins not previously associated with RFs. Three of five proteins selected for follow-up were confirmed enriched in the RF fraction by immunobloting of both the AxD mouse models and human patients: receptor for activated protein C kinase 1 (RACK1), G1/S-specific cyclin D2, and ATP-dependent RNA helicase DDX3X. Immunohistochemistry validated cyclin D2 as a new RF component, but results for RACK1 and DDX3X were equivocal. None of these was decreased in the non-RF fractions compared to controls. A similar result was obtained for the previously known RF component, alphaB-crystallin, which had been a candidate for sequestration. Thus, no support was obtained for the sequestration hypothesis for AxD. Providing possible insight into disease progression, the association of several of the RF proteins with stress granules suggests a role for stress granules in the origin of RFs.
Collapse
Affiliation(s)
- Michael R. Heaven
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama 35294
| | - Daniel Flint
- Department of Neurobiology and the Civitan International Research Center, Center for Glial Biology in Medicine, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Shan M. Randall
- Keck Fourier Transform Mass Spectrometry Laboratory, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695
| | | | - Landon Wilson
- Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - James E. Goldman
- Department of Pathology & Cell Biology, Columbia University, New York, New York, 10032
| | - David C. Muddiman
- Keck Fourier Transform Mass Spectrometry Laboratory, Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695
| | - Michael Brenner
- Department of Neurobiology and the Civitan International Research Center, Center for Glial Biology in Medicine, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|