1
|
Chen F, Wang Q, Li Y, Li F, Zhang L, Gu X. TGF-β1-induced apoptosis in retinal endothelial cells is implicated in retinal vein occlusion. Exp Eye Res 2025; 250:110168. [PMID: 39577604 DOI: 10.1016/j.exer.2024.110168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/21/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Retinal vein occlusion (RVO) is a serious vascular condition that impairs vision due to retinal endothelial cell injury and apoptosis. This study aimed to identify key molecular pathways and therapeutic targets involved in RVO pathogenesis. Transcriptomic analysis of the retinal tissues from a mouse RVO model was performed to identify differentially expressed genes and co-expression modules associated with RVO. Protein-protein interaction network analysis pinpointed putative hub genes. In vitro experiments using human retinal microvascular endothelial cells (HRMECs) validated the involvement of identified genes/pathways in apoptosis induced by oxygen-glucose deprivation/reperfusion (OGD/R) and UV exposure. Gene expression was assessed by RT-qPCR, while protein levels and phosphorylation were measured by ELISA and Western blotting. Apoptosis was evaluated using flow cytometry, and reactive oxygen species (ROS) were quantified using a fluorescence-based assay. A total of 392 genes were identified as putatively involved in RVO-associated apoptosis, enriched in MAPK, TGF-β and other signaling pathways. Among top hub genes, TGF-β1 emerged as a central regulator whose expression and signaling (pSmad2/3) increased after OGD/R induction or UV exposure in HRMECs. TGF-β1-induced HRMEC apoptosis was mediated by p38/JNK activation. Similar effects were observed for OGD/R and UV triggering TGF-β1-dependent p38/JNK signaling and apoptosis. Pharmacological inhibition of TGF-β signaling attenuated the apoptotic and oxidative stress responses induced by OGD/R and UV exposure. This study elucidates TGF-β1 as a crucial mediator of retinal endothelial injury through p38/JNK-induced apoptosis, suggesting TGF-β1 pathway inhibition as a potential therapeutic strategy for RVO.
Collapse
Affiliation(s)
- Fengyu Chen
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qi Wang
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yujin Li
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Fen Li
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lin Zhang
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xuezhong Gu
- Department of Hematology, The First People's Hospital of Yunnan Province, Yunnan Province Clinical Research Center for Hematologic Disease, Yunnan Province Clinical Center for Hematologic Disease, Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China; Department of Hematology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Feng W, Guan Z, Ying WZ, Xing D, Ying KE, Sanders PW. Matrix metalloproteinase-9 regulates afferent arteriolar remodeling and function in hypertension-induced kidney disease. Kidney Int 2023; 104:740-753. [PMID: 37423509 PMCID: PMC10854403 DOI: 10.1016/j.kint.2023.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/01/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
This study tested if matrix metalloproteinase (MMP)-9 promoted microvascular pathology that initiates hypertensive (HT) kidney disease in salt-sensitive (SS) Dahl rats. SS rats lacking Mmp9 (Mmp9-/-) and littermate control SS rats were studied after one week on a normotensive 0.3% sodium chloride (Pre-HT SS and Pre-HT Mmp9-/-) or a hypertension-inducing diet containing 4.0% sodium chloride (HT SS and HT Mmp9-/-). Telemetry-monitored blood pressure of both the HT SS and HT Mmp9-/- rats increased and did not differ. Kidney microvessel transforming growth factor-beta 1 (Tgfb1) mRNA did not differ between Pre-HT SS and Pre-HT Mmp9-/- rats, but with hypertension and expression of Mmp9 and Tgfb1 increased in HT SS rats, along with phospho-Smad2 labeling of nuclei of vascular smooth muscle cells, and with peri-arteriolar fibronectin deposition. Loss of MMP-9 prevented hypertension-induced phenotypic transformation of microvascular smooth muscle cells and the expected increased microvascular expression of pro-inflammatory molecules. Loss of MMP-9 in vascular smooth muscle cells in vitro prevented cyclic strain-induced production of active TGF-β1 and phospho-Smad2/3 stimulation. Afferent arteriolar autoregulation was impaired in HT SS rats but not in HT Mmp9-/- rats or the HT SS rats treated with doxycycline, an MMP inhibitor. HT SS but not HT Mmp9-/- rats showed decreased glomerular Wilms Tumor 1 protein-positive cells (a marker of podocytes) along with increased urinary podocin and nephrin mRNA excretion, all indicative of glomerular damage. Thus, our findings support an active role for MMP-9 in a hypertension-induced kidney microvascular remodeling process that promotes glomerular epithelial cell injury in SS rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhengrong Guan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei-Zhong Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kai Er Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA.
| |
Collapse
|
3
|
Fan J, Lin B, Fan M, Niu T, Gao F, Tan B, Du X. Research progress on the mechanism of radiation enteritis. Front Oncol 2022; 12:888962. [PMID: 36132154 PMCID: PMC9483210 DOI: 10.3389/fonc.2022.888962] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/10/2022] [Indexed: 12/12/2022] Open
Abstract
Radiation enteritis (Re) is one of the most common complications of radiation therapy for abdominal tumors. The efficacy of cancer treatment by radiation is often limited by the side effects of Re. Re can be acute or chronic. Treatment of acute Re is essentially symptomatic. However, chronic Re usually requires surgical procedures. The underlying mechanisms of Re are complex and have not yet been elucidated. The purpose of this review is to provide an overview of the pathogenesis of Re. We reviewed the role of intestinal epithelial cells, intestinal stem cells (ISCs), vascular endothelial cells (ECs), intestinal microflora, and other mediators of Re, noting that a better understanding of the pathogenesis of Re may lead to better treatment modalities.
Collapse
Affiliation(s)
- Jinjia Fan
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong, China
| | - Binwei Lin
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
| | - Mi Fan
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong, China
| | - Tintin Niu
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong, China
| | - Feng Gao
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
| | - Bangxian Tan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong, China
| | - Xiaobo Du
- Departmant of Oncology, National Health Commission Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology, Mianyang, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nan Chong, China
- *Correspondence: Xiaobo Du,
| |
Collapse
|
4
|
Ying KE, Feng W, Ying WZ, Li X, Xing D, Sun Y, Chen Y, Sanders PW. Dietary salt initiates redox signaling between endothelium and vascular smooth muscle through NADPH oxidase 4. Redox Biol 2022; 52:102296. [PMID: 35378363 PMCID: PMC8980891 DOI: 10.1016/j.redox.2022.102296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Prevention of phenotype switching of vascular smooth muscle cells is an important determinant of normal vascular physiology. Hydrogen peroxide (H2O2) promotes osteogenic differentiation of vascular smooth muscle cells through expression of Runt related transcription factor 2 (Runx2). In this study, an increase in dietary NaCl increased endothelial H2O2 generation through NOX4, a NAD(P)H oxidase. The production of H2O2 was sufficient to increase Runx2, osteopontin and osteocalcin in adjacent vascular smooth muscle cells from control littermate mice but was inhibited in mice lacking endothelial Nox4. A vascular smooth muscle cell culture model confirmed the direct involvement of the activation of protein kinase B (Akt) with inactivation of FoxO1 and FoxO3a observed in the control mice on the high NaCl diet. The present study also showed a reduction of catalase activity in aortas during high NaCl intake. The findings demonstrated an interesting cell-cell communication in the vascular wall that was initiated with H2O2 production by endothelium and was regulated by dietary NaCl intake. A better understanding of how dietary salt intake alters vascular biology may improve treatment of vascular disease that involves activation of Runx2.
Collapse
Affiliation(s)
- Kai Er Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Wei-Zhong Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Xingsheng Li
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Dongqi Xing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Birmingham Department of Veterans Affairs Health Care System, Birmingham, AL, 35233, USA
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294-0007, USA; Birmingham Department of Veterans Affairs Health Care System, Birmingham, AL, 35233, USA.
| |
Collapse
|
5
|
Kurtz T, Pravenec M, DiCarlo S. Mechanism-based strategies to prevent salt sensitivity and salt-induced hypertension. Clin Sci (Lond) 2022; 136:599-620. [PMID: 35452099 PMCID: PMC9069470 DOI: 10.1042/cs20210566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
High-salt diets are a major cause of hypertension and cardiovascular (CV) disease. Many governments are interested in using food salt reduction programs to reduce the risk for salt-induced increases in blood pressure and CV events. It is assumed that reducing the salt concentration of processed foods will substantially reduce mean salt intake in the general population. However, contrary to expectations, reducing the sodium density of nearly all foods consumed in England by 21% had little or no effect on salt intake in the general population. This may be due to the fact that in England, as in other countries including the U.S.A., mean salt intake is already close to the lower normal physiologic limit for mean salt intake of free-living populations. Thus, mechanism-based strategies for preventing salt-induced increases in blood pressure that do not solely depend on reducing salt intake merit attention. It is now recognized that the initiation of salt-induced increases in blood pressure often involves a combination of normal increases in sodium balance, blood volume and cardiac output together with abnormal vascular resistance responses to increased salt intake. Therefore, preventing either the normal increases in sodium balance and cardiac output, or the abnormal vascular resistance responses to salt, can prevent salt-induced increases in blood pressure. Suboptimal nutrient intake is a common cause of the hemodynamic disturbances mediating salt-induced hypertension. Accordingly, efforts to identify and correct the nutrient deficiencies that promote salt sensitivity hold promise for decreasing population risk of salt-induced hypertension without requiring reductions in salt intake.
Collapse
Affiliation(s)
- Theodore W. Kurtz
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94017-0134, U.S.A
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Stephen E. DiCarlo
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, U.S.A
| |
Collapse
|
6
|
Stimulation of Epithelial Sodium Channels in Endothelial Cells by Bone Morphogenetic Protein-4 Contributes to Salt-Sensitive Hypertension in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3921897. [PMID: 33194000 PMCID: PMC7641672 DOI: 10.1155/2020/3921897] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 01/03/2023]
Abstract
Previous studies have shown that high salt induces artery stiffness by causing endothelial dysfunction via increased sodium influx. We used our unique split-open artery technique combined with protein biochemistry and in vitro measurement of vascular tone to test a hypothesis that bone morphogenetic protein 4 (BMP4) mediates high salt-induced loss of vascular relaxation by stimulating the epithelial sodium channel (ENaC) in endothelial cells. The data show that high salt intake increased BMP4 both in endothelial cells and in the serum and that exogenous BMP4 stimulated ENaC in endothelial cells. The data also show that the stimulation is mediated by p38 mitogen-activated protein kinases (p38 MAPK) and serum and glucocorticoid-regulated kinase 1 (Sgk1)/neural precursor cell expressed developmentally downregulated gene 4-2 (Nedd4-2) (Sgk1/Nedd4-2). Furthermore, BMP4 decreased mesenteric artery relaxation in a benzamil-sensitive manner. These results suggest that high salt intake stimulates endothelial cells to express and release BMP4 and that the released BMP4 reduces artery relaxation by stimulating ENaC in endothelial cells. Therefore, stimulation of ENaC in endothelial cells by BMP4 may serve as another pathway to participate in the complex mechanism of salt-sensitive (SS) hypertension.
Collapse
|
7
|
Feng W, Guan Z, Xing D, Li X, Ying WZ, Remedies CE, Inscho EW, Sanders PW. Avian erythroblastosis virus E26 oncogene homolog-1 (ETS-1) plays a role in renal microvascular pathophysiology in the Dahl salt-sensitive rat. Kidney Int 2020; 97:528-537. [PMID: 31932071 PMCID: PMC7039742 DOI: 10.1016/j.kint.2019.09.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/27/2019] [Accepted: 09/05/2019] [Indexed: 12/29/2022]
Abstract
Prior studies reported that haploinsufficiency of the transcription factor ETS-1 is renoprotective in Dahl salt-sensitive rats, but the mechanism is unclear. Here, we tested whether ETS-1 is involved in hypertension-induced renal microvascular pathology and autoregulatory impairment. Hypertension was induced in salt-sensitive rats and salt-sensitive rats that are heterozygous with 1 wild-type or reference allele of Ets1 (SSEts1+/-) by feeding a diet containing 4% sodium chloride for 1 week. Increases in blood pressure did not differ. However, phosphorylated ETS-1 increased in afferent arterioles of hypertensive salt-sensitive rats, but not in hypertensive SSEts1+/- rats. Afferent arterioles of hypertensive salt-sensitive rats showed increased monocyte chemotactic protein-1 expression and infiltration of CD68 positive monocytes/macrophages. Isolated kidney microvessels showed increased mRNA expression of vascular cell adhesion molecule, intercellular adhesion molecule, P-selectin, fibronectin, transforming growth factor-β, and collagen I in hypertensive salt-sensitive rats compared with hypertensive SSEts1+/- rats. Using the in vitro blood-perfused juxtamedullary nephron preparation, pressure-mediated afferent arteriolar responses were significantly blunted in hypertensive salt-sensitive rats compared to hypertensive SSEts1+/- rats. Over a 65-170 mm Hg pressure range tested baseline arteriolar diameters averaged 15.1 μm and remained between 107% and 89% of baseline diameter in hypertensive salt-sensitive rats vs. 114% and 73% in hypertensive SSEts1+/- rats (significantly different). Thus, ETS-1 participates in renal arteriolar pathology and autoregulation and thereby is involved in hypertension-mediated kidney injury in salt-sensitive rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Zhengrong Guan
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Division of Pulmonary, Allergy & Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xingsheng Li
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei-Zhong Ying
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Colton E Remedies
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Edward W Inscho
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paul W Sanders
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Veterans Affairs Medical Center, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Zahid KR, Raza U, Chen J, Raj UJ, Gou D. Pathobiology of pulmonary artery hypertension: role of long non-coding RNAs. Cardiovasc Res 2020; 116:1937-1947. [PMID: 32109276 DOI: 10.1093/cvr/cvaa050] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/30/2019] [Accepted: 02/25/2020] [Indexed: 12/30/2022] Open
Abstract
Abstract
Pulmonary arterial hypertension (PAH) is a disease with complex pathobiology, significant morbidity and mortality, and remains without a cure. It is characterized by vascular remodelling associated with uncontrolled proliferation of pulmonary artery smooth muscle cells, endothelial cell proliferation and dysfunction, and endothelial-to-mesenchymal transition, leading to narrowing of the vascular lumen, increased vascular resistance and pulmonary arterial pressure, which inevitably results in right heart failure and death. There are multiple molecules and signalling pathways that are involved in the vascular remodelling, including non-coding RNAs, i.e. microRNAs and long non-coding RNAs (lncRNAs). It is only in recent years that the role of lncRNAs in the pathobiology of pulmonary vascular remodelling and right ventricular dysfunction is being vigorously investigated. In this review, we have summarized the current state of knowledge about the role of lncRNAs as key drivers and gatekeepers in regulating major cellular and molecular trafficking involved in the pathogenesis of PAH. In addition, we have discussed the limitations and challenges in translating lncRNA research in vivo and in therapeutic applications of lncRNAs in PAH.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices, Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Khadim Abid Majeed Road, Rawalpindi, Pakistan
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| | - Usha J Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| |
Collapse
|
9
|
Hammad ASA, Ahmed ASF, Heeba GH, Taye A. Heme oxygenase-1 contributes to the protective effect of resveratrol against endothelial dysfunction in STZ-induced diabetes in rats. Life Sci 2019; 239:117065. [PMID: 31751579 DOI: 10.1016/j.lfs.2019.117065] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/28/2022]
Abstract
Endothelial dysfunction is a common complication of diabetes that mainly stems from increased reactive oxygen species, which makes antioxidants of great benefit. Resveratrol (RSV) is an antioxidant that shows protective effects in a variety of disease models where the ameliorative effect appears to be mediated, in part, via heme oxygenase-1 (HO-1) induction. However, the pathophysiological relevance of HO-1 in the ameliorative response of RSV in endothelial dysfunction is not clearly defined. The present study was conducted to investigate whether HO-1 plays a role in diabetes-induced vascular dysfunction. Streptozotocin-diabetic rats were treated with RSV (10 mg/kg) in presence or absence of an HO-1 blocker, Zinc protoporphyrin (ZnPP) to assess vascular function and indicators of disease status. We found that RSV treatment significantly abrogated diabetes induced vascular dysfunction. This improvement was associated with the ability of RSV to decrease oxidative stress markers alongside a reduction in the aortic TGF-β expression, elevation of NOS3 expression and aortic nitrite concentration as well as HO activity. These ameliorative effects were diminished when ZnPP was administered prior to RSV. Our results clearly demonstrate the protective effects of RSV in diabetes-associated endothelial dysfunction and verified a causal role of HO-1 in this setting.
Collapse
Affiliation(s)
- Asmaa S A Hammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt
| | - Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt.
| | - Gehan H Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt
| | - Ashraf Taye
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Egypt
| |
Collapse
|
10
|
Dietary Sodium Restriction Reduces Arterial Stiffness, Vascular TGF-β-Dependent Fibrosis and Marinobufagenin in Young Normotensive Rats. Int J Mol Sci 2018; 19:ijms19103168. [PMID: 30326586 PMCID: PMC6214093 DOI: 10.3390/ijms19103168] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/14/2018] [Accepted: 10/10/2018] [Indexed: 12/27/2022] Open
Abstract
High salt (HS) intake stimulates the production of marinobufagenin (MBG), an endogenous steroidal Na/K-ATPase ligand, which activates profibrotic signaling. HS is accompanied by a blood pressure (BP) increase in salt-sensitive hypertension, but not in normotensive animals. Here, we investigated whether HS stimulates MBG production and activates transforming growth factor-beta (TGF-β) profibrotic signaling in young normotensive rats, and whether these changes can be reversed by reducing salt to a normal salt (NS) level. Three-month old male Sprague–Dawley rats received NS for 4 and 8 weeks (0.5% NaCl; NS4 and NS8), or HS for 4 and 8 weeks (4% NaCl; HS4 and HS8), or HS for 4 weeks followed by NS for 4 weeks (HS4/NS4), n = 8/group. Systolic BP (SBP), pulse wave velocity (PWV), MBG excretion, aortic collagen 1α2, collagen 4α1 and TGF-β, Smad2, Smad3, Fli-1 mRNA, and total collagen abundance were measured at baseline (BL), and on weeks 4 and 8. Statistical analysis was performed using one-way ANOVA. SBP was not affected by HS (125 ± 5 and 126 ± 6 vs. 128 ± 7 mmHg, HS4 and HS8 vs. BL, p > 0.05). HS increased MBG (164 ± 19 vs. 103 ± 19 pmol/24 h/kg, HS4 vs. BL, p < 0.05) and PWV (3.7 ± 0.2 vs. 2.7 ± 0.2 m/s, HS4 vs. NS4, p < 0.05). HS8 was associated with a further increase in MBG and PWV, with an increase in aortic Col1a2 80%), Col4a1 (50%), Tgfb1 (30%), Smad2 (30%) and Smad3 (45%) mRNAs, and aortic wall collagen (180%) vs. NS8 (all p < 0.05). NS following HS downregulated HS-induced factors: in HS4/NS4, the MBG level was 91 ± 12 pmol/24 h/kg (twofold lower than HS8, p < 0.01), PWV was 3.7 ± 0.3 vs. 4.7 ± 0.2 m/s (HS4/NS4 vs. HS8, p < 0.05), aortic wall Tgfb1, Col1a2, Col4a1, Smad2, Smad3 mRNAs, and collagen abundance were reversed by salt reduction to the BL levels (p < 0.05). HS was associated with an activation of TGF-β signaling, aortic fibrosis and aortic stiffness accompanied by an MBG increase in the absence of SBP changes in young normotensive rats. The reduction of dietary salt following HS decreased MBG, PWV, aortic wall collagen and TGF-β. Thus, HS-induced aortic stiffness in normotensive animals occurred in the context of elevated MBG, which may activate SMAD-dependent TGF-β pro-fibrotic signaling. This data suggests that a decrease in salt consumption could help to restore aortic elasticity and diminish the risk of cardiovascular disease by reducing the production of the pro-fibrotic factor MBG.
Collapse
|
11
|
Sebastian A, Cordain L, Frassetto L, Banerjee T, Morris RC. Postulating the major environmental condition resulting in the expression of essential hypertension and its associated cardiovascular diseases: Dietary imprudence in daily selection of foods in respect of their potassium and sodium content resulting in oxidative stress-induced dysfunction of the vascular endothelium, vascular smooth muscle, and perivascular tissues. Med Hypotheses 2018; 119:110-119. [PMID: 30122481 DOI: 10.1016/j.mehy.2018.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 07/29/2018] [Accepted: 08/04/2018] [Indexed: 01/05/2023]
Abstract
We hypothesize that the major environmental determinant of the expression of essential hypertension in America and other Westernized countries is dietary imprudence in respect of the consumption of daily combinations of foods containing suboptimal amounts of potassium and blood pressure-lowering phytochemicals, and supraphysiological amounts of sodium. We offer as premise that Americans on average consume suboptimal amounts of potassium and blood pressure-lowering phytochemicals, and physiologically excessive amounts of sodium, and that such dietary imprudence leads to essential hypertension through oxidative stress-induced vascular endothelial and smooth muscle dysfunction. Such dysfunctions restrict nitric oxide bioavailability, impairing endothelial cell-mediated relaxation of the underlying vascular smooth muscle, initiating and maintaining inappropriately increased peripheral and renal vascular resistance. The biochemical steps from oxidative stress to vascular endothelial dysfunction and its pernicious cardiovascular consequences are well established and generally accepted. The unique aspect of our hypothesis resides in the contention that Americans' habitual consumption of foods resulting in suboptimal dietary intake of potassium and supraphysiological intake of sodium result in oxidative stress, the degree of which, we suggest, will correlate with the degree of deviation of potassium and sodium intake from optimal. Because suboptimal intakes of potassium reflect suboptimal intakes of fruits and vegetables, associated contributors to oxidative stress include suboptimal intakes of magnesium, nitrate, polyphenols, carotenoids, and other phytochemical antioxidants for which fruits and vegetables contain abundant amounts. Currently Americans consume potassium-to-sodium in molar ratios of less than or close to 1.0 and the Institute of Medicine (IOM) recommends a molar ratio of 1.2. Ancestral diets to which we are physiologically adapted range from molar ratios of 5.0 to 10.0 or higher. Accordingly, we suggest that the average American is usually afflicted with oxidative stress-induced vascular endothelial dysfunction, and therefore the standards for normal blood pressure and pre-hypertension often reflect a degree of clinically significant hypertension. In this article, we provide support for those contentions, and indicate the findings that the hypothesis predicts.
Collapse
Affiliation(s)
- Anthony Sebastian
- Division of Nephrology, Department of Medicine, School of Medicine, University of California, San Francisco, CA, USA.
| | - Loren Cordain
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, USA
| | - Lynda Frassetto
- Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Tanushree Banerjee
- University of California, San Francisco, School of Medicine, Department of Medicine, San Francisco, CA, USA
| | - R Curtis Morris
- Division of Nephrology, Department of Medicine, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
12
|
The American Heart Association Scientific Statement on salt sensitivity of blood pressure: Prompting consideration of alternative conceptual frameworks for the pathogenesis of salt sensitivity? J Hypertens 2018. [PMID: 28650918 DOI: 10.1097/hjh.0000000000001458] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
: Recently, the American Heart Association (AHA) published a scientific statement on salt sensitivity of blood pressure which emphasized a decades old conceptual framework for the pathogenesis of this common disorder. Here we examine the extent to which the conceptual framework for salt sensitivity emphasized in the AHA Statement accommodates contemporary findings and views of the broader scientific community on the pathogenesis of salt sensitivity. In addition, we highlight alternative conceptual frameworks and important contemporary theories of salt sensitivity that are little discussed in the AHA Statement. We suggest that greater consideration of conceptual frameworks and theories for salt sensitivity beyond those emphasized in the AHA Statement may help to advance understanding of the pathogenesis of salt-induced increases in blood pressure and, in consequence, may lead to improved approaches to preventing and treating this common disorder.
Collapse
|
13
|
Tao J, Dong B, Yang LX, Xu KQ, Ma S, Lu J. TGF‑β1 expression in adults with non‑traumatic osteonecrosis of the femoral head. Mol Med Rep 2017; 16:9539-9544. [PMID: 29152655 DOI: 10.3892/mmr.2017.7817] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/01/2017] [Indexed: 11/05/2022] Open
Abstract
Non-traumatic osteonecrosis of the femoral head (NONFH) is a common clinical osteoarthropathy. The present study aimed to investigate the association between transforming growth factor β1 (TGF‑β1) and NONFH. Femoral head specimens were collected from patients with NONFH. Patients with traumatic osteonecrosis served as the control. Hematoxylin and eosin (H&E) staining was used to visualize the bone tissue architecture. Immunohistochemistry and densitometry were performed to quantify TGF‑β1 expression in tissues. Flow cytometry was used to detect cluster of differentiation (CD)3+, CD4+ and CD8+ cells, and the ratio of CD4+ to CD8+ T cells in the peripheral blood. H&E staining revealed osteonecrosis, disintegration of osteocytes with karyopyknosis and karyorrhexis, loss of osteocyte lacunae, aberrantly arranged circumferential lamellae, as well as dissolution of the lamellae and subtle osteogenesis in the experimental group, as opposed to the control group. Immunohistochemistry revealed that the expression of TGF‑β1 was significantly reduced in the experimental group (P<0.01). Further, the NONFH group had a decrease in the CD3+ and CD4+ cell populations (P<0.05 and P<0.01, respectively), an increase in the CD8+ cell population (P<0.05), as well as a reduction in the ratio of CD4+ to CD8+ cells (P<0.01). The present study indicated that TGF‑β1 expression was reduced in NONFH. This was associated with impaired repairing capacity of the femoral head and dysregulated subsets of T‑lymphocytes and possible immune functions.
Collapse
Affiliation(s)
- Jun Tao
- Department of Orthopaedics, Huainan No. 1 People's Hospital, Huainan, Anhui 232007, P.R. China
| | - Bin Dong
- Department of Orthopaedics, Huainan No. 1 People's Hospital, Huainan, Anhui 232007, P.R. China
| | - Li-Xin Yang
- Department of Orthopaedics, Huainan No. 1 People's Hospital, Huainan, Anhui 232007, P.R. China
| | - Ke-Qing Xu
- Department of Orthopaedics, Huainan No. 1 People's Hospital, Huainan, Anhui 232007, P.R. China
| | - Shuai Ma
- Department of Orthopaedics, Huainan No. 1 People's Hospital, Huainan, Anhui 232007, P.R. China
| | - Jun Lu
- Department of Pathogen Biology and Immunology, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| |
Collapse
|
14
|
Feng W, Chen B, Xing D, Li X, Fatima H, Jaimes EA, Sanders PW. Haploinsufficiency of the Transcription Factor Ets-1 Is Renoprotective in Dahl Salt-Sensitive Rats. J Am Soc Nephrol 2017; 28:3239-3250. [PMID: 28696249 DOI: 10.1681/asn.2017010085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/22/2017] [Indexed: 11/03/2022] Open
Abstract
Studies using Dahl salt-sensitive (SS) rats identified specific quantitative trait loci that predispose animals to hypertension-associated albuminuria and kidney injury. We explored the hypothesis that kidney-specific expression of the transcription factor Ets-1, located within one of these loci on chromosome 8, mediates glomerular injury in SS hypertension. During the first week on a high-salt diet, SS rats and SS rats with only one functioning Ets-1 gene (ES rats) demonstrated similar increases in BP. However, serum creatinine concentration, albuminuria, and glomerular expression of ETS-1 and two ETS-1 targets, MCP-1 and MMP2, did not increase as substantially in ES rats as in SS rats. Mean BP subsequently increased further in SS rats and remained higher than that of ES rats for the rest of the study. After 4 weeks of high-salt intake, ES rats still showed a lower mean serum creatinine concentration and less albuminuria, as well as less histologic evidence of glomerular injury and kidney fibrosis, than SS rats did. To investigate the specific contribution of renal Ets-1, we transplanted kidneys from ES or SS rats into salt-resistant SS-Chr 13BN/McwiCrl (SS-13BN) rats. Within 10 days on a high-salt diet, BP increased similarly in ES and SS allograft recipients, becoming significantly higher than the BP of control isograft recipients. However, mean serum creatinine concentration and albuminuria remained lower in ES allograft recipients than in SS allograft recipients at 2 weeks, and ES allografts showed less glomerular injury and interstitial fibrosis. In conclusion, reduced renal expression of ETS-1 prevented hypertension-associated kidney injury in SS rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine,
| | - Bo Chen
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dongqi Xing
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine
| | - Xingsheng Li
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine
| | - Huma Fatima
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Edgar A Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Paul W Sanders
- Divisions of Nephrology and Cardiovascular Disease, Departments of Medicine.,Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
15
|
Vanhoutte PM, Zhao Y, Xu A, Leung SWS. Thirty Years of Saying NO: Sources, Fate, Actions, and Misfortunes of the Endothelium-Derived Vasodilator Mediator. Circ Res 2017; 119:375-96. [PMID: 27390338 DOI: 10.1161/circresaha.116.306531] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/02/2016] [Indexed: 12/16/2022]
Abstract
Endothelial cells control vascular tone by releasing nitric oxide (NO) produced by endothelial NO synthase. The activity of endothelial NO synthase is modulated by the calcium concentration and by post-translational modifications (eg, phosphorylation). When NO reaches vascular smooth muscle, soluble guanylyl cyclase is its primary target producing cGMP. NO production is stimulated by circulating substances (eg, catecholamines), platelet products (eg, serotonin), autacoids formed in (eg, bradykinin) or near (eg, adiponectin) the vascular wall and physical factors (eg, shear stress). NO dysfunction can be caused, alone or in combination, by abnormal coupling of endothelial cell membrane receptors, insufficient supply of substrate (l-arginine) or cofactors (tetrahydrobiopterin), endogenous inhibitors (asymmetrical dimethyl arginine), reduced expression/presence/dimerization of endothelial NO synthase, inhibition of its enzymatic activity, accelerated disposition of NO by reactive oxygen species and abnormal responses (eg, biased soluble guanylyl cyclase activity producing cyclic inosine monophosphate) of the vascular smooth muscle. Major culprits causing endothelial dysfunction, irrespective of the underlying pathological process (aging, obesity, diabetes mellitus, and hypertension), include stimulation of mineralocorticoid receptors, activation of endothelial Rho-kinase, augmented presence of asymmetrical dimethyl arginine, and exaggerated oxidative stress. Genetic and pharmacological interventions improve dysfunctional NO-mediated vasodilatations if protecting the supply of substrate and cofactors for endothelial NO synthase, preserving the presence and activity of the enzyme and reducing reactive oxygen species generation. Common achievers of such improvement include maintained levels of estrogens and increased production of adiponectin and induction of silent mating-type information regulation 2 homologue 1. Obviously, endothelium-dependent relaxations are not the only beneficial action of NO in the vascular wall. Thus, reduced NO-mediated responses precede and initiate the atherosclerotic process.
Collapse
Affiliation(s)
- Paul M Vanhoutte
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Yingzi Zhao
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Susan W S Leung
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Abstract
Salt resistance/sensitivity refers specifically to the effect of dietary sodium chloride (salt) intake on BP. Increased dietary salt intake promotes an early and uniform expansion of extracellular fluid volume and increased cardiac output. To compensate for these hemodynamic changes and maintain constant BP in salt resistance, renal and peripheral vascular resistance falls and is associated with an increase in production of nitric oxide. In contrast, the decline in peripheral vascular resistance and the increase in nitric oxide are impaired or absent in salt sensitivity, promoting an increase in BP in these individuals. Endothelial dysfunction may pose a particularly significant risk factor in the development of salt sensitivity and subsequent hypertension. Vulnerable salt-sensitive populations may have in common underlying endothelial dysfunction due to genetic or environmental influences. These individuals may be very sensitive to the hemodynamic stress of increased effective blood volume, setting in motion untoward molecular and biochemical events that lead to overproduction of TGF-β, oxidative stress, and limited bioavailable nitric oxide. Finally, chronic high-salt ingestion produces endothelial dysfunction, even in salt-resistant subjects. Thus, the complex syndrome of salt sensitivity may be a function of the endothelium, which is integrally involved in the vascular responses to high salt intake.
Collapse
Affiliation(s)
| | - Louis J Dell'Italia
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
| | - Paul W Sanders
- Departments of Medicine and
- Department of Medicine, Veterans Affairs Medical Center, Birmingham, Alabama
- Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
17
|
Randell A, Chokshi K, Kane B, Chang H, Naiel S, Dickhout JG, Daneshtalab N. Alterations to the middle cerebral artery of the hypertensive-arthritic rat model potentiates intracerebral hemorrhage. PeerJ 2016; 4:e2608. [PMID: 27833798 PMCID: PMC5101607 DOI: 10.7717/peerj.2608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/25/2016] [Indexed: 12/13/2022] Open
Abstract
Aims We have recently created an age-dependent hypertensive-mono-arthritic animal model from the stroke-resistant spontaneously hypertensive rat to model populations with autoimmune disease who are hypertensive and are prone to stroke. The model exhibits signs of hemorrhagic stroke (HS) subsequent to chronic inflammation and hypertension. HS is also associated with the inability of middle cerebral arteries to undergo pressure dependent constriction (PDC). We investigated alterations in the cerebrovasculature of our hypertensive mono-arthritic animals that develop stroke. Main Methods Animals were fed either a high salt diet (HSD) (4% NaCl) or Purina chow (0.58% NaCl) from weaning. Complete Freund’s Adjuvant (CFA) was injected into the left hind paw at 21–28 weeks; controls received saline and histological and functional studies were performed. Results Brain damage was more prominent with the high salt, with inflammation exacerbating the damage. High salt alone significantly decreased middle cerebral artery’s (MCA’s) ability to undergo PDC. Inflammation significantly decreased the ability of cerebrovasculature to respond to pressure step in the regular salt diet. The responses to vasoactive peptides were also significantly attenuated in both inflamed groups regardless of diet. Conclusion Induction of chronic systemic inflammation increases brain damage, and affect the MCA’s vasogenic function, decreasing its ability to respond to intraluminal pressure. HSD further exacerbates organ damage associated with chronic inflammation, further compromising cerebrovascular function, and likely increasing the incidence of intracerebral hemorrhage and injury.
Collapse
Affiliation(s)
- Amy Randell
- School of Pharmacy, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| | - Killol Chokshi
- School of Pharmacy, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| | - Brittany Kane
- School of Pharmacy, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| | - Hilary Chang
- School of Pharmacy, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| | - Safaa Naiel
- Department of Medicine, Division of Nephrology, McMaster University , Hamilton, Ontario , Canada
| | - Jeffrey G Dickhout
- Department of Medicine, Division of Nephrology, McMaster University , Hamilton, Ontario , Canada
| | - Noriko Daneshtalab
- School of Pharmacy, Memorial University of Newfoundland , St. John's, Newfoundland and Labrador , Canada
| |
Collapse
|
18
|
Pathak R, Wang J, Garg S, Aykin-Burns N, Petersen KU, Hauer-Jensen M. Recombinant Thrombomodulin (Solulin) Ameliorates Early Intestinal Radiation Toxicity in a Preclinical Rat Model. Radiat Res 2016; 186:112-20. [PMID: 27459702 DOI: 10.1667/rr14408.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intestinal radiation toxicity occurs during and after abdominopelvic radiotherapy. Endothelial cells play a significant role in modulating radiation-induced intestinal damage. We demonstrated that the endothelial cell surface receptor thrombomodulin (TM), a protein with anticoagulant, anti-inflammatory and antioxidant properties, mitigates radiation-induced lethality in mice. The goal of this study was to determine whether recombinant TM (Solulin) can protect the intestine from toxicity in a clinically relevant rat model. A 4 cm loop of rat small bowel was exposed to fractionated 5 Gy X radiation for 9 consecutive days. The animals were randomly assigned to receive daily subcutaneous injections of vehicle or Solulin (3 mg/kg/day or 10 mg/kg/day) for 27 days starting 4 days before irradiation. Early intestinal injury was assessed two weeks after irradiation by quantitative histology, morphometry, immunohistochemistry and luminol bioluminescence imaging. Solulin treatment significantly ameliorated intestinal radiation injury, made evident by a decrease in myeloperoxidase (MPO) activity, transforming growth factor beta (TGF-β) immunoreactivity, collagen-I deposition, radiation injury score (RIS) and intestinal serosal thickening. These findings indicate the need for further development of Solulin as a prophylactic and/or therapeutic agent to mitigate radiation-induced intestinal damage.
Collapse
Affiliation(s)
- Rupak Pathak
- a Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Junru Wang
- a Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sarita Garg
- a Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nukhet Aykin-Burns
- a Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Martin Hauer-Jensen
- a Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas;,c Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|