1
|
Falero-Diaz G, Barboza CDA, Vazquez-Padron RI, Velazquez OC, Lassance-Soares RM. Loss of c-Kit in Endothelial Cells Protects against Hindlimb Ischemia. Biomedicines 2024; 12:1358. [PMID: 38927565 PMCID: PMC11201387 DOI: 10.3390/biomedicines12061358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is the end stage of peripheral artery disease (PAD), and around 30% of CLI patients are ineligible for current treatments. The angiogenic benefits of c-Kit have been reported in the ischemia scenario; however, the present study demonstrates the effects of specific endothelial c-Kit signaling in arteriogenesis during hindlimb ischemia. METHODS We created conditional knockout mouse models that decrease c-Kit (c-Kit VE-Cadherin CreERT2-c-Kit) or its ligand (SCF VE-Cadherin CreERT2-SCF) specifically in endothelial cells (ECs) after tamoxifen treatment. These mice and a control group (wild-type VE-Cadherin CreERT2-WT) were subjected to hindlimb ischemia or aortic crush to evaluate perfusion/arteriogenesis and endothelial barrier permeability, respectively. RESULTS Our data confirmed the lower gene expression of c-Kit and SCF in the ECs of c-Kit and SCF mice, respectively. In addition, we confirmed the lower percentage of ECs positive for c-Kit in c-Kit mice. Further, we found that c-Kit and SCF mice had better limb perfusion and arteriogenesis compared to WT mice. We also demonstrated that c-Kit and SCF mice had a preserved endothelial barrier after aortic crush compared to WT. CONCLUSIONS Our data demonstrate the deleterious effects of endothelial SCF/c-Kit signaling on arteriogenesis and endothelial barrier integrity.
Collapse
Affiliation(s)
- Gustavo Falero-Diaz
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Catarina de A. Barboza
- Department of Medicine, Miller School of Medicine, University of Miami, 1580 NW 10th Ave, Batchelor Building, Miami, FL 33136, USA;
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Omaida C. Velazquez
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Roberta M. Lassance-Soares
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| |
Collapse
|
2
|
Du J, Li Y, Jia X, Kong X, Liang L, Wang D, Li A, Chen Q, Su H, Li W, Xu D. Elevated c-kit expression in failed autologous arteriovenous fistulas in end-stage renal disease patients. J Vasc Access 2024; 25:423-431. [PMID: 35855563 DOI: 10.1177/11297298221112541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Serum stem cell factor is elevated in end-stage renal disease (ESRD) patients. This study aimed to investigate the expression of the c-kit receptor, which is the specific membrane receptor of stem cell factor, in failed autologous arteriovenous fistulas (AVFs) in end-stage renal disease patients. METHODS A total of 14 ESRD patients with initial AVFs creation and 16 ESRD patients with reconstruction were enrolled in this study. Hematoxylin and eosin (H&E) and elastic Verhoeff-Van Gieson (EVG) staining were used for histomorphometric analyses. Immunohistochemistry was used to examine the expression of c-kit in the intima, and a correlation analysis was performed with the intimal area and the percentage of area stenosis. A double-label immunofluorescence method was used to explore the colocalization of c-kit with α-smooth muscle actin (α-SMA) and CD31. The expression of c-kit and the related PI3K/Akt signaling axis, including PI3K, P-PI3K, Akt, P-Akt473, P-Akt308, and mTOR, was measured by western blotting. RESULTS Internal elastic lamina (IEL) area, intimal area, percentage of area stenosis, and average optical density (AOD) of c-kit in the intima were significantly higher in the failed group than in the preoperative group (p ⩽ 0.001). The AOD of c-kit in the intima was positively correlated with the intimal area and the percentage of stenosis (intimal area: R = 0.744, p < 0.001; the percentage of stenosis: R = 0.923, p < 0.001). C-kit colocalized with α-SMA but not with CD31 in studies of c-kit target cells. Moreover, the levels of c-kit and P-PI3K, P-Akt473 and mTOR in the PI3K/Akt axis were also higher in the failed group than in the initial group (p < 0.05). CONCLUSIONS C-kit and related proteins associated with the PI3K/Akt pathway were elevated in failed AVFs among ESRD patients and that the expression level of c-kit in the intima correlates with the degree of AVF stenosis.
Collapse
Affiliation(s)
- Jing Du
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
- Department of Blood Purification Center, Weifang People's Hospital, Weifang, Shandong, China
| | - Yang Li
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| | - Xiaoyan Jia
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| | - Xianglei Kong
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| | - Liming Liang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| | - Dongmei Wang
- Department of Blood Purification Center, Weifang People's Hospital, Weifang, Shandong, China
| | - Anzhuang Li
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Qinlan Chen
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Hong Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenbin Li
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| | - Dongmei Xu
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Nephrology Research Institute of Shandong Province, Jinan, Shandong, China
| |
Collapse
|
3
|
Li J, Liu F, Mo K, Ni H, Yin Y. Effects of weaning on intestinal longitudinal muscle-myenteric plexus function in piglets. SCIENCE CHINA. LIFE SCIENCES 2024; 67:379-390. [PMID: 37824029 DOI: 10.1007/s11427-022-2391-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/18/2023] [Indexed: 10/13/2023]
Abstract
Weaning piglets usually suffer from severe diarrhea (commonly known as postweaning diarrhea, PWD) along with intestinal motility disorder. Intestinal peristalsis is mainly regulated by the longitudinal muscle-myenteric plexus (LM-MP). To understand the relationship between intestinal LM-MP function and the development of PWD, we compared the intestinal electrical activity, and the transcriptional profile of the LM-MP between 21-day-old piglets (just weaned, n=7) and 24-day-old piglets (suffered the most severe weaning stress, n=7). The results showed that 24-day-old piglets exhibited different degrees of diarrhea. A significant increase in the slow-wave frequency in the ileum and colon was observed in 24-day-old piglets, while c-kit expression in the intestinal LM-MPs was significantly decreased, indicating that PWD caused by elevated slow-wave frequency may be associated with loss of c-kit. The real-time quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) showed that intestinal LM-MPs in 24-day-old piglets may undergo inflammation and oxidative stress. Significant increases in 8-hydroxy-2'-deoxyguanosine and decreases in thioredoxin suggest that weaning may lead to DNA damage in the LM-MP of 24-day-old piglets. In addition, activating transcription factor 3 was significantly upregulated, indicating nerve damage in the LM-MP of 24-day-old piglets. The transcriptomic results showed that most of the differentially expressed genes in the ileal LM-MP after weaning were downregulated and closely related to the cell cycle process. Subsequent RT-qPCR analysis showed that the relative expression of p21 was upregulated, while the expression of cyclin A2, cyclin B1, and proliferating cell nuclear antigen was downregulated in the ileal and colonic LM-MP of 24-day-old piglets, suggesting that weaning may inhibit cell proliferation and cause G1/S cell cycle arrest in ileal and colonic LM-MP. In conclusion, weaning may lead to cell cycle arrest by causing DNA damage in the LM-MP, impairing intestinal motility regulation, and ultimately leading to diarrhea in piglets.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Fenfen Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Kaibin Mo
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hengjia Ni
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| |
Collapse
|
4
|
Steffen BT, Pankow JS, Norby FL, Lutsey PL, Demmer RT, Guan W, Pankratz N, Li A, Liu G, Matsushita K, Tin A, Tang W. Proteomics Analysis of Genetic Liability of Abdominal Aortic Aneurysm Identifies Plasma Neogenin and Kit Ligand: The ARIC Study. Arterioscler Thromb Vasc Biol 2023; 43:367-378. [PMID: 36579647 PMCID: PMC9995137 DOI: 10.1161/atvbaha.122.317984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Genome-wide association studies have reported 23 gene loci related to abdominal aortic aneurysm (AAA)-a potentially lethal condition characterized by a weakened dilated vessel wall. This study aimed to identify proteomic signatures and pathways related to these risk loci to better characterize AAA genetic susceptibility. METHODS Plasma concentrations of 4870 proteins were determined using a DNA aptamer-based array. Linear regression analysis estimated the associations between the 23 risk alleles and plasma protein levels with adjustments for potential confounders in a race-stratified analysis of 1671 Black and 7241 White participants. Significant proteins were then evaluated for their prediction of clinical AAA (454 AAA events in 11 064 individuals), and those significantly associated with AAA were further interrogated using Mendelian randomization analysis. RESULTS Risk variants proximal to PSRC1-CELSR2-SORT1, PCIF1-ZNF335-MMP9, RP11-136O12.2/TRIB1, ZNF259/APOA5, IL6R, PCSK9, LPA, and APOE were associated with 118 plasma proteins in Whites and 59 were replicated in Black participants. Novel associations with clinical AAA incidence were observed for kit ligand (HR, 0.59 [95% CI, 0.42-0.82] for top versus first quintiles) and neogenin (HR, 0.64 [95% CI, 0.46-0.88]) over a median 21.2-year follow-up; neogenin was also associated with ultrasound-detected asymptomatic AAA (N=4295; 57 asymptomatic AAA cases). Mendelian randomization inverse variance weighted estimates suggested that AAA risk is promoted by lower levels of kit ligand (OR per SD=0.67; P=1.4×10-5) and neogenin (OR per SD=0.50; P=0.03). CONCLUSIONS Low levels of neogenin and kit ligand may be novel risk factors for AAA development in potentially causal pathways. These findings provide insights and potential targets to reduce AAA susceptibility.
Collapse
Affiliation(s)
- Brian T. Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Division of Health Data Science, Department of Surgery, University of Minnesota, Minneapolis, MN 55455
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Faye L. Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA 90048
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, 55455
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Aixin Li
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| | - Guning Liu
- Division of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center, School of Public Health, Houston, TX 77030
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD 21205
| | - Adrienne Tin
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN 55454
| |
Collapse
|
5
|
Xu J, Wang J, Chen Y, Hou Y, Hu J, Wang G. Recent advances of natural and bioengineered extracellular vesicles and their application in vascular regeneration. Regen Biomater 2022; 9:rbac064. [PMID: 36176713 PMCID: PMC9514852 DOI: 10.1093/rb/rbac064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
The progression of cardiovascular diseases such as atherosclerosis and myocardial infarction leads to serious vascular injury, highlighting the urgent need for targeted regenerative therapy. Extracellular vesicles (EVs) composed of a lipid bilayer containing nuclear and cytosolic materials are relevant to the progression of cardiovascular diseases. Moreover, EVs can deliver bioactive cargo in pathological cardiovascular and regulate the biological function of recipient cells, such as inflammation, proliferation, angiogenesis and polarization. However, because the targeting and bioactivity of natural EVs are subject to several limitations, bioengineered EVs have achieved wide advancements in biomedicine. Bioengineered EVs involve three main ways to acquire including (i) modification of the EVs after isolation; (ii) modification of producer cells before EVs’ isolation; (iii) synthesize EVs using natural or modified cell membranes, and encapsulating drugs or bioactive molecules into EVs. In this review, we first summarize the cardiovascular injury-related disease and describe the role of different cells and EVs in vascular regeneration. We also discuss the application of bioengineered EVs from different producer cells to cardiovascular diseases. Finally, we summarize the surface modification on EVs which can specifically target abnormal cells in injured vascular.
Collapse
Affiliation(s)
| | | | - Yidan Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Yuanfang Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering Modern Life Science Experiment Teaching Center of Bioengineering College, Chongqing University, Chongqing 400030, China
| | - Jianjun Hu
- Correspondence address. E-mail: (G.W.); (J.H.)
| | - Guixue Wang
- Correspondence address. E-mail: (G.W.); (J.H.)
| |
Collapse
|
6
|
c-Kit expression in smooth muscle cells reduces atherosclerosis burden in hyperlipidemic mice. Atherosclerosis 2021; 324:133-140. [PMID: 33781566 DOI: 10.1016/j.atherosclerosis.2021.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 01/13/2021] [Accepted: 03/04/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Increased receptor tyrosine kinase (RTK) activity has been historically linked to atherosclerosis. Paradoxically, we recently found that global deficiency in c-Kit function increased atherosclerosis in hyperlipidemic mice. This study aimed to investigate if such unusual atheroprotective phenotype depends upon c-Kit's function in smooth muscle cells (SMC). METHODS We studied atherosclerosis in a SMC-specific conditional knockout mice (KitSMC) and control littermate. Tamoxifen (TAM) and vehicle treated mice were fed high fat diet for 16 weeks before atherosclerosis assessment in the whole aorta using oil red staining. Smooth muscle cells were traced within the aortic sinus of conditional c-Kit tracing mice (KitSMC eYFP) and their control littermates (KitWT eYFP) by immunofluorescent confocal microscopy. We then performed RNA sequencing on primary SMC from c-Kit deficient and control mice, and identified significantly altered genes and pathways as a result of c-Kit deficiency in SMC. RESULTS Atherosclerosis significantly increased in KitSMC mice with respect to control groups. In addition, the loss of c-Kit in SMC increased plaque size and necrotic core area in the aortic sinus of hyperlipidemic mice. Smooth muscle cells from KitSMC eYFP mice were more prone to migrate and express foam cell markers (e.g., Mac2 and MCAM) than those from control littermate animals. RNAseq analysis showed a significant upregulation in genes associated with cell proliferation, migration, lipid metabolism, and inflammation secondary to the loss of Kit function in primary SMCs. CONCLUSIONS Loss of c-Kit increases SMC migration, proliferation, and expression of foam cell markers in atherosclerotic plaques from hyperlipidemic mice.
Collapse
|
7
|
Yan W, Li T, Yin T, Hou Z, Qu K, Wang N, Durkan C, Dong L, Qiu J, Gregersen H, Wang G. M2 macrophage-derived exosomes promote the c-KIT phenotype of vascular smooth muscle cells during vascular tissue repair after intravascular stent implantation. Theranostics 2020; 10:10712-10728. [PMID: 32929376 PMCID: PMC7482821 DOI: 10.7150/thno.46143] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: For intravascular stent implantation to be successful, the processes of vascular tissue repair and therapy are considered to be critical. However, the mechanisms underlying the eventual fate of vascular smooth muscle cells (VSMCs) during vascular tissue repair remains elusive. In this study, we hypothesized that M2 macrophage-derived exosomes to mediate cell-to-cell crosstalk and induce dedifferentiation phenotypes in VSMCs. Methods: In vivo, 316L bare metal stents (BMS) were implanted from the left iliac artery into the abdominal aorta of 12-week-old male Sprague-Dawley (SD) rats for 7 and 28 days. Hematoxylin and eosin (HE) were used to stain the neointimal lesions. En-face immunofluorescence staining of smooth muscle 22 alpha (SM22α) and CD68 showed the rat aorta smooth muscle cells (RASMCs) and macrophages. Immunohistochemical staining of total galactose-specific lectin 3 (MAC-2) and total chitinase 3-like 3 (YM-1) showed the total macrophages and M2 macrophages. In vitro, exosomes derived from IL-4+IL-13-treated macrophages (M2Es) were isolated by ultracentrifugation and characterized based on their specific morphology. Ki-67 staining was conducted to assess the effects of the M2Es on the proliferation of RASMCs. An atomic force microscope (AFM) was used to detect the stiffness of the VSMCs. GW4869 was used to inhibit exosome release. RNA-seq was performed to determine the mRNA profiles of the RASMCs and M2Es-treated RASMCs. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of the mRNAs. Western blotting was used to detect the candidate protein expression levels. T-5224 was used to inhibit the DNA binding activity of AP-1 in RASMCs. Results: M2Es promote c-KIT expression and softening of nearby VSMCs, hence accelerating the vascular tissue repair process. VSMCs co-cultured in vitro with M2 macrophages presented an increased capacity for de-differentiation and softening, which was exosome dependent. In addition, the isolated M2Es helped to promote VSMC dedifferentiation and softening. Furthermore, the M2Es enhanced vascular tissue repair potency by upregulation of VSMCs c-KIT expression via activation of the c-Jun/activator protein 1 (AP-1) signaling pathway. Conclusions: The findings of this study emphasize the prominent role of M2Es during VSMC dedifferentiation and vascular tissue repair via activation of the c-Jun/AP-1 signaling pathway, which has a profound impact on the therapeutic strategies of coronary stenting techniques.
Collapse
|
8
|
Hernandez DR, Rojas MG, Martinez L, Rodriguez BL, Zigmond ZM, Vazquez-Padron RI, Lassance-Soares RM. c-Kit deficiency impairs nitric oxide signaling in smooth muscle cells. Biochem Biophys Res Commun 2019; 518:227-232. [PMID: 31416613 PMCID: PMC6732002 DOI: 10.1016/j.bbrc.2019.08.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Receptor tyrosine kinases have been implicated in various vascular remodeling processes and cardiovascular disease. However, their role in the regulation of vascular tone is poorly understood. Herein, we evaluate the contribution of c-Kit signaling to vasoactive responses. METHODS The vascular reactivity of mesenteric arteries was assessed under isobaric conditions in c-Kit deficient (KitW/W-v) and littermate control mice (Kit+/+) using pressure myography. Protein levels of soluble guanylyl cyclase beta 1 (sGCβ1) were quantified by Western blot. Mean arterial pressure was measured after high salt (8% NaCl) diet treatment using the tail-cuff method. RESULTS Smooth muscle cells (SMCs) from c-Kit deficient mice showed a 5-fold downregulation of sGCβ1 compared to controls. Endothelium-dependent relaxation of mesenteric arteries demonstrated a predominance of prostanoid vs. nitric oxide (NO) signaling in both animal groups. The dependence on prostanoid-induced dilation was higher in c-Kit mutant mice than in controls, as indicated by a significant impairment in vasorelaxation with indomethacin with respect to the latter. Endothelium-independent relaxation showed significant dysfunction of NO signaling in c-Kit deficient SMCs compared to controls. Mesenteric artery dilation was rescued by addition of a cGMP analog, but not with a NO donor, indicating a deficiency in cGMP production in c-Kit deficient SMCs. Finally, c-Kit deficient mice developed higher blood pressure on an 8% NaCl diet compared to their control littermates. CONCLUSION c-Kit deficiency inhibits NO signaling in SMCs. The existence of this c-Kit/sGC signaling axis may be relevant for vascular reactivity and remodeling.
Collapse
Affiliation(s)
- Diana R Hernandez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Miguel G Rojas
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Laisel Martinez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Boris L Rodriguez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Zachary M Zigmond
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | | | | |
Collapse
|
9
|
Song L, Zigmond ZM, Martinez L, Lassance-Soares RM, Macias AE, Velazquez OC, Liu ZJ, Salama A, Webster KA, Vazquez-Padron RI. c-Kit suppresses atherosclerosis in hyperlipidemic mice. Am J Physiol Heart Circ Physiol 2019; 317:H867-H876. [PMID: 31441677 PMCID: PMC6843012 DOI: 10.1152/ajpheart.00062.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/18/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is the most common underlying cause of cardiovascular morbidity and mortality worldwide. c-Kit (CD117) is a member of the receptor tyrosine kinase family, which regulates differentiation, proliferation, and survival of multiple cell types. Recent studies have shown that c-Kit and its ligand stem cell factor (SCF) are present in arterial endothelial cells and smooth muscle cells (SMCs). The role of c-Kit in cardiovascular disease remains unclear. The aim of the current study is to determine the role of c-Kit in atherogenesis. For this purpose, atherosclerotic plaques were quantified in c-Kit-deficient mice (KitMut) after they were fed a high-fat diet (HFD) for 16 wk. KitMut mice demonstrated substantially greater atherosclerosis compared with control (KitWT) littermates (P < 0.01). Transplantation of c-Kit-positive bone marrow cells into KitMut mice failed to rescue the atherogenic phenotype, an indication that increased atherosclerosis was associated with reduced arterial c-Kit. To investigate the mechanism, SMC organization and morphology were analyzed in the aorta by histopathology and electron microscopy. SMCs were more abundant, disorganized, and vacuolated in aortas of c-Kit mutant mice compared with controls (P < 0.05). Markers of the "contractile" SMC phenotype (calponin, SM22α) were downregulated with pharmacological and genetic c-Kit inhibition (P < 0.05). The absence of c-Kit increased lipid accumulation and significantly reduced the expression of the ATP-binding cassette transporter G1 (ABCG1) necessary for lipid efflux in SMCs. Reconstitution of c-Kit in cultured KitMut SMCs resulted in increased spindle-shaped morphology, reduced proliferation, and elevated levels of contractile markers, all indicators of their restored contractile phenotype (P < 0.05).NEW & NOTEWORTHY This study describes the novel vasculoprotective role of c-Kit against atherosclerosis and its function in the preservation of the SMC contractile phenotype.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Animals
- Aorta/metabolism
- Aorta/ultrastructure
- Aortic Diseases/etiology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Disease Models, Animal
- Foam Cells/metabolism
- Foam Cells/pathology
- Humans
- Hyperlipidemias/complications
- Hyperlipidemias/metabolism
- Mice, Knockout, ApoE
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Mutation
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/ultrastructure
- Phenotype
- Plaque, Atherosclerotic
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-kit/metabolism
- Signal Transduction
- Calponins
Collapse
Affiliation(s)
- Lei Song
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Zachary M Zigmond
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Laisel Martinez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | | | - Alejandro E Macias
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | - Omaida C Velazquez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | - Zhao-Jun Liu
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | - Alghidak Salama
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Roberto I Vazquez-Padron
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
10
|
Debreczeni ML, Németh Z, Kajdácsi E, Schwaner E, Makó V, Masszi A, Doleschall Z, Rigó J, Walter FR, Deli MA, Pál G, Dobó J, Gál P, Cervenak L. MASP-1 Increases Endothelial Permeability. Front Immunol 2019; 10:991. [PMID: 31130964 PMCID: PMC6509239 DOI: 10.3389/fimmu.2019.00991] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/17/2019] [Indexed: 01/13/2023] Open
Abstract
Pathologically increased vascular permeability is an important dysfunction in the pathomechanism of life-threatening conditions, such as sepsis, ischemia/reperfusion, or hereditary angioedema (HAE), diseases accompanied by uncontrolled activation of the complement system. HAE for example is caused by the deficiency of C1-inhibitor (the main regulator of early complement activation), which leads to edematous attacks threatening with circulatory collapse. We have previously reported that endothelial cells become activated during HAE attacks. A natural target of C1-inhibitor is mannan-binding lectin-associated serine protease-1 (MASP-1), a multifunctional serine protease, which plays a key role in the activation of complement lectin pathway. We have previously shown that MASP-1 induces the pro-inflammatory activation of endothelial cells and in this study we investigated whether MASP-1 can directly affect endothelial permeability. All experiments were performed on human umbilical vein endothelial cells (HUVECs). Real-time micro electric sensing revealed that MASP-1 decreases the impedance of HUVEC monolayers and in a recently developed permeability test (XperT), MASP-1 dose-dependently increased endothelial paracellular transport. We show that protease activated receptor-1 mediated intracellular Ca2+-mobilization, Rho-kinase activation dependent myosin light chain (MLC) phosphorylation, cytoskeletal actin rearrangement, and disruption of interendothelial junctions are underlying this phenomenon. Furthermore, in a whole-transcriptome microarray analysis MASP-1 significantly changed the expression of 25 permeability-related genes in HUVECs-for example it up-regulated bradykinin B2 receptor expression. According to our results, MASP-1 has potent permeability increasing effects. During infections or injuries MASP-1 may help eliminate the microbes and/or tissue debris by enhancing the extravasation of soluble and cellular components of the immune system, however, it may also play a role in the pathomechanism of diseases, where edema formation and complement lectin pathway activation are simultaneously present. Our findings also raise the possibility that MASP-1 may be a promising target of anti-edema drug development.
Collapse
Affiliation(s)
- Márta L. Debreczeni
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Németh
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Erika Kajdácsi
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Endre Schwaner
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Veronika Makó
- MTA-SE Research Group of Immunology and Hematology, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - András Masszi
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltán Doleschall
- Department of Pathogenetics, National Institute of Oncology, Budapest, Hungary
| | - János Rigó
- First Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Fruzsina R. Walter
- Biological Research Centre, Institute of Biophysics, Hungarian Academy of Sciences, Szeged, Hungary
| | - Mária A. Deli
- Biological Research Centre, Institute of Biophysics, Hungarian Academy of Sciences, Szeged, Hungary
| | - Gábor Pál
- Department of Biochemistry, Eötvös Loránd University, Budapest, Hungary
| | - József Dobó
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Péter Gál
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Cervenak
- Research Laboratory, 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
11
|
Baek SJ, Chun JM, Kang TW, Seo YS, Kim SB, Seong B, Jang Y, Shin GH, Kim C. Identification of Epigenetic Mechanisms Involved in the Anti-Asthmatic Effects of Descurainia sophia Seed Extract Based on a Multi-Omics Approach. Molecules 2018; 23:molecules23112879. [PMID: 30400597 PMCID: PMC6278437 DOI: 10.3390/molecules23112879] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 01/17/2023] Open
Abstract
Asthma, a heterogeneous disease of the airways, is common around the world, but little is known about the molecular mechanisms underlying the interactions between DNA methylation and gene expression in relation to this disease. The seeds of Descurainia sophia are traditionally used to treat coughs, asthma and edema, but their effects on asthma have not been investigated by multi-omics analysis. We undertook this study to assess the epigenetic effects of ethanol extract of D. sophia seeds (DSE) in an ovalbumin (OVA)-induced mouse model of asthma. We profiled genome-wide DNA methylation by Methyl-seq and characterized the transcriptome by RNA-seq in mouse lung tissue under three conditions: saline control, OVA-induced, and DSE-treated. In total, 1995 differentially methylated regions (DMRs) were identified in association with anti-asthmatic effects, most in promoter and coding regions. Among them, 25 DMRs were negatively correlated with the expression of the corresponding 18 genes. These genes were related to development of the lung, respiratory tube and respiratory system. Our findings provide insights into the anti-asthmatic effects of D. sophia seeds and reveal the epigenetic targets of anti-inflammatory processes in mice.
Collapse
Affiliation(s)
- Su-Jin Baek
- Bioinformatics Group, R&D Center, Insilicogen Corporation, 35, Techno 9-ro, Yuseong-gu, Daejeon 34027, Korea.
| | - Jin Mi Chun
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| | - Tae-Wook Kang
- Bioinformatics Group, R&D Center, Insilicogen Corporation, 35, Techno 9-ro, Yuseong-gu, Daejeon 34027, Korea.
| | - Yun-Soo Seo
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| | - Sung-Bae Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| | - Boseok Seong
- Future Medicine Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| | - Yunji Jang
- Future Medicine Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| | - Ga-Hee Shin
- Bioinformatics Group, R&D Center, Insilicogen Corporation, 35, Techno 9-ro, Yuseong-gu, Daejeon 34027, Korea.
| | - Chul Kim
- Future Medicine Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea.
| |
Collapse
|