1
|
Naser IH, Hamza AA, Alhili A, Faisal AN, Ali MS, Kadhim NA, Suliman M, Alshahrani MY, Alawadi A. Atypical chemokine receptor 4 (ACKR4/CCX-CKR): A comprehensive exploration across physiological and pathological landscapes in contemporary research. Cell Biochem Funct 2024; 42:e4009. [PMID: 38597217 DOI: 10.1002/cbf.4009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/02/2024] [Accepted: 03/31/2024] [Indexed: 04/11/2024]
Abstract
Atypical chemokine receptor 4 (ACKR4), also known as CCX-CKR, is a member of the chemokine receptor family that lacks typical G protein signaling activity. Instead, ACKR4 functions as a scavenger receptor that can bind and internalize a wide range of chemokines, influencing their availability and activity in the body. ACKR4 is involved in various physiological processes, such as immune cell trafficking and the development of thymus, spleen, and lymph nodes. Moreover, ACKR4 has been implicated in several pathological conditions, including cancer, heart and lung diseases. In cancer, ACKR4 plays a complex role, acting as a tumor suppressor or promoter depending on the type of cancer and the stage of the disease. For instance, ACKR4 may inhibit the growth and metastasis of breast cancer, but it may also promote the progression of hepatocellular carcinoma and gastric cancer. In inflammatory situations, ACKR4 has been found to modulate the recruitment and activation of immune cells, contributing to the pathogenesis of diseases such as myocardial infraction and pulmonary sarcoidosis. The study of ACKR4 is still ongoing, and further research is needed to fully understand its role in different physiological and pathological contexts. Nonetheless, ACKR4 represents a promising target for the development of novel therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University College, Hillah, Babil, Iraq
| | - Asia Ali Hamza
- Department of Pharmaceutics, Faculty of pharmacy, University of Al-Ameed, Karbala, Iraq
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | | | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Gonçalves TL, de Araújo LP, Pereira Ferrer V. Tamoxifen as a modulator of CXCL12-CXCR4-CXCR7 chemokine axis: A breast cancer and glioblastoma view. Cytokine 2023; 170:156344. [PMID: 37639844 DOI: 10.1016/j.cyto.2023.156344] [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: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
The chemokine stromal cell-derived-factor 1 (SDF)-1/CXCL12 acts by binding to its receptors, the CXC-4 chemokine receptor (CXCR4) and the CXC-7 chemokine receptor (CXCR7). The binding of CXCL12 to its receptors results in downstream signaling that leads to cell survival, proliferation and migration of tumor cells. CXCL12 and CXCR4 are highly expressed in breast cancer (BC) and glioblastoma (GBM) compared to normal cells. High expression of this chemokine axis correlates with increased therapy resistance and grade, tumor spread and poorer prognosis in these tumors. Tamoxifen (TMX) is a selective estrogen receptor modulator (SERM) that inhibits the expression of estrogen-regulated genes, including growth and angiogenic factors secreted by tumor cells. Additionally, TMX targets several proteins, such as protein kinase C (PKC), phospholipase C (PLC), P-glycoprotein (PgP), phosphatidylinositol-3-kinase (PI3K) and ion channels. This drug showed promising antitumor activity against both BC and GBM cells. In this review, we discuss the role of the CXCL12-CXCR4-CXCR7 chemokine axis in BC and GBM tumor biology and propose TMX as a potential modulator of this axis in these tumors. TMX modulates the CXCL12-CXCR4-CXCR7 axis in BC, however, there are no studies on this in GBM. We propose that studying this axis in GBM cells/patients treated with TMX might be beneficial for these patients. TMX inhibits important signaling pathways in these tumors and the activation of this chemokine axis is associated with increased therapy resistance.
Collapse
Affiliation(s)
- Thaynan Lopes Gonçalves
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Luanna Prudencio de Araújo
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Wang K, Sun S, Zhang G, Lu Z, Chen H, Fan X, Gu C, Pan X, Lin Q, Chen O, Cai L, Dai X, Wang X, Lu C, Yan X, Tan Y. CXCR7 Agonist TC14012 Improves Angiogenic Function of Endothelial Progenitor Cells via Activating Akt/eNOS Pathway and Promotes Ischemic Angiogenesis in Diabetic Limb Ischemia. Cardiovasc Drugs Ther 2023; 37:849-863. [PMID: 35471717 PMCID: PMC10926281 DOI: 10.1007/s10557-022-07337-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Endothelial progenitor cells (EPCs) play a critical role in repairing damaged vessels and triggering ischemic angiogenesis, but their number is reduced and function is impaired under diabetic conditions. Improving EPC function has been considered a promising strategy to ameliorate diabetic vascular complications. In the present study, we aim to investigate whether and how CXCR7 agonist TC14012 promotes the angiogenic function of diabetic EPCs. METHODS High glucose (HG) treatment was used to mimic the hyperglycemia in diabetes. Tube formation, cell scratch recovery and transwell assay, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and cleaved-caspase3 expression were used to evaluate the angiogenic capability, cell migration, and apoptosis of EPCs, respectively. Hind limb ischemia (HLI) model was used to appraise the ability of TC14012 in promoting diabetic ischemic angiogenesis in vivo. RESULTS HG treatment impaired EPC tube formation and migration, and induced EPC apoptosis and oxidative damage, while TC14012 rescued tube formation and migration, and prevented HG-induced apoptosis and oxidative damage of EPCs. Furthermore, these beneficial effects of TC14012 on EPCs were attenuated by specific siRNAs against CXCR7, validating that CXCR7 is a functional target of TC14012 in EPCs. Mechanistic studies demonstrated that HG treatment reduced CXCR7 expression in EPCs, and impaired Akt and endothelial nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) production; similarly, these signal impairments in HG-exposed EPCs could be rescued by TC14012. However, the protective effects of TC14012 on tube formation and migration, Akt and eNOS phosphorylation, and NO production in HG-treated EPCs were almost completely abolished by siRNAs against CXCR7 or Akt specific inhibitor wortmannin. More importantly, in vivo study showed that TC14012 administration enhanced blood perfusion recovery and angiogenesis in the ischemic hind limb and increased the EPC number in peripheral circulation of db/db mice, demonstrating the capability of TC14012 in promoting EPC mobilization and ischemia angiogenic function. CONCLUSION TC14012 can prevent EPCs from HG-induced dysfunction and apoptosis, improve eNOS activity and NO production via CXCR7/Akt signal pathway, and promote EPC mobilization and diabetic ischemia angiogenesis.
Collapse
Affiliation(s)
- Kai Wang
- Department of Pediatrics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, 570 South Preston Street, Baxter-I Building Suite 304E, Louisville, KY, 40202, USA
| | - Shiyue Sun
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guigui Zhang
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zixian Lu
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Chen
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xia Fan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chunjie Gu
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaohong Pan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qian Lin
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, 570 South Preston Street, Baxter-I Building Suite 304E, Louisville, KY, 40202, USA
| | - Oscar Chen
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, 570 South Preston Street, Baxter-I Building Suite 304E, Louisville, KY, 40202, USA
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, 570 South Preston Street, Baxter-I Building Suite 304E, Louisville, KY, 40202, USA
| | - Xiaozhen Dai
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiao Wang
- Department of Obstetrics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaosheng Lu
- Department of Pediatrics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, 570 South Preston Street, Baxter-I Building Suite 304E, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Cai F, Chen W, Zhao R, Liu Y. Mechanisms of Nrf2 and NF-κB pathways in diabetic wound and potential treatment strategies. Mol Biol Rep 2023; 50:5355-5367. [PMID: 37029875 DOI: 10.1007/s11033-023-08392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/15/2023] [Indexed: 04/09/2023]
Abstract
The issue of delayed wound healing or nonhealing in diabetic patients presents a challenge for modern medicine. A number of attempts have been made to understand the mechanisms behind diabetic wound. In a hyperglycemic environment, increased intracellular reactive oxygen species (ROS) disturb the balance between oxidation and antioxidant, causing the wound environment to deteriorate. It has been established that the nuclear factor E2-related factor 2 (Nrf2) and nuclear factor-kappa B (NF-κB) pathways play an important role in regulating inflammation and oxidative stress. Several potential treatment strategies involving Nrf2 and/or NF-κB pathways have been explored in previous studies. Hence, we analyzed mechanisms and changes in Nrf2 and NF-κB pathways in response to oxidative stress and inflammation in diabetic environment. Additionally, we reviewed potential treatment strategies from the past five years for diabetic wound by Nrf2 and/or NF-κB pathways, including receptor agonists, vitamins, hormones, exosomes, drugs, plants, and biomaterials. It may be useful to develop drugs to promote diabetic wound healing.
Collapse
Affiliation(s)
- Feiyu Cai
- Department of Burns and Plastic Surgery & Wound Repair Surgery, the Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Wenjiao Chen
- Department of Burns and Plastic Surgery & Wound Repair Surgery, the Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Ruomei Zhao
- Department of Burns and Plastic Surgery & Wound Repair Surgery, the Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yi Liu
- Department of Burns and Plastic Surgery & Wound Repair Surgery, the Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
5
|
Liraglutide Improves the Angiogenic Capability of EPC and Promotes Ischemic Angiogenesis in Mice under Diabetic Conditions through an Nrf2-Dependent Mechanism. Cells 2022; 11:cells11233821. [PMID: 36497087 PMCID: PMC9736458 DOI: 10.3390/cells11233821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 12/05/2022] Open
Abstract
The impairment in endothelial progenitor cell (EPC) functions results in dysregulation of vascular homeostasis and dysfunction of the endothelium under diabetic conditions. Improving EPC function has been considered as a promising strategy for ameliorating diabetic vascular complications. Liraglutide has been widely used as a therapeutic agent for diabetes. However, the effects and mechanisms of liraglutide on EPC dysfunction remain unclear. The capability of liraglutide in promoting blood perfusion and angiogenesis under diabetic conditions was evaluated in the hind limb ischemia model of diabetic mice. The effect of liraglutide on the angiogenic function of EPC was evaluated by cell scratch recovery assay, tube formation assay, and nitric oxide production. RNA sequencing was performed to assess the underlying mechanisms. Liraglutide enhanced blood perfusion and angiogenesis in the ischemic hindlimb of db/db mice and streptozotocin-induced type 1 diabetic mice. Additionally, liraglutide improved tube formation, cell migration, and nitric oxide production of high glucose (HG)-treated EPC. Assessment of liraglutide target pathways revealed a network of genes involved in antioxidant activity. Further mechanism study showed that liraglutide decreased the production of reactive oxygen species and increased the activity of nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 deficiency attenuated the beneficial effects of liraglutide on improving EPC function and promoting ischemic angiogenesis under diabetic conditions. Moreover, liraglutide activates Nrf2 through an AKT/GSK3β/Fyn pathway, and inhibiting this pathway abolished liraglutide-induced Nrf2 activation and EPC function improvement. Overall, these results suggest that Liraglutide represents therapeutic potential in promoting EPC function and ameliorating ischemic angiogenesis under diabetic conditions, and these beneficial effects relied on Nrf2 activation.
Collapse
|
6
|
Dai X, Wang K, Fan J, Liu H, Fan X, Lin Q, Chen Y, Chen H, Li Y, Liu H, Chen O, Chen J, Li X, Ren D, Li J, Conklin DJ, Wintergerst KA, Li Y, Cai L, Deng Z, Yan X, Tan Y. Nrf2 transcriptional upregulation of IDH2 to tune mitochondrial dynamics and rescue angiogenic function of diabetic EPCs. Redox Biol 2022; 56:102449. [PMID: 36063728 PMCID: PMC9463384 DOI: 10.1016/j.redox.2022.102449] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 07/30/2022] [Accepted: 08/15/2022] [Indexed: 01/11/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are reduced in number and impaired in function in diabetic patients. Whether and how Nrf2 regulates the function of diabetic EPCs remains unclear. In this study, we found that the expression of Nrf2 and its downstream genes were decreased in EPCs from both diabetic patients and db/db mice. Survival ability and angiogenic function of EPCs from diabetic patients and db/db mice also were impaired. Gain- and loss-of-function studies, respectively, showed that knockdown of Nrf2 increased apoptosis and impaired tube formation in EPCs from healthy donors and wild-type mice, while Nrf2 overexpression decreased apoptosis and rescued tube formation in EPCs from diabetic patients and db/db mice. Additionally, proangiogenic function of Nrf2-manipulated mouse EPCs was validated in db/db mice with hind limb ischemia. Mechanistic studies demonstrated that diabetes induced mitochondrial fragmentation and dysfunction of EPCs by dysregulating the abundance of proteins controlling mitochondrial dynamics; upregulating Nrf2 expression attenuated diabetes-induced mitochondrial fragmentation and dysfunction and rectified the abundance of proteins controlling mitochondrial dynamics. Further RNA-sequencing analysis demonstrated that Nrf2 specifically upregulated the transcription of isocitrate dehydrogenase 2 (IDH2), a key enzyme regulating tricarboxylic acid cycle and mitochondrial function. Overexpression of IDH2 rectified Nrf2 knockdown- or diabetes-induced mitochondrial fragmentation and EPC dysfunction. In a therapeutic approach, supplementation of an Nrf2 activator sulforaphane enhanced angiogenesis and blood perfusion recovery in db/db mice with hind limb ischemia. Collectively, these findings indicate that Nrf2 is a potential therapeutic target for improving diabetic EPC function. Thus, elevating Nrf2 expression enhances EPC resistance to diabetes-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia likely via transcriptional upregulating IDH2 expression and improving mitochondrial function of diabetic EPCs.
Collapse
Affiliation(s)
- Xiaozhen Dai
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China; Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiawei Fan
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China
| | - Hanjie Liu
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Xia Fan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qian Lin
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yuhang Chen
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Hu Chen
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yao Li
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Hairong Liu
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Oscar Chen
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jing Chen
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Xiaohong Li
- Kentucky IDeA Network for Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, USA
| | - Di Ren
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Daniel J Conklin
- Department of Medicine and Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Kupper A Wintergerst
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA; Division of Endocrinology, Department of Pediatrics, University of Louisville, Norton Children's Hospital, Louisville, KY, USA; Wendy L. Novak Diabetes Care Center, Norton Children's Hospital, Louisville, KY, USA
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lu Cai
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA; Wendy L. Novak Diabetes Care Center, Norton Children's Hospital, Louisville, KY, USA
| | - Zhongbin Deng
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Yi Tan
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA; Wendy L. Novak Diabetes Care Center, Norton Children's Hospital, Louisville, KY, USA.
| |
Collapse
|
7
|
Huang H, Huang W. Regulation of Endothelial Progenitor Cell Functions in Ischemic Heart Disease: New Therapeutic Targets for Cardiac Remodeling and Repair. Front Cardiovasc Med 2022; 9:896782. [PMID: 35677696 PMCID: PMC9167961 DOI: 10.3389/fcvm.2022.896782] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022] Open
Abstract
Ischemic heart disease (IHD) is the leading cause of morbidity and mortality worldwide. Ischemia and hypoxia following myocardial infarction (MI) cause subsequent cardiomyocyte (CM) loss, cardiac remodeling, and heart failure. Endothelial progenitor cells (EPCs) are involved in vasculogenesis, angiogenesis and paracrine effects and thus have important clinical value in alternative processes for repairing damaged hearts. In fact, this study showed that the endogenous repair of EPCs may not be limited to a single cell type. EPC interactions with cardiac cell populations and mesenchymal stem cells (MSCs) in ischemic heart disease can attenuate cardiac inflammation and oxidative stress in a microenvironment, regulate cell survival and apoptosis, nourish CMs, enhance mature neovascularization, alleviate adverse ventricular remodeling after infarction and enhance ventricular function. In this review, we introduce the definition and discuss the origin and biological characteristics of EPCs and summarize the mechanisms of EPC recruitment in ischemic heart disease. We focus on the crosstalk between EPCs and endothelial cells (ECs), smooth muscle cells (SMCs), CMs, cardiac fibroblasts (CFs), cardiac progenitor cells (CPCs), and MSCs during cardiac remodeling and repair. Finally, we discuss the translation of EPC therapy to the clinic and treatment strategies.
Collapse
|
8
|
Rodríguez-Esparragón F, Torres-Mata LB, López-Fernández JC, Cappiello L, González-Martín JM, Clavo B, Serna-Gómez JA, Estupiñán-Quintana L, Torres-Ascensión C, Villar J. Clinical relevance of circulating angiogenic cells in patients with ischemic stroke. BMC Cardiovasc Disord 2022; 22:118. [PMID: 35313809 PMCID: PMC8939119 DOI: 10.1186/s12872-021-02421-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/12/2021] [Indexed: 11/30/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) are circulating angiogenic cells with endothelial features associated with risk for stroke. We aimed to delve into their functional characteristics. EPCs were isolated and cultured from Ischemic Stroke (IS) patients and predictors of their variance evaluated. Methods This is a single-center observational study evaluating 187 consecutively hospitalized patients with IS. EPCs were isolated from blood samples. The number of circulating angiogenic cells (CACs), colony-forming units (CFU-ECs) and the emergence of late outgrowths endothelial cells (LOECs) were counted. We collected clinical variables and measured the stromal cell-derived factor 1 alpha (SDF1α) serum levels. We also examined the relative telomere length and the expression of osteogenic gene markers in CACs. Results CACs counts and CFU-ECs colony numbers were positively correlated (rho = 0.41, p < 0.001, n = 187). We found significant differences according to whether thrombolytic treatment was performed in the distribution of CFU-ECs (odds ratio (OR) = 2.5; 95% confidence interval (CI) 1.01–6.35; p = 0.042) and CACs (OR = 4.45; 95% IC 1.2–15.5; p = 0.012). The main determinants of CACs variation were the number of risks factors, thrombolysis treatment, arterial hypertension, LOECs occurrence, and the vascular endothelial growth factor expression, whereas CFU-ECs variations depended on hemoglobin content and the relative reduction in the National Institutes of Health Stroke Scale (NIHSS) criteria. The main predictors of LOECs appearance were thrombolysis and length of hospital stay. Conclusions Our study supports the relevance of patient risk factors and treatments in the analysis of the functional properties of EPCs.
Collapse
Affiliation(s)
- Francisco Rodríguez-Esparragón
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain.
| | - Laura B Torres-Mata
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain
| | | | - Laura Cappiello
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain
| | - Jesús M González-Martín
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain
| | - Bernardino Clavo
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain.,Chronic Pain Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain.,Radiation Oncology Department, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain.,Universitary Institute for Research in Biomedicine and Health (iUIBS), Molecular and Translational Pharmacology Group, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.,Spanish Group of Clinical Research in Radiation Oncology (GICOR), 28029, Madrid, Spain.,Research Network On Health Services in Chronic Diseases (REDISSEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaime A Serna-Gómez
- Department of Cardiovascular Surgery, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Lidia Estupiñán-Quintana
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain
| | - Cristina Torres-Ascensión
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain
| | - Jesús Villar
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Barranco de La Ballena S/N, 35019, Las Palmas de Gran Canaria, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Li Ka Shing Knowledge Institute at St Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
9
|
Song B, Chen D, Liu Z, Cheng Y, Zhang Z, Han W, Zhang R, Gong Y. Stromal cell-derived factor-1 exerts opposing roles through CXCR4 and CXCR7 in angiotensin II-induced adventitial remodeling. Biochem Biophys Res Commun 2022; 594:38-45. [PMID: 35066378 DOI: 10.1016/j.bbrc.2022.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/29/2021] [Accepted: 01/09/2022] [Indexed: 11/02/2022]
Abstract
Recent studies have emphasized the role of vascular adventitia inflammation and immune response in hypertension. It has been reported that stromal cell-derived factor-1 (SDF-1) plays various biological functions through its receptors C-X-C motif chemokine receptor 4 (CXCR4) and CXCR7 in tumor growth and tissue repair. However, it is unclear that whether SDF-1/CXCR4/CXCR7 axis is involved in hypertensive vascular remodeling. In the present study, the involvement of SDF-1/CXCR4/CXCR7 axis was evaluated with lentivirus-mediated shRNA of SDF-1 and CXCR7, CXCR4 antagonist AMD3100 and CXCR7 agonist VUF11207 in angiotensin II (AngII)-induced hypertensive mice and in cultured adventitial fibroblasts (AFs). Results showed that AngII infusion markedly increased SDF-1 expressed in vascular adventitia, but not in media and endothelium. Importantly, blockade of SDF-1/CXCR4 axis strikingly potentiated AngII-induced adventitial thickening and fibrosis, as indicated by enhanced collagen I deposition. In contrast, CXCR7 shRNA largely attenuated AngII-induced adventitial thickness and fibrosis, whereas CXCR7 activation with VUF11207 significantly potentiated AngII-induced adventitial thickening and fibrosis. In consistent with these in vivo study, CXCR4 inhibition with AMD3100 and CXCR7 activation with VUF11207 aggravated AngII-induced inflammation, proliferation and migration in cultured AFs. In summary, these results suggested that SDF-1 exerted opposing effects through CXCR4 and CXCR7 in AngII-induced vascular adventitial remodeling.
Collapse
Affiliation(s)
- Bei Song
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongrui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zixiong Liu
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuwen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zebei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Han
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yanchun Gong
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Regulation of endothelial progenitor cell functions during hyperglycemia: new therapeutic targets in diabetic wound healing. J Mol Med (Berl) 2022; 100:485-498. [PMID: 34997250 DOI: 10.1007/s00109-021-02172-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Diabetes is primarily characterized by hyperglycemia, and its high incidence is often very costly to patients, their families, and national economies. Unsurprisingly, the number and function of endothelial progenitor cells (EPCs) decrease in patients resulting in diabetic wound non-healing. As precursors of endothelial cells (ECs), these cells were discovered in 1997 and found to play an essential role in wound healing. Their function, number, and role in wound healing has been widely investigated. Hitherto, a lot of complex molecular mechanisms have been discovered. In this review, we summarize the mechanisms of how hyperglycemia affects the function and number of EPCs and how the affected cells impact wound healing. We aim to provide a complete summary of the relationship between diabetic hyperglycosemia, EPCs, and wound healing, as well as a better comprehensive platform for subsequent related research.
Collapse
|
11
|
Endothelial ACKR3 drives atherosclerosis by promoting immune cell adhesion to vascular endothelium. Basic Res Cardiol 2022; 117:30. [PMID: 35674847 PMCID: PMC9177477 DOI: 10.1007/s00395-022-00937-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 01/31/2023]
Abstract
Atherosclerosis is the foundation of potentially fatal cardiovascular diseases and it is characterized by plaque formation in large arteries. Current treatments aimed at reducing atherosclerotic risk factors still allow room for a large residual risk; therefore, novel therapeutic candidates targeting inflammation are needed. The endothelium is the starting point of vascular inflammation underlying atherosclerosis and we could previously demonstrate that the chemokine axis CXCL12-CXCR4 plays an important role in disease development. However, the role of ACKR3, the alternative and higher affinity receptor for CXCL12 remained to be elucidated. We studied the role of arterial ACKR3 in atherosclerosis using western diet-fed Apoe-/- mice lacking Ackr3 in arterial endothelial as well as smooth muscle cells. We show for the first time that arterial endothelial deficiency of ACKR3 attenuates atherosclerosis as a result of diminished arterial adhesion as well as invasion of immune cells. ACKR3 silencing in inflamed human coronary artery endothelial cells decreased adhesion molecule expression, establishing an initial human validation of ACKR3's role in endothelial adhesion. Concomitantly, ACKR3 silencing downregulated key mediators in the MAPK pathway, such as ERK1/2, as well as the phosphorylation of the NF-kB p65 subunit. Endothelial cells in atherosclerotic lesions also revealed decreased phospho-NF-kB p65 expression in ACKR3-deficient mice. Lack of smooth muscle cell-specific as well as hematopoietic ACKR3 did not impact atherosclerosis in mice. Collectively, our findings indicate that arterial endothelial ACKR3 fuels atherosclerosis by mediating endothelium-immune cell adhesion, most likely through inflammatory MAPK and NF-kB pathways.
Collapse
|
12
|
Duval V, Alayrac P, Silvestre JS, Levoye A. Emerging Roles of the Atypical Chemokine Receptor 3 (ACKR3) in Cardiovascular Diseases. Front Endocrinol (Lausanne) 2022; 13:906586. [PMID: 35846294 PMCID: PMC9276939 DOI: 10.3389/fendo.2022.906586] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/27/2022] [Indexed: 11/14/2022] Open
Abstract
Chemokines, and their receptors play a crucial role in the pathophysiology of cardiovascular diseases (CVD). Chemokines classically mediate their effects by binding to G-protein-coupled receptors. The discovery that chemokines can also bind to atypical chemokine receptors (ACKRs) and initiate alternative signaling pathways has changed the paradigm regarding chemokine-related functions. Among these ACKRs, several studies have highlighted the exclusive role of ACKR3, previously known as C-X-C chemokine receptor type 7 (CXCR7), in CVD. Indeed, ACKR3 exert atheroprotective, cardioprotective and anti-thrombotic effects through a wide range of cells including endothelial cells, platelets, inflammatory cells, fibroblasts, vascular smooth muscle cells and cardiomyocytes. ACKR3 functions as a scavenger receptor notably for the pleiotropic chemokine CXCL12, but also as a activator of different pathways such as β-arrestin-mediated signaling or modulator of CXCR4 signaling through the formation of ACKR3-CXCR4 heterodimers. Hence, a better understanding of the precise roles of ACKR3 may pave the way towards the development of novel and improved therapeutic strategies for CVD. Here, we summarize the structural determinant characteristic of ACKR3, the molecules targeting this receptor and signaling pathways modulated by ACKR3. Finally, we present and discuss recent findings regarding the role of ACKR3 in CVD.
Collapse
Affiliation(s)
- Vincent Duval
- Université Paris Cité, Institut National de la Santé Et Recherche Médicale (INSERM), Paris Cardiovascular Research Center PARCC, Paris, France
| | - Paul Alayrac
- Université Paris Cité, Institut National de la Santé Et Recherche Médicale (INSERM), Paris Cardiovascular Research Center PARCC, Paris, France
| | - Jean-Sébastien Silvestre
- Université Paris Cité, Institut National de la Santé Et Recherche Médicale (INSERM), Paris Cardiovascular Research Center PARCC, Paris, France
| | - Angélique Levoye
- Université Paris Cité, Institut National de la Santé Et Recherche Médicale (INSERM), Paris Cardiovascular Research Center PARCC, Paris, France
- UFR Santé Médecine Biologie Humaine, Université Sorbonne Paris Nord, Bobigny, France
- *Correspondence: Angélique Levoye,
| |
Collapse
|
13
|
Murad HAS, Rafeeq MM, Alqurashi TMA. Role and implications of the CXCL12/CXCR4/CXCR7 axis in atherosclerosis: still a debate. Ann Med 2021; 53:1598-1612. [PMID: 34494495 PMCID: PMC8439212 DOI: 10.1080/07853890.2021.1974084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023] Open
Abstract
Atherosclerosis is one of the leading causes of mortality and morbidity worldwide. Chemokines and their receptors are implicated in the pathogenesis of atherosclerosis. CXCL12 is a member of the chemokine family exerting a myriad role in atherosclerosis through its classical CXCR4 and atypical ACKR3 (CXCR7) receptors. The modulatory and regulatory functional spectrum of CXCL12/CXCR4/ACKR3 axis in atherosclerosis spans from proatherogenic, prothrombotic and proinflammatory to atheroprotective, plaque stabilizer and dyslipidemia rectifier. This diverse continuum is executed in a wide range of biological units including endothelial cells (ECs), progenitor cells, macrophages, monocytes, platelets, lymphocytes, neutrophils and vascular smooth muscle cells (VSMCs) through complex heterogeneous and homogenous coupling of CXCR4 and ACKR3 receptors, employing different downstream signalling pathways, which often cross-talk among themselves and with other signalling interactomes. Hence, a better understanding of this structural and functional heterogeneity and complex phenomenon involving CXCL12/CXCR4/ACKR3 axis in atherosclerosis would not only help in formulation of novel therapeutics, but also in elucidation of the CXCL12 ligand and its receptors, as possible diagnostic and prognostic biomarkers.Key messagesThe role of CXCL12 per se is proatherogenic in atherosclerosis development and progression.The CXCL12 receptors, CXCR4 and ACKR3 perform both proatherogenic and athero-protective functions in various cell typesDue to functional heterogeneity and cross talk of CXCR4 and ACKR3 at receptor level and downstream pathways, regional boosting with specific temporal and spatial modulators of CXCL12, CXCR4 and ACKR3 need to be explored.
Collapse
Affiliation(s)
- Hussam A. S. Murad
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| | - Misbahuddin M. Rafeeq
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| | - Thamer M. A. Alqurashi
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| |
Collapse
|
14
|
Tian Y, Wang Z, Zheng X, Song W, Cai L, Rane M, Zhao Y. KLF15 negatively regulates cardiac fibrosis by which SDF-1β attenuates cardiac fibrosis in type 2 diabetic mice. Toxicol Appl Pharmacol 2021; 427:115654. [PMID: 34310909 DOI: 10.1016/j.taap.2021.115654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 02/08/2023]
Abstract
Diabetic cardiomyopathy (DCM) is a serious diabetic complication that lacks effective preventive or therapeutic approaches. Wild-type and Klf15 knockout (Klf15-KO) mice were fed with either high fat diet (HFD, 60% kcal from fat) or normal diet (ND, 10% kcal from fat) for 3 months and then injected with streptozotocin or vehicle, to induce type 2 diabetes (T2D). All T2D and age-matched control mice were treated with or without SDF-1β at 5 mg/kg body-weight twice a week and also continually received HFD or ND for 3 months. At the end of 6-month study, after cardiac functions were measured, mice were euthanized to collect heart tissue. For in vitro mechanistic study, H9c2 cells were exposed to palmitate to mimic in vivo condition of T2D. SDF-1β prevented T2D-induced cardiac dysfunction and fibrosis and T2D-down-regulated KLF15 expression in wild-type diabetic heart tissue. However, the preventive effects of SDF-1β on both KLF15 expression and fibrosis was abolished, with partial cardiac protection in Klf15-KO/T2D mice. These results demonstrate partial KLF15-dependence for SDF-1β's cardiac fibrotic protection from T2D, but not on SDF-1β's protective effects on T2D-induced cardiac dysfunction. Further study showed that SDF-1β inhibited palmitate-induced cardiomyocyte fibrosis through its receptor CXCR7-mediated activation of p38β MAPK signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Tian
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhenyu Wang
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiangyu Zheng
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Wenjing Song
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Lu Cai
- Pediatric Research Institute, Departments of Pediatrics, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Madhavi Rane
- Division Nephrology, Departments of Medicine, Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
15
|
Xinkeshu Improves Endothelial Function and Augments Reendothelialization Capacity in Coronary Artery Disease with Anxiety/Depression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5561272. [PMID: 34336100 PMCID: PMC8313340 DOI: 10.1155/2021/5561272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/21/2021] [Accepted: 06/11/2021] [Indexed: 11/21/2022]
Abstract
The disruption of endothelial homeostasis is the hallmark of coronary artery disease (CAD) and psychological disorders such as anxiety/depression. Xinkeshu (XKS), a traditional Chinese patent medicine, plays an essential role in CAD and psychological condition; however, the mechanisms underlying the effects of XKS on the endothelial function and endogenous endothelium-repair capacity in CAD patients with anxiety/depression remain elusive. In this study, endothelial function and endothelial progenitor cell- (EPC-) mediated reendothelialization capacity were compared among age-matched healthy subjects, CAD patients with or without anxiety/depression. Besides, CAD patients with anxiety/depression received 1-month XKS treatment. Anxiety/depression symptoms were evaluated by Generalized Anxiety Disorder 7-item (GAD-7)/Patient Health Questionnaire-9 (PHQ-9) score, endothelial function was tested by flow mediated dilation (FMD) measurement, and EPC-mediated reendothelialization capacity was evaluated by a carotid artery injury model in nude mouse (n = 6) with the injection of XKS-incubated EPCs from CAD patients with anxiety/depression. The results showed that FMD and EPC-mediated reendothelialization capacity of CAD patients with anxiety/depression were compromised compared to healthy subjects and CAD patients without anxiety/depression. After 1 month of XKS treatment, FMD increased from 4.29 ± 1.65 to 4.87 ± 1.58% (P < 0.05) in CAD patients with anxiety/depression, whereas it remained unchanged in the controls. Moreover, XKS decreased GAD-7 and PHQ-9 scores. Meanwhile, incubating XKS enhanced in vivo reendothelialization capacity and in vitro apoptosis of EPCs from CAD patients with anxiety/depression, which was associated with the upregulation of CXC-chemokine receptor 7 (CXCR7) and inhibition of phosphorylation of p38 signaling. CXCR7 knockdown abolished the beneficial effects of XKS, which was rescued by p38 inhibitor SB203580. Our data demonstrate for the first time that XKS improves endothelial function and enhances EPC-mediated reendothelialization through CXCR7/p38/cleaved casepase-3 signaling and provides novel insight into the detailed mechanism of XKS in maintaining endothelial homeostasis in CAD patients with anxiety/depression.
Collapse
|
16
|
João Pissarra A, Abreu C, Mansinho A, Lúcia Costa A, Dâmaso S, Lobo-Martins S, Martins M, Costa L. Landscape of Current Targeted Therapies for Advanced Colorectal Cancer. COLORECTAL CANCER 2021. [DOI: 10.5772/intechopen.93978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Colorectal cancer (CRC) is one of the most frequent and lethal cancer types worldwide. While surgery with chemotherapy and radiotherapy remains the only curative approach for localized CRC, for metastatic disease the therapeutic landscape has significantly evolved over the last years. Development and approval of novel targeted therapies, such as monoclonal antibodies against EGFR and VEGF, have significantly increased the median survival of patients with metastatic disease, with some trials reporting a benefit over 40 months. Increasing accessibility of high throughput sequencing has unraveled several new therapeutic targets. Actionable alterations, such as HER2 overexpression, BRAF mutations, and NTRK fusions, are currently available in metastatic disease, providing significant therapeutic opportunities for these patients, while new emerging agents, as immune checkpoint inhibitors, promise better treatment options in the near future. In this chapter, an overview of established and future CRC targeted therapies in the clinical setting is provided, as well as their mechanism of action, limitations, and future applicability.
Collapse
|
17
|
Liu X, Huang Z, Zhang Y, Shui X, Liu F, Wu Z, Xu S. Lacidipine Ameliorates the Endothelial Senescence and Inflammatory Injury Through CXCR7/P38/C/EBP-β Signaling Pathway. Front Cardiovasc Med 2021; 8:692540. [PMID: 34295928 PMCID: PMC8290057 DOI: 10.3389/fcvm.2021.692540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/31/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Lacidipine, a third-generation calcium channel blocker, exerts beneficial effects on the endothelium of hypertensive patients in addition to blood pressure lowering. However, the detailed mechanism underlying Lacidipine-related endothelial protection is still elusive. Methods: Sixteen spontaneous hypertensive rats (SHRs) were randomly divided into two groups: Lacidipine-treated SHR group and saline-treated control group. Tail systolic blood pressure was monitored for four consecutive weeks. Endothelial cells (ECs) were pretreated with Lacidipine prior to being stimulated with H2O2, bleomycin, or Lipopolysaccharides (LPS) in vitro. Then, cell activity, migration, and senescence were measured by Cell Counting Kit-8 assay, transwell assay, and β-galactosidase staining, respectively. The fluorescent probe 2′, 7′-dichlorofluorescein diacetate (DCFH-DA) was used to assess the intracellular reactive oxygen species (ROS). Related protein expression was detected by Western blotting and immunofluorescence. Results: Our data showed that Lacidipine treatment lowered the blood pressure of SHRs accompanied by the elevation of CXCR7 expression and suppression of P38 and CCAAT/enhancer-binding protein beta (C/EBP-β) compared with the control group. In vitro experiments further demonstrated that Lacidipine increased the cell viability and function of ECs under oxidative stress, cell senescence, and inflammatory activation via the CXCR7/P38/signaling pathway. Conclusions: Our results suggested that Lacidipine plays a protective role in EC senescence, oxidative stress, and inflammatory injury through the regulation of CXCR7/P38/C/EBP-β signaling pathway.
Collapse
Affiliation(s)
- Xing Liu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuoshan Huang
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Zhang
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xing Shui
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fanmao Liu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhen Wu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China.,National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Jiang C, Li R, Xiu C, Ma X, Hu H, Wei L, Tang Y, Tao M, Zhao J. Upregulating CXCR7 accelerates endothelial progenitor cell-mediated endothelial repair by activating Akt/Keap-1/Nrf2 signaling in diabetes mellitus. Stem Cell Res Ther 2021; 12:264. [PMID: 33941256 PMCID: PMC8091720 DOI: 10.1186/s13287-021-02324-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/02/2021] [Indexed: 11/15/2022] Open
Abstract
Background Endothelial progenitor cell (EPC) dysfunction contributes to vascular disease in diabetes mellitus. However, the molecular mechanism underlying EPC dysfunction and its contribution to delayed reendothelialization in diabetes mellitus remain unclear. Our study aimed to illustrate the potential molecular mechanism underlying diabetic EPC dysfunction in vivo and in vitro. Furthermore, we assessed the effect of EPC transplantation on endothelial regeneration in diabetic rats. Methods Late outgrowth EPCs were isolated from the bone marrow of rats for in vivo and in vitro studies. In vitro functional assays and Western blotting were conducted to reveal the association between C-X-C chemokine receptor type 7 (CXCR7) expression and diabetic EPC dysfunction. To confirm the association between cellular CXCR7 levels and EPC function, CXCR7 expression in EPCs was upregulated and downregulated via lentiviral transduction and RNA interference, respectively. Western blotting was used to reveal the potential molecular mechanism by which the Stromal-Derived Factor-1 (SDF-1)/CXCR7 axis regulates EPC function. To elucidate the role of the SDF-1/CXCR7 axis in EPC-mediated endothelial regeneration, a carotid artery injury model was established in diabetic rats. After the model was established, saline-treated, diabetic, normal, or CXCR7-primed EPCs were injected via the tail vein. Results Diabetic EPC dysfunction was associated with decreased CXCR7 expression. Furthermore, EPC dysfunction was mimicked by knockdown of CXCR7 in normal EPCs. However, upregulating CXCR7 expression reversed the dysfunction of diabetic EPCs. The SDF-1/CXCR7 axis positively regulated EPC function by activating the AKT-associated Kelch-like ECH-associated protein 1 (keap-1)/nuclear factor erythroid 2-related factor 2 (Nrf2) axis, which was reversed by blockade of AKT and Nrf2. Transplantation of CXCR7-EPCs accelerated endothelial repair and attenuated neointimal hyperplasia in diabetes mellitus more significantly than transplantation of diabetic or normal EPCs. However, the therapeutic effect of CXCR7-EPC transplantation on endothelial regeneration was reversed by knockdown of Nrf2 expression. Conclusions Dysfunction of diabetic EPCs is associated with decreased CXCR7 expression. Furthermore, the SDF-1/CXCR7 axis positively regulates EPC function by activating the AKT/keap-1/Nrf2 axis. CXCR7-primed EPCs might be useful for endothelial regeneration in diabetes-associated vascular disease.
Collapse
Affiliation(s)
- Chunyu Jiang
- Department of Interventional Therapy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200233, People's Republic of China
| | - Ruiting Li
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China
| | - Chaoyang Xiu
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China
| | - Xu Ma
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China
| | - Hui Hu
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China
| | - Liming Wei
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China
| | - Yihan Tang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Mingyang Tao
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Jungong Zhao
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
19
|
Santagata S, Ieranò C, Trotta AM, Capiluongo A, Auletta F, Guardascione G, Scala S. CXCR4 and CXCR7 Signaling Pathways: A Focus on the Cross-Talk Between Cancer Cells and Tumor Microenvironment. Front Oncol 2021; 11:591386. [PMID: 33937018 PMCID: PMC8082172 DOI: 10.3389/fonc.2021.591386] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor 4 (CXCR4) and 7 (CXCR7) are G-protein-coupled receptors (GPCRs) activated through their shared ligand CXCL12 in multiple human cancers. They play a key role in the tumor/tumor microenvironment (TME) promoting tumor progression, targeting cell proliferation and migration, while orchestrating the recruitment of immune and stromal cells within the TME. CXCL12 excludes T cells from TME through a concentration gradient that inhibits immunoactive cells access and promotes tumor vascularization. Thus, dual CXCR4/CXCR7 inhibition will target different cancer components. CXCR4/CXCR7 antagonism should prevent the development of metastases by interfering with tumor cell growth, migration and chemotaxis and favoring the frequency of T cells in TME. Herein, we discuss the current understanding on the role of CXCL12/CXCR4/CXCR7 cross-talk in tumor progression and immune cells recruitment providing support for a combined CXCR4/CXCR7 targeting therapy. In addition, we consider emerging approaches that coordinately target both immune checkpoints and CXCL12/CXCR4/CXCR7 axis.
Collapse
Affiliation(s)
- Sara Santagata
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Caterina Ieranò
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Maria Trotta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Capiluongo
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Federica Auletta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Giuseppe Guardascione
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Stefania Scala
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| |
Collapse
|
20
|
Nguyen HT, Reyes-Alcaraz A, Yong HJ, Nguyen LP, Park HK, Inoue A, Lee CS, Seong JY, Hwang JI. CXCR7: a β-arrestin-biased receptor that potentiates cell migration and recruits β-arrestin2 exclusively through Gβγ subunits and GRK2. Cell Biosci 2020; 10:134. [PMID: 33292475 PMCID: PMC7686738 DOI: 10.1186/s13578-020-00497-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Some chemokine receptors referred to as atypical chemokine receptors (ACKRs) are thought to non-signaling decoys because of their inability to activate typical G-protein signaling pathways. CXCR7, also known as ACKR3, binds to only two chemokines, SDF-1α and I-TAC, and recruits β-arrestins. SDF-1α also binds to its own conventional receptor, CXCR4, involving in homeostatic modulation such as development and immune surveillance as well as pathological conditions such as inflammation, ischemia, and cancers. Recently, CXCR7 is suggested as a key therapeutic target together with CXCR4 in such conditions. However, the molecular mechanisms underlying cellular responses and functional relation with CXCR7 and CXCR4 have not been elucidated, despite massive studies. Therefore, we aimed to reveal the molecular networks of CXCR7 and CXCR4 and compare their effects on cell migration. METHODS Base on structural complementation assay using NanoBiT technology, we characterized the distinct mechanisms underlying β-arrestin2 recruitment by both CXCR4 and CXCR7. Crosslinking and immunoprecipitation were conducted to analyze complex formation of the receptors. Gene deletion using CRISPR and reconstitution of the receptors were applied to analysis of ligand-dependent ERK phosphorylation and cell migration. All experiments were performed in triplicate and repeated more than three times. Unpaired Student's t-tests or ANOVA using PRISM5 software were employed for statistical analyses. RESULTS Ligand binding to CXCR7 does not result in activation of typical signaling pathways via Gα subunits but activation of GRK2 via βγ subunits and receptor phosphorylation with subsequent β-arrestin2 recruitment. In contrast, CXCR4 induced Gαi activation and recruited β-arrestin2 through C-terminal phosphorylation by both GRK2 and GRK5. SDF-1α-stimulated ERK phosphorylation was facilitated by CXCR4, but not CXCR7. Heterodimerization of CXCR4 and CXCR7 was not confirmed in this study, while homodimerization of them was verified by crosslinking experiment and NanoBiT assay. Regarding chemotaxis, SDF-1α-stimulated cell migration was mediated by both CXCR4 and CXCR7. CONCLUSION This study demonstrates that SDF-1α-stimulated CXCR7 mediates β-arrestin2 recruitment via different molecular networking from that of CXCR4. CXCR7 may be neither a simple scavenger nor auxiliary receptor but plays an essential role in cell migration through cooperation with CXCR4.
Collapse
Affiliation(s)
- Huong Thi Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | | | - Hyo Jeong Yong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Lan Phuong Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hee-Kyung Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Cheol Soon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Young Seong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jong-Ik Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Koch C, Engele J. Functions of the CXCL12 Receptor ACKR3/CXCR7-What Has Been Perceived and What Has Been Overlooked. Mol Pharmacol 2020; 98:577-585. [PMID: 32883765 DOI: 10.1124/molpharm.120.000056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
The CXCL12 system is central to the development of many organs and is further crucially engaged in pathophysiological processes underlying cancer, inflammation, and cardiovascular disorders. This disease-associated role presently focuses major interest on the two CXCL12 receptors, CXCR4 and atypical chemokine receptor 3 (ACKR3)/CXCR7, as promising therapeutic targets. Major obstacles in these ongoing efforts are confusing reports on the differential use of either ACKR3/CXCR7 and/or CXCR4 across various cells as well as on the specific function(s) of ACKR3/CXCR7. Although basically no doubts remain that CXCR4 represents a classic chemokine receptor, functions assigned to ACKR3/CXCR7 range from those of a strictly silent scavenger receptor eventually modulating CXCR4 signaling to an active and independent signaling receptor. In this review, we depict a thorough analysis of our present knowledge on different modes of organization and functions of the cellular CXCL12 system. We further highlight the potential role of ACKR3/CXCR7 as a "crosslinker" of different receptor systems. Finally, we discuss mechanisms with the potency to impinge on the cellular organization of the CXCL12 system and hence might represent additional future therapeutic targets. SIGNIFICANCE STATEMENT: Delineating the recognized functions of atypical chemokine receptor 3 and CXCR4 in CXCL12 signaling is central to the more detailed understanding of the role of the CXCL12 system in health and disease and will help to guide future research efforts.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| |
Collapse
|
22
|
Abdel Fattah IO, Nasr El-Din WA. Granulocyte-colony stimulating factor improves intervertebral disc degeneration in experimental adult male rats: A microscopic and radiological study. Anat Rec (Hoboken) 2020; 304:787-802. [PMID: 33015986 DOI: 10.1002/ar.24519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/22/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Abstract
Intervertebral disc degeneration (IVDD) is a major contributor to low back pain (LBP). Granulocyte-colony stimulating factor (GCSF) is known to mobilize hematopoietic stem cells (HSCs) that may be implicated in intervertebral disc (IVD) regeneration. Rats were divided into the following three groups: (i) control group; (ii) IVDD group-the rats underwent Co5/Co6 and Co7/Co8 IVDD operation; and (iii) GCSF-treated group-the rats received daily GCSF subcutaneous injections starting 6 weeks after the IVDD operation and continued for 5 days. All of the rats were euthanized after 8 weeks, and IVDs were assessed by tail X-ray and histopathological, immunohistochemical, and transmission electron microscopy (TEM) analyses. The X-rays showed disc narrowing in the IVDD group that was significantly widened in the GCSF-treated rats. Histologically, the IVDD group showed disarrangement of the annulus fibrosis lamellae, complete degeneration of the nucleus pulposus, and loss of proteoglycan content. These changes were improved after GCSF treatment. Vertebral endplate thickness and cellularity were significantly decreased with IVDD and significantly increased after GCSF treatment. Stromal cell-derived factor-1α (SDF-1α) immune expression was significantly increased in the IVDD group but decreased in the GCSF-treated group. However, the caspase-3 expression percentage showed no significant difference among the studied groups. TEM showed excessive collagen deposits around the notochordal cells in the IVDD group, which were attenuated in the GCSF-treated group. These results indicate that GCSF improves IVDD and promotes its recovery based on radiological, histological and TEM findings.
Collapse
Affiliation(s)
- Islam Omar Abdel Fattah
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Wael Amin Nasr El-Din
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,Department of Anatomy, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
23
|
Jiang C, Li R, Ma X, Hu H, Wei L, Zhao J. Plerixafor stimulates adhesive activity and endothelial regeneration of endothelial progenitor cells via elevating CXCR7 expression. J Diabetes Complications 2020; 34:107654. [PMID: 32741660 DOI: 10.1016/j.jdiacomp.2020.107654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/14/2020] [Accepted: 06/14/2020] [Indexed: 01/28/2023]
Abstract
AIMS To assess the effects of plerixafor on function and endothelial regeneration of endothelial progenitor cells (EPCs). METHODS The proliferation and adhesion capacity of EPCs were evaluated in vitro. Furthermore, the expression levels of CXC chemokine receptor-7 (CXCR7) were detected before and after treatment with plerixafor. The CXCR7 expression of EPCs was knocked-down by RNA interference to evaluate the role of CXCR7 in regulating function of EPCs. A rat carotid artery injury model was established to assess the influences of plerixafor on endothelial regeneration. RESULTS Plerixafor stimulated adhesion capacity of EPCs, associating with upregulation of CXCR7 and activation of LFA-1 and VLA-4 molecules. Knockdown of CXCR7 slightly impaired proliferation capacity but significantly attenuated adhesion capacity of EPCs. Plerixafor facilitated endothelial repair at 7 days, while reduced neointimal hyperplasia at 7 and 14 days via recruiting more EPCs participating in endothelial reparation. CONCLUSIONS Plerixafor can positively regulate adhesion capacity of EPCs to HUVECs via elevating the expression level of CXCR7 and stimulating LFA-1 and VLA-4 molecules activation. Treatment with plerixafor accelerated re-endothelialization and inhibited neointimal hyperplasia after endoth elial injury, indicating that it can to be used for endothelial regeneration.
Collapse
Affiliation(s)
- Chunyu Jiang
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Ruiting Li
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Xu Ma
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Hui Hu
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Liming Wei
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Jungong Zhao
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China..
| |
Collapse
|
24
|
Jiang C, Li R, Ma X, Hu H, Guo J, Zhao J. AMD3100 and SDF‑1 regulate cellular functions of endothelial progenitor cells and accelerate endothelial regeneration in a rat carotid artery injury model. Mol Med Rep 2020; 22:3201-3212. [PMID: 32945467 PMCID: PMC7453604 DOI: 10.3892/mmr.2020.11432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/22/2020] [Indexed: 11/12/2022] Open
Abstract
The present study was conducted to assess the effects of AMD3100 and stromal cell-derived factor 1 (SDF-1) on cellular functions and endothelial regeneration of endothelial progenitor cells (EPCs). The cell proliferation and adhesion capacity of EPCs were evaluated in vitro following treatment with AMD3100 and SDF-1 using a Cell Counting Kit-8 assay. Furthermore, the expression levels of C-X-C motif chemokine receptor 4 (CXCR4) and C-X-C motif chemokine receptor 7 (CXCR7) were detected before and after treatment with AMD3100 and SDF-1 to elucidate their possible role in regulating the cellular function of EPCs. A rat carotid artery injury model was established to assess the influences of AMD3100 and SDF-1 on endothelial regeneration. AMD3100 reduced the proliferation and adhesion capacity of EPCs to fibronectin (FN), whereas it increased the adhesion capacity of EPCs to human umbilical vein endothelial cells (HUVECs). However, SDF-1 stimulated the proliferation and cell adhesion capacity of EPCs to HUVECs and FN. Additionally, the expression levels of CXCR7 but not CXCR4 were upregulated following AMD3100 treatment, whereas the expression levels of both CXCR4 and CXCR7 were upregulated after SDF-1 treatment. In vivo results demonstrated that AMD3100 increased the number of EPCs in the peripheral blood and facilitated endothelial repair at 7 days after treatment. However, local administration of SDF-1 alone did not enhance reendothelialization 7 and 14 days after treatment. Importantly, the combination of AMD3100 with SDF-1 exhibited superior therapeutic effects compared with AMD3100 treatment alone, accelerated reendothelialization 7 days after treatment, and attenuated neointimal hyperplasia at day 7 and 14 by recruiting more EPCs to the injury site. In conclusion, AMD3100 could positively regulate the adhesion capacity of EPCs to HUVECs via elevation of the expression levels of CXCR7 but not CXCR4, whereas SDF-1 could stimulate the proliferation and adhesion capacity of EPCs to FN and HUVECs by elevating the expression levels of CXCR4 and CXCR7. AMD3100 combined with SDF-1 outperformed AMD3100 alone, promoted early reendothelialization and inhibited neointimal hyperplasia, indicating that early reendothelialization attenuated neointimal hypoplasia following endothelial injury.
Collapse
Affiliation(s)
- Chunyu Jiang
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Ruiting Li
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Xu Ma
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Hui Hu
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Juan Guo
- Department of Hematology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Jungong Zhao
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| |
Collapse
|
25
|
Liu J, Dou Q, Zhou C, Zhou L, Zhao F, Xu L, Xu Z, Ge Y, Wu R, Jia R. Low-energy shock wave pretreatment recruit circulating endothelial progenitor cells to attenuate renal ischaemia reperfusion injury. J Cell Mol Med 2020; 24:10589-10603. [PMID: 32761803 PMCID: PMC7521246 DOI: 10.1111/jcmm.15678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Low‐energy shock wave (LESW) has been recognized as a promising non‐invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme‐linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF‐1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.
Collapse
Affiliation(s)
- Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Quanliang Dou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuzheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ran Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Huang H, Xu Z, Qi Y, Zhang W, Zhang C, Jiang M, Deng S, Wang H. Exosomes from SIRT1-Overexpressing ADSCs Restore Cardiac Function by Improving Angiogenic Function of EPCs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:737-750. [PMID: 32771925 PMCID: PMC7412761 DOI: 10.1016/j.omtn.2020.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022]
Abstract
Acute myocardial infarction (AMI) is one of the leading causes of mortality in cardiovascular diseases. The aim of this study was to investigate whether exosomes from Sirtuin 1 (SIRT1)-overexpressing adipose-derived stem cells (ADSCs) had a protective effect on AMI. The expression of C-X-C chemokine receptor type 7 (CXCR7) was significantly downregulated in peripheral blood endothelial progenitor cells (EPCs) from AMI patients (AMI-EPCs) compared with that in healthy donors, which coincided with impaired tube formation. The exosomes from SIRT1 overexpression in ADSCs (ADSCs-SIRT1-Exos) increased the expression of C-X-C motif chemokine 12 (CXCL12) and nuclear factor E2 related factor 2 (Nrf2) in AMI-EPCs, which promoted migration and tube formation of AMI-EPCs, and overexpression of CXCR7 helped AMI-EPCs to restore the function of cell migration and tube formation. Moreover, CXCR7 was downregulated in the myocardium of AMI mice, and knockout of CXCR7 exacerbated AMI-induced impairment of cardiovascular function. Injection of ADSCs-SIRT1-Exos increased the survival and promoted the recovery of myocardial function with reduced infarct size and post-AMI left ventricular remodeling, induced vasculogenesis, and decreased AMI-induced myocardial inflammation. These findings showed that ADSCs-SIRT1-Exos may recruit EPCs to the repair area and that this recruitment may be mediated by Nrf2/CXCL12/CXCR7 signaling.
Collapse
Affiliation(s)
- Hui Huang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Zhenxing Xu
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Yuan Qi
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Wei Zhang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Chenjun Zhang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Mei Jiang
- Department of Neurology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Shengqiong Deng
- Department of Clinical Laboratory, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China
| | - Hairong Wang
- Department of Cardiology, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, P.R. China.
| |
Collapse
|
27
|
Xie W, Wu F, He J, Su C, Chen L, Zhang X, Sun J, Liu X, Tao J. PGC-1α gene transfer restores adhesion and reendothelialization of endothelial progenitor cells from patients with hypertension. J Hum Hypertens 2020; 35:510-516. [PMID: 32555356 DOI: 10.1038/s41371-020-0364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 11/09/2022]
Abstract
Endothelial progenitor cells (EPCs) play an important role in endothelial vascular development and endothelial repair. The upregulation of functional gene expression in EPCs may contribute to the maintenance of EPC-based endothelial repair. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a powerful regulator of mitochondrial biogenesis and antioxidant defense, but its impact on early EPCs remains poorly understood. This study was designed to investigate whether decline in PGC-1α expression impairs in vitro adhesion, migration, and in vivo reendothelialization capacity of early EPCs from patients with hypertension. We engineered an over-expression of PGC-1α within EPCs from hypertensive patients, which were transduced with an adenoviral vector encoding the human PGC-1α gene. Then we tested the migration and adhesion function of EPCs in vitro and endothelium-reparative capacity in vivo with a nude mouse model of carotid artery injury. Our data revealed for the first time that PGC-1α expression of EPCs is lower in hypertensive patients than that in healthy subjects. Meanwhile, the in vitro adhesion and migration function, in vivo endothelial repair capacity of early EPCs were significantly reduced in hypertensive patients compared with normal subjects. Furthermore, PGC-1α gene transfer contributes to increased adhesion in vitro and enhanced endothelium-reparative capacity in vivo of EPCs from hypertensive patients through the augmented expression of PGC-1α. Thus, upregulation of PGC-1α expression of EPCs may be a novel complementary therapeutic target to increase endothelium-reparative capacity in hypertensive patients.
Collapse
Affiliation(s)
- Wenli Xie
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Fang Wu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jiang He
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | | | - Long Chen
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaoyu Zhang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jiapan Sun
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xing Liu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
28
|
Atypical chemokine receptor ACKR3/CXCR7 controls postnatal vasculogenesis and arterial specification by mesenchymal stem cells via Notch signaling. Cell Death Dis 2020; 11:307. [PMID: 32366833 PMCID: PMC7198625 DOI: 10.1038/s41419-020-2512-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are known to play a role in postnatal vasculogenesis and hold great promise for vascular regeneration. However, the mechanisms by which the endothelial differentiation and specification of MSCs remain unclear. We examined the potential role and molecular mechanisms of atypical chemokine receptor ACKR3/CXCR7 in MSC-mediated endothelial cell differentiation and specification. Here, we showed that vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) activate CXCR7 expression on MSCs through PDGF receptors, PDGFRα and PDGFRβ-mediated phosphoinositide 3-kinase (PI3K)/Akt signaling. Genetic and pharmacologic blockage of CXCR7 on MSCs suppressed the VEGF or stromal cell-derived factor 1 (SDF)-1-induced the capacity for vasculogenesis in vitro and in vivo. Moreover, CXCR7 gain of function markedly promoted vasculogenesis by MSCs in vitro and in vivo and induced endothelial differentiation along the arterial endothelial cell lineage via upregulation of Notch signaling. However, blockade of Notch signaling inhibited CXCR7-induced vasculogensis by MSCs. These results indicate CXCR7 is a critical regulator of MSC-mediated postnatal vasculogenesis and arterial specification via Notch signaling.
Collapse
|
29
|
Wu SH, Zhang F, Yao S, Tang L, Zeng HT, Zhu LP, Yang Z. Shear Stress Triggers Angiogenesis of Late Endothelial Progenitor Cells via the PTEN/Akt/GTPCH/BH4 Pathway. Stem Cells Int 2020; 2020:5939530. [PMID: 32399044 PMCID: PMC7210539 DOI: 10.1155/2020/5939530] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/03/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Shear stress is an effective modulator of endothelial progenitor cells (EPCs) and has been suggested to play an important role in angiogenesis. The phosphatase and tensin homolog (PTEN)/Akt and guanosine triphosphate cyclohydrolase (GTPCH)/tetrahydrobiopterin (BH4) pathways regulate the function of early EPCs. However, the role of these pathways in the shear stress-induced angiogenesis of late EPCs remains poorly understood. Therefore, we aim to investigate whether shear stress could upregulate the angiogenesis capacity of late EPCs and to further explore the possible underlying mechanisms. METHODS Late EPCs were subjected to laminar shear stress (LSS), and their in vitro migration, proliferation, and tube formation capacity were determined. In addition, the in vivo angiogenesis capacity was explored, along with the expression of molecules involved in the PTEN/Akt and GTPCH/BH4 pathways. RESULTS LSS elevated the in vitro activities of late EPCs, which were accompanied by downregulated PTEN expression, accelerated Akt phosphorylation, and GTPCH/BH4 pathway activation (all P < 0.05). Following Akt inhibition, LSS-induced upregulated GTPCH expression, BH4, and NO level of EPCs were suppressed. LSS significantly improved the migration, proliferation, and tube formation ability (15 dyn/cm2 LSS vs. stationary: 72.2 ± 5.5 vs. 47.3 ± 7.3, 0.517 ± 0.05 vs. 0.367 ± 0.038, and 1.664 ± 0.315 vs. 1 ± 0, respectively; all P < 0.05) along with the in vivo angiogenesis capacity of late EPCs, contributing to the recovery of limb ischemia. These effects were also blocked by Akt inhibition or GTPCH knockdown (P < 0.05, respectively). CONCLUSIONS This study provides the first evidence that shear stress triggers angiogenesis in late EPCs via the PTEN/Akt/GTPCH/BH4 pathway, providing a potential nonpharmacologic therapeutic strategy for promoting angiogenesis in ischemia-related diseases.
Collapse
Affiliation(s)
- Shao-Hong Wu
- Department of Ultrasound, The First Affiliated Hospital, Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, China 510080
| | - Feng Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, China 410008
| | - Shun Yao
- Department of Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science, Guangzhou, China 510080
| | - Lu Tang
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, China 410008
| | - Hai-Tao Zeng
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, 58, 2nd Village Cross Road, Guangzhou, China 510080
| | - Ling-Ping Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, China 410008
| | - Zhen Yang
- Division of Emergency Medicine, Department of General Internal Medicine, Department of Emergency Intensive Care Unit & Department of Cardiology & Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yet-Sen University, 58 2nd Zhongshan Road, Guangzhou, China 510080
| |
Collapse
|
30
|
Fan X, He L, Dai Q, He J, Chen X, Dai X, Zhang C, Sun D, Meng X, Sun S, Huang J, Chen J, Lin L, Chen L, Tan Y, Yan X. Interleukin-1β augments the angiogenesis of endothelial progenitor cells in an NF-κB/CXCR7-dependent manner. J Cell Mol Med 2020; 24:5605-5614. [PMID: 32239650 PMCID: PMC7214148 DOI: 10.1111/jcmm.15220] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/01/2019] [Accepted: 09/11/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are able to trigger angiogenesis, and pro‐inflammatory cytokines have beneficial effects on angiogenesis under physiological and pathological conditions. C‐X‐C chemokine receptor type 7 (CXCR‐7), receptor for stromal cell‐derived factor‐1, plays a critical role in enhancing EPC angiogenic function. Here, we examined whether CXCR7 mediates the pro‐angiogenic effects of the inflammatory cytokine interleukin‐1β (IL‐1β) in EPCs. EPCs were isolated by density gradient centrifugation and angiogenic capability was evaluated in vitro by Matrigel capillary formation assay and fibrin gel bead assay. IL‐1β elevated CXCR7 expression at both the transcriptional and translational levels in a dose‐ and time‐dependent manner, and blockade of the nuclear translocation of NF‐κB dramatically attenuated the IL‐1β‐mediated up‐regulation of CXCR7 expression. IL‐1β stimulation significantly promoted EPCs tube formation and this effect was largely impaired by CXCR7‐siRNA transfection. IL‐1β treatment stimulated extracellular signal‐regulated kinase 1/2 (Erk1/2) phosphorylation, and inhibition of Erk1/2 phosphorylation partially impaired IL‐1β‐induced tube formation of EPCs but without significant effects on CXCR7 expression. Moreover, blocking NF‐κB had no significant effects on IL‐1β‐stimulated Erk1/2 phosphorylation. These findings indicate that CXCR7 plays an important role in the IL‐1β‐enhanced angiogenic capability of EPCs and antagonizing CXCR7 is a potential strategy for inhibiting angiogenesis under inflammatory conditions.
Collapse
Affiliation(s)
- Xia Fan
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China.,Department of Endocrinology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luqing He
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Qiaoxia Dai
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Junhong He
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Xiangjuan Chen
- Department of Obstetrics and Gynecology, The first affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaozhen Dai
- School of Biomedicine, Chengdu Medical College, Chengdu, China
| | - Chi Zhang
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China.,Department of Endocrinology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Da Sun
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Xue Meng
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Shiyue Sun
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Jiameng Huang
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China.,School of Nursing, Wenzhou Medical University, Wenzhou, China
| | - Lin Lin
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liangmiao Chen
- Department of Endocrinology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Tan
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| | - Xiaoqing Yan
- Department of Pharmacy, Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
31
|
Santopaolo M, Sambataro M, Spinetti G, Madeddu P. Bone marrow as a target and accomplice of vascular complications in diabetes. Diabetes Metab Res Rev 2020; 36 Suppl 1:e3240. [PMID: 31840418 DOI: 10.1002/dmrr.3240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022]
Abstract
Peripheral vascular complications are common in diabetic patients. While pathogenic mechanisms have received much consideration, only recently regenerative processes captured attention. There is now a consensus that the bone marrow is a source of reparative cells and that this healing mechanism is lost in people with diabetes, especially in those suffering from ischemic complications. This failure was thought to occur due to a negative impact of diabetes on the mobilization of stem/progenitor cells with angiogenic properties from the bone marrow to the circulation. Moreover, those patients showing severely reduced bone marrow cell mobilization also bared a very high risk for adverse cardiovascular events. More recently, the structural integrity of the bone marrow was recognized to be altered because of the rarefaction of local microvasculature and innervation, thus mirroring anatomical features that typically occur in peripheral tissues. Ensuing hypoxia, nutrient starvation, and creation of an acidic and oxidative environment concur in causing the depletion of stem/progenitor cells in the endosteal niche and in forcing stromal cells to activate an adipogenesis program. Moreover, stem/progenitor cells acquire a pathogenic phenotype and, once mobilized, can pass harmful signalling molecules to vascular cells of peripheral tissues thereby contributing to ischemic complications. These new pieces of evidence indicate that the bone marrow should deserve more attention in the current care of critical limb ischemia and diabetic foot. Owing to powerful reserve capacities, the bone marrow integrity could be preserved and even rescued using rehabilitation programs and pharmacological treatments with consequent benefit for local and whole-organism homeostasis.
Collapse
Affiliation(s)
- Marianna Santopaolo
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| | - Maria Sambataro
- Endocrine, Metabolism, and Nutrition Disease Unit, Santa Maria di Ca' Foncello Hospital, Treviso, Italy
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Paolo Madeddu
- Bristol Heart Institute, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Bristol, UK
| |
Collapse
|
32
|
Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, Shamseddine A. Resistance Mechanisms to Anti-angiogenic Therapies in Cancer. Front Oncol 2020; 10:221. [PMID: 32175278 PMCID: PMC7056882 DOI: 10.3389/fonc.2020.00221] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor growth and metastasis rely on tumor vascular network for the adequate supply of oxygen and nutrients. Tumor angiogenesis relies on a highly complex program of growth factor signaling, endothelial cell (EC) proliferation, extracellular matrix (ECM) remodeling, and stromal cell interactions. Numerous pro-angiogenic drivers have been identified, the most important of which is the vascular endothelial growth factor (VEGF). The importance of pro-angiogenic inducers in tumor growth, invasion and extravasation make them an excellent therapeutic target in several types of cancers. Hence, the number of anti-angiogenic agents developed for cancer treatment has risen over the past decade, with at least eighty drugs being investigated in preclinical studies and phase I-III clinical trials. To date, the most common approaches to the inhibition of the VEGF axis include the blockade of VEGF receptors (VEGFRs) or ligands by neutralizing antibodies, as well as the inhibition of receptor tyrosine kinase (RTK) enzymes. Despite promising preclinical results, anti-angiogenic monotherapies led only to mild clinical benefits. The minimal benefits could be secondary to primary or acquired resistance, through the activation of alternative mechanisms that sustain tumor vascularization and growth. Mechanisms of resistance are categorized into VEGF-dependent alterations, non-VEGF pathways and stromal cell interactions. Thus, complementary approaches such as the combination of these inhibitors with agents targeting alternative mechanisms of blood vessel formation are urgently needed. This review provides an updated overview on the pathophysiology of angiogenesis during tumor growth. It also sheds light on the different pro-angiogenic and anti-angiogenic agents that have been developed to date. Finally, it highlights the preclinical evidence for mechanisms of angiogenic resistance and suggests novel therapeutic approaches that might be exploited with the ultimate aim of overcoming resistance and improving clinical outcomes for patients with cancer.
Collapse
Affiliation(s)
- Yolla Haibe
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Malek Kreidieh
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Hiba El Hajj
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ibrahim Khalifeh
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| |
Collapse
|
33
|
Kong L, Zuo R, Wang M, Wang W, Xu J, Chai Y, Guan J, Kang Q. Silencing MicroRNA-137-3p, which Targets RUNX2 and CXCL12 Prevents Steroid-induced Osteonecrosis of the Femoral Head by Facilitating Osteogenesis and Angiogenesis. Int J Biol Sci 2020; 16:655-670. [PMID: 32025213 PMCID: PMC6990928 DOI: 10.7150/ijbs.38713] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022] Open
Abstract
The main pathogenesis of steroid-induced osteonecrosis of the femoral head (SONFH) includes decreased osteogenic capacity of bone marrow-derived mesenchymal stem cells (BMSCs) and damaged blood supply to the femoral head. MicroRNAs (miRNAs) have been shown to play prominent roles in SONFH development. However, there is no report that a specific miRNA targeting two genes in two different pathogenic pathways has been applied to this disease. The present study investigated the effects of transplantation of miR-137-3p-silenced BMSCs on the prevention and early treatment of SONFH. First, western blotting and dual luciferase assays were employed to verify that miR-137-3p directly targets Runx2 and CXCL12. Then, silencing of miR-137-3p was found to facilitate osteogenic differentiation of BMSCs, which was confirmed by alkaline phosphatase (ALP) staining, alizarin red staining and qRT-PCR. Silencing of miR-137-3p also promoted angiogenesis by human umbilical vein endothelial cells (HUVECs) in the presence or absence of glucocorticoids. Thereafter, overexpression of Runx2 and CXCL12 without the 3′ untranslated region (3′UTR) partially rescued the effects of miR-137-3p on osteogenesis and angiogenesis, respectively. This finding further supported the hypothesis that miR-137-3p exerts its functions partly by regulating the genes, Runx2 and CXCL12. We also demonstrated that SONFH was partially prevented by transplantation of miR-137-3p-silenced BMSCs into a rat model. Micro-CT and histology showed that the transplantation of miR-137-3p-silenced BMSCs significantly improved bone regeneration. Additionally, the results of enzyme-linked immunosorbent assays (ELISA) and flow cytometry suggested that stromal cell-derived factor-1α (SDF-1α) and endothelial progenitor cells (EPCs) participated in the process of vascular repair. Taken together, these findings show that silencing of miR-137-3p directly targets the genes, Runx2 and CXCL12, which can play critical roles in SONFH repair by facilitating osteogenic differentiation and mobilizing EPCs.
Collapse
Affiliation(s)
- Lingchi Kong
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Rongtai Zuo
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Mengwei Wang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Wenbo Wang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jia Xu
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yimin Chai
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Junjie Guan
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Qinglin Kang
- Department of Orthopedic surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
34
|
Sabbah N, Tamari T, Elimelech R, Doppelt O, Rudich U, Zigdon-Giladi H. Predicting Angiogenesis by Endothelial Progenitor Cells Relying on In-Vitro Function Assays and VEGFR-2 Expression Levels. Biomolecules 2019; 9:biom9110717. [PMID: 31717420 PMCID: PMC6921061 DOI: 10.3390/biom9110717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Clinical trials have demonstrated the safety and efficacy of autologous endothelial progenitor cell (EPC) therapy in various diseases. Since EPCs' functions are influenced by genetic, systemic and environmental factors, the therapeutic potential of each individual EPCs is unknown and may affect treatment outcome. Therefore, our aim was to compare EPCs function among healthy donors in order to predict blood vessel formation (angiogenesis) before autologous EPC transplantation. Human EPCs were isolated from the blood of ten volunteers. EPCs proliferation rate, chemoattractant ability, and CXCR4 mRNA levels were different among donors (p < 0.0001, p < 0.01, p < 0.001, respectively). A positive correlation was found between SDF-1, CXCR4, and EPCs proliferation (R = 0.736, p < 0.05 and R = 0.8, p < 0.01, respectively). In-vivo, blood vessels were counted ten days after EPCs transplantation in a subcutaneous mouse model. Mean vessel density was different among donors (p = 0.0001); nevertheless, donors with the lowest vessel densities were higher compared to control (p < 0.05). Finally, using a linear regression model, a mathematical equation was generated to predict blood vessel density relying on: (i) EPCs chemoattractivity, and (ii) VEGFR-2 mRNA levels. Results reveal differences in EPCs functions among healthy individuals, emphasizing the need for a potency assay to pave the way for standardized research and clinical use of human EPCs.
Collapse
Affiliation(s)
- Nadin Sabbah
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
| | - Rina Elimelech
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- Department of Periodontology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Ofri Doppelt
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Utai Rudich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Hadar Zigdon-Giladi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
- Department of Periodontology, Rambam Health Care Campus, Haifa 3109601, Israel
- Correspondence: ; Tel.: +972-4-854-3606
| |
Collapse
|
35
|
Huang HT, Liu ZC, Wu KQ, Gu SR, Lu TC, Zhong CJ, Zhou YX. MiR-92a regulates endothelial progenitor cells (EPCs) by targeting GDF11 via activate SMAD2/3/FAK/Akt/eNOS pathway. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:563. [PMID: 31807544 DOI: 10.21037/atm.2019.09.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background The effects of miR-92a on EPCs are still poorly elucidated. This study aimed to investigate the effects of miR-92a on EPCs (Endothelial progenitor cells) in a model of hypoxia (HO) or high glucose (HG)-induced EPCs injury by targeting GDF11 (Differentiation growth factor 11). Methods The effects of miR-92a on EPCs subjected to HO or HG were investigated firstly. Subsequently, the action mechanism of miR-92a on EPCs by targeting GDF11 was elucidated. Proliferation, apoptosis, migration, angiogenesis was measured with MTT, flow cytometry, transwell, tube formation respectively. After 24 h, levels of reactive oxygen species (ROS) were measured by fluorescence intensity. LDH and NO (nitric oxide) levels were determined by ELISA. The expression of FLK-1 (fetal liver kinase 1) and vWF (von Willebrand factor) was detected by immunofluorescence. mRNA and protein expression levels were examined using PCR and western blotting respectively. The interaction between miR-92a and GDF11 was evaluated by dual-luciferase reporter assay. Results Our results showed that HO or HG increased apoptosis, production of LDH and generation of ROS, but decreased the ability of migration and tube formation and generation of NO in EPCs; inhibiting of miR-92a decreased HO or HG-induced injury of EPCs, whereas miR-92a over-expression had the opposite effect; the protective effects induced by inhibiting of miR-92a on EPCs could be reversed by GDF11 siRNA and the harmful effects induced by over-expression of miR-92a could be rescued by over-expression of GDF11, which showed that the harmful effects of miR-92a be related to its inhibition of GDF11 and subsequent inactivation of the SMAD2/3/FAK/Akt/eNOS signaling pathway. Conclusions Inhibiting miR-92a can protect EPCs from HO or HG-induced injury. The effect of miR-92a on EPCs are mediated by regulating of GDF11 and downstream SMAD2/3/FAK/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Hai-Tao Huang
- Department of Thoracic and Cardiovascular Surgery, Nantong First People's Hospital, Nantong 226001, China
| | - Zhen-Chuan Liu
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Kai-Qin Wu
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shao-Rui Gu
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tian-Cheng Lu
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chong-Jun Zhong
- Department of Thoracic and Cardiovascular Surgery, Nantong First People's Hospital, Nantong 226001, China
| | - Yong-Xin Zhou
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
36
|
Issa Bhaloo S, Wu Y, Le Bras A, Yu B, Gu W, Xie Y, Deng J, Wang Z, Zhang Z, Kong D, Hu Y, Qu A, Zhao Q, Xu Q. Binding of Dickkopf-3 to CXCR7 Enhances Vascular Progenitor Cell Migration and Degradable Graft Regeneration. Circ Res 2019; 123:451-466. [PMID: 29980568 PMCID: PMC6092110 DOI: 10.1161/circresaha.118.312945] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Vascular progenitor cells play key roles in physiological and pathological vascular remodeling—a process that is crucial for the regeneration of acellular biodegradable scaffolds engineered as vital strategies against the limited availability of healthy autologous vessels for bypass grafting. Therefore, understanding the mechanisms driving vascular progenitor cells recruitment and differentiation could help the development of new strategies to improve tissue-engineered vessel grafts and design drug-targeted therapy for vessel regeneration. Objective: In this study, we sought to investigate the role of Dkk3 (dickkopf-3), recently identified as a cytokine promotor of endothelial repair and smooth muscle cell differentiation, on vascular progenitor cells cell migration and vascular regeneration and to identify its functional receptor that remains unknown. Methods and Results: Vascular stem/progenitor cells were isolated from murine aortic adventitia and selected for the Sca-1 (stem cell antigen-1) marker. Dkk3 induced the chemotaxis of Sca-1+ cells in vitro in transwell and wound healing assays and ex vivo in the aortic ring assay. Functional studies to identify Dkk3 receptor revealed that overexpression or knockdown of chemokine receptor CXCR7 (C-X-C chemokine receptor type 7) in Sca-1+ cells resulted in alterations in cell migration. Coimmunoprecipitation experiments using Sca-1+ cell extracts treated with Dkk3 showed the physical interaction between DKK3 and CXCR7, and specific saturation binding assays identified a high-affinity Dkk3-CXCR7 binding with a dissociation constant of 14.14 nmol/L. Binding of CXCR7 by Dkk3 triggered the subsequent activation of ERK1/2 (extracellular signal-regulated kinases 1/2)-, PI3K (phosphatidylinositol 3-kinase)/AKT (protein kinase B)-, Rac1 (Ras-related C3 botulinum toxin substrate 1)-, and RhoA (Ras homolog gene family, member A)-signaling pathways involved in Sca-1+ cell migration. Tissue-engineered vessel grafts were fabricated with or without Dkk3 and implanted to replace the rat abdominal aorta. Dkk3-loaded tissue-engineered vessel grafts showed efficient endothelization and recruitment of vascular progenitor cells, which had acquired characteristics of mature smooth muscle cells. CXCR7 blocking using specific antibodies in this vessel graft model hampered stem/progenitor cell recruitment into the vessel wall, thus compromising vascular remodeling. Conclusions: We provide a novel and solid evidence that CXCR7 serves as Dkk3 receptor, which mediates Dkk3-induced vascular progenitor migration in vitro and in tissue-engineered vessels, hence harnessing patent grafts resembling native blood vessels.
Collapse
Affiliation(s)
- Shirin Issa Bhaloo
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Yifan Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China (Y.W., Z.W., D.K., Q.Z.)
| | - Alexandra Le Bras
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (B.Y., A.Q.)
| | - Wenduo Gu
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Yao Xie
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Jiacheng Deng
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Zhihong Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China (Y.W., Z.W., D.K., Q.Z.)
| | - Zhongyi Zhang
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China (Y.W., Z.W., D.K., Q.Z.)
| | - Yanhua Hu
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (B.Y., A.Q.)
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China (Y.W., Z.W., D.K., Q.Z.)
| | - Qingbo Xu
- From the School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (S.I.B., A.L.B., W.G., Y.X., J.D., Z.Z., Y.H., Q.X.)
| |
Collapse
|
37
|
Ding P, Zhang W, Tan Q, Yao C, Lin S. Impairment of circulating endothelial progenitor cells (EPCs) in patients with glucocorticoid-induced avascular necrosis of the femoral head and changes of EPCs after glucocorticoid treatment in vitro. J Orthop Surg Res 2019; 14:226. [PMID: 31324193 PMCID: PMC6642597 DOI: 10.1186/s13018-019-1279-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Background Avascular necrosis of the femoral head (ANFH) is a severe complication after high-dose glucocorticoid (GC) administration. The pathogenesis of GC-induced ANFH remains unclear. Though the important role of endothelial progenitor cells (EPCs) in the progression of GC-induced ANFH has been noticed, the effects of GCs on EPCs and the underlying mechanism still need further study. Methods Circulating EPCs were obtained from the peripheral blood of ANFH patients and healthy controls by Ficoll-density gradient centrifugation. CD133+CD34+ cells with DiI-Ac-LDL uptake and FITC-UEA-1 binding were considered as EPCs. Number and functions of EPCs were analyzed by flow cytometry, chemotaxis assay, and tube formation assay. EPCs from healthy controls were also treated by different concentrations of methylprednisolone and prednisolone in vitro, and cell growth and angiogenic function were evaluated. Expression of CXCR7 and its downstream Akt/GSK-3β/Fyn pathway were also analyzed by western blots after cells treated by methylprednisolone in vitro. Results The number and functions of EPCs in patients with GC-induced ANFH were significantly decreased. In vitro study showed for the first time that except extremely high concentrations, low to medium concentrations of GCs did not have significant effects on EPCs’ growth. Methylprednisolone and prednisolone both inhibited angiogenesis of EPCs even at low concentrations. Mechanism studies found CXCR7 was downregulated in EPCs after methylprednisolone treatment in vitro. Expression and phosphorylation of Akt and GSK-3β were also decreased with an upregulation of Fyn expression after steroid treatment. Conclusions Our study showed that GC-induced ANFH patients have reduced the number and impaired functions of circulating EPCs. GCs did not show a significant effect on the growth of EPCs in vitro except extremely high concentrations of GCs. However, GCs significantly impaired EPC angiogenic function in vitro, even at low concentrations. Our study also suggested that downregulation of CXCR7 and its downstream Akt/GSK-3β/Fyn pathway in EPCs might be a novel mechanism of how GCs suppress EPCs’ angiogenesis.
Collapse
Affiliation(s)
- Peng Ding
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Wen Zhang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Qiyuan Tan
- Department of Endocrinology, Shanghai Jiaotong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Chen Yao
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Sen Lin
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
38
|
Liu L, Leng S, Yue J, Lu Q, Xu W, Yi X, Huang D, Zhang L. EDTA Enhances Stromal Cell–derived Factor 1α–induced Migration of Dental Pulp Cells by Up-regulating Chemokine Receptor 4 Expression. J Endod 2019; 45:599-605.e1. [DOI: 10.1016/j.joen.2019.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 12/18/2022]
|
39
|
Wang P, Zhang H, Li Z, Liu X, Jin Y, Lei M, Jiao Z, Bi Y, Guo W. Low-Dose Radiation Promotes the Proliferation and Migration of AGE-Treated Endothelial Progenitor Cells Derived from Bone Marrow via Activating SDF-1/CXCR4/ERK Signaling Pathway. Radiat Res 2019; 191:518-526. [PMID: 30925138 DOI: 10.1667/rr15200.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Low-dose radiation (LDR) has been confirmed to mobilize bone marrow-derived endothelial progenitor cells (EPCs) and promote diabetic wound healing. But it is unclear whether LDR acts directly on EPCs and promotes their proliferation and migration. Given the key role of advanced glycosylation end products (AGE) in the pathogenesis of diabetes, we used AGE to induce EPC damage. We then investigated the effect of LDR on the proliferation and migration of AGE-treated EPCs and explored the underlying mechanisms. EPCs cultured in vitro were treated with different concentrations of AGE, and the cells were then exposed to different low doses and treated with a specific antagonist for CXCR4, AMD3100 (1 lmol/l). The proliferation and migration abilities of EPCs were detected using the CCK-8 and wound healing assays, respectively. The mRNA and protein expression of SDF-1 and CXCR4 in AGE-treated EPCs were measured using qPCR and Western blot analysis, respectively. The expressions of ERK and phosphorylated ERK (pERK) were detected using Western blot analysis. The results showed that 200 mg/l and 400 mg/l AGE had an inhibitory effect on the proliferation of EPCs, and this inhibitory effect was exerted in a dose- and time-dependent manner. AGE significantly reduced the migration ability of EPCs cultured in vitro. After the cells received either 50 or 75 mGy low-dose irradiation, the proliferation of EPCs and AGE-treated EPCs was clearly increased; in addition, LDR also enhanced cell migration ability, but this enhancement was counteracted by AMD3100. Results from qPCR and Western blot analysis showed that LDR increased the mRNA and protein expression of SDF-1/ CXCR4. LDR also upregulated pERK expression in EPCs and AGE-treated EPCs, but LDR-induced upregulation of pERK expression was inhibited by AMD3100. These findings indicate that LDR can directly activate the SDF-1/CXCR4 biological axis and downstream ERK signaling pathway, and promote the proliferation and migration abilities of EPCs by increasing the expression of SDF-1, CXCR4 and pERK in EPCs.
Collapse
Affiliation(s)
- Ping Wang
- Departments of a Otolaryngology-Head and Neck Surgery
| | | | - Zhuo Li
- c Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Xiaobo Liu
- Departments of a Otolaryngology-Head and Neck Surgery
| | - Yingli Jin
- d Department of Pharmacology, College of Basic Medical Science, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Manman Lei
- c Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Zixuan Jiao
- c Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Yaru Bi
- c Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Weiying Guo
- c Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| |
Collapse
|
40
|
Gu X, Wang XQ, Lin MJ, Liang H, Fan SY, Wang L, Yan X, Liu W, Shen FX. Molecular interplay between microRNA-130a and PTEN in palmitic acid-mediated impaired function of endothelial progenitor cells: Effects of metformin. Int J Mol Med 2019; 43:2187-2198. [PMID: 30896786 DOI: 10.3892/ijmm.2019.4140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Metformin serves an important role in improving the functions of endothelial progenitor cells (EPCs). MicroRNAs (miRNAs), small non‑coding RNAs, have been investigated as significant regulators of EPC vascular functions. The present study investigated the molecular crosstalk between metformin and miRNA‑130a (miR‑130a) in the functions of EPCs exposed to palmitic acid (PA). Isolated EPCs were treated with metformin, PA, and metformin + PA, respectively. Cell Counting Kit‑8, Transwell and Matrigel assays were performed to detect the proliferation, migration and tube formation ability of EPCs following different treatments. The expression of miR‑130a, phosphatase and tensin homolog (PTEN) and phosphorylated‑AKT was analyzed by reverse transcription‑quantitative polymerase chain reaction and western blotting. The specific mechanism underlying the function of metformin in EPCs was further elucidated by transfecting miR‑130a mimics and inhibitor to overexpress and inhibit the expression of miR‑130a in EPCs, respectively. EPCs exhibited impaired functions of proliferation (P<0.01 compared with the control), migration (P<0.01 compared with the control) and tube formation (P<0.01 compared with the control) following treatment with PA, and the expression levels of miR‑130a and PTEN were decreased and increased, respectively. However, the presence of metformin, or the overexpression of miR‑130a using miR‑130a mimic alleviated the impairment of angiogenesis and proliferation, decreased the expression of PTEN and activated the phosphoinositide‑3 kinase/AKT pathway in EPCs exposed to PA. By contrast, downregulating the expression of miR‑130a with a miR‑130a inhibitor reversed the metformin‑mediated protection. These results demonstrate the beneficial effect of miR‑130a/PTEN on EPC functions, which can be regulated by metformin. The effects of metformin on improving PA‑induced EPC dysfunction are mediated by miR‑130a and PTEN, which may assist in the prevention and/or treatment of diabetic vascular disease.
Collapse
Affiliation(s)
- Xuemei Gu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiao-Qian Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Min-Jie Lin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Haili Liang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Shi-Yan Fan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Luyin Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoqing Yan
- School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wenyue Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fei-Xia Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
41
|
Li JS, Li B. Renal Injury Repair: How About the Role of Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:661-670. [PMID: 31399989 DOI: 10.1007/978-981-13-8871-2_32] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Renal failure is one of the most important causes of mortality and morbidity all over the world. Acute kidney injury (AKI) is a major clinical problem that affects up to 5% of all hospitalized patients. Although the kidney has a remarkable capacity for regeneration after acute injury, the mortality among patients with severe AKI remains dismally high, and in clinical practice, most patients cannot be cured completely and suffer from chronic kidney disease (CKD). Recently, the incidence and prevalence of CKD have increased, largely as a result of the enhanced prevalence of diabetes and obesity. The progressive nature of CKD and the ensuing end-stage renal disease (ESRD) place a substantial burden on global healthcare resources. Currently, dialysis and transplantation remain the only treatment options. Finding new therapeutic methods to fight AKI and CKD remains an ongoing quest. Although the human renal histological structure is complex, stem cell therapies have been applied to repair injured kidneys. The curative effects of mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and nephron progenitor cells (NPCs) on renal repair have also been reported by researchers. This review focuses on stem cell therapy and mechanisms for renal injury repair.
Collapse
Affiliation(s)
- Jian-Si Li
- Department of Nephrology, 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bing Li
- Department of Nephrology, 2nd Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
42
|
Darvishi B, Majidzadeh-A K, Ghadirian R, Mosayebzadeh M, Farahmand L. Recruited bone marrow derived cells, local stromal cells and IL-17 at the front line of resistance development to anti-VEGF targeted therapies. Life Sci 2018; 217:34-40. [PMID: 30472294 DOI: 10.1016/j.lfs.2018.11.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022]
Abstract
Although anti-angiogenic agents targeting VEGF have shown affordable beneficial outcomes in several human cancer types, in most pre-clinical and clinical studies, these effects are transient and followed by rapid relapse and tumor regrowth. Recently, it has been suggested that recruited bone marrow derived cells (BMDCs) to the tumor-microenvironment together with stromal cells play an important role in development of resistance to anti-VEGF therapies. Additionally, acquired resistance to anti-VEGF therapies has shown to be mediated partly through overexpression of different pro-angiogenic cytokines and growth factors including G-CSF, IL-6, IL-8, VEGF and FGF by these cells. Alongside, IL-17, a pro-inflammatory cytokine, mostly secreted by infiltrated CD4+ T helper cells, has shown to mediate resistance to anti-VEGF therapies, through recruiting BMDCs and modulating stromal cells activities including endothelial cells, tumor associated macrophages and cancer associated fibroblasts. Here, we examined the role of BMDCs, tumor stromal cells, IL-17 and their negotiation in development of resistance to anti-VEGF targeted therapies.
Collapse
Affiliation(s)
- Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Tasnim Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reihane Ghadirian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Marjan Mosayebzadeh
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
43
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
44
|
Wang C, Chen W, Shen J. CXCR7 Targeting and Its Major Disease Relevance. Front Pharmacol 2018; 9:641. [PMID: 29977203 PMCID: PMC6021539 DOI: 10.3389/fphar.2018.00641] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Chemokine receptors are the target of small peptide chemokines. They play various important roles in physiological and pathological processes. CXCR7, later renamed ACKR3, is a non-classical seven transmembrane-spanning receptor whose function as a signaling or non-signaling scavenger/decoy receptor is currently under debate. Even for cell signaling mechanisms, there has been inconsistency on whether CXCR7 couples to G-proteins or β-arrestins. Several reasons may contribute to this uncertainty or controversy. In one hand, it has been neglected that CXCR7 has more than five natural ligands and unfortunately, most of the prior research only studied SDF-1 (CXCL12) and/or I-TAC (CXCL11); on the other hand, there are mounting evidence supporting ligand and tissue bias for receptor signaling, but limited such information is available for CXCR7. In this review we focus on summarizing the endogenous and exogenous ligands of CXCR7, the main diseases related to CXCR7 and the biased signaling events happening on CXCR7. These three aspects of CXCR7 pharmacologic properties may explain why the contradicting opinions of whether CXCR7 is a signaling or non-signaling receptor exist. Further, potential new direction and perspective for the study of CXCR7 biology and pharmacology are highlighted.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Weilin Chen
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jianzhong Shen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| |
Collapse
|
45
|
The 5T4 oncofetal glycoprotein does not act as a general organizer of the CXCL12 system in cancer cells. Exp Cell Res 2018; 364:175-183. [PMID: 29408206 DOI: 10.1016/j.yexcr.2018.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/07/2023]
Abstract
The chemokine, CXCL12, promotes cancer growth and metastasis through interaction with either CXCR4 and/or CXCR7. This tumor-specific organization of the CXCL12 system obscures current therapeutic approaches, aiming at the selective inactivation of CXCL12 receptors. Since it has been previously suggested that the cellular use of CXCR4 or CXCR7 is dictated by the 5T4 oncofetal glycoprotein, we have now tested whether 5T4 would represent a general and reliable marker for the organization of the CXCL12 system in cancer cells. The CXCR4 antagonist, AMD3100, as well as the CXCR7 antagonist, CCX771, demonstrated that the cancer cell lines A549, C33A, DLD-1, MDA-231, and PC-3 use either CXCR7 and/or CXCR4 for mediating CXCL12-induced chemotaxis and cell proliferation. The use of CXCL12 receptors as well as their subcellular localization remained unchanged in most cell lines following siRNA-mediated depletion of 5T4. In distinct cell lines, inhibition of 5T4 expression, however, modulated tumor cell migration and proliferation per se. Collectively our analyses fail to demonstrate general organizational influences of 5T4 of the CXCL12 system in different cancer cell lines, and, hence, dismiss its future use as a diagnostic marker.
Collapse
|
46
|
Dai X, Zeng J, Yan X, Lin Q, Wang K, Chen J, Shen F, Gu X, Wang Y, Chen J, Pan K, Cai L, Wintergerst KA, Tan Y. Sitagliptin-mediated preservation of endothelial progenitor cell function via augmenting autophagy enhances ischaemic angiogenesis in diabetes. J Cell Mol Med 2018; 22:89-100. [PMID: 28799229 PMCID: PMC5742710 DOI: 10.1111/jcmm.13296] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 05/27/2017] [Indexed: 01/13/2023] Open
Abstract
Recently, the dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin, a major anti-hyperglycaemic agent, has received substantial attention as a therapeutic target for cardiovascular diseases via enhancing the number of circulating endothelial progenitor cells (EPCs). However, the direct effects of sitagliptin on EPC function remain elusive. In this study, we evaluated the proangiogenic effects of sitagliptin on a diabetic hind limb ischaemia (HLI) model in vivo and on EPC culture in vitro. Treatment of db/db mice with sitagliptin (Januvia) after HLI surgery efficiently enhanced ischaemic angiogenesis and blood perfusion, which was accompanied by significant increases in circulating EPC numbers. EPCs derived from the bone marrow of normal mice were treated with high glucose to mimic diabetic hyperglycaemia. We found that high glucose treatment induced EPC apoptosis and tube formation impairment, which were significantly prevented by sitagliptin pretreatment. A mechanistic study found that high glucose treatment of EPCs induced dramatic increases in oxidative stress and apoptosis; pretreatment of EPCs with sitagliptin significantly attenuated high glucose-induced apoptosis, tube formation impairment and oxidative stress. Furthermore, we found that sitagliptin restored the basal autophagy of EPCs that was impaired by high glucose via activating the AMP-activated protein kinase/unc-51-like autophagy activating kinase 1 signalling pathway, although an autophagy inhibitor abolished the protective effects of sitagliptin on EPCs. Altogether, the results indicate that sitagliptin-induced preservation of EPC angiogenic function results in an improvement of diabetic ischaemia angiogenesis and blood perfusion, which are most likely mediated by sitagliptin-induced prevention of EPC apoptosis via augmenting autophagy.
Collapse
Affiliation(s)
- Xiaozhen Dai
- Chinese‐American Research Institute for Diabetic ComplicationsSchool of Pharmaceutical SciencesSchool of Nursing at the Wenzhou Medical UniversityWenzhouChina
- School of BiomedicineChengdu Medical CollegeChengduChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
| | - Jun Zeng
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
- Department of Medical Genetics and Cell BiologySchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Xiaoqing Yan
- Chinese‐American Research Institute for Diabetic ComplicationsSchool of Pharmaceutical SciencesSchool of Nursing at the Wenzhou Medical UniversityWenzhouChina
| | - Qian Lin
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
- Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleUSA
| | - Kai Wang
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
- Departments of Pediatrics, Endocrinology and Metabolismthe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jing Chen
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
| | - Feixia Shen
- Departments of Pediatrics, Endocrinology and Metabolismthe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xuemei Gu
- Departments of Pediatrics, Endocrinology and Metabolismthe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yuehui Wang
- Departments of Geriatricsthe First Hospital of Jilin UniversityChangchunChina
| | - Jun Chen
- Chinese‐American Research Institute for Diabetic ComplicationsSchool of Pharmaceutical SciencesSchool of Nursing at the Wenzhou Medical UniversityWenzhouChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
| | - Kejian Pan
- School of BiomedicineChengdu Medical CollegeChengduChina
| | - Lu Cai
- Chinese‐American Research Institute for Diabetic ComplicationsSchool of Pharmaceutical SciencesSchool of Nursing at the Wenzhou Medical UniversityWenzhouChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
- Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleUSA
| | - Kupper A. Wintergerst
- Department of PediatricsDivision of EndocrinologyWendy L. Novak Diabetes Care CenterUniversity of LouisvilleLouisvilleKYUSA
| | - Yi Tan
- Chinese‐American Research Institute for Diabetic ComplicationsSchool of Pharmaceutical SciencesSchool of Nursing at the Wenzhou Medical UniversityWenzhouChina
- Pediatric Research InstituteDepartment of PediatricsUniversity of Louisville School of MedicineLouisvilleUSA
- Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleUSA
| |
Collapse
|
47
|
Chang PY, Zhang BY, Cui S, Qu C, Shao LH, Xu TK, Qu YQ, Dong LH, Wang J. MSC-derived cytokines repair radiation-induced intra-villi microvascular injury. Oncotarget 2017; 8:87821-87836. [PMID: 29152123 PMCID: PMC5675675 DOI: 10.18632/oncotarget.21236] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/26/2017] [Indexed: 02/06/2023] Open
Abstract
Microvascular injury initiates the pathogenesis of radiation enteropathy. As previously demonstrated, the secretome from mesenchymal stem cells contains various angiogenic cytokines that exhibited therapeutic potential for ischemic lesions. As such, the present study aimed to investigate whether cytokines derived from mesenchymal stem cells can repair endothelial injuries from irradiated intestine. Here, serum-free medium was conditioned by human adipose-derived mesenchymal stem cells, and we found that there were several angiogenic cytokines in the medium, including IL-8, angiogenin, HGF and VEGF. This medium promoted the formation of tubules between human umbilical cord vein endothelial cells and protected these cells against radiation-induced apoptosis in vitro. Likewise, our in vivo results revealed that repeated injections of mesenchymal stem cell-conditioned medium could accelerate the recovery of irradiated mice by reducing the serum levels of pro-inflammatory cytokines, including IL-1α, IL-6 and TNF-α, and promoting intra-villi angiogenesis. Herein, intervention by conditioned medium could increase the number of circulating endothelial progenitors, whereas neutralizing SDF-1α and/or inhibiting PI3K would hamper the recruitment of endothelial progenitors to the injured sites. Such results suggested that SDF-1α and PI3K-mediated phosphorylation were required for intra-villi angiogenesis. To illustrate this, we found that conditioned medium enabled endothelial cells to increase intracellular levels of phosphorylated Akt Ser473, both under irradiated and steady state conditions, and to up-regulate the expression of the CXCR4 and CXCR7 genes. Collectively, the present results revealed the therapeutic effects of mesenchymal stem cell-derived cytokines on microvascular injury of irradiated intestine.
Collapse
Affiliation(s)
- Peng-Yu Chang
- State Key Laboratory of Electroanalytical Chemistry, Chinese Academy of Sciences, Changchun Jilin 130022, P.R. China.,Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Bo-Yin Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Shuang Cui
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Chao Qu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Li-Hong Shao
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Tian-Kai Xu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Ya-Qin Qu
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Li-Hua Dong
- Department of Radiation Oncology, First Bethune Hospital of Jilin University, Changchun 130021, P.R. China
| | - Jin Wang
- State Key Laboratory of Electroanalytical Chemistry, Chinese Academy of Sciences, Changchun Jilin 130022, P.R. China.,Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA
| |
Collapse
|
48
|
CXCL12 enhances angiogenesis through CXCR7 activation in human umbilical vein endothelial cells. Sci Rep 2017; 7:8289. [PMID: 28811579 PMCID: PMC5557870 DOI: 10.1038/s41598-017-08840-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is the process by which new vessels form from existing vascular networks. Human umbilical vein endothelial cells (HUVECs) may contribute to the study of vascular repair and angiogenesis. The chemokine CXCL12 regulates multiple cell functions, including angiogenesis, mainly through its receptor CXCR4. In contrast to CXCL12/CXCR4, few studies have described roles for CXCR7 in vascular biology, and the downstream mechanism of CXCR7 in angiogenesis remains unclear. The results of the present study showed that CXCL12 dose-dependently enhanced angiogenesis in chorioallantoic membranes (CAMs) and HUVECs. The specific activation of CXCR7 with TC14012 (a CXCR7 agonist) resulted in the significant induction of tube formation in HUVECs and in vivo. Further evidence suggested that CXCL12 induced directional polarization and migration in the HUVECs, which is necessary for tube formation. Moreover, CXCR7 translocalization was observed during the polarization of HUVECs in stripe assays. Finally, treatment with TC14012 also significantly increased PI3K/Akt phosphorylation, and tube formation was blocked by treating HUVECs with an Akt inhibitor. Overall, this study indicated that CXCL12-stimulated CXCR7 acts as a functional receptor to activate Akt for angiogenesis in HUVECs and that CXCR7 may be a potential target molecule for endothelial regeneration and repair after vascular injury.
Collapse
|
49
|
Bai YP, Xiao S, Tang YB, Tan Z, Tang H, Ren Z, Zeng H, Yang Z. Shear stress-mediated upregulation of GTP cyclohydrolase/tetrahydrobiopterin pathway ameliorates hypertension-related decline in reendothelialization capacity of endothelial progenitor cells. J Hypertens 2017; 35:784-797. [PMID: 28033126 DOI: 10.1097/hjh.0000000000001216] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Guanosine triphosphate cyclohydrolase/tetrahydrobiopterin (GTPCH)/(BH4) pathway has been proved to regulate the function of endothelial progenitor cells (EPCs) in deoxycorticosterone acetate-salt hypertensive mice, indicating that GTPCH/BH4 pathway may be an important repair target for hypertension-related endothelial injury. Shear stress is an important nonpharmacologic strategy to modulate the function of EPCs. Here, we investigated the effects of laminar shear stress on the GTPCH/BH4 pathway and endothelial repair capacity of circulating EPCs in hypertension. METHOD Laminar shear stress was loaded on the human EPCs from hypertensive patients and normotensive patients. The in-vitro function, in-vivo reendothelialization capacity and GTPCH/BH4 pathway of human EPCs were evaluated. RESULTS Both in-vitro function and reendothelialization capacity of EPCs were lower in hypertensive patients than that in normotensive patients. The GTPCH/BH4 pathway of EPCs was downregulated in hypertensive patients. Shear stress increased in-vitro function and reendothelialization capacity of EPCs from hypertensive patients and normotensive patients. Furthermore, shear stress upregulated the expression of GTPCH I and levels of BH4, nitric oxide, and cGMP of EPCs, and reduced thrombospondin-1 expression. With treatment of GTPCH knockdown or nitroarginine methyl ester inhibition, shear stress-induced increased levels of BH4, nitric oxide and cGMP of EPCs was suppressed. When GTPCH/BH4 pathway of EPCs was blocked, the effects of shear stress on in-vitro function and reendothelialization capacity of EPCs were inhibited. CONCLUSION The study demonstrates for the first time that shear stress-induced upregulation of the GTPCH/BH4 pathway ameliorates hypertension-related decline in endothelial repair capacity of EPCs. These findings provide novel nonpharmacologic therapeutic approach for hypertension-related endothelial repair.
Collapse
Affiliation(s)
- Yong-Ping Bai
- aDepartment of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan bDepartment of Neurology, Sun Yat-Sen Memorial Hospital cDepartment of Pharmacology, Zhongshan School of Medicine dDepartment of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University eSun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine fCenter for Reproductive Medicine, The Sixth Affiliated Hospital gDepartment of Hypertension & Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Dai X, Yan X, Zeng J, Chen J, Wang Y, Chen J, Li Y, Barati MT, Wintergerst KA, Pan K, Nystoriak MA, Conklin DJ, Rokosh G, Epstein PN, Li X, Tan Y. Elevating CXCR7 Improves Angiogenic Function of EPCs via Akt/GSK-3β/Fyn-Mediated Nrf2 Activation in Diabetic Limb Ischemia. Circ Res 2017; 120:e7-e23. [PMID: 28137917 DOI: 10.1161/circresaha.117.310619] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Endothelial progenitor cells (EPCs) respond to stromal cell-derived factor 1 (SDF-1) through chemokine receptors CXCR7 and CXCR4. Whether SDF-1 receptors involves in diabetes mellitus-induced EPCs dysfunction remains unknown. OBJECTIVE To determine the role of SDF-1 receptors in diabetic EPCs dysfunction. METHODS AND RESULTS CXCR7 expression, but not CXCR4 was reduced in EPCs from db/db mice, which coincided with impaired tube formation. Knockdown of CXCR7 impaired tube formation of EPCs from normal mice, whereas upregulation of CXCR7 rescued angiogenic function of EPCs from db/db mice. In normal EPCs treated with oxidized low-density lipoprotein or high glucose also reduced CXCR7 expression, impaired tube formation, and increased oxidative stress and apoptosis. The damaging effects of oxidized low-density lipoprotein or high glucose were markedly reduced by SDF-1 pretreatment in EPCs transduced with CXCR7 lentivirus but not in EPCs transduced with control lentivirus. Most importantly, EPCs transduced with CXCR7 lentivirus were superior to EPCs transduced with control lentivirus for therapy of ischemic limbs in db/db mice. Mechanistic studies demonstrated that oxidized low-density lipoprotein or high glucose inhibited protein kinase B and glycogen synthase kinase-3β phosphorylation, nuclear export of Fyn and nuclear localization of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), blunting Nrf2 downstream target genes heme oxygenase-1, NAD(P)H dehydrogenase (quinone 1) and catalase, and inducing an increase in EPC oxidative stress. This destructive cascade was blocked by SDF-1 treatment in EPCs transduced with CXCR7 lentivirus. Furthermore, inhibition of phosphatidylinositol 3-kinase/protein kinase B prevented SDF-1/CXCR7-mediated Nrf2 activation and blocked angiogenic repair. Moreover, Nrf2 knockdown almost completely abolished the protective effects of SDF-1/CXCR7 on EPC function in vitro and in vivo. CONCLUSIONS Elevated expression of CXCR7 enhances EPC resistance to diabetes mellitus-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia. The benefits of CXCR7 are mediated predominantly by a protein kinase B/glycogen synthase kinase-3β/Fyn pathway via increased activity of Nrf2.
Collapse
Affiliation(s)
- Xiaozhen Dai
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Xiaoqing Yan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jun Zeng
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jing Chen
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yuehui Wang
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Jun Chen
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yan Li
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Michelle T Barati
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Kupper A Wintergerst
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Kejian Pan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Matthew A Nystoriak
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Daniel J Conklin
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Gregg Rokosh
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Paul N Epstein
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Xiaokun Li
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.)
| | - Yi Tan
- From the Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences & School of Nursing at the Wenzhou Medical University, Wenzhou, China (X.D., X.Y., Jun Chen, X.L., Y.T.); School of Biomedicine, Chengdu Medical College, China (X.D., K.P.); Department of Pediatrics, Children's Hospital Research Institute, School of Medicine (X.D., J.Z., Jing Chen, Jun Chen, P.N.E., Y.T.), Department of Surgery (Y.L.), Department of Medicine (M.T.B., M.A.N., D.J.C.), Division of Endocrinology, Department of Pediatrics, Wendy L. Novak Diabetes Care Center (K.A.W.), and Diabetes and Obesity Center (D.J.C.), University of Louisville, KY; Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China (Y.W.); and Division of Cardiovascular Disease, University of Alabama at Birmingham (G.R.).
| |
Collapse
|