1
|
Jahankhani K, Ahangari F, Adcock IM, Mortaz E. Possible cancer-causing capacity of COVID-19: Is SARS-CoV-2 an oncogenic agent? Biochimie 2023; 213:130-138. [PMID: 37230238 PMCID: PMC10202899 DOI: 10.1016/j.biochi.2023.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/24/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has shown diverse life-threatening effects, most of which are considered short-term. In addition to its short-term effects, which has claimed many millions of lives since 2019, the long-term complications of this virus are still under investigation. Similar to many oncogenic viruses, it has been hypothesized that SARS-CoV-2 employs various strategies to cause cancer in different organs. These include leveraging the renin angiotensin system, altering tumor suppressing pathways by means of its nonstructural proteins, and triggering inflammatory cascades by enhancing cytokine production in the form of a "cytokine storm" paving the way for the emergence of cancer stem cells in target organs. Since infection with SARS-CoV-2 occurs in several organs either directly or indirectly, it is expected that cancer stem cells may develop in multiple organs. Thus, we have reviewed the impact of coronavirus disease 2019 (COVID-19) on the vulnerability and susceptibility of specific organs to cancer development. It is important to note that the cancer-related effects of SARS-CoV-2 proposed in this article are based on the ability of the virus and its proteins to cause cancer but that the long-term consequences of this infection will only be illustrated in the long run.
Collapse
Affiliation(s)
- Kasra Jahankhani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Immune Health Program at Hunter Medical Research Institute and the College of Health and Medicine at the University of Newcastle, Australia
| | - Esmaeil Mortaz
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Afsar B, Afsar RE. Hypoxia-inducible factors and essential hypertension: narrative review of experimental and clinical data. Pharmacol Rep 2023:10.1007/s43440-023-00497-x. [PMID: 37210694 DOI: 10.1007/s43440-023-00497-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023]
Abstract
Hypoxia-inducible factor (HIFs) is a new class of drug developed for the management of anemia in chronic kidney disease (CKD) patients. HIFs increase the production of erythropoietin in the kidney and liver, enhance the absorption and utilization of iron, and stimulate the maturation and proliferation of erythroid progenitor cells. Besides, HIFs regulate many physiologic processes by orchestrating the transcription of hundreds of genes. Essential hypertension (HT) is an epidemic worldwide. HIFs play a role in many biological processes involved in the regulation of blood pressure (BP). In the current review, we summarize pre-clinical and clinical studies investigating the relationship between HIFs and BP regulation in patients with CKD, conflicting issues, and discuss future potential strategies.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
3
|
Papadopoulos KI, Papadopoulou A, Aw TC. Beauty and the beast: host microRNA-155 versus SARS-CoV-2. Hum Cell 2023; 36:908-922. [PMID: 36847920 PMCID: PMC9969954 DOI: 10.1007/s13577-023-00867-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/29/2023] [Indexed: 02/28/2023]
Abstract
Severe acute respiratory coronavirus 2 (SARS-CoV-2) infection in the young and healthy usually results in an asymptomatic or mild viral syndrome, possibly through an erythropoietin (EPO)-dependent, protective evolutionary landscape. In the old and in the presence of co-morbidities, however, a potentially lethal coronavirus disease 2019 (COVID-19) cytokine storm, through unrestrained renin-angiotensin aldosterone system (RAAS) hyperactivity, has been described. Multifunctional microRNA-155 (miR-155) elevation in malaria, dengue virus (DENV), the thalassemias, and SARS-CoV-1/2, plays critical antiviral and cardiovascular roles through its targeted translational repression of over 140 genes. In the present review, we propose a plausible miR-155-dependent mechanism whereby the translational repression of AGRT1, Arginase-2 and Ets-1, reshapes RAAS towards Angiotensin II (Ang II) type 2 (AT2R)-mediated balanced, tolerable, and SARS-CoV-2-protective cardiovascular phenotypes. In addition, it enhances EPO secretion and endothelial nitric oxide synthase activation and substrate availability, and negates proinflammatory Ang II effects. Disrupted miR-155 repression of AT1R + 1166C-allele, significantly associated with adverse cardiovascular and COVID-19 outcomes, manifests its decisive role in RAAS modulation. BACH1 and SOCS1 repression creates an anti-inflammatory and cytoprotective milieu, robustly inducing antiviral interferons. MiR-155 dysregulation in the elderly, and in comorbidities, allows unimpeded RAAS hyperactivity to progress towards a particularly aggressive COVID-19 course. Elevated miR-155 in thalassemia plausibly engenders a favorable cardiovascular profile and protection against malaria, DENV, and SARS-CoV-2. MiR-155 modulating pharmaceutical approaches could offer novel therapeutic options in COVID-19.
Collapse
Affiliation(s)
- K. I. Papadopoulos
- THAI StemLife, 566/3 Soi Ramkhamhaeng 39 (Thepleela 1), Prachaouthit Rd., Wangthonglang, Bangkok, 10310 Thailand
| | - A. Papadopoulou
- Occupational and Environmental Health Services, Feelgood Lund, Ideon Science Park, Scheelevägen 17, 223 63 Lund, Sweden
| | - T. C. Aw
- Department of Laboratory Medicine, Changi General Hospital, 2 Simei Street 3, Singapore, 529889 Singapore
- Department of Medicine, National University of Singapore, Singapore, 119228 Singapore
| |
Collapse
|
4
|
Devaux CA, Raoult D. The impact of COVID-19 on populations living at high altitude: Role of hypoxia-inducible factors (HIFs) signaling pathway in SARS-CoV-2 infection and replication. Front Physiol 2022; 13:960308. [PMID: 36091390 PMCID: PMC9454615 DOI: 10.3389/fphys.2022.960308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cases of coronavirus disease 2019 (COVID-19) have been reported worldwide. However, one epidemiological report has claimed a lower incidence of the disease in people living at high altitude (>2,500 m), proposing the hypothesis that adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection. This publication was initially greeted with skepticism, because social, genetic, or environmental parametric variables could underlie a difference in susceptibility to the virus for people living in chronic hypobaric hypoxia atmospheres. Moreover, in some patients positive for SARS-CoV-2, early post-infection ‘happy hypoxia” requires immediate ventilation, since it is associated with poor clinical outcome. If, however, we accept to consider the hypothesis according to which the adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection, identification of the molecular rational behind it is needed. Among several possibilities, HIF-1 regulation appears to be a molecular hub from which different signaling pathways linking hypoxia and COVID-19 are controlled. Interestingly, HIF-1α was reported to inhibit the infection of lung cells by SARS-CoV-2 by reducing ACE2 viral receptor expression. Moreover, an association of the rs11549465 variant of HIF-1α with COVID-19 susceptibility was recently discovered. Here, we review the evidence for a link between HIF-1α, ACE2 and AT1R expression, and the incidence/severity of COVID-19. We highlight the central role played by the HIF-1α signaling pathway in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
- *Correspondence: Christian Albert Devaux,
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
5
|
Cho SN, Choi JA, Lee J, Son SH, Lee SA, Nguyen TD, Choi SY, Song CH. Ang II-Induced Hypertension Exacerbates the Pathogenesis of Tuberculosis. Cells 2021; 10:cells10092478. [PMID: 34572127 PMCID: PMC8465031 DOI: 10.3390/cells10092478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
It has been known that infection plays a role in the development of hypertension. However, the role of hypertension in the progression of infectious diseases remain unknown. Many countries with high rates of hypertension show geographical overlaps with those showing high incidence rates of tuberculosis (TB). To explore the role of hypertension in tuberculosis, we compared the effects of hypertension during mycobacterial infection, we infected both hypertensive Angiotensin II (Ang II) and control mice with Mycobacterium tuberculosis (Mtb) strain H37Ra by intratracheal injection. Ang II-induced hypertension promotes cell death through both apoptosis and necrosis in Mtb H37Ra infected mouse lungs. Interestingly, we found that lipid accumulation in pulmonary tissues was significantly increased in the hypertension group compared to the normal controls. Ang II-induced hypertension increases the formation of foamy macrophages during Mtb infection and it leads to cell death. Moreover, the hypertension group showed more severe granuloma formation and fibrotic lesions in comparison with the control group. Finally, we observed that the total number of mycobacteria was increased in the lungs in the hypertension group compared to the normal controls. Taken together, these results suggest that hypertension increases intracellular survival of Mtb through formation of foamy macrophages, resulting in severe pathogenesis of TB.
Collapse
Affiliation(s)
- Soo-Na Cho
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Ji-Ae Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Junghwan Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Sang-Hun Son
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Seong-Ahn Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Tam-Doan Nguyen
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Song-Yi Choi
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
- Translational Immunology Institute, Chungnam National University, Daejeon 34134, Korea
| | - Chang-Hwa Song
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Translational Immunology Institute, Chungnam National University, Daejeon 34134, Korea
- Correspondence: ; Tel.: +82-42-580-8245; Fax: +82-42-585-3686
| |
Collapse
|
6
|
Rebolledo DL, Lipson KE, Brandan E. Driving fibrosis in neuromuscular diseases: Role and regulation of Connective tissue growth factor (CCN2/CTGF). Matrix Biol Plus 2021; 11:100059. [PMID: 34435178 PMCID: PMC8377001 DOI: 10.1016/j.mbplus.2021.100059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Connective tissue growth factor or cellular communication network 2 (CCN2/CTGF) is a matricellular protein member of the CCN family involved in several crucial biological processes. In skeletal muscle, CCN2/CTGF abundance is elevated in human muscle biopsies and/or animal models for diverse neuromuscular pathologies, including muscular dystrophies, neurodegenerative disorders, muscle denervation, and muscle overuse. In this context, CCN2/CTGF is deeply involved in extracellular matrix (ECM) modulation, acting as a strong pro-fibrotic factor that promotes excessive ECM accumulation. Reducing CCN2/CTGF levels or biological activity in pathological conditions can decrease fibrosis, improve muscle architecture and function. In this work, we summarize information about the role of CCN2/CTGF in fibrosis associated with neuromuscular pathologies and the mechanisms and signaling pathways that regulate their expression in skeletal muscle.
Collapse
Affiliation(s)
- Daniela L Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | | | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| |
Collapse
|
7
|
Saad MAE, Fahmy MIM, Sayed RH, El-Yamany MF, El-Naggar R, Hegazy AAE, Al-Shorbagy M. Eprosartan: A closer insight into its neuroprotective activity in rats with focal cerebral ischemia-reperfusion injury. J Biochem Mol Toxicol 2021; 35:e22796. [PMID: 33942446 DOI: 10.1002/jbt.22796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022]
Abstract
Eprosartan (EPRO), an angiotensin receptor type-1 (AT-1) blocker, exhibited neuroprotective activities in ischemic stroke resulting from focal cerebral ischemia in rats. The current study aimed to clarify the neuroprotective role of EPRO in middle carotid artery occlusion (MCAO)-induced ischemic stroke in rats. Fifty-six male Wistar rats were divided into four groups (n = 14 per group): sham-operated group, sham receiving EPRO (60 mg/kg/day, po) group, ischemia-reperfusion (IR) group, and IR receiving EPRO (60 mg/kg/day, po) group. MCAO led to a remarkable impairment in motor function together with stimulation of inflammatory and apoptotic pathways in the hippocampus of rats. After MCAO, the AT1 receptor in the brain was stimulated, resulting in activation of Janus kinase 2/signal transducers and activators of transcription 3 signaling generating more neuroinflammatory milieu and destructive actions on the hippocampus. Augmentation of caspase-3 level by MCAO enhanced neuronal apoptosis synchronized with neurodegenerative effects of oxidative stress biomarkers. Pretreatment with EPRO opposed motor impairment and decreased oxidative and apoptotic mediators in the hippocampus of rats. The anti-inflammatory activity of EPRO was revealed by downregulation of nuclear factor-kappa B and tumor necrosis factor-β levels and (C-X-C motif) ligand 1 messenger RNA (mRNA) expression. Moreover, the study confirmed the role of EPRO against a unique pathway of hypoxia-inducible factor-1α and its subsequent inflammatory mediators. Furthermore, upregulation of caveolin-1 mRNA level was also observed along with decreased oxidative stress marker levels and brain edema. Therefore, EPRO showed neuroprotective effects in MCAO-induced cerebral ischemia in rats via attenuation of oxidative, apoptotic, and inflammatory pathways.
Collapse
Affiliation(s)
- Muhammad A E Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, New Giza University, Giza, Egypt
| | - Mohamed I M Fahmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Muhammad F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Reham El-Naggar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Ahmed A E Hegazy
- Department of Neurosurgery, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Muhammad Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, New Giza University, Giza, Egypt
| |
Collapse
|
8
|
Abstract
Neuroanatomic and functional studies show the paraventricular (PVN) of the hypothalamus to have a central role in the autonomic control that supports cardiovascular regulation. Direct and indirect projections from the PVN preautonomic neurons to the sympathetic preganglionic neurons in the spinal cord modulate sympathetic activity. The preautonomic neurons of the PVN adjust their level of activation in response to afferent signals arising from peripheral viscerosensory receptors relayed through the nucleus tractus solitarius. The prevailing sympathetic tone is a balance between excitatory and inhibitory influences that arises from the preautonomic PVN neurons. Under physiologic conditions, tonic sympathetic inhibition driven by a nitric oxide-γ-aminobutyric acid-mediated mechanism is dominant, but in pathologic situation such as heart failure there is a switch from inhibition to sympathoexcitation driven by glutamate and angiotensin II. Angiotensin II, reactive oxygen species, and hypoxia as a result of myocardial infarction/ischemia alter the tightly regulated posttranslational protein-protein interaction of CAPON (carboxy-terminal postsynaptic density protein ligand of neuronal nitric oxide synthase (NOS1)) and PIN (protein inhibitor of NOS1) signaling mechanism. Within the preautonomic neurons of the PVN, the disruption of CAPON and PIN signaling leads to a downregulation of NOS1 expression and reduced NO bioavailability. These data support the notion that CAPON-PIN dysregulation of NO bioavailability is a major contributor to the pathogenesis of sympathoexcitation in heart failure.
Collapse
Affiliation(s)
- Susan Pyner
- Department of Biosciences, Durham University, Durham, United Kingdom.
| |
Collapse
|
9
|
Sharma NM, Haibara AS, Katsurada K, Nandi SS, Liu X, Zheng H, Patel KP. Central Ang II (Angiotensin II)-Mediated Sympathoexcitation: Role for HIF-1α (Hypoxia-Inducible Factor-1α) Facilitated Glutamatergic Tone in the Paraventricular Nucleus of the Hypothalamus. Hypertension 2020; 77:147-157. [PMID: 33296248 PMCID: PMC7720881 DOI: 10.1161/hypertensionaha.120.16002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central infusion of Ang II (angiotensin II) has been associated with increased sympathetic outflow resulting in neurogenic hypertension. In the present study, we appraised whether the chronic increase in central Ang II activates the paraventricular nucleus of the hypothalamus (PVN) resulting in elevated sympathetic tone and altered baro- and chemoreflexes. Further, we evaluated the contribution of HIF-1α (hypoxia-inducible factor-1α), a transcription factor involved in enhancing the expression of N-methyl-D-aspartate receptors and thus glutamatergic-mediated sympathetic tone from the PVN. Ang II infusion (20 ng/minute, intracerebroventricular, 14 days) increased mean arterial pressure (126±9 versus 84±4 mm Hg), cardiac sympathetic tone (96±7 versus 75±6 bpm), and decreased cardiac parasympathetic tone (16±2 versus 36±3 versus bpm) compared with saline-infused controls in conscious rats. The Ang II-infused group also showed an impaired baroreflex control of heart rate (-1.50±0.1 versus -2.50±0.3 bpm/mm Hg), potentiation of the chemoreflex pressor response (53±7 versus 30±7 mm Hg) and increased number of FosB-labeled cells (53±3 versus 19±4) in the PVN. Concomitant with the activation of the PVN, there was an increased expression of HIF-1α and N-Methyl-D-Aspartate-type1 receptors in the PVN. Further, Ang II-infusion showed increased renal sympathetic nerve activity (20.5±2.3% versus 6.4±1.9% of Max) and 3-fold enhanced renal sympathetic nerve activity responses to microinjection of N-methyl-D-aspartate (200 pmol) into the PVN of anesthetized rats. Further, silencing of HIF-1α in NG108 cells abrogated the expression of N-methyl-D-aspartate-N-methyl-D-aspartate-type1 induced by Ang II. Taken together, our studies suggest a novel Ang II-HIF-1α-N-methyl-D-aspartate receptor-mediated activation of preautonomic neurons in the PVN, resulting in increased sympathetic outflow and alterations in baro- and chemoreflexes.
Collapse
Affiliation(s)
- Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (N.M.S., A.S.H., K.K., S.S.N., K.P.P.)
| | - Andréa S Haibara
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (N.M.S., A.S.H., K.K., S.S.N., K.P.P.)
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (N.M.S., A.S.H., K.K., S.S.N., K.P.P.)
| | - Shyam S Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (N.M.S., A.S.H., K.K., S.S.N., K.P.P.)
| | - Xuefei Liu
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (X.L., H.Z.)
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (X.L., H.Z.)
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (N.M.S., A.S.H., K.K., S.S.N., K.P.P.)
| |
Collapse
|
10
|
Marchesi N, Barbieri A, Fahmideh F, Govoni S, Ghidoni A, Parati G, Vanoli E, Pascale A, Calvillo L. Use of dual-flow bioreactor to develop a simplified model of nervous-cardiovascular systems crosstalk: A preliminary assessment. PLoS One 2020; 15:e0242627. [PMID: 33253266 PMCID: PMC7703955 DOI: 10.1371/journal.pone.0242627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic conditions requiring long-term rehabilitation therapies, such as hypertension, stroke, or cancer, involve complex interactions between various systems/organs of the body and mutual influences, thus implicating a multiorgan approach. The dual-flow IVTech LiveBox2 bioreactor is a recently developed inter-connected dynamic cell culture model able to mimic organ crosstalk, since cells belonging to different organs can be connected and grown under flow conditions in a more physiological environment. This study aims to setup for the first time a 2-way connected culture of human neuroblastoma cells, SH-SY5Y, and Human Coronary Artery Smooth Muscle Cells, HCASMC through a dual-flow IVTech LiveBox2 bioreactor, in order to represent a simplified model of nervous-cardiovascular systems crosstalk, possibly relevant for the above-mentioned diseases. The system was tested by treating the cells with 10nM angiotensin II (AngII) inducing PKCβII/HuR/VEGF pathway activation, since AngII and PKCβII/HuR/VEGF pathway are relevant in cardiovascular and neuroscience research. Three different conditions were applied: 1- HCASMC and SH-SY5Y separately seeded in petri dishes (static condition); 2- the two cell lines separately seeded under flow (dynamic condition); 3- the two lines, seeded in dynamic conditions, connected, each maintaining its own medium, with a membrane as interface for biohumoral changes between the two mediums, and then treated. We detected that only in condition 3 there was a synergic AngII-dependent VEGF production in SH-SY5Y cells coupled to an AngII-dependent PKCβII/HuR/VEGF pathway activation in HCASMC, consistent with the observed physiological response in vivo. HCASMC response to AngII seems therefore to be generated by/derived from the reciprocal cell crosstalk under the dynamic inter-connection ensured by the dual flow LiveBox 2 bioreactor. This system can represent a useful tool for studying the crosstalk between organs, helpful for instance in rehabilitation research or when investigating chronic diseases; further, it offers the advantageous opportunity of cultivating each cell line in its own medium, thus mimicking, at least in part, distinct tissue milieu.
Collapse
Affiliation(s)
- Nicoletta Marchesi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Annalisa Barbieri
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Foroogh Fahmideh
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Alice Ghidoni
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Gianfranco Parati
- Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Emilio Vanoli
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Cardiovascular Department, IRCCS Multimedica, Sesto San Giovanni, Italy
| | - Alessia Pascale
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Laura Calvillo
- Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
11
|
Role of hypoxia in skeletal muscle fibrosis: Synergism between hypoxia and TGF-β signaling upregulates CCN2/CTGF expression specifically in muscle fibers. Matrix Biol 2019; 87:48-65. [PMID: 31669521 DOI: 10.1016/j.matbio.2019.09.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023]
Abstract
Several skeletal muscle diseases are characterized by fibrosis, the excessive accumulation of extracellular matrix. Transforming growth factor-β (TGF-β) and connective tissue growth factor (CCN2/CTGF) are two profibrotic factors augmented in fibrotic skeletal muscle, together with signs of reduced vasculature that implies a decrease in oxygen supply. We observed that fibrotic muscles are characterized by the presence of positive nuclei for hypoxia-inducible factor-1α (HIF-1α), a key mediator of the hypoxia response. However, it is not clear how a hypoxic environment could contribute to the fibrotic phenotype in skeletal muscle. We evaluated the role of hypoxia and TGF-β on CCN2 expression in vitro. Fibroblasts, myoblasts and differentiated myotubes were incubated with TGF-β1 under hypoxic conditions. Hypoxia and TGF-β1 induced CCN2 expression synergistically in myotubes but not in fibroblasts or undifferentiated muscle progenitors. This induction requires HIF-1α and the Smad-independent TGF-β signaling pathway. We performed in vivo experiments using pharmacological stabilization of HIF-1α or hypoxia-induced via hindlimb ischemia together with intramuscular injections of TGF-β1, and we found increased CCN2 expression. These observations suggest that hypoxic signaling together with TGF-β signaling, which are both characteristics of a fibrotic skeletal muscle environment, induce the expression of CCN2 in skeletal muscle fibers and myotubes.
Collapse
|
12
|
Saavedra JM, Armando I. Angiotensin II AT2 Receptors Contribute to Regulate the Sympathoadrenal and Hormonal Reaction to Stress Stimuli. Cell Mol Neurobiol 2018; 38:85-108. [PMID: 28884431 PMCID: PMC6668356 DOI: 10.1007/s10571-017-0533-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Angiotensin II, through AT1 receptor stimulation, mediates multiple cardiovascular, metabolic, and behavioral functions including the response to stressors. Conversely, the function of Angiotensin II AT2 receptors has not been totally clarified. In adult rodents, AT2 receptor distribution is very limited but it is particularly high in the adrenal medulla. Recent results strongly indicate that AT2 receptors contribute to the regulation of the response to stress stimuli. This occurs in association with AT1 receptors, both receptor types reciprocally influencing their expression and therefore their function. AT2 receptors appear to influence the response to many types of stressors and in all components of the hypothalamic-pituitary-adrenal axis. The molecular mechanisms involved in AT2 receptor activation, the complex interactions with AT1 receptors, and additional factors participating in the control of AT2 receptor regulation and activity in response to stressors are only partially understood. Further research is necessary to close this knowledge gap and to clarify whether AT2 receptor activation may carry the potential of a major translational advance.
Collapse
Affiliation(s)
- J M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road, Bldg. D, Room 287, Washington, DC, 20007, USA.
| | - I Armando
- The George Washington University School of Medicine and Health Sciences, Ross Hall Suite 738 2300 Eye Street, Washington, DC, USA
| |
Collapse
|
13
|
Sharma NM, Cunningham CJ, Zheng H, Liu X, Patel KP. Hypoxia-Inducible Factor-1α Mediates Increased Sympathoexcitation via Glutamatergic N-Methyl-d-Aspartate Receptors in the Paraventricular Nucleus of Rats With Chronic Heart Failure. Circ Heart Fail 2017; 9:CIRCHEARTFAILURE.116.003423. [PMID: 27810863 DOI: 10.1161/circheartfailure.116.003423] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/13/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Increased sympathetic outflow is a major contributor to the progression of chronic heart failure (CHF). Potentiation of glutamatergic tone has been causally related to the sympathoexcitation in CHF. Specifically, an increase in the N-methyl-d-aspartate-type 1 receptor (NMDA-NR1) expression within the paraventricular nucleus (PVN) is critically linked to the increased sympathoexcitation during CHF. However, the molecular mechanism(s) for the upregulation of NMDA-NR1 remains unexplored. We hypothesized that hypoxia via hypoxia-inducible factor 1α (HIF-1α) might contribute to the augmentation of the NMDA-NR1-mediated sympathoexcitatory responses from the PVN in CHF. METHODS AND RESULTS Immunohistochemistry staining, mRNA, and protein for hypoxia-inducible factor 1α were upregulated within the PVN of left coronary artery-ligated CHF rats. In neuronal cell line (NG108-15) in vitro, hypoxia caused a significant increase in mRNA and protein for HIF-1α (2-fold) with the concomitant increase in NMDA-NR1 mRNA, protein levels, and glutamate-induced Ca+ influx. Chromatin immunoprecipitation assay identified HIF-1α binding to NMDA-NR1 promoter during hypoxia. Silencing of HIF-1α in NG108 cells leads to a significant decrease in expression of NMDA-NR1, suggesting that expression of HIF-1α is necessary for the upregulation of NMDA-NR1. Consistent with these observations, HIF-1α silencing within the PVN abrogated the increased basal sympathetic tone and sympathoexcitatory responses to microinjection of NMDA in the PVN of rats with CHF. CONCLUSIONS These results uncover a critical role for HIF-1 in the upregulation of NMDA-NR1 to mediate sympathoexcitation in CHF. We conclude that subtle hypoxia within the PVN may act as a metabolic cue to modulate sympathoexcitation during CHF.
Collapse
Affiliation(s)
- Neeru M Sharma
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Craig J Cunningham
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Hong Zheng
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Xuefei Liu
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha
| | - Kaushik P Patel
- From the Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha.
| |
Collapse
|
14
|
Movafagh S, Raj D, Sanaei-Ardekani M, Bhatia D, Vo K, Mahmoudieh M, Rahman R, Kim EH, Harralson AF. Hypoxia Inducible Factor 1: A Urinary Biomarker of Kidney Disease. Clin Transl Sci 2017; 10:201-207. [PMID: 28181420 PMCID: PMC5421733 DOI: 10.1111/cts.12445] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/03/2017] [Indexed: 01/22/2023] Open
Abstract
Identifying noninvasive biomarkers of kidney disease is valuable for diagnostic and therapeutic purposes. Hypoxia inducible factor 1 (HIF-1) expression is known to be elevated in the kidneys in several renal disease pathologies. We hypothesized that the urinary HIF-1a mRNA level may be a suitable biomarker for expression of this protein in chronic kidney disease (CKD). We compared HIF-1a mRNA levels from urine pellets of CKD and healthy subjects. To ensure that urinary HIF-1a mRNA is of kidney origin, we examined colocalization of HIF-1a mRNA with two kidney specific markers in urine cells. We found that HIF-1a mRNA is readily quantifiable in urine pellets and its expression was significantly higher in CKD patients compared with healthy adults. We also showed that the urinary HIF-1a mRNA comes primarily from cells of renal origin. Our data suggest that urinary HIF-1a mRNA is a potential biomarker in CKD and can be noninvasively assessed in patients.
Collapse
Affiliation(s)
- S Movafagh
- Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, Virginia, USA
| | - D Raj
- Department of Nephrology, George Washington University Division of Kidney Diseases and Hypertension, Washington, DC, USA
| | | | - D Bhatia
- Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, Virginia, USA
| | - K Vo
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, Virginia, USA
| | - M Mahmoudieh
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, Virginia, USA
| | - R Rahman
- Kidney and Hypertension Specialists, Manassas, Virginia, USA
| | - E H Kim
- Kidney and Hypertension Specialists, Manassas, Virginia, USA
| | - A F Harralson
- Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, Virginia, USA
| |
Collapse
|
15
|
Yilmaz TU, Yazihan N, Dalgic A, Kaya EE, Salman B, Kocak M, Akcil E. Role of ATP-dependent K channels in the effects of erythropoietin in renal ischaemia injury. Indian J Med Res 2016. [PMID: 26205024 PMCID: PMC4525406 DOI: 10.4103/0971-5916.160713] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background & objectives: Erythropoietin (EPO) has cytoprotective and anti-apoptotic effects in pathological conditions, including hypoxia and ischaemia-reperfusion injury. One of the targets to protect against injury is ATP-dependent potassium (KATP) channels. These channels could be involved in EPO induced ischaemic preconditoning like a protective effect. We evaluated the cell cytoprotective effects of EPO in relation to KATP channel activation in the renal tubular cell culture model under hypoxic/normoxic conditions. Methods: Dose and time dependent effects of EPO, KATP channel blocker glibenclamide and KATP channel opener diazoxide on cellular proliferation were evaluated by colorimetric assay MTT [3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide] under normoxic and hypoxic conditions in human renal proximal tubular cell line (CRL-2830). Evaluation of the dose and time dependent effects of EPO, glibenclamide and diazoxide on apoptosis was done by caspase-3 activity levels. Hypoxia inducible factor-1 alpha (HIF-1 α) mRNA levels were measured by semi-quantative reverse transcription polymerase chain reaction (RT)-PCR. Kir 6.1 protein expresion was evalutaed by Western blot. Results: Glibenclamide treatment decreased the number of living cells in a time and dose dependent manner, whereas EPO and diazoxide treatments increased. Glibenclamide (100 μM) treatment significantly blocked the anti-apoptotic effects of EPO (10 IU/ml) under both normoxic and hypoxic conditions. EPO (10 IU/ml) and diazoxide (100 μM) treatments significantly increased (P<0.01) whereas glibenclamide decreased (P<0.05) HIF-1 α mRNA expression. Glibenclamide significantly (P<0.01) decreased EPO induced HIF-1 α mRNA expression when compared with the EPO alone group. Interpretation & conclusions: Our results showed that the cell proliferative, cytoprotective and anti-apoptotic effects of EPO were associated with KATP channels in the renal tubular cell culture model under hypoxic/normal conditions.
Collapse
Affiliation(s)
- Tonguc Utku Yilmaz
- School of Medicine, Department of General Surgery, Kocaeli University, Kocaeli, Turkey
| | | | | | | | | | | | | |
Collapse
|
16
|
Silva AP, Mendes F, Fragoso A, Jeronimo T, Pimentel A, Gundlach K, Büchel J, Santos N, Neves PL. Altered serum levels of FGF-23 and magnesium are independent risk factors for an increased albumin-to-creatinine ratio in type 2 diabetics with chronic kidney disease. J Diabetes Complications 2016; 30:275-80. [PMID: 26750742 DOI: 10.1016/j.jdiacomp.2015.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/12/2022]
Abstract
AIMS To investigate the role of FGF-23 and magnesium in relation to the albumin-to-creatinine ratio in type 2 diabetics with chronic kidney disease (CKD) stages 2-4. METHODS In a cross-sectional study we included all eligible type 2 diabetic patients with CKD stages 2-4, followed in our outpatient Diabetic Kidney clinic. We used descriptive statistics, the Student'st-test, ANOVA and the chi-square tests. Our population was divided according to the UACR (G1 30-300 mg/g and G2≥300 mg/g), and compared these groups regarding several biological and laboratorial parameters. We employed a multiple regression model to identify risk factors of increased UACR. RESULTS The patients in G2 displayed a lower eGFR (p=0.0001) and, had lower levels of magnesium (p=0.004) as well as higher levels of FGF-23 (p=0.043) compared to patients in G1. FGF-23 (β=0.562, P=0.0001) and the magnesium (β=- 8.916, p=0.0001) were associated with increased UACR. CONCLUSIONS A dysregulation of mineral metabolism, reflected by altered levels of magnesium and FGF-23, correlates with an increased UACR in type 2 diabetic patients with CKD stages 2-4.
Collapse
Affiliation(s)
- Ana Paula Silva
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal.
| | - Filipa Mendes
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | - André Fragoso
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | | | - Ana Pimentel
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | - Kristina Gundlach
- Fresenius Medical Care Deutschland GmbH, Bad Homburg vor der Höhe, Germany
| | - Janine Büchel
- Fresenius Medical Care Deutschland GmbH, Bad Homburg vor der Höhe, Germany
| | - Nélio Santos
- Pathology Clinic, Centro Hospitalar do Algarve, Faro, Portugal
| | - Pedro Leão Neves
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| |
Collapse
|
17
|
Calò LA, Davis PA, Maiolino G, Pagnin E, Ravarotto V, Naso E, Carraro G, Naso A. Assessing the Relationship of Angiotensin II Type 1 Receptors with Erythropoietin in a Human Model of Endogenous Angiotensin II Type 1 Receptor Antagonism. Cardiorenal Med 2015; 6:16-24. [PMID: 27194993 DOI: 10.1159/000439183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/28/2015] [Indexed: 12/26/2022] Open
Abstract
HYPOTHESIS/INTRODUCTION Angiotensin II (Ang II) has been shown to control erythropoietin (EPO) synthesis as Ang II type 1 receptor (AT1R) blockers block Ang-II-induced EPO oversecretion. To further explore the involvement of AT1R in processes controlling EPO levels, plasma EPO and mononuclear cell NADPH oxidase 4 (NOX4) - a NOX family member involved in oxygen sensing, which is a process central to controlling EPO levels - were assessed in Bartter's/Gitelman's syndrome (BS/GS) patients, a human model of endogenous AT1R antagonism and healthy subjects. Heme oxygenase (HO)-1, antioxidant and anti-inflammatory factor related to NOX4 activation, and the relationship of EPO and NOX4 to HO-1 were also assessed. MATERIALS AND METHODS EPO was measured by chemiluminescent immunoassay, HO-1 by sandwich immunoassay and NOX4 protein expression by Western blot. RESULTS EPO was increased in BS/GS patients compared to healthy subjects (7.64 ± 2.47 vs. 5.23 ± 1.07 U/l; p = 0.025), whereas NOX4 did not differ between BS/GS and healthy subjects (1.76 ± 0.61 vs. 1.65 ± 0.54 densitometric units; p = n.s.), and HO-1 was increased in BS/GS patients compared to healthy subjects (9.58 ± 3.07 vs. 5.49 ± 1.04 ng/ml; p = 0.003). NOX4 positively correlated with HO-1 only in BS/GS patients; no correlation was found between EPO and either NOX4 or HO-1 in those two groups. CONCLUSIONS The effect of the renin-angiotensin system on EPO cannot be solely mediated by Ang II via AT1R signaling, but rather, EPO levels are also determined by a complex interrelated set of signals that involve AT2R, nitric oxide levels, NOX4 and HO-1 activity.
Collapse
Affiliation(s)
- Lorenzo A Calò
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Paul A Davis
- Department of Nutrition, University of California, Davis, Calif., USA
| | - Giuseppe Maiolino
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Elisa Pagnin
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Verdiana Ravarotto
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Elena Naso
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Gianni Carraro
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| | - Agostino Naso
- Department of Medicine, Nephrology and Hypertension, University of Padova, Padova, Italy
| |
Collapse
|
18
|
Güzel D, Dursun AD, Fıçıcılar H, Tekin D, Tanyeli A, Akat F, Topal Çelikkan F, Sabuncuoğlu B, Baştuğ M. Effect of intermittent hypoxia on the cardiac HIF-1/VEGF pathway in experimental type 1 diabetes mellitus. Anatol J Cardiol 2015; 16:76-83. [PMID: 26467365 PMCID: PMC5336740 DOI: 10.5152/akd.2015.5925] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE High altitude and hypoxic preconditioning have cardioprotective effects by increasing coronary vascularity, reducing post-ischemic injury, and improving cardiac function. Our purpose was to examine if intermittent hypoxia treatment has any restoring effects related to the possible role of the HIF-1/VEGF pathway on diabetic cardiomyopathy. METHODS Wistar Albino male rats (n=34) were divided into four groups: control (C), intermittent hypoxia (IH), diabetes mellitus (DM), and diabetes mellitus plus intermittent hypoxia (DM+IH). Following a streptozotocin (STZ) injection (50 mg/kg, i.p.), blood glucose levels of 250 mg/dL and above were considered as DM. IH and DM+IH groups were exposed to hypoxia 6 h/day for 42 days at a pressure corresponding to 3000 m altitude. Twenty-four hours after the IH protocol, hearts were excised. Hematoxylin and eosin-stained apical parts of the left ventricles were evaluated. Hypoxia inducible factor-1 (HIF-1), vascular endothelial growth factor 164 (VEGF164), and VEGF188 polymerase chain reaction products were run in agarose gel electrophoresis. Band density analysis of UV camera images was performed using Image J. The data were compared by one-way ANOVA, repeated measures two-way ANOVA, and the Kruskal-Wallis test. RESULTS The percent weight change was lower in the DM group than in the controls (p=0.004). The tissue injury was the highest in the DM group and the least in the IH group. Diabetes decreased, whereas the IH treatment increased the vascularity. A decrease was observed in the VEGF188 mRNA levels in the DM+IH group compared with the C group, but there were no difference in HIF-1α and VEGF164 mRNA levels between the groups. CONCLUSION The IH treatment restored the diabetic effects on the heart by reducing tissue injury and increasing the capillarity without transcriptional changes in HIF-1/VEGF correspondingly.
Collapse
Affiliation(s)
- Derya Güzel
- Department of Physiology, Faculty of Medicine, Sakarya University; Sakarya-Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Loeffler I, Wolf G. The role of hypoxia and Morg1 in renal injury. Eur J Clin Invest 2015; 45:294-302. [PMID: 25615026 DOI: 10.1111/eci.12405] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/19/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Renal hypoxia is known to play an important role in the pathophysiology of acute renal injury as well as in chronic kidney diseases. The mediators of hypoxia are the transcription factors HIF (hypoxia-inducible factors), that are highly regulated. Under normoxic conditions constitutively expressed HIF-α subunits are hydroxylated by prolyl hydroxylases (PHD1, PHD2, and PHD3) and subsequently degraded by proteasomes. MATERIALS AND METHODS This narrative review is based on the material searched for and obtained via PubMed and MEDLINE up to January 2015. RESULTS The MAPK organizer 1 (Morg1) has been identified to act as a scaffold protein of PHD3 and suppression of Morg1 leads to the stabilization of HIF-α, which forms in the absence of oxygen a heterodimer with HIF-β, translocates to the nucleus and promotes the transcription of HIF target genes. CONCLUSIONS This review summarizes the current knowledge regarding the role of hypoxia, HIF signalling, and Morg1 in acute and chronic renal injury.
Collapse
Affiliation(s)
- Ivonne Loeffler
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
20
|
Umschweif G, Shabashov D, Alexandrovich AG, Trembovler V, Horowitz M, Shohami E. Neuroprotection after traumatic brain injury in heat-acclimated mice involves induced neurogenesis and activation of angiotensin receptor type 2 signaling. J Cereb Blood Flow Metab 2014; 34:1381-90. [PMID: 24849663 PMCID: PMC4126099 DOI: 10.1038/jcbfm.2014.93] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 04/23/2014] [Accepted: 04/30/2014] [Indexed: 12/18/2022]
Abstract
Long-term exposure of mice to mild heat (34°C±1°C) confers neuroprotection against traumatic brain injury (TBI); however, the underling mechanisms are not fully understood. Heat acclimation (HA) increases hypothalamic angiotensin II receptor type 2 (AT2) expression and hypothalamic neurogenesis. Accumulating data suggest that activation of the brain AT2 receptor confers protection against several types of brain pathologies, including ischemia, a hallmark of the secondary injury occurring following TBI. As AT2 activates the same pro-survival pathways involved in HA-mediated neuroprotection (e.g., Akt phosphorylation, hypoxia-inducible factor 1α (HIF-1α), and brain-derived neurotrophic factor (BDNF)), we examined the role of AT2 in HA-mediated neuroprotection after TBI. Using an AT2-specific antagonist PD123319, we found that the improvements in motor and cognitive recovery as well as reduced lesion volume and neurogenesis seen in HA mice were all diminished by AT2 inhibition, whereas no significant alternations were observed in control mice. We also found that nerve growth factor/tropomyosin-related kinase receptor A (TrkA), BDNF/TrkB, and HIF-1α pathways are upregulated by HA and inhibited on PD123319 administration, suggesting that these pathways play a role in AT2 signaling in HA mice. In conclusion, AT2 is involved in HA-mediated neuroprotection, and AT2 activation may be protective and should be considered a novel drug target in the treatment of TBI patients.
Collapse
Affiliation(s)
- Gali Umschweif
- 1] Department of Pharmacology, School of Pharmacy, Institute of Drug Research, Jerusalem, Israel [2] Laboratory of Environmental Physiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dalia Shabashov
- Department of Pharmacology, School of Pharmacy, Institute of Drug Research, Jerusalem, Israel
| | | | - Victoria Trembovler
- Department of Pharmacology, School of Pharmacy, Institute of Drug Research, Jerusalem, Israel
| | - Michal Horowitz
- Laboratory of Environmental Physiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Shohami
- Department of Pharmacology, School of Pharmacy, Institute of Drug Research, Jerusalem, Israel
| |
Collapse
|
21
|
Li G, Lu WH, Ai R, Yang JH, Chen F, Tang ZZ. The relationship between serum hypoxia-inducible factor 1α and coronary artery calcification in asymptomatic type 2 diabetic patients. Cardiovasc Diabetol 2014; 13:52. [PMID: 24564828 PMCID: PMC3938975 DOI: 10.1186/1475-2840-13-52] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/20/2014] [Indexed: 01/02/2023] Open
Abstract
Background Hypoxia-inducible factor 1 (HIF-1), a master regulator of oxygen homeostasis, is a heterodimer consisting of HIF-1α and HIF-1β subunits, and is implicated in calcification of cartilage and vasculature. The goal of this study was to determine the relationship between serum HIF-1α with coronary artery calcification (CAC) in patients with type 2 diabetes. Methods The subjects were 405 (262 males, 143 females, age 51.3 ± 6.4 years) asymptomatic patients with type 2 diabetes mellitus. Serum HIF-1α and interleukin-6 (IL-6) levels were measured by ELISA. CAC scores were assessed by a 320-slice CT scanner. The subjects were divided into 4 quartiles depending on serum HIF-1α levels. Results Average serum HIF-1α was 184.4 ± 66.7 pg/ml. Among patients with higher CAC scores, HIF-1α levels were also significantly increased (p <0.001). HIF-1α levels positively correlated with CRP, IL-6, UKPDS risk score, HbA1c, FBG, and CACS, but did not correlate with diabetes duration, age, and LDL. According to the multivariate analysis, HIF-1α levels significantly and independently predict the presence of CAC. ROC curve analysis showed that the serum HIF-1α level can predict the extent of CAC, but the specificity was lower than the traditional risk factors UKPDS and HbA1c. Conclusion As a marker of hypoxia, serum HIF-1α level may be an independent risk factor for the presence of CAC. These findings indicate that elevated serum HIF-1α may be involved in vascular calcification in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Gang Li
- Emergency Department, Wuhan General Hospital of Guangzhou Military Command, Wu Luo Road, Hong Shan, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
22
|
Ranasinghe WKB, Sengupta S, Williams S, Chang M, Shulkes A, Bolton DM, Baldwin G, Patel O. The effects of nonspecific HIF1α inhibitors on development of castrate resistance and metastases in prostate cancer. Cancer Med 2014; 3:245-51. [PMID: 24464861 PMCID: PMC3987074 DOI: 10.1002/cam4.189] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 11/23/2013] [Accepted: 12/23/2013] [Indexed: 01/22/2023] Open
Abstract
Expression of hypoxia-inducible factor (HIF)1α increases the risk of castrate-resistant prostate cancer (CRPC) and metastases in patients on androgen deprivation therapy (ADT) for prostate cancer (PC). We aimed to investigate the effects of nonspecific HIF1α inhibitors (Digoxin, metformin, and angiotensin-2 receptor blockers) on development of CRPC and metastases while on ADT. A retrospective review of prospectively collected medical records was conducted of all men who had continuous ADT as first-line therapy for CRPC at the Austin Hospital from 1983 to 2011. Association between HIF1α inhibitor medications and time to develop CRPC was investigated using actuarial statistics. Ninety-eight patients meeting the criteria were identified. Eighteen patients (21.4%) were treated with the nonspecific HIF1α inhibitors. Both groups had similar characteristics, apart from patients on HIF1α inhibitors being older (70 years vs. 63.9 years). The median CRPC-free survival was longer in men using HIF1α inhibitors compared to those not on inhibitors (6.7 years vs. 2.7 years, P = 0.01) and there was a 71% reduction in the risk of developing CRPC (HR 0.29 [95% CI 0.10-0.78] P = 0.02) after adjustment for Gleason score, age, and prostate-specific antigen (PSA). The median metastasis-free survival in men on HIF1α inhibitors was also significantly longer compared to those on no inhibitors (5.1 years vs. 2.6 years, P = 0.01) with an 81% reduction in the risk of developing metastases (HR 0.19 [CI 0.05-0.76] P = 0.02) after adjustment for Gleason score, age, and PSA. Nonspecific HIF1α inhibitors appear to increase the progression-free survival and reduce the risk of developing CRPC and metastases in patients on continuous ADT.
Collapse
Affiliation(s)
- Weranja K B Ranasinghe
- Department of Urology, Austin Health/University of Melbourne, Heidelberg, Victoria, Australia; Department of Surgery, Austin Health/University of Melbourne, Heidelberg, Victoria, Australia; Royal Melbourne Hospital, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Stahr A, Frahm C, Kretz A, Bondeva T, Witte O, Wolf G. Morg1+/− heterozygous mice are protected from experimentally induced focal cerebral ischemia. Brain Res 2012; 1482:22-31. [DOI: 10.1016/j.brainres.2012.09.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 08/25/2012] [Accepted: 09/10/2012] [Indexed: 01/13/2023]
|
24
|
Bondeva T, Heinzig J, Franke S, Wolf G. Angiotensin II differentially regulates Morg1 expression in kidney cells. Am J Nephrol 2012; 35:442-55. [PMID: 22555025 DOI: 10.1159/000337922] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/08/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND The mitogen-activated protein kinase organizer 1 (Morg1) belongs to the WD-40 repeat protein family and is a scaffold molecule for the extracellular regulated kinase signaling pathway. Morg1 also binds to prolyl-hydroxylase 3 (PHD3) and regulates the hypoxia-inducible factor-1α (HIF-1α) expression via PHD3 stabilization. Morg1 has been detected in the kidney as well as in other cell tissues but its expression in renal cells has not been well investigated. It has been widely shown that angiotensin II (ANG II) mediates renal damage. We have previously shown that ANG II downregulates the expression of PHD3 in PC12 cells. The aim of this study was to analyze whether ANG II regulates Morg1 expression in mouse mesangial cells (MMC), mouse proximal tubular cells (MTC) and in differentiated podocytes. The correlation between the expression of Morg1 and PHD3 activity was also addressed. METHODS Effect of ANG II on the Morg1 mRNA expression level was assessed by real-time PCR. Morg1 and HIF-1α cellular localization was analyzed by immunohistochemistry. HIF-1α promoter activity was investigated using a reporter gene system. PHD3 hydroxylase activity test was measured with a hydroxylation-coupled decarboxylation assay. RESULTS ANG II differentially regulates Morg1 expression in MMC, MTC and differentiated podocytes. We detected a biphasic effect of ANG II on Morg1 mRNA expression which was time dependent. While 9-hour ANG II treatment downregulated Morg1 expression in MMC, it induced Morg1 expression in MTC. Conversely, 24-hour ANG II stimulation upregulated the expression of Morg1 mRNA in MMC, but showed an opposite effect in MTC and differentiated podocytes. In addition, we found that ANG II signals mostly through the AT(1) receptor subtype in MMC and via the AT(2) subtype in MTC. PHD3 activity correlated to Morg1 expression patterns. Our data also demonstrate that HIF-1α transcriptional activity in MTC contrasted to PHD3 activity at 9 and 24 h, whereas in the MMC and in podocytes we did not find any correlation between PHD3 HIF-1α hydroxylation ability and HIF-1α transcriptional activation, suggesting a different mechanism of regulation in these cell types. Interestingly, the reduced expression of Morg1 in mesangial cells isolated from Morg1 (+/-) heterozygous mice correlated with a reduced PHD3 enzymatic activity and an increased HIF-1α transcriptional activity compared with mesangial cells originated from wild-type (Morg1 +/+) mice. CONCLUSIONS We show for the first time in various renal cells that ANG II modulates Morg1 expression and HIF-1α transcriptional activity via cell type-specific mechanisms, demonstrating a novel mechanism by which ANG II may contribute to renal disease.
Collapse
Affiliation(s)
- Tzvetanka Bondeva
- Department of Internal Medicine III, Friedrich Schiller University, Jena, Germany
| | | | | | | |
Collapse
|
25
|
|
26
|
Rüster C, Wolf G. Angiotensin II as a morphogenic cytokine stimulating renal fibrogenesis. J Am Soc Nephrol 2011; 22:1189-99. [PMID: 21719784 DOI: 10.1681/asn.2010040384] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inhibitors of the renin-angiotensin-aldosterone system attenuate glomerulosclerosis and interstitial fibrosis. Although the mechanisms underlying their antifibrotic effects are complex, angiotensin II (Ang II) emerges as a major profibrogenic cytokine. Ang II modulates renal cell growth, extracellular matrix synthesis, and degradation by multiple fibrotic pathways. One of the main targets of Ang II in renal fibrosis is TGFβ. Many, but not all, of the stimulatory effects of Ang II on fibrogenesis depend on the induction of TGFβ and its downstream mediators of matrix accumulation, inflammation, and apoptosis. However because of the difficulty in targeting TGFβ, connective tissue growth factor β (CTGF), a downstream mediator of TGFβ, has become a more promising antifibrotic target. Ang II can directly induce expression of renal CTGF and mediate epithelial-mesenchymal transition. Other profibrotic factors stimulated by Ang II include endothelin-1, plasminogen activator inhibitor-1, matrix metalloproteinase (MMP)-2, and a tissue inhibitor of metalloproteinase-2. Finally, connections among Ang II, hypoxia, and the induction of hypoxia-inducible factor-1α contribute to fibrogenesis. A better understanding of the multiple morphogenic effects of Ang II may be necessary to develop better strategies to halt the progression of renal disease.
Collapse
Affiliation(s)
- Christiane Rüster
- Department of Internal Medicine III, Friedrich Schiller University, Erlanger-Allee 101, D-07740 Jena, Germany
| | | |
Collapse
|
27
|
Kristensen PL, Pedersen-Bjergaard U, Schalkwijk C, Olsen NV, Thorsteinsson B. Erythropoietin and vascular endothelial growth factor as risk markers for severe hypoglycaemia in type 1 diabetes. Eur J Endocrinol 2010; 163:391-8. [PMID: 20566589 DOI: 10.1530/eje-10-0464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Circulating erythropoietin (EPO) and vascular endothelial growth factor (VEGF) increase during hypoglycaemia and may represent protective hormonal counter-regulatory responses. We tested the hypothesis that low levels of EPO and VEGF are associated with a higher frequency of severe hypoglycaemia in a cohort of patients with type 1 diabetes. DESIGN Prospective observational follow-up study. METHODS Totally 219 patients with type 1 diabetes (41% females, age 46+/-13 years (mean+/-s.d.), duration of diabetes 21+/-12 years, and HbAlc 8.5+/-1.1%) were followed in a 1-year observational study. Plasma EPO and serum VEGF levels were measured at baseline with ELISA. Events of severe hypoglycaemia defined by third party assistance were recorded and validated in telephone interviews within 24 h. RESULTS Totally 235 episodes of severe hypoglycaemia (1.1 episodes per patient-year) were reported by 82 patients (37%). At baseline, plasma EPO was 8.6 (3.1-34.3) U/l (median (range)), and serum VEGF was 52.2 (6.6-337) pg/ml. The levels of EPO and VEGF were not associated with frequency of severe and mild hypoglycaemia. The levels of EPO were not associated with age, sex, duration of diabetes, body mass index, HbAlc, C-peptide level or hypoglycaemia awareness status. The levels of VEGF were positively associated with age and female sex. CONCLUSIONS Although several studies suggest that VEGF and EPO may affect brain function during hypoglycaemia, this study does not support random VEGF or EPO levels to determine future risk of severe hypoglycaemia in people with type 1 diabetes.
Collapse
Affiliation(s)
- P L Kristensen
- Endocrinology Section, Department of Cardiology and Endocrinology, Hillerød Hospital, Dyrehavevej 29, DK-3400 Hillerød, Denmark.
| | | | | | | | | |
Collapse
|
28
|
Bibeau K, Battista MC, Houde V, Brochu M. Fetal adrenal gland alterations in a rat model of adverse intrauterine environment. Am J Physiol Regul Integr Comp Physiol 2009; 298:R899-911. [PMID: 19923365 DOI: 10.1152/ajpregu.00238.2009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
By feeding a low-sodium diet to dams over the last third of gestation, we have developed an animal model of intrauterine growth restriction (IUGR). Given that fetal adrenal development and maturation occur during late gestation in rats, the aim of this study was to evaluate the expression of proteins and enzymes involved in steroidogenesis and catecholamine synthesis in adrenal glands from IUGR fetuses. A gene microarray was performed to investigate for alteration in the pathways participating in hormone production. Results show that increased aldosterone serum concentrations in IUGR fetuses were associated with higher mRNA adrenal levels of angiotensin II receptor type 1 (AT(1)R) and cytochrome P450 aldosterone synthase in response to decreased serum sodium content. Conversely, reduced serum corticosterone concentrations in these fetuses appear to result from alterations in gene expression involved in cholesterol metabolism, such as the augmented apolipoprotein E levels, and in steroidogenesis, like the decreased levels of cytochrome P45011beta-hydroxylase. Furthermore, increased AT(2)R expression and the presence of hypoxia and oxidative stress may, in turn, explain the higher adrenal mRNA levels of enzymes involved in catecholamine synthesis. Despite this increase, catecholamine adrenal content was reduced in males and was similar in females compared with sex-matched controls, suggesting higher catecholamine secretion. This could be associated with the induction of genes involved in inflammation-related, acute-phase response in IUGR fetuses. All of these alterations could have long-lasting health effects and may, hence, be implicated in the pathogenesis of increased blood pressure and cardiac hypertrophy observed in IUGR adult animals from this model.
Collapse
Affiliation(s)
- Karine Bibeau
- Research Centre, Centre Hospitalier Universitaire Sainte-Justine, 3175 Côte Ste-Catherine, Montréal, QC, Canada H3T 1C5
| | | | | | | |
Collapse
|
29
|
Saito M, Shinohara Y, Sasaki H, Netsu Y, Yoshida M, Nakahata N. Type 1 angiotensin receptor (AT1-R)-mediated decrease in type 2 angiotensin receptor mRNA level is dependent on Gq and extracellular signal-regulated kinase 1//2 in AT1-R-transfected PC12 cells. J Neuroendocrinol 2008; 20:299-308. [PMID: 18208547 DOI: 10.1111/j.1365-2826.2008.01646.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Angiotensin II (Ang II) functions through two major Ang II receptor subtypes, type 1 (AT1-R) and type 2 (AT2-R), both of which are classified to be G protein-coupled receptors. AT2-R is highly expressed at the fetal stage, and in heart remodelling and brain ischaemia; therefore, it is important to clarify the regulatory mechanisms of AT2-R expression. Although AT1-R is generally believed to modulate AT2-R expression in some tissues or cells, the exact mechanism remains to be clarified. In the present study, we examined the effect of AT1-R stimulation on expression of endogenous rat AT2-R (rAT2-R) in AT1-R-transfected PC12 cells. rAT2-R mRNA and protein expression were decreased by Ang II in PC12 cells transfected with rAT1A-R in a time-dependent manner, mediated through a decline in mRNA stability. The C-terminus of G protein-coupled receptor (GPCR) is important for GPCR-mediated signal transduction. Therefore, we used C-terminus-deleted human AT1-R (hAT1-327STOP), which is thought to be a nondesensitised mutant of hAT1-R. As a result, Ang II decreased rAT2-R mRNA expression to a greater extent in cells transfected with hAT1-327STOP than with wild-type hAT1-R. The decrease was completely reversed by AT1-R antagonist candesartan, G(q) inhibitor YM254980, and mitogen-activated protein kinase (MAPK) kinase 1/2 inhibitor U0126, but not by pertussis toxin, which uncouples the receptor with G(i), or p38 MAPK inhibitor SB203580. We suggest, possibly for the first time, that the hAT1-R/G(q)/extracellular signal-regulated kinase 1/2 pathway is involved in the down-regulation of AT2-R using PC12 cells transfected with AT1-R.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Down-Regulation
- GTP-Binding Protein alpha Subunits, Gq-G11/physiology
- Gene Expression Regulation, Neoplastic/drug effects
- Mitogen-Activated Protein Kinase 1/physiology
- Mitogen-Activated Protein Kinase 3/physiology
- PC12 Cells
- Protein Structure, Tertiary/physiology
- RNA Stability/drug effects
- RNA, Messenger/metabolism
- Rats
- Receptor, Angiotensin, Type 1/chemistry
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Signal Transduction/physiology
- Transfection
Collapse
Affiliation(s)
- M Saito
- Department of Cellular Signalling, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Angiotensin II (AngII) is an important mediator in renal injury. Accumulating evidence suggests that AngII stimulates intracellular formation of reactive oxygen species (ROS) such as the superoxide anion and hydrogen peroxide. AngII activates several subunits of the membrane-bound multicomponent NAD(P)H oxidase and also increases ROS formation in the mitochondria. Some of these effects may be induced by aldosterone and not directly by AngII. The superoxide anion and hydrogen peroxide influence other downstream signaling pathways, such as transcription factors, tyrosine kinases/phosphatases, ion channels, and mitogen-activated protein kinases. Through these signaling pathways, ROS have distinct functional effects on renal cells. They are transducers of cell growth, apoptosis, and cell migration and affect expression of inflammatory and extracellular matrix genes. For example, AngII-mediated expression of p27(Kip1), a cell-cycle regulatory protein, and induction of tubular hypertrophy depend on the generation of ROS. The effects of ROS generated within different renal cells ultimately depend on the locally generated concentrations and the balance of pro- and antioxidant pathways. Although the concept that AngII mediates oxidative stress in the kidney has been validated in experimental models, the exact role is still incompletely understood in human renal diseases.
Collapse
Affiliation(s)
- Anja Sachse
- Department of Medicine, University of Jena, Jena, Germany
| | | |
Collapse
|
31
|
Munk VC, Sanchez de Miguel L, Petrimpol M, Butz N, Banfi A, Eriksson U, Hein L, Humar R, Battegay EJ. Angiotensin II Induces Angiogenesis in the Hypoxic Adult Mouse Heart In Vitro Through an AT
2
–B2 Receptor Pathway. Hypertension 2007; 49:1178-85. [PMID: 17339539 DOI: 10.1161/hypertensionaha.106.080242] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiotensin II is a vasoactive peptide that may affect vascularization of the ischemic heart via angiogenesis. In this study we aimed at studying the mechanisms underlying the angiogenic effects of angiotensin II under hypoxia in the mouse heart in vitro. Endothelial sprout formation from pieces of mouse hearts was assessed under normoxia (21% O
2
) and hypoxia (1% O
2
) during a 7-day period of in vitro culture. Only under hypoxia did angiotensin II dose-dependently induce endothelial sprout formation, peaking at 10
−7
mol/L of angiotensin II. Angiotensin II type 1 (AT
1
) receptor blockade by losartan did not affect angiotensin II–induced sprouting in wild-type mice. Conversely, the angiotensin II type 2 (AT
2
) receptor antagonist PD 123319 blocked this response. In hearts from AT
1
−/−
mice, angiotensin II–elicited sprouting was preserved but blocked again by AT
2
receptor antagonism. In contrast, no angiotensin II–induced sprouting was found in preparations from hearts of AT
2
−/−
mice. Angiotensin II–mediated angiogenesis was also abolished by a specific inhibitor of the B2 kinin receptor in both wild-type and AT
1
−/−
mice. Furthermore, angiotensin II failed to induce endothelial sprout formation in hearts from B2
−/−
mice. Finally, NO inhibition completely blunted sprouting in hearts from wild-type mice, whereas NO donors could restore sprouting in AT
2
−/−
and B2
−/−
hearts. This in vitro study suggests the obligatory role of hypoxia in the angiogenic effect of angiotensin II in the mouse heart via the AT
2
receptor through a mechanism that involves bradykinin, its B2 receptor, and NO as a downstream effector.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Angiotensin II/pharmacology
- Animals
- Coronary Vessels/drug effects
- Coronary Vessels/physiopathology
- Dose-Response Relationship, Drug
- Hypoxia/metabolism
- Hypoxia/physiopathology
- In Vitro Techniques
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Physiologic
- Nitric Oxide/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Bradykinin B2/deficiency
- Receptor, Bradykinin B2/metabolism
Collapse
Affiliation(s)
- Veronica C Munk
- Department of Research, Laboratory of Vascular Biology, University Hospital, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Eberhardt W, Doller A, Akool ES, Pfeilschifter J. Modulation of mRNA stability as a novel therapeutic approach. Pharmacol Ther 2007; 114:56-73. [PMID: 17320967 DOI: 10.1016/j.pharmthera.2007.01.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 01/10/2007] [Accepted: 01/10/2007] [Indexed: 12/21/2022]
Abstract
During the last decade evidence has accumulated that modulation of mRNA stability plays a central role in cellular homeostasis, including cell differentiation, proliferation and adaptation to external stimuli. The functional relevance of posttranscriptional gene regulation is highlighted by many pathologies, wherein occurrence tightly correlates with a dysregulation in mRNA stability, including chronic inflammation, cardiovascular diseases and cancer. Most commonly, the cis-regulatory elements of mRNA decay are represented by the adenylate- and uridylate (AU)-rich elements (ARE) which are specifically bound by trans-acting RNA binding proteins, which finally determine whether mRNA decay is delayed or facilitated. Regulation of mRNA decay by RNA stabilizing and RNA destabilizing factors is furthermore controlled by different intrinsic and environmental stimuli. The modulation of mRNA binding proteins, therefore, illuminates a promising approach for the pharmacotherapy of those key pathologies mentioned above and characterized by a posttranscriptional dysregulation. Most promisingly, intracellular trafficking of many of the mRNA stability regulating factors is, in turn, regulated by some major signaling pathways, including the mitogen-activated protein kinase (MAPK) cascade, the AMP-activated kinase (AMPK) and the protein kinase (PK) C (PKC) family. In this review, we present timely examples of genes regulated by mRNA stability with a special focus on signaling pathways involved in the ARE-dependent mRNA decay. A better understanding of these processes may form the basis for the development of novel therapeutics to treat major human diseases.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
33
|
Rüster C, Wolf G. Renin-angiotensin-aldosterone system and progression of renal disease. J Am Soc Nephrol 2006; 17:2985-91. [PMID: 17035613 DOI: 10.1681/asn.2006040356] [Citation(s) in RCA: 310] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Inhibition of the renin-angiotensin-aldosterone system (RAAS) is one of the most powerful maneuvers to slow progression of renal disease. Angiotensin II (AngII) has emerged in the past decade as a multifunctional cytokine that exhibits many nonhemodynamic properties, such as acting as a growth factor and profibrogenic cytokine, and even having proinflammatory properties. Many of these deleterious functions are mediated by other factors, such as TGF-beta and chemoattractants that are induced in the kidney by AngII. Moreover, understanding of the RAAS has become much more complex in recent years with the identification of novel peptides (e.g., AngIV) that could bind to specific receptors, elucidating deleterious effects, and non-angiotensin-converting enzyme (ACE)-mediated generation of AngII. The ability of renal cells to produce AngII in a concentration that is much higher than what is found in the systemic circulation and the observation that aldosterone may be engaged directly in profibrogenic processes independent of hypertension have added to the complexity of the RAAS. Even renin has now been identified to have a "life on its own" and mediates profibrotic effects via binding to specific receptors. Finally, drugs that are used to block the RAAS, such as ACE inhibitors or certain AngII type 1 receptor antagonists, may have properties on cells independent of AngII (ACE inhibitor-mediated outside-inside signaling and peroxisome proliferator-activated receptor-gamma stimulatory effects of certain sartanes). Although blockade of the RAAS with ACE inhibitors, AngII type 1 receptor antagonists, or the combination of both should be part of every strategy to slow progression of renal disease, a better understanding of the novel aspects of the RAAS should contribute to the development of innovative strategies not only to completely halt progression but also to induce regression of human renal disease.
Collapse
Affiliation(s)
- Christiane Rüster
- Department of Internal Medicine III, Friedrich-Schiller-University, Jena, Germany
| | | |
Collapse
|
34
|
Hopfer U, Hopfer H, Jablonski K, Stahl RAK, Wolf G. The Novel WD-repeat Protein Morg1 Acts as a Molecular Scaffold for Hypoxia-inducible Factor Prolyl Hydroxylase 3 (PHD3). J Biol Chem 2006; 281:8645-55. [PMID: 16407229 DOI: 10.1074/jbc.m513751200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1), a transcriptional complex composed of an oxygen-sensitive alpha- and a beta-subunit, plays a pivotal role in cellular adaptation to low oxygen availability. Under normoxia, the alpha-subunit of HIF-1 is hydroxylated by a family of prolyl hydroxylases (PHDs) and consequently targeted for proteasomal degradation. Three different PHDs have been identified, but the difference among their in vivo roles remain unclear. PHD3 is strikingly expressed by hypoxia, displays high substrate specificity, and has been identified in other signaling pathways. PHD3 may therefore hydroxylate divergent substrates and/or connect divergent cellular responses with HIF. We identified a novel WD-repeat protein, recently designated Morg1 (MAPK organizer 1), by screening a cDNA library with yeast two-hybrid assays. The interaction between PHD3 and Morg1 was confirmed in vitro and in vivo. We found seven WD-repeat domains by cloning the full-length cDNA of Morg1. By confocal microscopy both proteins co-localize within the cytoplasm and the nucleus and display a similar tissue expression pattern in Northern blots. Binding occurs at a conserved region predicted to the top surface of one propeller blade. Finally, HIF-mediated reporter gene activity is decreased by Morg1 and reduced to basal levels when Morg1 is co-expressed with PHD3. Suppression of Morg1 or PHD3 by stealth RNA leads to a marked increase of HIF-1 activity. These results indicate that Morg1 specifically interacts with PHD3 most likely by acting as a molecular scaffold. This interaction may provide a molecular framework between HIF regulation and other signaling pathways.
Collapse
Affiliation(s)
- Ulrike Hopfer
- Department of Medicine, University of Hamburg, Martinistr. 52, D-20246 Hamburg.
| | | | | | | | | |
Collapse
|
35
|
Wolf G, Chen S, Ziyadeh FN. From the periphery of the glomerular capillary wall toward the center of disease: podocyte injury comes of age in diabetic nephropathy. Diabetes 2005; 54:1626-34. [PMID: 15919782 DOI: 10.2337/diabetes.54.6.1626] [Citation(s) in RCA: 445] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nephropathy is a major complication of diabetes. Alterations of mesangial cells have traditionally been the focus of research in deciphering molecular mechanisms of diabetic nephropathy. Injury of podocytes, if recognized at all, has been considered a late consequence caused by increasing proteinuria rather than an event inciting diabetic nephropathy. However, recent biopsy studies in humans have provided evidence that podocytes are functionally and structurally injured very early in the natural history of diabetic nephropathy. The diabetic milieu, represented by hyperglycemia, nonenzymatically glycated proteins, and mechanical stress associated with hypertension, causes downregulation of nephrin, an important protein of the slit diaphragm with antiapoptotic signaling properties. The loss of nephrin leads to foot process effacement of podocytes and increased proteinuria. A key mediator of nephrin suppression is angiotensin II (ANG II), which can activate other cytokine pathways such as transforming growth factor-beta (TGF-beta) and vascular endothelial growth factor (VEGF) systems. TGF-beta1 causes an increase in mesangial matrix deposition and glomerular basement membrane (GBM) thickening and may promote podocyte apoptosis or detachment. As a result, the denuded GBM adheres to Bowman's capsule, initiating the development of glomerulosclerosis. VEGF is both produced by and acts upon the podocyte in an autocrine manner to modulate podocyte function, including the synthesis of GBM components. Through its effects on podocyte biology, glomerular hemodynamics, and capillary endothelial permeability, VEGF likely plays an important role in diabetic albuminuria. The mainstays of therapy, glycemic control and inhibition of ANG II, are key measures to prevent early podocyte injury and the subsequent development of diabetic nephropathy.
Collapse
Affiliation(s)
- Gunter Wolf
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania, 700 Clinical Research Building, 415 Curie Blvd., Philadelphia, PA 19104-4218, USA
| | | | | |
Collapse
|
36
|
Krick S, Hänze J, Eul B, Savai R, Seay U, Grimminger F, Lohmeyer J, Klepetko W, Seeger W, Rose F. Hypoxia-driven proliferation of human pulmonary artery fibroblasts: cross-talk between HIF-1alpha and an autocrine angiotensin system. FASEB J 2005; 19:857-9. [PMID: 15718424 DOI: 10.1096/fj.04-2890fje] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pulmonary artery adventitial fibroblasts (FBPA) may play a central role in lung vascular remodeling under conditions of hypoxia and inflammation, the result being pulmonary hypertension and cor pulmonale. In cultured human FBPA, both angiotensin II (Ang II) and hypoxia promoted cell cycle progression and cell proliferation and suppressed apoptosis. These effects were further enhanced when both stimuli were applied simultaneously. Hypoxia elevated the expression of hypoxia-inducible factor 1alpha (HIF-1alpha) and increased the expression of genes regulated by the hypoxia-responsive element (HRE). Up-regulation of both angiotensin-converting enzyme (ACE) and Ang II receptor type 1 (AT1) was also observed. Exogenous Ang II further increased HIF/HRE-dependent signaling in FBPA, whereas suppression of the autocrine ACE-Ang II-AT1 loop with inhibitors of ACE, AT1, and phosphatidylinositol 3-kinase (PI3K) reduced the proliferative response to both hypoxia and exogenous Ang II. Overexpression of HIF-1alpha by transient transfection caused the same proliferative effect and up-regulation of AT1 expression that were observed under hypoxic conditions. In contrast, small interfering RNA targeting HIF-1alpha inhibited hypoxia-induced ACE and AT1 expression. Our studies indicate that the ACE-Ang II-AT1 system serves as a positive feedback loop and fosters FBPA proliferation under hypoxic conditions, with the PI3K-HIF-HRE axis as the central effector pathway. This pathway may thus facilitate vascular remodeling under hypoxic conditions.
Collapse
Affiliation(s)
- Stefanie Krick
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, Justus-Liebig-University Giessen, Giessen, Germany. stefanie
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kirito K, Fox N, Komatsu N, Kaushansky K. Thrombopoietin enhances expression of vascular endothelial growth factor (VEGF) in primitive hematopoietic cells through induction of HIF-1alpha. Blood 2005; 105:4258-63. [PMID: 15705785 PMCID: PMC1895043 DOI: 10.1182/blood-2004-07-2712] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thrombopoietin (TPO), the primary regulator of thrombopoiesis, is also an important, nonredundant mediator of hematopoietic stem cell (HSC) development. For example, following transplantation, HSC expansion is approximately 15-fold more robust in normal than in Tpo(-/-) mice. Vascular endothelial growth factor (VEGF) also plays an important role in HSC development, where it acts in an intracellular autocrine fashion to promote cell survival. Thus, we tested the hypothesis that TPO affects the autocrine production of VEGF to account for its favorable effects on HSCs. We found that VEGF transcripts are reduced in purified sca-1(+)/c-kit(+)/Gr-1(-) marrow cells derived from Tpo(-/-) mice and that TPO induces VEGF transcripts in these primitive hematopoietic cells. Additional studies determined that TPO induces VEGF expression by increasing the level of its primary transcription factor, hypoxia-inducible factor 1alpha (HIF-1alpha), by enhancing its protein stability. Moreover, VEGF expression is important for the TPO effect on primitive hematopoietic cells because blockade of the VEGF receptor with a specific inhibitor substantially blunts TPO-induced growth of single sca-1(+)/c-kit(+)/Gr-1(-) marrow cells in serum-free cultures. Along with previous findings that TPO affects Hox transcription factors that regulate HSC proliferation, these data contribute to our growing understanding of the mechanisms by which a hormone can influence stem cell development.
Collapse
Affiliation(s)
- Keita Kirito
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, San Diego, CA, USA
| | | | | | | |
Collapse
|
38
|
Abstract
Although debated for many years whether haemodynamic or structural changes are more important in the development of diabetic nephropathy, it is now clear that these processes are interwoven and present two sides of one coin. On a molecular level, hyperglycaemia and proteins altered by high blood glucose such as Amadori products and advanced glycation end-products (AGEs) are key players in the development of diabetic nephropathy. Recent evidence suggests that an increase in reactive oxygen species (ROS) formation induced by high glucose-mediated activation of the mitochondrial electron-transport chain is an early event in the development of diabetic complications. A variety of growth factors and cytokines are then induced through complex signal transduction pathways involving protein kinase C, mitogen-activated protein kinases, and the transcription factor NF-kappaB. High glucose, AGEs, and ROS act in concert to induce growth factors and cytokines. Particularly, TGF-beta is important in the development of renal hypertrophy and accumulation of extracellular matrix components. Activation of the renin-angiotensin system by high glucose, mechanical stress, and proteinuria with an increase in local formation of angiotensin II (ANG II) causes many of the pathophysiological changes associated with diabetic nephropathy. In fact, it has been shown that angiotensin II is involved in almost every pathophysiological process implicated in the development of diabetic nephropathy (haemodynamic changes, hypertrophy, extracellular matrix accumulation, growth factor/cytokine induction, ROS formation, podocyte damage, proteinuria, interstitial inflammation). Consequently, blocking these deleterious effects of ANG II is an essential part of every therapeutic regiment to prevent and treat diabetic nephropathy. Recent evidence suggests that regression of diabetic nephropathy could be achieved under certain circumstances.
Collapse
Affiliation(s)
- G Wolf
- Department of Medicine, Division of Nephrology, Osteology and Rheumatology, University of Hamburg, Hamburg, Germany.
| |
Collapse
|