1
|
Gyorffy VJ, Dwivedi DJ, Liaw PC, Fox-Robichaud AE, Tsang JLY, Binnie A. Impact of sample processing delays on plasma markers of inflammation, chemotaxis, cell death, and blood coagulation. PLoS One 2024; 19:e0311921. [PMID: 39480839 PMCID: PMC11527306 DOI: 10.1371/journal.pone.0311921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Biosampling studies in critically ill patients traditionally involve bedside collection of samples followed by local processing (ie. centrifugation, aliquotting, and freezing) and storage. However, community hospitals, which care for the majority of Canadian patients, often lack the infrastructure for local processing and storage of specimens. A potential solution is a "simplified" biosampling protocol whereby blood samples are collected at the bedside and then shipped to a central site for processing and storage. One potential limitation of this approach is that delayed processing may alter sample characteristics. OBJECTIVE To determine whether delays in blood sample processing affect the stability of cytokines (IL-6, TNF, IL-10, IFN-γ), chemokines (IL-8, IP-10, MCP-1, MCP-4, MIP-1α, MIP-1β), cell-free DNA (cfDNA) (released by dying cells), and blood clotting potential in human blood samples. METHODS Venous blood was collected into EDTA and citrate sample tubes and stored at room temperature (RT) or 4°C for progressive intervals up to 72 hours, prior to processing. Plasma cytokines and chemokines were quantified using single or multiplex immunoassays. cfDNA was measured using Picogreen DNA Quantification. Blood clotting potential was measured using a thrombin generation assay. RESULTS Blood samples were collected from 9 intensive care unit (ICU) patients and 7 healthy volunteers. Admission diagnoses for the ICU patients included sepsis, trauma, ruptured abdominal aortic aneurysm, intracranial hemorrhage, gastrointestinal bleed, and hyperkalemia. After pre-processing delays of up to 72 hours at RT or 4°C, no significant changes were observed in plasma cytokines, chemokines, cfDNA, or thrombin formation. CONCLUSIONS Delayed sample processing for up to 72 hours at either RT or 4°C did not significantly affect cytokines, chemokines, cfDNA, or blood clotting potential in plasma samples from healthy volunteers and ICU patients. A "simplified" biosampling protocol is a feasible solution for conducting biosampling research at hospitals without local processing capacity.
Collapse
Affiliation(s)
- Vanessa J. Gyorffy
- Faculty of Arts and Science, McMaster University, Hamilton, ON, Canada
- Niagara Health Knowledge Institute, St. Catharines, ON, Canada
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
| | - Dhruva J. Dwivedi
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Patricia C. Liaw
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alison E. Fox-Robichaud
- Thrombosis and Atherosclerosis Research Institute (TaARI), McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer L. Y. Tsang
- Niagara Health Knowledge Institute, St. Catharines, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexandra Binnie
- Critical Care Department, William Osler Health System, Etobicoke, ON, Canada
| |
Collapse
|
2
|
ElMenshawy N, Ibrahim Fouda M, Mofreh M, Hisham El-Etriby H, O Elnenaei M, Eissa M. Impact of CD34/CD309 expression in circulating endothelial progenitor cells on prognosis and response to bortezomib therapy in multiple myeloma. J Immunoassay Immunochem 2024; 45:481-491. [PMID: 39135454 DOI: 10.1080/15321819.2024.2388614] [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] [Indexed: 09/03/2024]
Abstract
Multiple myeloma (MM) is a prevalent yet incurable hematologic malignancy. Despite the proven efficacy of proteasome inhibitors in treating MM, resistance to Bortezomib-based treatments persists in a subset of patients. This case control study explores the potential of circulating endothelial progenitor cells (EPCs) as biomarkers for predicting response to Proteasome Inhibitor based therapy combined with Dexamethasone in MM patients. This study was conducted on 105 MM patients receiving bortezomib plus dexamethasone therapy and 90 healthy individuals as a control group. Utilizing 8-color multi-parameter flow cytometry, we assessed the levels of circulating EPCs, identified through CD34 FITC and CD309 PE markers at diagnosis and after one treatment cycle (4 weeks). Our findings revealed that patients exhibiting poor response to therapy showed significantly higher CD34/CD309 values than those with a good response (p < 0.001). The delineation of response based on CD34/CD309 expression was established with a cutoff ≤ 0.9 for percentage (yielding 100% sensitivity and 94.1% specificity) and ≤ 12.5 for absolute value (also with 100% sensitivity and 94.1% specificity). These results underscore the potential of EPC population levels, as quantified by CD34/CD309, to serve as a predictive biomarker for immunomodulatory treatment in MM patients undergoing Proteasome Inhibitor and Dexamethasone therapy.
Collapse
Affiliation(s)
- Nadia ElMenshawy
- Clinical Pathology Department, Hematology Unit, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| | - Manal Ibrahim Fouda
- Clinical Pathology Department, Hematology Unit, Mansoura Medical School, Mansoura University, Mansoura, Egypt
| | - Mohamed Mofreh
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Manal O Elnenaei
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mohamed Eissa
- College of Medicine, King Khalid University, KSA and Clinical Pathology Department, Faculty of Medicine, Zagazig University, Egypt
| |
Collapse
|
3
|
Wang X, Chen Y, Meng H, Ruan J, Meng F. SREBP-1-mediated lipogenesis confers resistance to ferroptosis and improves endothelial injury. FASEB J 2024; 38:e23806. [PMID: 38970404 DOI: 10.1096/fj.202400721r] [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: 04/01/2024] [Revised: 06/15/2024] [Accepted: 06/26/2024] [Indexed: 07/08/2024]
Abstract
Atherosclerosis refers to a disease characterized by the formation of lipid plaque deposits within arterial walls, leading to reduced blood flow or blockage of blood outflow. The process of endothelial injury induced by oxidized low-density lipoprotein (ox-LDL) is considered the initial stage of atherosclerosis. Ferroptosis is a form of iron-dependent, non-apoptotic cell death, and current research suggests its association with coronary artery disease (CAD). In this study, we observed a correlation between reduced expression of SREBP-1 and the occurrence of stable CAD. Additionally, during the process of endothelial injury induced by ox-LDL, we also noted decreased expression of the SREBP-1/SCD1/FADS2 and involvement in the ferroptosis process. Mechanistically, ox-LDL induced endothelial injury by inhibiting the lipid biosynthesis process mediated by the SREBP-1/SCD1/FADS2, thereby inducing lipid peroxidation and ferroptosis. On the contrary, overexpression of SREBP-1 or supplementation with monounsaturated fatty acids counteracted iron accumulation, mitochondrial damage, and lipid peroxidation-induced ferroptosis, thereby improving endothelial injury. Our study indicated that the decreased expression of peripheral blood SREBP-1 mRNA is an independent risk factor for stable CAD. Furthermore, in endothelial cells, the lipid biosynthesis process mediated by SREBP-1 could ameliorate endothelial injury by resisting ferroptosis. The study has been registered with the Chinese Clinical Trial Registry, which serves as a primary registry in the World Health Organization International Clinical Trials Registry Platform (ChiCTR2300074315, August 3rd, 2023).
Collapse
Affiliation(s)
- Xue Wang
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yanqiu Chen
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Heyu Meng
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianjun Ruan
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fanbo Meng
- China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Ramirez-Velandia F, Mensah E, Salih M, Wadhwa A, Young M, Muram S, Taussky P, Ogilvy CS. Endothelial Progenitor Cells: A Review of Molecular Mechanisms in the Pathogenesis and Endovascular Treatment of Intracranial Aneurysms. Neuromolecular Med 2024; 26:25. [PMID: 38886284 DOI: 10.1007/s12017-024-08791-4] [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: 02/19/2024] [Accepted: 05/09/2024] [Indexed: 06/20/2024]
Abstract
This comprehensive review explores the multifaceted role of endothelial progenitor cells (EPCs) in vascular diseases, focusing on their involvement in the pathogenesis and their contributions to enhancing the efficacy of endovascular treatments for intracranial aneurysms (IAs). Initially discovered as CD34+ bone marrow-derived cells implicated in angiogenesis, EPCs have been linked to vascular repair, vasculogenesis, and angiogenic microenvironments. The origin and differentiation of EPCs have been subject to debate, challenging the conventional notion of bone marrow origin. Quantification methods, including CD34+ , CD133+ , and various assays, reveal the influence of factors, like age, gender, and comorbidities on EPC levels. Cellular mechanisms highlight the interplay between bone marrow and angiogenic microenvironments, involving growth factors, matrix metalloproteinases, and signaling pathways, such as phosphatidylinositol-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK). In the context of the pathogenesis of IAs, EPCs play a role in maintaining vascular integrity by replacing injured and dysfunctional endothelial cells. Recent research has also suggested the therapeutic potential of EPCs after coil embolization and flow diversion, and this has led the development of device surface modifications aimed to enhance endothelialization. The comprehensive insights underscore the importance of further research on EPCs as both therapeutic targets and biomarkers in IAs.
Collapse
Affiliation(s)
- Felipe Ramirez-Velandia
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Emmanuel Mensah
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Mira Salih
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Aryan Wadhwa
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
| | - Michael Young
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Sandeep Muram
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Philipp Taussky
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, USA
| | - Christopher S Ogilvy
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, 110 Francis Street, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Tian W, Feng B, Zhang L, Dai G, Lin L, Jiang W, Wang Y. Tibial transverse transport induces mobilization of endothelial progenitor cells to accelerate angiogenesis and ulcer wound healing through the VEGFA/CXCL12 pathway. Biochem Biophys Res Commun 2024; 709:149853. [PMID: 38555838 DOI: 10.1016/j.bbrc.2024.149853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Tibial transverse transport (TTT) can promote the healing of chronic foot ulcers, but the specific cellular and molecular mechanisms by which TTT promotes wound healing remain unclear. METHODS New Zealand White rabbits were selected to induce foot ulcer models. The treatment included unilateral TTT surgery and bilateral TTT surgery. Observation of tissue neovascularization structure by HE staining and CD31 immunofluorescence detection. Collagen fiber formation was detected through the Masson staining. The mobilization of endothelial progenitor cell (EPCs) were analyzed by VEGFR2 immunofluorescence detection and flow cytometry detection of the number of VEGFR2/Tie-2-positive cells in peripheral blood. ELISA and qPCR assay were performed to detect VEGFA and CXCL12 levels. RESULTS The complete healing time of ulcer surfaces in sham, unilateral and bilateral TTT groups was about 22 days, 17 days and 13 days, respectively. TTT treatment significantly increased the deposition of granulation tissue and epithelialization of wounds. It also led to an increase in collagen fiber content and the level of the microvascular marker CD31. Furthermore, TTT treatment upregulated the levels of VEGFA and CXCL12 in peripheral blood and wound tissues, as well as increased the expression of VEGFR2 in wound tissues and the proportion of VEGFR2/Tie-2 in peripheral blood. Moreover, these effects of TTT treatment in the bilateral group was more significant than that in the unilateral group. CONCLUSIONS TTT may facilitate wound fibroblasts to release VEGFA and CXCL12, causing EPC mobilization, thus promoting angiogenesis and ulcer wound healing.
Collapse
Affiliation(s)
- Weiqing Tian
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Bo Feng
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Lan Zhang
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Guangming Dai
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Ligong Lin
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Wei Jiang
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China
| | - Yongjun Wang
- Department of Orthopedics, Baogang Hospital of Inner Mongolia Autonomous Region, Baotou, 014000, Inner Mongolia Autonomous Region, PR China.
| |
Collapse
|
6
|
Cao Y, Qi J, Wang J, Chen L, Wang Y, Long Y, Li B, Lai J, Yao Y, Meng Y, Yu X, Chen X, Ng LG, Li X, Lu Y, Cheng X, Cui W, Sun Y. Injectable "Homing-Like" Bioactive Short-Fibers for Endometrial Repair and Efficient Live Births. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306507. [PMID: 38504456 PMCID: PMC11132084 DOI: 10.1002/advs.202306507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/14/2023] [Indexed: 03/21/2024]
Abstract
The prevalence of infertility caused by endometrial defects is steadily increasing, posing a significant challenge to women's reproductive health. In this study, injectable "homing-like" bioactive decellularized extracellular matrix short-fibers (DEFs) of porcine skin origin are innovatively designed for endometrial and fertility restoration. The DEFs can effectively bind to endometrial cells through noncovalent dipole interactions and release bioactive growth factors in situ. In vitro, the DEFs effectively attracted endometrial cells through the "homing-like" effect, enabling cell adhesion, spreading, and proliferation on their surface. Furthermore, the DEFs effectively facilitated the proliferation and angiogenesis of human primary endometrial stromal cells (HESCs) and human umbilical vein endothelial cells (HUVECs), and inhibited fibrosis of pretreated HESCs. In vivo, the DEFs significantly accelerated endometrial restoration, angiogenesis, and receptivity. Notably, the deposition of endometrial collagen decreased from 41.19 ± 2.16% to 14.15 ± 1.70% with DEFs treatment. Most importantly, in endometrium-injured rats, the use of DEFs increased the live birth rate from 30% to an impressive 90%, and the number and development of live births close to normal rats. The injectable "homing-like" bioactive DEFs system can achieve efficient live births and intrauterine injection of DEFs provides a new promising clinical strategy for endometrial factor infertility.
Collapse
Affiliation(s)
- Yumeng Cao
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Jia Qi
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Liang Chen
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Yuan Wang
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Yijing Long
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Boyu Li
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Junliang Lai
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Yejie Yao
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Yiwen Meng
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Xiaohua Yu
- Hangzhou Phil Stone Biotech Co., Ltd.HangzhouZhejiang311215P. R. China
| | - Xiao‐Dong Chen
- Department of Comprehensive DentistryUniversity of Texas Health Science Center at San AntonioSan AntonioTX78229USA
- Research ServiceSouth Texas Veterans Health Care SystemAudie Murphy VA Medical CenterSan AntonioTX78229USA
| | - Lai Guan Ng
- Shanghai Immune Therapy InstituteShanghai Jiao Tong University School of Medicine affiliated Renji HospitalShanghai200127P. R. China
| | - Xinyu Li
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Yao Lu
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Xiaoyue Cheng
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025P. R. China
| | - Yun Sun
- Department of Reproductive Medicine, Ren Ji HospitalShanghai Jiao Tong University School of MedicineShanghai200135P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive GeneticsShanghai200135P. R. China
| |
Collapse
|
7
|
Huang C, Huang W, Meng Y, Zhou C, Wang X, Zhang C, Tian Y, Wei W, Li Y, Zhou Q, Chen W, Tang Y. T1-weighted MRI of targeting atherosclerotic plaque based on CD40 expression on engulfed USPIO's cell surface. Biomed Mater 2024; 19:025019. [PMID: 38215489 DOI: 10.1088/1748-605x/ad1df6] [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: 09/13/2023] [Accepted: 01/12/2024] [Indexed: 01/14/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of cholesterol within the arterial wall. Its progression can be monitored via magnetic resonance imaging (MRI). Ultrasmall Superparamagnetic Particles of Iron Oxide (USPIO) (<5 nm) have been employed as T1 contrast agents for MRI applications. In this study, we synthesized USPIO with an average surface carboxylation of approximately 5.28 nm and a zeta potential of -47.8 mV. These particles were phagocytosed by mouse aortic endothelial cells (USPIO-MAECs) and endothelial progenitor cells (USPIO-EPCs), suggesting that they can be utilized as potential contrast agent and delivery vehicle for the early detection of atherosclerosis. However, the mechanism by which this contrast agent is delivered to the plaque remains undetermined. Our results demonstrated that with increasing USPIO concentration during 10-100 μg ml-1, consistent change appeared in signal enhancement on T1-weighted MRI. Similarly, T1-weighted MRI of MAECs and EPCs treated with these concentrations exhibited a regular change in signal enhancement. Prussian blue staining of USPIO revealed substantial absorption into MAECs and EPCs after treatment with 50 μg ml-1USPIO for 24 h. The iron content in USPIO-EPCs was much higher (5 pg Fe/cell) than in USPIO-MAECs (0.8 pg Fe/cell). In order to substantiate our hypothesis that CD40 protein on the cell surface facilitates migration towards inflammatory cells, we utilized AuNPs-PEI (gold nanoparticles-polyethylenimine) carrying siRNACD40to knockout CD40 expression in MAECs. It has been documented that gold nanoparticle-oligonucleotide complexes could be employed as intracellular gene regulation agents for the control of protein level in cells. Our results confirmed that macrophages are more likely to bind to MAECs treated with AuNPs-PEI-siRNANC(control) for 72 h than to MAECs treated with AuNPs-PEI-siRNACD40(reduced CD40 expression), thus confirming CD40 targeting at the cellular level. When USPIO-MAECs and MAECs (control) were delivered to mice (high-fat-fed) via tail vein injection respectively, we observed a higher iron accumulation in plaques on blood vessels in high-fat-fed mice treated with USPIO-MAECs. We also demonstrated that USPIO-EPCs, when delivered to high-fat-fed mice via tail vein injection, could indeed label plaques by generating higher T1-weighted MRI signals 72 h post injection compared to controls (PBS, USPIO and EPCs alone). In conclusion, we synthesized a USPIO suitable for T1-weighted MRI. Our results have confirmed separately at the cellular and tissue andin vivolevel, that USPIO-MAECs or USPIO-EPCs are more accessible to atherosclerotic plaques in a mouse model. Furthermore, the high expression of CD40 on the cell surface is a key factor for targeting and USPIO-EPCs may have potential therapeutic effects.
Collapse
Affiliation(s)
- Chen Huang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Medical Imaging Institute of Panyu District, Guangzhou 511400, People's Republic of China
| | - Wentao Huang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, People's Republic of China
| | - Yixuan Meng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, People's Republic of China
| | - Chengqian Zhou
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America
| | - Xiaozhuan Wang
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, People's Republic of China
| | - Chunyu Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, People's Republic of China
| | - Yuzhen Tian
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, People's Republic of China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou 510663, People's Republic of China
| | - Yongsheng Li
- Guangdong Cord Blood Bank, Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou 510663, People's Republic of China
| | - Quan Zhou
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, People's Republic of China
| | - Wenli Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, People's Republic of China
| | - Yukuan Tang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central Hospital, Medical Imaging Institute of Panyu District, Guangzhou 511400, People's Republic of China
| |
Collapse
|
8
|
Dhindsa DS, Desai SR, Jin Q, Sandesara PB, Mehta A, Liu C, Tahhan AS, Nayak A, Ejaz K, Hooda A, Moazzami K, Islam SJ, Rogers SC, Almuwaqqat Z, Mokhtari A, Hesaroieh I, Ko YA, Sperling LS, Waller EK, Quyyumi AA. Circulating progenitor cells and outcomes in patients with coronary artery disease. Int J Cardiol 2023; 373:7-16. [PMID: 36460208 PMCID: PMC9840693 DOI: 10.1016/j.ijcard.2022.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Low quantities of circulating progenitor cells (CPCs), specifically CD34+ populations, reflect impairment of intrinsic regenerative capacity. This study investigates the relationship between subsets of CPCs and adverse outcomes. METHODS 1366 individuals undergoing angiography for evaluation of coronary artery disease (CAD) were enrolled into the Emory Cardiovascular Biobank. Flow cytometry identified CPCs as CD45med blood mononuclear cells expressing the CD34 epitope, with further enumeration of hematopoietic CPCs as CD133+/CXCR4+ cells and endothelial CPCs as vascular endothelial growth factor receptor-2 (VEGFR2+) cells. Adjusted Cox or Fine and Gray's sub-distribution hazard regression models analyzed the relationship between CPCs and 1) all-cause death and 2) a composite of cardiovascular death and non-fatal myocardial infarction (MI). RESULTS Over a median 3.1-year follow-up period (IQR 1.3-4.9), there were 221 (16.6%) all-cause deaths and 172 (12.9%) cardiovascular deaths/MIs. Hematopoietic CPCs were highly correlated, and the CD34+/CXCR4+ subset was the best independent predictor. Lower counts (≤median) of CD34+/CXCR4+ and CD34+/VEGFR2+ cells independently predicted all-cause mortality (HR 1.46 [95% CI 1.06-2.01], p = 0.02 and 1.59 [95% CI 1.15-2.18], p = 0.004) and cardiovascular death/MI (HR 1.50 [95% CI 1.04-2.17], p = 0.03 and 1.47 [95% CI 1.01-2.03], p = 0.04). A combination of low CD34+/CXCR4+ and CD34+/VEGFR2+ CPCs predicted all-cause death (HR 2.1, 95% CI 1.4-3.0; p = 0.0002) and cardiovascular death/MI (HR 2.0, 95% CI 1.3-3.2; p = 0.002) compared to those with both lineages above the cut-offs. CONCLUSIONS Lower levels of hematopoietic and endothelial CPCs indicate diminished endogenous regenerative capacity and independently correlate with greater mortality and cardiovascular risk in patients with CAD.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Qingchun Jin
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Anurag Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman S Tahhan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Aditi Nayak
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kiran Ejaz
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ananya Hooda
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kasra Moazzami
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shabatun J Islam
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Rogers
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zakaria Almuwaqqat
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ali Mokhtari
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Iraj Hesaroieh
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Laurence S Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
9
|
Shi X, Seidle KA, Simms KJ, Dong F, Chilian WM, Zhang P. Endothelial progenitor cells in the host defense response. Pharmacol Ther 2023; 241:108315. [PMID: 36436689 PMCID: PMC9944665 DOI: 10.1016/j.pharmthera.2022.108315] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Extensive injury of endothelial cells in blood vasculature, especially in the microcirculatory system, frequently occurs in hosts suffering from sepsis and the accompanied systemic inflammation. Pathological factors, including toxic components derived from invading microbes, oxidative stress associated with tissue ischemia/reperfusion, and vessel active mediators generated during the inflammatory response, are known to play important roles in mediating endothelial injury. Collapse of microcirculation and tissue edema developed from the failure of endothelial barrier function in vital organ systems, including the lung, brain, and kidney, are detrimental, which often predict fatal outcomes. The host body possesses a substantial capacity for maintaining vascular homeostasis and repairing endothelial damage. Bone marrow and vascular wall niches house endothelial progenitor cells (EPCs). In response to septic challenges, EPCs in their niche environment are rapidly activated for proliferation and angiogenic differentiation. In the meantime, release of EPCs from their niches into the blood stream and homing of these vascular precursors to tissue sites of injury are markedly increased. The recruited EPCs actively participate in host defense against endothelial injury and repair of damage in blood vasculature via direct differentiation into endothelial cells for re-endothelialization as well as production of vessel active mediators to exert paracrine and autocrine effects on angiogenesis/vasculogenesis. In recent years, investigations on significance of EPCs in host defense and molecular signaling mechanisms underlying regulation of the EPC response have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches for effective prevention and treatment of sepsis-induced vascular injury as well as vital organ system failure.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kelly A Seidle
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kevin J Simms
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Ping Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America.
| |
Collapse
|
10
|
Moazzami K, Mehta A, Young A, Dhindsa DS, Martin G, Mokhtari A, Hesaroieh IG, Shah A, Bremner JD, Vaccarino V, Waller EK, Quyyumi AA. The association between baseline circulating progenitor cells and vascular function: The role of aging and risk factors. Vasc Med 2022; 27:532-541. [PMID: 36062298 PMCID: PMC10150400 DOI: 10.1177/1358863x221116411] [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] [Indexed: 01/24/2023]
Abstract
BACKGROUND To investigate the cross-sectional and longitudinal relationships between vascular function and circulating progenitor cell (CPC) counts with respect to aging and exposure to risk factors. METHODS In 797 adult participants, CPCs were enumerated by flow cytometry as CD45med mononuclear cells expressing CD34 epitope and its subsets co-expressing CD133, and chemokine C-X-C motif receptor 4 (CXCR4+). Arterial stiffness was evaluated by tonometry-derived pulse wave velocity (PWV) and microvascular function was assessed as digital reactive hyperemia index (RHI). RESULTS In cross-sectional analyses, for every doubling in CD34+ cell counts, PWV was 15% higher and RHI was 9% lower, after adjusting for baseline characteristics and risk factors (p for all < 0.01). There were significant CPC-by-age-by-risk factor interactions (p <0.05) for both vascular measures. Among younger subjects (< 48 years), CPC counts were higher in those with risk factors and vascular function was better in those with higher compared to those with lower CPC counts (p for all < 0.0l). In contrast, in older participants, CPCs were not higher in those with risk factors, and vascular function was worse compared to the younger age group. A lower CPC count at baseline was an independent predictor of worsening vascular function during 2-year follow-up. CONCLUSION A higher CPC count in the presence of risk factors is associated with better vascular function among younger individuals. There is no increase in CPC count with risk factors in older individuals who have worse vascular function. Moreover, a higher CPC count is associated with less vascular dysfunction with aging.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Anurag Mehta
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - An Young
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Devinder Singh Dhindsa
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg Martin
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Ali Mokhtari
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Iraj Ghaini Hesaroieh
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - J Douglas Bremner
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Edmund K Waller
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
11
|
Dynamics of endothelial progenitor cells in patients with advanced hepatocellular carcinoma. Dig Liver Dis 2022; 54:911-917. [PMID: 34876355 DOI: 10.1016/j.dld.2021.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Circulating endothelial progenitor cells (EPC) predict tumor vascularization and disease progression, but limited information is available on their dynamics in hepatocellular carcinoma (HCC) undergoing systemic treatment. METHODS We prospectively analyzed different populations of EPC in 16 patients with advanced HCC receiving sorafenib. Patients were studied before therapy (T0, n = 16) and after two (T2, n = 12) and eight weeks (T8, n = 8), using high-performance flow-cytometry. The tumor response at T8 was categorized as progressive disease (PD) or clinical benefit (CB, all other responses). RESULTS At T0, higher levels of CD34+CD133+KDR+ and CD34+KDR+ were observed in patients with alpha-fetoprotein ≥400 ng/ml or non-viral liver disease, whereas CD34+CD133+KDR+ cells were virtually absent in patients with vascular invasion. CD34+KDR+ and CD34+CD133+KDR+ were directly correlated with platelet count. Frequencies of all populations of EPC declined in patients receiving sorafenib. Levels of CD34+CD133+ were higher at T0 in patients with CB compared to patients with PD. In patients belonging to the CB group CD34+KDR+ cells at T0 were directly correlated to platelet count. CONCLUSION In patients with advanced HCC, EPC are directly correlated with platelet count, suggesting a common activation of selected bone marrow pathways. Levels of a CD34+KDR+ are higher at baseline in patients responding to sorafenib.
Collapse
|
12
|
Canjuga D, Steinle H, Mayer J, Uhde AK, Klein G, Wendel HP, Schlensak C, Avci-Adali M. Homing of mRNA-Modified Endothelial Progenitor Cells to Inflamed Endothelium. Pharmaceutics 2022; 14:pharmaceutics14061194. [PMID: 35745767 PMCID: PMC9229815 DOI: 10.3390/pharmaceutics14061194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 01/25/2023] Open
Abstract
Endothelial progenitor cells (EPCs) are one of the most important stem cells for the neovascularization of tissues damaged by ischemic diseases such as myocardial infarction, ischemic stroke, or critical limb ischemia. However, their low homing efficiency in the treatment of ischemic tissues limits their potential clinical applications. The use of synthetic messenger RNA (mRNA) for cell engineering represents a novel and promising technology for the modulation of cell behavior and tissue regeneration. To improve the therapeutic potential of EPCs, in this study, murine EPCs were engineered with synthetic mRNAs encoding C-X-C chemokine receptor 4 (CXCR4) and P-selectin glycoprotein ligand 1 (PSGL-1) to increase the homing and migration efficiency of EPCs to inflamed endothelium. Flow cytometric measurements revealed that the transfection of EPCs with CXCR4 and PSGL-1 mRNA resulted in increased expressions of CXCR4 and PSGL-1 on the cell surface compared with the unmodified EPCs. The transfection of EPCs with mRNAs did not affect cell viability. CXCR4-mRNA-modified EPCs showed significantly higher migration potential than unmodified cells in a chemotactic migration assay. The binding strength of the EPCs to inflamed endothelium was determined with single-cell atomic force microscopy (AFM). This showed that the mRNA-modified EPCs required a three-fold higher detachment force to be released from the TNF-α-activated endothelium than unmodified EPCs. Furthermore, in a dynamic flow model, significantly increased binding of the mRNA-modified EPCs to inflamed endothelium was detected. This study showed that the engineering of EPCs with homing factors encoding synthetic mRNAs increases the homing and migration potentials of these stem cells to inflamed endothelium. Thus, this strategy represents a promising strategy to increase the therapeutic potential of EPCs for the treatment of ischemic tissues.
Collapse
Affiliation(s)
- Denis Canjuga
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Heidrun Steinle
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Jana Mayer
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Ann-Kristin Uhde
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Gerd Klein
- Center for Medical Research, Department of Medicine II, University of Tuebingen, Waldhörnlestraße 22, 72072 Tuebingen, Germany;
| | - Hans Peter Wendel
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Christian Schlensak
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany; (D.C.); (H.S.); (J.M.); (A.-K.U.); (H.P.W.); (C.S.)
- Correspondence: ; Tel.: +49-7071-29-86605; Fax: +49-7071-29-3617
| |
Collapse
|
13
|
Post-COVID-19 patients show an increased endothelial progenitor cell production. Transl Res 2022; 243:14-20. [PMID: 35085821 PMCID: PMC8785258 DOI: 10.1016/j.trsl.2022.01.004] [Citation(s) in RCA: 10] [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: 10/04/2021] [Revised: 12/24/2021] [Accepted: 01/20/2022] [Indexed: 12/20/2022]
Abstract
SARS-CoV-2, the cause of COVID-19, has generated a global emergency. The endothelium is a target of SARS-CoV-2, generating endothelial dysfunction, an essential step for the development of cardiovascular complications. The number of endothelial progenitor cells acts as an indicator of vascular damage. However, its role in SARS-CoV-2 is unknown. The aim of this study was to quantify the number of endothelial colony forming cells (ECFCs) and assess for the first time if there is a significant increase after SARS-CoV-2 infection. This study also evaluates whether the number of ECFC is related to the presence of pulmonary embolism (PE), and if this increase correlates with any of the clinical parameters studied. A total of 63 subjects were recruited including 32 subjects 3-months after overcoming COVID-19 and 31 healthy controls. The results confirm the presence of vascular sequelae in post-COVID-19 patients, with an abnormal increase in the number of ECFCs in blood circulation compared to controls (2.81 ± 2.33 vs 1.23 ± 1.86, P = 0.001). There was no difference in ECFC production in COVID-19 who presented acute PE compared to those that did not (3.21 ± 2.49 vs 2.50 ± 2.23, P > 0.05). The appearance of ECFC colonies in COVID-19 patients was significantly related to male gender (P = 0.003), the presence of systemic hypertension (P = 0.01) and elevated hemoglobin levels (P = 0.02) at the time of ECFC isolation and lower PaO2 levels (P = 0.01) at admission. Whether these results indicate a prompt response of the patient to repair the damaged endothelium or reflect a postinfection injury that will persist in time is not known.
Collapse
Key Words
- 6mwt, 6-minute walk test
- ace-2, angiotensin-converting enzyme 2
- bmi, body mass index
- crp, c-reactive protein
- dd, dimer-d
- dlco, carbon monoxide diffusing capacity
- dlp, dyslipidemia
- dm, diabetes mellitus
- ecfc, endothelial colony forming cell
- epc, endothelial progenitor cell
- epo, erythropoietin
- fev, forced expiratory volume
- fib, fibrinogen
- fvc, forced vital capacity
- hb, hemoglobin
- hta, arterial hypertension
- htc, hematocrit
- ldh, lactate dehydrogenase
- lym, lymphocytes
- mf, maximum ferritin
- pao2, partial pressure of oxygen
- pbmc's, peripheral blood mononuclear cells
- pe, pulmonary embolism
- rv, residual volume
- tlc, total lung capacity
Collapse
|
14
|
Dubey R, Prabhakar PK, Gupta J. Epigenetics: key to improve delayed wound healing in type 2 diabetes. Mol Cell Biochem 2022; 477:371-383. [PMID: 34739665 DOI: 10.1007/s11010-021-04285-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/23/2021] [Indexed: 12/13/2022]
Abstract
Diabetes-related delayed wound healing is a multifactorial, nuanced, and intertwined complication that causes substantial clinical morbidity. The etiology of diabetes and its related microvascular complications is affected by genes, diet, and lifestyle factors. Epigenetic modifications such as DNA methylation, histone modifications, and post-transcriptional RNA regulation (microRNAs) are subsequently recognized as key facilitators of the complicated interaction between genes and the environment. Current research suggests that diabetes-persuaded dysfunction of epigenetic pathways, which results in changed expression of genes in target cells and cause diabetes-related complications including cardiomyopathy, nephropathy, retinopathy, delayed wound healing, etc., which are foremost drivers to diabetes-related adverse outcomes. In this paper, we discuss the role of epigenetic mechanisms in controlling tissue repair, angiogenesis, and expression of growth factors, as well as recent findings that show the alteration of epigenetic events during diabetic wound healing.
Collapse
Affiliation(s)
- Rupal Dubey
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, 144411, Phagwara, Punjab, India
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, School of Physiotherapy and Paramedical Sciences, Lovely Professional University, 144411, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Jalandhar-Delhi G.T. Road, 144411, Phagwara, Punjab, India.
| |
Collapse
|
15
|
Camps-Renom P, Jiménez-Xarrié E, Soler M, Puig N, Aguilera-Simón A, Marín R, Prats-Sánchez L, Delgado-Mederos R, Martínez-Domeño A, Guisado-Alonso D, Guasch-Jiménez M, Martí-Fàbregas J. Endothelial Progenitor Cells Count after Acute Ischemic Stroke Predicts Functional Outcome in Patients with Carotid Atherosclerosis. J Stroke Cerebrovasc Dis 2021; 30:106144. [PMID: 34649037 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Circulating Endothelial Progenitor Cells (EPCs) predict cardiovascular outcomes in patients with coronary disease. However, the predictive value of EPCs after ischemic stroke is not well established. We aimed to study the prognostic role of EPCs in patients with acute ischemic stroke and carotid atherosclerosis, focusing on post-stroke functional outcome and stroke recurrences. MATERIALS AND METHODS We studied consecutive adult patients with an acute (<7 days) anterior circulation ischemic stroke and carotid atherosclerosis. Cardioembolic strokes were excluded. We measured circulating EPCs by flow cytometry (CD34+/CD133+/KDR+) at inclusion (7±1 days after stroke) and at one year of follow-up. At three months and at one year we registered the modified Rankin Scale score, stroke recurrences and coronary syndromes during the follow-up. RESULTS We studied 80 patients with a mean age of 74.3±10.4 years. We divided the population in tertiles according to the EPCs count. At three months we observed a favorable outcome in 25/36 (69.4%) patients in the lowest, 19/22 (86.4%) in the medium and 21/22 (95.5%) in the highest tercile (p=0.037). In the multivariable analysis a higher EPCs count was associated with favorable functional outcome after adjusting for age and baseline NIHSS score (OR=3.61, 95%CI 1.34-9.76; p=0.011). This association persisted at one year of follow-up. We did not find association between counts of EPCs and stroke recurrence. CONCLUSIONS In patients with acute ischemic stroke and carotid atherosclerosis, a higher count of EPCs was associated with favorable functional outcome in the mid and long-term follow-up. Counts of EPCs did not predict stroke recurrences.
Collapse
Affiliation(s)
- Pol Camps-Renom
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain..
| | - Elena Jiménez-Xarrié
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Marta Soler
- Facility of Cytometry, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona, Spain
| | - Núria Puig
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Ana Aguilera-Simón
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Rebeca Marín
- Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Luis Prats-Sánchez
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Raquel Delgado-Mederos
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Alejandro Martínez-Domeño
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Daniel Guisado-Alonso
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Marina Guasch-Jiménez
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| | - Joan Martí-Fàbregas
- Stroke Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona (Department of Medicine), C/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.; Cerebrovascular Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), C/Sant Quintí 77, 08041, Barcelona Spain
| |
Collapse
|
16
|
Moon S, Chang MS, Koh SH, Choi YK. Repair Mechanisms of the Neurovascular Unit after Ischemic Stroke with a Focus on VEGF. Int J Mol Sci 2021; 22:ijms22168543. [PMID: 34445248 PMCID: PMC8395233 DOI: 10.3390/ijms22168543] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/22/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022] Open
Abstract
The functional neural circuits are partially repaired after an ischemic stroke in the central nervous system (CNS). In the CNS, neurovascular units, including neurons, endothelial cells, astrocytes, pericytes, microglia, and oligodendrocytes maintain homeostasis; however, these cellular networks are damaged after an ischemic stroke. The present review discusses the repair potential of stem cells (i.e., mesenchymal stem cells, endothelial precursor cells, and neural stem cells) and gaseous molecules (i.e., nitric oxide and carbon monoxide) with respect to neuroprotection in the acute phase and regeneration in the late phase after an ischemic stroke. Commonly shared molecular mechanisms in the neurovascular unit are associated with the vascular endothelial growth factor (VEGF) and its related factors. Stem cells and gaseous molecules may exert therapeutic effects by diminishing VEGF-mediated vascular leakage and facilitating VEGF-mediated regenerative capacity. This review presents an in-depth discussion of the regeneration ability by which endogenous neural stem cells and endothelial cells produce neurons and vessels capable of replacing injured neurons and vessels in the CNS.
Collapse
Affiliation(s)
- Sunhong Moon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Korea;
| | - Mi-Sook Chang
- Department of Oral Anatomy, Seoul National University School of Dentistry, Seoul 03080, Korea;
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea;
| | - Yoon Kyung Choi
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul 05029, Korea;
- Correspondence: ; Tel.: +82-2-450-0558; Fax: +82-2-444-3490
| |
Collapse
|
17
|
Barrera JA, Trotsyuk AA, Maan ZN, Bonham CA, Larson MR, Mittermiller PA, Henn D, Chen K, Mays CJ, Mittal S, Mermin-Bunnell AM, Sivaraj D, Jing S, Rodrigues M, Kwon SH, Noishiki C, Padmanabhan J, Jiang Y, Niu S, Inayathullah M, Rajadas J, Januszyk M, Gurtner GC. Adipose-Derived Stromal Cells Seeded in Pullulan-Collagen Hydrogels Improve Healing in Murine Burns. Tissue Eng Part A 2021; 27:844-856. [PMID: 33789446 DOI: 10.1089/ten.tea.2020.0320] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Burn scars and scar contractures cause significant morbidity for patients. Recently, cell-based therapies have been proposed as an option for improving healing and reducing scarring after burn injury, through their known proangiogenic and immunomodulatory paracrine effects. Our laboratory has developed a pullulan-collagen hydrogel that, when seeded with mesenchymal stem cells (MSCs), improves cell viability and augments their proangiogenic capacity in vivo. Concurrently, recent research suggests that prospective isolation of cell subpopulations with desirable transcriptional profiles can be used to further improve cell-based therapies. In this study, we examined whether adipose-derived stem cell (ASC)-seeded hydrogels could improve wound healing following thermal injury using a murine contact burn model. Partial thickness contact burns were created on the dorsum of mice. On days 5 and 10 following injury, burns were debrided and received either ASC hydrogel, ASC injection alone, hydrogel alone, or no treatment. On days 10 and 25, burns were harvested for histologic and molecular analysis. This experiment was repeated using CD26+/CD55+ FACS-enriched ASCs to further evaluate the regenerative potential of ASCs in wound healing. ASC hydrogel-treated burns demonstrated accelerated time to reepithelialization, greater vascularity, and increased expression of the proangiogenic genes MCP-1, VEGF, and SDF-1 at both the mRNA and protein level. Expression of the profibrotic gene Timp1 and proinflammatory gene Tnfa was downregulated in ASC hydrogel-treated burns. ASC hydrogel-treated burns exhibited reduced scar area compared to hydrogel-treated and control wounds, with equivalent scar density. CD26+/CD55+ ASC hydrogel treatment resulted in accelerated healing, increased dermal appendage count, and improved scar quality with a more reticular collagen pattern. Here we find that ASC hydrogel therapy is effective for treating burns, with demonstrated proangiogenic, fibromodulatory, and immunomodulatory effects. Enrichment for CD26+/CD55+ ASCs has additive benefits for tissue architecture and collagen remodeling postburn injury. Research is ongoing to further facilitate clinical translation of this promising therapeutic approach. Impact statement Burns remain a significant public health burden. Stem cell therapy has gained attention as a promising approach for treating burns. We have developed a pullulan-collagen biomimetic hydrogel scaffold that can be seeded with adipose-derived stem cells (ASCs). We assessed the delivery and activity of our scaffold in a murine contact burn model. Our results suggest that localized delivery of ASC hydrogel treatment is a promising approach for the treatment of burn wounds, with the potential for rapid clinical translation. We believe our work will have broad implications for both hydrogel therapeutics and regenerative medicine and will be of interest to the general scientific community.
Collapse
Affiliation(s)
- Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Zeshaan N Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Clark A Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Madelyn R Larson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Paul A Mittermiller
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Chyna J Mays
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Smiti Mittal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Alana M Mermin-Bunnell
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Serena Jing
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Melanie Rodrigues
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Chikage Noishiki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yuanwen Jiang
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Simiao Niu
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Mohammed Inayathullah
- Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Center, Stanford University, Stanford, California, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
18
|
Erkek ET, Nazligul E, Nalcaci M, Yenerel MN. Circulating Endothelial Progenitor Cells and Their Relation to Thrombosis in Paroxysmal Nocturnal Hemoglobinuria and Aplastic Anemia. Indian J Hematol Blood Transfus 2021; 38:319-326. [PMID: 35496969 PMCID: PMC9001756 DOI: 10.1007/s12288-021-01445-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 05/05/2021] [Indexed: 10/21/2022] Open
Abstract
Thrombosis is a leading cause of morbidity and mortality in paroxysmal nocturnal hemoglobinuria (PNH). Multiple factors are responsible for the thrombotic tendency in these patients. Endothelial progenitorcells (EPCs) originate from primitive hematopoietic stem cells. The EPC count is considered indicative of potential damage and restoration capacity in vascular disease; lower EPC counts are deemed as a risk factor in cardiovascular diseases. We aimed to investigate the count of circulating EPCs in PNH and aplastic anemia (AA) patients receiving eculizumab treatment or not receiving treatment and their relationship with thrombosis. Seventeen PNH patients, 18 AA patients, and 10 healthy volunteers were included in the study. The CD309, CD133, and CD34 antibodies were used to determine counts of circulating EPCs using flowcytometry. EPC levels were compared between the PNH, AA, and healthy control groups. Kolmogorov-Smirnov test. ANOVA, Kruskal-Wallis, and Mann-Whitney U testswereperformedto analyze the quantitative data, while χ2 testing was performed to analyze the qualitative data. Therewasnosignificantdifference in EPC levelsbetweenpatientswithandwithout a history of thrombosis (P > 0.05). Further, therewasnosignificantdifference in thelevels of EPCsbetweenthe AA and PNH groups (P > 0.05). However, there was a significant positive correlation between levels of EPCs and lactate dehydrogenase (LDH) in multivariate analysis (P < 0.05). The study findings suggest that hemolysis promotes vascular endothelial and new blood vessel formation. Increased EPCs in PNH may indirectly indicatevascular endothelial damage in PNH.
Collapse
|
19
|
Abstract
ABSTRACT Norepinephrine is used in the acute care setting to establish and maintain hemodynamic stability in patients with hypotension. Although it is often a lifesaving medication, norepinephrine may lead to profound vascular insufficiency in the extremities, resulting in dry gangrene and skin necrosis. The purpose of this article is to present a case series of skin complications related to treatment with norepinephrine and review the pathophysiology behind these complications. The authors also explore risk stratification as it relates to history and clinical presentation with subsequent focus on contingencies to mitigate the adverse effects of vasoconstriction on peripheral tissues.
Collapse
|
20
|
Alwjwaj M, Kadir RRA, Bayraktutan U. The secretome of endothelial progenitor cells: a potential therapeutic strategy for ischemic stroke. Neural Regen Res 2021; 16:1483-1489. [PMID: 33433461 PMCID: PMC8323700 DOI: 10.4103/1673-5374.303012] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke continues to be a leading cause of mortality and morbidity in the world. Despite recent advances in the field of stroke medicine, thrombolysis with recombinant tissue plasminogen activator remains as the only pharmacological therapy for stroke patients. However, due to short therapeutic window (4.5 hours of stroke onset) and increased risk of hemorrhage beyond this point, each year globally less than 1% of stroke patients receive this therapy which necessitate the discovery of safe and efficacious therapeutics that can be used beyond the acute phase of stroke. Accumulating evidence indicates that endothelial progenitor cells (EPCs), equipped with an inherent capacity to migrate, proliferate and differentiate, may be one such therapeutics. However, the limited availability of EPCs in peripheral blood and early senescence of few isolated cells in culture conditions adversely affect their application as effective therapeutics. Given that much of the EPC-mediated reparative effects on neurovasculature is realized by a wide range of biologically active substances released by these cells, it is possible that EPC-secretome may serve as an important therapeutic after an ischemic stroke. In light of this assumption, this review paper firstly discusses the main constituents of EPC-secretome that may exert the beneficial effects of EPCs on neurovasculature, and then reviews the currently scant literature that focuses on its therapeutic capacity.
Collapse
Affiliation(s)
- Mansour Alwjwaj
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Rais Reskiawan A Kadir
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, Clinical Sciences Building, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
21
|
Nandi S, Potunuru UR, Kumari C, Nathan AA, Gopal J, Menon GI, Siddharthan R, Dixit M, Thangaraj PR. Altered kinetics of circulating progenitor cells in cardiopulmonary bypass (CPB) associated vasoplegic patients: A pilot study. PLoS One 2020; 15:e0242375. [PMID: 33211740 PMCID: PMC7676651 DOI: 10.1371/journal.pone.0242375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/31/2020] [Indexed: 11/19/2022] Open
Abstract
Vasoplegia observed post cardiopulmonary bypass (CPB) is associated with substantial morbidity, multiple organ failure and mortality. Circulating counts of hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPC) are potential markers of neo-vascularization and vascular repair. However, the significance of changes in the circulating levels of these progenitors in perioperative CPB, and their association with post-CPB vasoplegia, are currently unexplored. We enumerated HSC and EPC counts, via flow cytometry, at different time-points during CPB in 19 individuals who underwent elective cardiac surgery. These 19 individuals were categorized into two groups based on severity of post-operative vasoplegia, a clinically insignificant vasoplegic Group 1 (G1) and a clinically significant vasoplegic Group 2 (G2). Differential changes in progenitor cell counts during different stages of surgery were compared across these two groups. Machine-learning classifiers (logistic regression and gradient boosting) were employed to determine if differential changes in progenitor counts could aid the classification of individuals into these groups. Enumerating progenitor cells revealed an early and significant increase in the circulating counts of CD34+ and CD34+CD133+ hematopoietic stem cells (HSC) in G1 individuals, while these counts were attenuated in G2 individuals. Additionally, EPCs (CD34+VEGFR2+) were lower in G2 individuals compared to G1. Gradient boosting outperformed logistic regression in assessing the vasoplegia grouping based on the fold change in circulating CD 34+ levels. Our findings indicate that a lack of early response of CD34+ cells and CD34+CD133+ HSCs might serve as an early marker for development of clinically significant vasoplegia after CPB.
Collapse
Affiliation(s)
- Sanhita Nandi
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Uma Rani Potunuru
- Apollo Hospitals Educational and Research Foundation, Chennai, India
| | | | - Abel Arul Nathan
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Jayashree Gopal
- Department of Endocrinology and Diabetology, Apollo Hospitals, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| | - Gautam I. Menon
- The Institute of Mathematical Sciences (HBNI), Chennai, India
- Departments of Physics and Biology, Ashoka University, Sonepat, India
| | | | - Madhulika Dixit
- Laboratory of Vascular Biology, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| | - Paul Ramesh Thangaraj
- Department of Cardiothoracic Surgery, Apollo Hospitals, Chennai, India
- Department of Mechanical Engineering, Indian Institute of Technology Madras, Chennai, India
- * E-mail: (JG); (MD); (PRT)
| |
Collapse
|
22
|
Kwon SH, Barrera JA, Noishiki C, Chen K, Henn D, Sheckter CC, Gurtner GC. Current and Emerging Topical Scar Mitigation Therapies for Craniofacial Burn Wound Healing. Front Physiol 2020; 11:916. [PMID: 32848859 PMCID: PMC7403506 DOI: 10.3389/fphys.2020.00916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/09/2020] [Indexed: 01/22/2023] Open
Abstract
Burn injury in the craniofacial region causes significant health and psychosocial consequences and presents unique reconstructive challenges. Healing of severely burned skin and underlying soft tissue is a dynamic process involving many pathophysiological factors, often leading to devastating outcomes such as the formation of hypertrophic scars and debilitating contractures. There are limited treatment options currently used for post-burn scar mitigation but recent advances in our knowledge of the cellular and molecular wound and scar pathophysiology have allowed for development of new treatment concepts. Clinical effectiveness of these experimental therapies is currently being evaluated. In this review, we discuss current topical therapies for craniofacial burn injuries and emerging new therapeutic concepts that are highly translational.
Collapse
Affiliation(s)
- Sun Hyung Kwon
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Janos A Barrera
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Chikage Noishiki
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Kellen Chen
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Dominic Henn
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Clifford C Sheckter
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
23
|
Leal V, Ribeiro CF, Oliveiros B, António N, Silva S. Intrinsic Vascular Repair by Endothelial Progenitor Cells in Acute Coronary Syndromes: an Update Overview. Stem Cell Rev Rep 2020; 15:35-47. [PMID: 30345477 DOI: 10.1007/s12015-018-9857-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone marrow-derived endothelial progenitor cells (EPCs) play a key role in the maintenance of endothelial homeostasis and endothelial repair at areas of vascular damage. The quantification of EPCs in peripheral blood by flow cytometry is a strategy to assess this reparative capacity. The number of circulating EPCs is inversely correlated with the number of cardiovascular risk factors and to the occurrence of cardiovascular events. Therefore, monitoring EPCs levels may provide an accurate assessment of susceptibility to cardiovascular injury, greatly improving risk stratification of patients with high cardiovascular risk, such as those with an acute myocardial infarction. However, there are many issues in the field of EPC identification and quantification that remain unsolved. In fact, there have been conflicting protocols used to the phenotypic identification of EPCs and there is still no consensual immunophenotypical profile that corresponds exactly to EPCs. In this paper we aim to give an overview on EPCs-mediated vascular repair with special focus on acute coronary syndromes and to discuss the different phenotypic profiles that have been used to identify and quantify circulating EPCs in several clinical studies. Finally, we will synthesize evidence on the prognostic role of EPCs in patients with high cardiovascular risk.
Collapse
Affiliation(s)
- Vânia Leal
- Group of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | - Carlos Fontes Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Bárbara Oliveiros
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Natália António
- Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Cardiology Department, Coimbra Hospital and Universitary Centre, Coimbra, Portugal
| | - Sónia Silva
- Group of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.,Institute of Pharmacology and Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
24
|
Tripathi DM, Hassan M, Siddiqui H, Kaur I, Rawal P, Bihari C, Kaur S, Sarin SK. Cirrhotic Endothelial Progenitor Cells Enhance Liver Angiogenesis and Fibrosis and Aggravate Portal Hypertension in Bile Duct-Ligated Cirrhotic Rats. Front Physiol 2020; 11:617. [PMID: 32595520 PMCID: PMC7300214 DOI: 10.3389/fphys.2020.00617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
Background Circulating cirrhotic endothelial progenitor cells (EPC) interact with both liver sinusoidal endothelial cells (LSEC) and hepatic stellate cells (HSC) and promote angiogenesis in vitro. This study evaluated the effect of cirrhotic and control EPCs on hepatic angiogenesis, microcirculation, and fibrosis in vivo in rat models of cirrhosis. Methodology Animal models of cirrhosis were prepared by bile duct ligation (BDL). Circulating EPCs isolated from healthy human and cirrhotic blood were characterized by flow cytometry, cultured and administered through the tail vein in BDL rats after 2 weeks of ligation. The cells were given thrice a week for 2 weeks. The untreated group of BDL rats received only saline. Fibrosis was evaluated by Masson’s trichrome staining. Dedifferentiated LSECs were identified by the expression of CD31, and activated HSCs were marked as alpha-SMA-positive cells and were studied by immunohistochemistry and western blotting in saline-, healthy EPC-, and cirrhotic EPC-treated rats. In vivo, hepatic and systemic hemodynamic parameters were evaluated. Liver functions were evaluated. Results In comparison to controls, BDL rats revealed an increase of fibrosis and angiogenesis. Among the treated rats, cirrhotic EPC-treated rats had increased fibrosis grade as compared to healthy EPC-treated and saline-treated rats. There was an increase of both fibrosis and angiogenesis markers, alpha-SMA and CD31 in cirrhotic EPC-treated rats as compared to healthy EPC-treated and saline-treated rats in immunohistochemistry and western blot studies. Cirrhotic EPC-treated BDL rats had high portal pressure and portal blood flow with significantly elevated hepatic vascular resistance in comparison with healthy EPC- and saline-treated BDL animals, without significant differences in mean arterial pressure. Cirrhotic EPC-treated BDL rats also showed a substantial increase in the hepatic expression of angiogenic receptors, VEGFR2 and CXCR4 in comparison with saline-treated rats. Conclusion The study suggests that transplantation of cirrhotic EPCs enhances LSEC differentiation and angiogenesis, activates HSCs and worsens fibrosis, thus resulting in hepatic hemodynamic derangements in BDL-induced cirrhosis.
Collapse
Affiliation(s)
- Dinesh Mani Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Mohsin Hassan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Hamda Siddiqui
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Chaggan Bihari
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.,Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
25
|
Jarajapu YPR. Targeting Angiotensin-Converting Enzyme-2/Angiotensin-(1-7)/Mas Receptor Axis in the Vascular Progenitor Cells for Cardiovascular Diseases. Mol Pharmacol 2020; 99:29-38. [PMID: 32321734 DOI: 10.1124/mol.119.117580] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Bone marrow-derived hematopoietic stem/progenitor cells are vasculogenic and play an important role in endothelial health and vascular homeostasis by participating in postnatal vasculogenesis. Progenitor cells are mobilized from bone marrow niches in response to remote ischemic injury and migrate to the areas of damage and stimulate revascularization largely by paracrine activation of angiogenic functions in the peri-ischemic vasculature. This innate vasoprotective mechanism is impaired in certain chronic clinical conditions, which leads to the development of cardiovascular complications. Members of the renin-angiotensin system-angiotensin-converting enzymes (ACEs) ACE and ACE2, angiotensin II (Ang II), Ang-(1-7), and receptors AT1 and Mas-are expressed in vasculogenic progenitor cells derived from humans and rodents. Ang-(1-7), generated by ACE2, is known to produce cardiovascular protective effects by acting on Mas receptor and is considered as a counter-regulatory mechanism to the detrimental effects of Ang II. Evidence has now been accumulating in support of the activation of the ACE2/Ang-(1-7)/Mas receptor pathway by pharmacologic or molecular maneuvers, which stimulates mobilization of progenitor cells from bone marrow, migration to areas of vascular damage, and revascularization of ischemic areas in pathologic conditions. This minireview summarizes recent studies that have enhanced our understanding of the physiology and pharmacology of vasoprotective axis in bone marrow-derived progenitor cells in health and disease. SIGNIFICANCE STATEMENT: Hematopoietic stem progenitor cells (HSPCs) stimulate revascularization of ischemic areas. However, the reparative potential is diminished in certain chronic clinical conditions, leading to the development of cardiovascular diseases. ACE2 and Mas receptor are key members of the alternative axis of the renin-angiotensin system and are expressed in HSPCs. Accumulating evidence points to activation of ACE2 or Mas receptor as a promising approach for restoring the reparative potential, thereby preventing the development of ischemic vascular diseases.
Collapse
Affiliation(s)
- Yagna P R Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
26
|
Ruscitto A, Morel MM, Shawber CJ, Reeve G, Lecholop MK, Bonthius D, Yao H, Embree MC. Evidence of vasculature and chondrocyte to osteoblast transdifferentiation in craniofacial synovial joints: Implications for osteoarthritis diagnosis and therapy. FASEB J 2020; 34:4445-4461. [PMID: 32030828 DOI: 10.1096/fj.201902287r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
Temporomandibular joint osteoarthritis (TMJ OA) leads to permanent cartilage destruction, jaw dysfunction, and compromises the quality of life. However, the pathological mechanisms governing TMJ OA are poorly understood. Unlike appendicular articular cartilage, the TMJ has two distinct functions as the synovial joint of the craniofacial complex and also as the site for endochondral jaw bone growth. The established dogma of endochondral bone ossification is that hypertrophic chondrocytes undergo apoptosis, while invading vasculature with osteoprogenitors replace cartilage with bone. However, contemporary murine genetic studies support the direct differentiation of chondrocytes into osteoblasts and osteocytes in the TMJ. Here we sought to characterize putative vasculature and cartilage to bone transdifferentiation using healthy and diseased TMJ tissues from miniature pigs and humans. During endochondral ossification, the presence of fully formed vasculature expressing CD31+ endothelial cells and α-SMA+ vascular smooth muscle cells were detected within all cellular zones in growing miniature pigs. Arterial, endothelial, venous, angiogenic, and mural cell markers were significantly upregulated in miniature pig TMJ tissues relative to donor matched knee meniscus fibrocartilage tissue. Upon surgically creating TMJ OA in miniature pigs, we discovered increased vasculature and putative chondrocyte to osteoblast transformation dually marked by COL2 and BSP or RUNX2 within the vascular bundles. Pathological human TMJ tissues also exhibited increased vasculature, while isolated diseased human TMJ cells exhibited marked increased in vasculature markers relative to control 293T cells. Our study provides evidence to suggest that the TMJ in higher order species are in fact vascularized. There have been no reports of cartilage to bone transdifferentiation or vasculature in human-relevant TMJ OA large animal models or in human TMJ tissues and cells. Therefore, these findings may potentially alter the clinical management of TMJ OA by defining new drugs that target angiogenesis or block the cartilage to bone transformation.
Collapse
Affiliation(s)
- Angela Ruscitto
- Cartilage Biology and Regenerative Medicine Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mallory M Morel
- Cartilage Biology and Regenerative Medicine Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Carrie J Shawber
- Department of OB/GYN, Division of Reproductive Sciences, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gwendolyn Reeve
- Division of Oral and Maxillofacial Surgery, New York Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Michael K Lecholop
- Department of Oral and Maxillofacial Surgery, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel Bonthius
- Clemson-MUSC Bioengineering Program, Department of Bioengineering, Clemson University, Greenville, SC, USA
| | - Hai Yao
- Clemson-MUSC Bioengineering Program, Department of Bioengineering, Clemson University, Greenville, SC, USA.,Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Mildred C Embree
- Cartilage Biology and Regenerative Medicine Laboratory, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Yellowley CE, Toupadakis CA, Vapniarsky N, Wong A. Circulating progenitor cells and the expression of Cxcl12, Cxcr4 and angiopoietin-like 4 during wound healing in the murine ear. PLoS One 2019; 14:e0222462. [PMID: 31513647 PMCID: PMC6742462 DOI: 10.1371/journal.pone.0222462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/29/2019] [Indexed: 01/16/2023] Open
Abstract
Migration of cells from both local and systemic sources is essential for the inflammatory and regenerative processes that occur during normal wound healing. CXCL12 is considered a critical regulator of CXCR4-positive cell migration during tissue regeneration. In this study, we investigated the expression of Cxcl12 and Cxcr4 during healing of a murine full thickness ear wound. We also investigated the expression of angiopoietin-like 4, which has been shown to participate in wound angiogenesis and reepithelialization. At time points up to 48hrs, complete blood counts were performed using automated hematology analysis, and the numbers of circulating stem and progenitor cells quantified using flow cytometry. Expression of both Cxcr4 and Angptl4 was significantly elevated within 3 days of wounding, and both were strongly expressed in cells of the epidermis. ANGPTL4 protein expression remained elevated in the epithelium through day 14. Cxcl12 expression was increased significantly at day 3, and remained elevated through day 21. Faint Cxcl12 staining was detectable in the epithelium at day 1, and thereafter staining was faint and more generalized. There were significantly fewer circulating total white blood cells and lymphocytes 1hr following ear punching. Similarly, there was a significant early (1hr) reduction in the number of circulating endothelial progenitor cells. Further studies are warranted to investigate whether ANGPTL4 and CXCL12/CXCR4 interact or synergize to facilitate cell recruitment and migration, and to potentiate reepithelialization and wound healing.
Collapse
Affiliation(s)
- Clare E Yellowley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Chrisoula A Toupadakis
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Natalia Vapniarsky
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Alice Wong
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
28
|
Expression of B2 Receptor on Circulating CD34-Positive Cells and Outcomes of Myocardial Infarction. DISEASE MARKERS 2019; 2019:7816438. [PMID: 31360266 PMCID: PMC6644252 DOI: 10.1155/2019/7816438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 11/17/2022]
Abstract
Background Bradykinin B2 receptor (B2R) is a widely expressed cell surface receptor. The relationship between B2R expression on circulating CD34+ cells and prognosis of myocardial infarction remains unknown. Methods We analyzed the expression of B2R on circulating CD34-positive cells and plasma VEGF concentration in 174 myocardial infarction patients. All involved patients were divided into two groups: high B2R group and low B2R group according to the median B2R expression percentage. 48 months of follow-up was performed. The endpoints were heart failure and revascularization. Results The plasma level of VEGF in the low B2R group is 67 ± 12 pg/mL, whereas the high B2R group has significantly elevated VEGF levels of 145 ± 27 pg/mL (P < 0.001). The concentration of VEGF has correlated with expression of B2R (r = 0.574, P < 0.001). During the 48 months of follow-up, low expression of B2 receptor on circulating CD34-positive cells indicates the high incidence of heart failure (hazard ratio: 2.247; 95% confidence interval: 1.110-4.547; P = 0.024) and revascularization (hazard ratio: 2.335; 95% confidence interval: 1.075-5.074; P = 0.032). Kaplan-Meier survival analysis showed that the cumulative hazard of heart failure (P = 0.014) and revascularization (P = 0.032) has significant differences between low B2R and high B2R. Conclusion Low expression of B2R on circulating progenitor cells indicated the poor outcomes of myocardial infarction.
Collapse
|
29
|
Circulating Proangiogenic Cells and Proteins in Patients with Glioma and Acute Myocardial Infarction: Differences in Neovascularization between Neoplasia and Tissue Regeneration. JOURNAL OF ONCOLOGY 2019; 2019:3560830. [PMID: 31428150 PMCID: PMC6679840 DOI: 10.1155/2019/3560830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/06/2019] [Indexed: 01/10/2023]
Abstract
Although extensive angiogenesis takes place in glial tumors, antiangiogenic therapies have remained without the expected success. In the peripheral circulation of glioma patients, increased numbers of endothelial precursor cells (EPCs) are present, potentially offering targets for antiangiogenic therapy. However, for an antiangiogenic therapy to be successful, the therapy should specifically target glioma-related EPC subsets and secreted factors only. Here, we compared the EPC subsets and plasma factors in the peripheral circulation of patients with gliomas to acute myocardial infarctions. We investigated the five most important EPC subsets and 21 angiogenesis-related plasma factors in peripheral blood samples of 29 patients with glioma, 14 patients with myocardial infarction, and 20 healthy people as controls, by FACS and Luminex assay. In GBM patients, all EPC subsets were elevated as compared to healthy subjects. In addition, HPC and KDR+ cell fractions were higher than in MI, while CD133+ and KDR+CD133+ cell fractions were lower. There were differences in relative EPC fractions between the groups: KDR+ cells were the largest fraction in GBM, while CD133+ cells were the largest fraction in MI. An increase in glioma malignancy grade coincided with an increase in the KDR+ fraction, while the CD133+ cell fraction decreased relatively. Most plasma angiogenic factors were higher in GBM than in MI patients. In both MI and GBM, the ratio of CD133+ HPCs correlated significantly with elevated levels of MMP9. In the GBM patients, MMP9 correlated strongly with levels of all HPCs. In conclusion, the data demonstrate that EPC traffic in patients with glioma, representing neoplasia, is different from that in myocardial infarction, representing tissue regeneration. Glioma patients may benefit from therapies aimed at lowering KDR+ cells and HPCs.
Collapse
|
30
|
Abstract
Accumulating evidence demonstrates that pre-vascularization of tissue-engineered constructs can significantly enhance their survival and engraftment upon transplantation. Endothelial cells (ECs), the basic component of vasculatures, are indispensable to the entire process of pre-vascularization. However, the source of ECs still poses an issue. Recent studies confirmed that diverse approaches are available in the derivation of ECs for tissue engineering, such as direct isolation of autologous ECs, reprogramming of somatic cells, and induced differentiation of stem cells in typology. Herein, we discussed a variety of human stem cells (i.e., totipotent, pluripotent, multipotent, oligopotent, and unipotent stem cells), which can be induced to differentiate into ECs and reviewed the multifarious approaches for EC generation, such as 3D EB formation for embryonic stem cells (ESCs), stem cell-somatic cell co-culture, and directed endothelial differentiation with growth factors in conventional 2D culture.
Collapse
Affiliation(s)
- Min Xu
- Key Laboratory of Oral Diseases Research of Anhui Province, Stomatological Hospital and College, Anhui Medical University, 69 Meishan Road, Hefei, 230032 Anhui Province China
| | - Jiacai He
- Key Laboratory of Oral Diseases Research of Anhui Province, Stomatological Hospital and College, Anhui Medical University, 69 Meishan Road, Hefei, 230032 Anhui Province China
| | - Chengfei Zhang
- Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Jianguang Xu
- Key Laboratory of Oral Diseases Research of Anhui Province, Stomatological Hospital and College, Anhui Medical University, 69 Meishan Road, Hefei, 230032 Anhui Province China
- Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Yuanyin Wang
- Key Laboratory of Oral Diseases Research of Anhui Province, Stomatological Hospital and College, Anhui Medical University, 69 Meishan Road, Hefei, 230032 Anhui Province China
| |
Collapse
|
31
|
Tal R, Dong D, Shaikh S, Mamillapalli R, Taylor HS. Bone-marrow-derived endothelial progenitor cells contribute to vasculogenesis of pregnant mouse uterus†. Biol Reprod 2019; 100:1228-1237. [PMID: 30601943 PMCID: PMC6497522 DOI: 10.1093/biolre/ioy265] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/02/2018] [Accepted: 01/01/2019] [Indexed: 01/01/2023] Open
Abstract
Angiogenesis is essential for cyclic endometrial growth, implantation, and pregnancy maintenance. Vasculogenesis, the formation of new blood vessels by bone marrow (BM)-derived endothelial progenitor cells (EPCs), has been shown to contribute to endometrial vasculature. However, it is unknown whether vasculogenesis occurs in neovascularization of the decidua during pregnancy. To investigate the contribution of BM-derived EPCs to vascularization of the pregnant uterus, we induced non-gonadotoxic submyeloablation by 5-fluorouracil administration to wild-type FVB/N female mice recipients followed by BM transplantation from transgenic mice expressing green fluorescent protein (GFP) under regulation of Tie2 endothelial-specific promoter. Following 1 month, Tie2-GFP BM-transplanted mice were bred and sacrificed at various gestational days (ED6.5, ED10.5, ED13.5, ED18.5, and postpartum). Bone-marrow-transplanted non-pregnant and saline-injected pregnant mice served as controls (n = 5-6/group). Implantation sites were analyzed by flow cytometry, immunohistochemistry, and immunofluorescence. While no GFP-positive EPCs were found in non-pregnant or early pregnant uteri of BM-transplanted mice, GFP-positive EPCs were first detected in pregnant uterus on ED10.5 (0.12%) and increased as the pregnancy progressed (1.14% on ED13.5), peaking on ED18.5 (1.42%) followed by decrease in the postpartum (0.9%). The percentage of endothelial cells that were BM-derived out of the total endothelial cell population in the implantation sites (GFP+CD31+/CD31+) were 9.3%, 15.8%, and 6.1% on ED13.5, ED18.5, and postpartum, respectively. Immunohistochemistry demonstrated that EPCs incorporated into decidual vasculature, and immunofluorescence showed that GFP-positive EPCs colocalized with CD31 in vascular endothelium of uterine implantation sites, confirming their endothelial lineage. Our findings indicate that BM-derived EPCs contribute to vasculogenesis of the pregnant mouse decidua.
Collapse
Affiliation(s)
- Reshef Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Dirong Dong
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shafiq Shaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Sella G, Kracoff SL. Unusual intercoronary communication of possible traumatic origin: a case report. EUROPEAN HEART JOURNAL-CASE REPORTS 2019; 2:yty070. [PMID: 31020147 PMCID: PMC6177065 DOI: 10.1093/ehjcr/yty070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/22/2018] [Indexed: 11/20/2022]
Abstract
Introduction The normal coronary circulation is physiologically interconnected by intercoronary and intracoronary anastomoses that are functionally insignificant and cannot be visualized by conventional coronary angiography due to their small size. The development of significant coronary stenosis increases the flow through these anastomoses and hence their size, making them readily visible. Large intercoronary communications in the absence of obstructive coronary artery disease constitute a very rare coronary artery anomaly, which is thought to be congenital in origin and located in specific anatomic locations. Case presentation A 62-year-old man was admitted to our department due to typical anginotic pain. Coronary angiography revealed a subtotal occlusion of the proximal circumflex and a very unusual tortuous intercoronary communication between the left main coronary artery and the left anterior descending artery. A drug eluting stent was successfully deployed at the circumflex and the patient’s symptoms resolved. Medical history was retaken revealing that he suffered a strong blunt trauma of the chest as an adolescent, which we believe could be a possible alternative explanation for the formation of the intercoronary communication. Comparison to a previous angiogram performed 4 years earlier showed that the intercoronary communication was already present and remained unchanged over the years. Discussion It is suggested that intercoronary communication arising from unusual locations and developing in disorganized fashion can be suspected as being of traumatic origin. We believe that there is no need for intervention to treat this type of anomaly.
Collapse
Affiliation(s)
- Gal Sella
- Department of Cardiology, Kaplan Medical Center, Pasternak St. POB 1, Rehovot, Israel
| | - Sharon L Kracoff
- Department of Plastic Surgery, Kaplan Medical Center, Pasternak St. POB 1, Rehovot, Israel
| |
Collapse
|
33
|
Aquila I, Frati G, Sciarretta S, Dellegrottaglie S, Torella D, Torella M. New imaging techniques project the cellular and molecular alterations underlying bicuspid aortic valve development. J Mol Cell Cardiol 2019; 129:197-207. [PMID: 30826295 DOI: 10.1016/j.yjmcc.2019.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/29/2022]
Abstract
Bicuspid aortic valve (BAV) disease is the most common congenital cardiac malformation associated with an increased lifetime risk and a high rate of surgically-relevant valve deterioration and aortic dilatation. Genomic data revealed that different genes are associated with BAV. A dominant genetic factor for the recent past was the basis to the recommendation for a more extensive aortic intervention. However very recent evidence that hemodynamic stressors and alterations of wall shear stress play an important role independent from the genetic trait led to more conservative treatment recommendations. Therefore, there is a current need to improve the ability to risk stratify BAV patients in order to obtain an early detection of valvulopathy and aortopathy while also to predict valve dysfunction and/or aortic disease development. Imaging studies based on new cutting-edge technologies, such us 4-dimensional (4D) flow magnetic resonance imaging (MRI), two-dimensional (2D) or three-dimensional (3D) speckle-tracking imaging (STI) and computation fluid dynamics, combined with studies demonstrating new gene mutations, specific signal pathways alterations, hemodynamic influences, circulating biomarkers modifications, endothelial progenitor cell impairment and immune/inflammatory response, all detected BAV valvulopathy progression and aortic wall abnormality. Overall, the main purpose of this review article is to merge the evidences of imaging and basic science studies in a coherent hypothesis that underlies and thus projects the development of both BAV during embryogenesis and BAV-associated aortopathy and its complications in the adult life, with the final goal to identifying aneurysm formation/rupture susceptibility to improve diagnosis and management of patients with BAV-related aortopathy.
Collapse
Affiliation(s)
- Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; IRCCS NEUROMED, Pozzilli, IS, Italy.
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; IRCCS NEUROMED, Pozzilli, IS, Italy
| | - Santo Dellegrottaglie
- Division of Cardiology, Ospedale Accreditato Villa dei Fiori, Acerra, Naples 80011, Italy; The Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro 88100, Italy.
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
34
|
Bali P, Bammidi S, Banik A, Nehru B, Anand A. CD34 and CD117 Stemness of Lineage-Negative Cells Reverses Memory Loss Induced by Amyloid Beta in Mouse Model. Front Behav Neurosci 2018; 12:222. [PMID: 30443207 PMCID: PMC6222267 DOI: 10.3389/fnbeh.2018.00222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
A majority of the neurodegenerative disorders including Alzheimer's disease are untreatable and occur primarily due to aging and rapidly changing lifestyles. The rodent Alzheimer's disease models are critical for investigating the underlying disease pathology and screening of novel therapeutic targets in preclinical settings. We aimed to characterize the stemness properties of human umbilical cord blood (hUCB) derived lineage-negative (Lin−) stem cells based on CD34 and CD117 expression as well as surface morphology using flow cytometry and scanning electron microscopy, respectively. The efficacy of the stem cells was tested by its capacity to rescue the injury caused by intrahippocampal delivery of varying doses of amyloid beta. The hUCB Lin− stem cells reversed memory loss due to Aβ42-induced injury more effectively at micromolar concentration, and not picomolar concentration. More studies are required to delineate the underlying molecular events associated with hUCB Lin− stem cells.
Collapse
Affiliation(s)
- Parul Bali
- Department of Biophysics, Panjab University, Chandigarh, India.,Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sridhar Bammidi
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Avijit Banik
- Department of Pharmacology, Rollins Research Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Bimla Nehru
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
35
|
Lidgerwood GE, Pitson SM, Bonder C, Pébay A. Roles of lysophosphatidic acid and sphingosine-1-phosphate in stem cell biology. Prog Lipid Res 2018; 72:42-54. [PMID: 30196008 DOI: 10.1016/j.plipres.2018.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/15/2018] [Accepted: 09/05/2018] [Indexed: 02/06/2023]
Abstract
Stem cells are unique in their ability to self-renew and differentiate into various cell types. Because of these features, stem cells are key to the formation of organisms and play fundamental roles in tissue regeneration and repair. Mechanisms controlling their fate are thus fundamental to the development and homeostasis of tissues and organs. Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are bioactive phospholipids that play a wide range of roles in multiple cell types, during developmental and pathophysiological events. Considerable evidence now demonstrates the potent roles of LPA and S1P in the biology of pluripotent and adult stem cells, from maintenance to repair. Here we review their roles for each main category of stem cells and explore how those effects impact development and physiopathology.
Collapse
Affiliation(s)
- Grace E Lidgerwood
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Claudine Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Alice Pébay
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; Ophthalmology, Department of Surgery, the University of Melbourne, Melbourne, Australia.
| |
Collapse
|
36
|
Del Papa N, Pignataro F. The Role of Endothelial Progenitors in the Repair of Vascular Damage in Systemic Sclerosis. Front Immunol 2018; 9:1383. [PMID: 29967618 PMCID: PMC6015881 DOI: 10.3389/fimmu.2018.01383] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/04/2018] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by a complex pathological process where the main scenario is represented by progressive loss of microvascular bed, with the consequent progressive fibrotic changes in involved organ and tissues. Although most aspects of vascular injury in scleroderma are poorly understood, recent data suggest that the scleroderma impairment of neovascularization could be related to both angiogenesis and vasculogenesis failure. Particularly, compensatory angiogenesis does not occur normally in spite of an important increase in many angiogenic factors either in SSc skin or serum. Besides insufficient angiogenesis, the contribution of defective vasculogenesis to SSc vasculopathy has been extensively studied. Over the last decades, our understanding of the processes responsible for the formation of new vessels after tissue ischemia has increased. In the past, adult neovascularization was thought to depend mainly on angiogenesis (a process by which new vessels are formed by the proliferation and migration of mature endothelial cells). More recently, increased evidence suggests that stem cells mobilize from the bone marrow into the peripheral blood (PB), differentiate in circulating endothelial progenitors (EPCs), and home to site of ischemia to contribute to de novo vessel formation. Significant advances have been made in understanding the biology of EPCs, and molecular mechanisms regulating EPC function. Autologous EPCs now are becoming a novel treatment option for therapeutic vascularization and vascular repair, mainly in ischemic diseases. However, different diseases, such as cardiovascular diseases, diabetes, and peripheral artery ischemia are related to EPC dysfunction. Several studies have shown that EPCs can be detected in the PB of patients with SSc and are impaired in their function. Based on an online literature search (PubMed, EMBASE, and Web of Science, last updated December 2017) using keywords related to “endothelial progenitor cells” and “Systemic Sclerosis,” “scleroderma vasculopathy,” “angiogenesis,” “vasculogenesis,” this review gives an overview on the large body of data of current research in this issue, including controversies over the identity and functions of EPCs, their meaning as biomarker of SSc microangiopathy and their clinical potency.
Collapse
|
37
|
Gao W, Li F, Liu L, Xu X, Zhang B, Wu Y, Yin D, Zhou S, Sun D, Huang Y, Zhang J. Endothelial colony-forming cell-derived exosomes restore blood-brain barrier continuity in mice subjected to traumatic brain injury. Exp Neurol 2018; 307:99-108. [PMID: 29883579 DOI: 10.1016/j.expneurol.2018.06.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/01/2018] [Accepted: 06/04/2018] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) tends to cause disruption of the blood-brain barrier (BBB). Previous studies have shown that intravenously or intracerebroventricularly infused human umbilical cord blood-derived endothelial colony-forming cells (ECFCs) can home to injury sites and improve outcomes in mice subjected to experimental TBI. Several reports have demonstrated that these cells did not incorporate directly into newly formed vasculature but instead stimulated the proliferation and migration of tissue-resident endothelial cells (ECs) via paracrine mechanisms. In the present study, exosomes, which range from 30 to 150 nm in diameter, were isolated from ECFC-conditioned medium. The exosomes were labeled with PKH67 ex vivo, and we observed that they were taken up by ECs with high efficiency after 12 h of incubation. Pretreatment with ECFC-derived exosomes promoted the migration of ECs subjected to scratch injury, and incubating ECs exposed to hypoxia with ECFC-derived exosomes decreased PTEN expression, stimulated AKT phosphorylation and increased tight junction (TJ) protein expression in the cells. Furthermore, in vivo delivery of ECFC-derived exosomes into TBI mice also inhibited PTEN expression and increased AKT expression, changes accompanied by reductions in Evans blue (EB) dye extravasation, brain edema and TJ degradation. These data demonstrated that ECFC-derived exosomes have beneficial effects on BBB integrity in mice with TBI.
Collapse
Affiliation(s)
- Weiwei Gao
- Department of Neurology, Tianjin Huanhu Hospital, 6 Jizhao Road, Tianjin, China
| | - Fei Li
- Department of Neurosurgery, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin 301800, PR China
| | - Li Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Xin Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Baoliang Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Yingang Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Dongpei Yin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Shuai Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Dongdong Sun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China
| | - Ying Huang
- Department of Neurosurgery, Tianjin Huanhu Hospital, 6 Jizhao Road, Tianjin, PR China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin 300052, PR China; Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in the Central Nervous System, Ministry of Education, Tianjin City 300052, PR China.
| |
Collapse
|
38
|
Samman Tahhan A, Hammadah M, Raad M, Almuwaqqat Z, Alkhoder A, Sandesara PB, Mohamed-Kelli H, Hayek SS, Kim JH, O'Neal WT, Topel ML, Grant AJ, Sabbak N, Heinl RE, Gafeer MM, Obideen M, Kaseer B, Abdelhadi N, Ko YA, Liu C, Hesaroieh I, Mahar EA, Vaccarino V, Waller EK, Quyyumi AA. Progenitor Cells and Clinical Outcomes in Patients With Acute Coronary Syndromes. Circ Res 2018; 122:1565-1575. [PMID: 29514830 PMCID: PMC5970041 DOI: 10.1161/circresaha.118.312821] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 02/27/2018] [Accepted: 03/02/2018] [Indexed: 11/16/2022]
Abstract
RATIONALE Circulating progenitor cells (CPCs) mobilize in response to ischemic injury, but their predictive value remains unknown in acute coronary syndrome (ACS). OBJECTIVE We aimed to investigate the number of CPCs in ACS compared with those with stable coronary artery disease (CAD), relationship between bone marrow PCs and CPCs, and whether CPC counts predict mortality in patients with ACS. METHODS AND RESULTS In 2028 patients, 346 had unstable angina, 183 had an acute myocardial infarction (AMI), and the remaining 1499 patients had stable CAD. Patients with ACS were followed for the primary end point of all-cause death. CPCs were enumerated by flow cytometry as mononuclear cells expressing a combination of CD34+, CD133+, vascular endothelial growth factor receptor 2+, or chemokine (C-X-C motif) receptor 4+. CPC counts were higher in subjects with AMI compared those with stable CAD even after adjustment for age, sex, race, body mass index, renal function, hypertension, diabetes mellitus, hyperlipidemia, and smoking; CD34+, CD34+/CD133+, CD34+/CXCR4+, and CD34+/VEGFR2+ CPC counts were 19%, 25%, 28%, and 142% higher in those with AMI, respectively, compared with stable CAD. There were strong correlations between the concentrations of CPCs and the PC counts in bone marrow aspirates in 20 patients with AMI. During a 2 (interquartile range, 1.31-2.86)-year follow-up period of 529 patients with ACS, 12.4% died. In Cox regression models adjusted for age, sex, body mass index, heart failure history, estimated glomerular filtration rate, and AMI, subjects with low CD34+ cell counts had a 2.46-fold (95% confidence interval, 1.18-5.13) increase in all-cause mortality, P=0.01. CD34+/CD133+ and CD34+/CXCR4+, but not CD34+/VEGFR2+ PC counts, had similar associations with mortality. Results were validated in a separate cohort of 238 patients with ACS. CONCLUSIONS CPC levels are significantly higher in patients after an AMI compared with those with stable CAD and reflect bone marrow PC content. Among patients with ACS, a lower number of hematopoietic-enriched CPCs are associated with a higher mortality.
Collapse
Affiliation(s)
- Ayman Samman Tahhan
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Muhammad Hammadah
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Mohamad Raad
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Zakaria Almuwaqqat
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Ayman Alkhoder
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Pratik B Sandesara
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Heval Mohamed-Kelli
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Salim S Hayek
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Jeong Hwan Kim
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Wesley T O'Neal
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Matthew L Topel
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Aubrey J Grant
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Nabil Sabbak
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Robert E Heinl
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Mohamad Mazen Gafeer
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Malik Obideen
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Belal Kaseer
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Nasser Abdelhadi
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Yi-An Ko
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Chang Liu
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Iraj Hesaroieh
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Ernestine A Mahar
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Viola Vaccarino
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute (E.K.K.), Emory University, Atlanta, GA
| | - Arshed A Quyyumi
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| |
Collapse
|
39
|
Ozkok A, Yildiz A. Endothelial Progenitor Cells and Kidney Diseases. Kidney Blood Press Res 2018; 43:701-718. [PMID: 29763891 DOI: 10.1159/000489745] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/03/2018] [Indexed: 01/12/2023] Open
Abstract
Endothelial progenitor cells (EPC) are bone marrow derived or tissue-resident cells that play major roles in the maintenance of vascular integrity and repair of endothelial damage. Although EPCs may be capable of directly engrafting and regenerating the endothelium, the most important effects of EPCs seem to be depended on paracrine effects. In recent studies, specific microvesicles and mRNAs have been found to mediate the pro-angiogenic and regenerative effects of EPCs on endothelium. EPC counts have important prognostic implications in cardiovascular diseases (CVD). Uremia and inflammation are associated with lower EPC counts which probably contribute to increased CVD risks in patients with chronic kidney disease. Beneficial effects of the EPC therapies have been shown in studies performed on different models of CVD and kidney diseases such as acute and chronic kidney diseases and glomerulonephritis. However, lack of a clear definition and specific marker of EPCs is the most important problem causing difficulties in interpretation of the results of the studies investigating EPCs.
Collapse
Affiliation(s)
- Abdullah Ozkok
- University of Health Sciences, Umraniye Training and Research Hospital, Department of Nephrology, Istanbul, Turkey,
| | - Alaattin Yildiz
- Istanbul University, Istanbul Faculty of Medicine, Department of Nephrology, Istanbul, Turkey
| |
Collapse
|
40
|
Increased Endothelial Progenitor Cell Number in Early Stage of Endometrial Cancer. Int J Gynecol Cancer 2018; 27:947-952. [PMID: 28498245 DOI: 10.1097/igc.0000000000000961] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES It is generally believed that circulating endothelial cells (CECs) and endothelial progenitor cells (EPCs) reflect the state of the endothelium, its injury and/or repair possibilities. In different types of cancers, increased numbers of CECs and EPCs were found, suggesting their participation in cancer angiogenesis. The objective of this study was to determine whether, in the blood circulation of women with early endometrial cancer, CEC and EPC levels differ from those of healthy women of similar age. METHODS For CEC number evaluation, samples of peripheral blood cells of women with endometrial carcinoma and control subjects were labeled with anti-CD31 and anti-CD45 antibodies; for EPCs, with anti-VEGFR2 (vascular-endothelium growth factor receptor 2)/KDR and anti-CD34 antibodies. The CEC and EPC cells were then quantified by flow cytometry. RESULTS Endothelial progenitor cell numbers (CD34, VEGFR2/KDR) in the peripheral blood of women with endometrial carcinoma were significantly augmented as compared with those of control healthy women and CEC numbers (CD31, CD45) were similar in both groups. Cancer patients were divided according to the grading into G1 and G2 groups and according to the stage into International Federation of Gynecology and Obstetrics (FIGO) stage IA and FIGO IB groups. Statistically significant augmented EPC numbers were demonstrated only in G1 and FIGO IA patients. CONCLUSIONS These results strongly suggest new vessel formation from recruited endothelial precursors as being involved mainly at the early stages of tumor progression.
Collapse
|
41
|
Abstract
BACKGROUND Burn blister fluid contains several angiogenic factors to promote wound neovascularization. In our previous study, we found that deep partial-thickness burn (DPTB) wounds showed higher expression levels of angiogenin to enhance vascularization compared with superficial partial-thickness burn wounds. Neovascularization is a complex process that involves an interaction between circulating angiogenic cells and mediators. We hypothesized that in addition to angiogenic factors burn blisters may contain specific cell types. The aim of the present study was to characterize the specific cells present in burn blisters. METHODS Twenty-four burn blister fluid samples were obtained with informed consent from patients with superficial partial-thickness burn (n = 16) or DPTB (n = 8) wounds. Blister cells were isolated from individual intact blisters and characterized with flow cytometry analysis using CD14, CD34, vascular endothelial growth factor receptor 2, and CD133 markers. CD14 and CD34 blister cells were also isolated using a magnetic-activated cell sorting system to examine their potential for endothelial differentiation. Angiogenin levels in the burn blister fluids were evaluated with enzyme-linked immunosorbent assay. RESULTS CD14 cells were the most highly represented cell type in the burn fluids of both groups, although a significantly greater percentage of CD14 cells were observed in DPTB fluids. CD14 blister cells had a higher potency to differentiate into functional endothelial cells as compared with CD34 cells. The proportion of CD14 cells gradually increased after burn injury. In contrast to CD14 cells, angiogenin showed the highest expression levels at day 1 postburn. With regard to burn wound neovascularization, angiogenin expression was partially correlated with CD14 blister cells in the burn fluids. CONCLUSIONS We provide the first report on the characterization of blister cells in burn fluids. Our data suggest that CD14 blister cells may play a role in burn wound neovascularization. Measurement of CD14 blister cells serves as a possible tool for assessing burn wound status.
Collapse
|
42
|
Wang D, Li LK, Dai T, Wang A, Li S. Adult Stem Cells in Vascular Remodeling. Am J Cancer Res 2018; 8:815-829. [PMID: 29344309 PMCID: PMC5771096 DOI: 10.7150/thno.19577] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/01/2017] [Indexed: 01/03/2023] Open
Abstract
Understanding the contribution of vascular cells to blood vessel remodeling is critical for the development of new therapeutic approaches to cure cardiovascular diseases (CVDs) and regenerate blood vessels. Recent findings suggest that neointimal formation and atherosclerotic lesions involve not only inflammatory cells, endothelial cells, and smooth muscle cells, but also several types of stem cells or progenitors in arterial walls and the circulation. Some of these stem cells also participate in the remodeling of vascular grafts, microvessel regeneration, and formation of fibrotic tissue around biomaterial implants. Here we review the recent findings on how adult stem cells participate in CVD development and regeneration as well as the current state of clinical trials in the field, which may lead to new approaches for cardiovascular therapies and tissue engineering.
Collapse
|
43
|
Peng BY, Dubey NK, Mishra VK, Tsai FC, Dubey R, Deng WP, Wei HJ. Addressing Stem Cell Therapeutic Approaches in Pathobiology of Diabetes and Its Complications. J Diabetes Res 2018; 2018:7806435. [PMID: 30046616 PMCID: PMC6036791 DOI: 10.1155/2018/7806435] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 04/19/2018] [Accepted: 05/27/2018] [Indexed: 12/14/2022] Open
Abstract
High morbidity and mortality of diabetes mellitus (DM) throughout the human population is a serious threat which needs to be addressed cautiously. Type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM) are most prevalent forms. Disruption in insulin regulation and resistance leads to increased formation and accumulation of advanced end products (AGEs), which further enhance oxidative and nitrosative stress leading to microvascular (retinopathy, neuropathy, and nephropathy) and macrovascular complications. These complications affect the normal function of organ and tissues and may cause life-threatening disorders, if hyperglycemia persists and improperly controlled. Current and traditional treatment procedures are only focused on to regulate the insulin level and do not cure the diabetic complications. Pancreatic transplantation seemed a viable alternative; however, it is limited due to lack of donors. Cell-based therapy such as stem cells is considered as a promising therapeutic agent against DM and diabetic complications owing to their multilineage differentiation and regeneration potential. Previous studies have demonstrated the various impacts of both pluripotent and multipotent stem cells on DM and its micro- and macrovascular complications. Therefore, this review summarizes the potential of stem cells to treat DM and its related complications.
Collapse
Affiliation(s)
- Bou-Yue Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Department of Dentistry, Taipei Medical University Hospital, Taipei City 110, Taiwan
| | - Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Viraj Krishna Mishra
- Applied Biotech Engineering Centre (ABEC), Department of Biotechnology, Ambala College of Engineering and Applied Research, Ambala, India
| | - Feng-Chou Tsai
- Department of Stem Cell Research, Cosmetic Clinic Group, Taipei City 110, Taiwan
| | - Rajni Dubey
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei City 106, Taiwan
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Hong-Jian Wei
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
| |
Collapse
|
44
|
Szpera-Goździewicz A, Majcherek M, Boruczkowski M, Goździewicz T, Dworacki G, Wicherek L, Bręborowicz GH. Circulating endothelial cells, circulating endothelial progenitor cells, and von Willebrand factor in pregnancies complicated by hypertensive disorders. Am J Reprod Immunol 2017; 77. [PMID: 28224722 DOI: 10.1111/aji.12625] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022] Open
Abstract
PROBLEM We tested the hypothesis that the number of both CECs and CEPCs as well as the vWf blood plasma concentration are altered in pregnant women with hypertensive disorders. METHOD OF STUDY Seventy-five pregnant women were enrolled in our study. We used multicolor flow cytometry for CEC and CEPC analysis and the commercial human VWF ELISA kit to measure vWf blood plasma concentration. RESULTS The highest number of CECs was found in the chronic hypertension group and the lowest number in the healthy pregnant control group. The highest number of CEPCs was found in the control group and the lowest number in the chronic hypertension group. The vWf blood plasma concentration was the highest in the pre-eclampsia group. The CEPC/CEC ratio reached its lowest value in the chronic hypertension group and its highest value in the control group. CONCLUSION The number of both CECs and CEPCs as well as the vWf blood plasma concentration depends on the type of hypertension complicating the pregnancy.
Collapse
Affiliation(s)
- Agata Szpera-Goździewicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Majcherek
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Boruczkowski
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Tomasz Goździewicz
- Department of Gynecology, Poznan University of Medical Sciences, Poznan, Poland
| | - Grzegorz Dworacki
- Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Lukasz Wicherek
- Department of Oncology, Radiotherapy and Gynecological Oncology, Ludwik Rydygier Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | | |
Collapse
|
45
|
RNA-sequencing-based comparative analysis of human hepatic progenitor cells and their niche from alcoholic steatohepatitis livers. Cell Death Dis 2017; 8:e3164. [PMID: 29095436 PMCID: PMC5775409 DOI: 10.1038/cddis.2017.543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/17/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023]
Abstract
Hepatic progenitor cells (HPCs) are small cells with a relative large oval nucleus and a scanty cytoplasm situated in the canals of Hering that express markers of (immature) hepatocytes and cholangiocytes. HPCs are present in large numbers in alcoholic steatohepatitis (ASH), one of the leading causes of chronic liver disease. To date, the mechanisms responsible for proliferation and differentiation of human HPCs are still poorly understood and the role of HPCs in ASH development is unknown. In this study, we aimed to characterise human HPCs and their interactions with other cells through comparison, on both protein and RNA level, of HPC-enriched cell populations from adult human liver tissue using different isolation methods. Fresh human liver tissue was collected from ASH explant livers and HPC-enriched cell populations were obtained via four different isolation methods: side population (SP), epithelial cell adhesion molecule (EpCAM) and trophoblast antigen 2 (TROP-2) membrane marker isolation and laser capture microdissection. Gene expression profiles of fluorescent-activated cell-sorted HPCs, whole liver extracts and laser microdissected HPC niches were determined by RNA-sequencing. Immunohistochemical evaluation of the isolated populations indicated the enrichment of HPCs in the SP, EpCAM+ and TROP-2+ cell populations. Pathway analysis of the transcription profiles of human HPCs showed an enrichment and activation of known HPC pathways like Wnt/β-catenin, TWEAK and HGF. Integration of the HPC niche profile suggests autocrine signalling by HPCs (TNFα, PDGFB and VEGFA) as well as paracrine signalling from the surrounding niche cells including MIF and IGF-1. In addition, we identified IL-17 A signalling as a potentially novel pathway in HPC biology. In conclusion, we provide the first RNA-seq-based, comparative transcriptome analysis of isolated human HPCs from ASH patients and revealed active signalling between HPCs and their surrounding niche cells in ASH livers and suggest that HPCs can actively contribute to liver inflammation.
Collapse
|
46
|
Mitchell A, Fujisawa T, Mills NL, Brittan M, Newby DE, Cruden NLM. Endothelial Progenitor Cell Biology and Vascular Recovery Following Transradial Cardiac Catheterization. J Am Heart Assoc 2017; 6:e006610. [PMID: 29080864 PMCID: PMC5721759 DOI: 10.1161/jaha.117.006610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Transradial catheterization is associated with radial artery injury and vasomotor dysfunction and represents an accessible model of acute vascular injury in humans. We characterized vascular injury and functional recovery to understand the role of circulating endothelial progenitor cells in vascular repair. METHODS AND RESULTS In 50 patients (aged 64±10 years, 70% male) undergoing transradial cardiac catheterization, radial artery injury was assessed by optical coherence tomography and examination of explanted vascular sheaths. Flow- and nitrate-mediated dilatation of the radial artery was assessed in both arms at baseline, at 24 hours, and at 1, 4, and 12 weeks. Circulating endothelial progenitor cell populations were quantified using flow cytometry. Late endothelial outgrowth colonies were isolated and examined in vitro. Optical coherence tomography identified macroscopic injury in 12 of 50 patients (24%), but endothelial cells (1.9±1.2×104 cells) were isolated from all arterial sheaths examined. Compared with the noncatheterized radial artery, flow-mediated vasodilatation was impaired in the catheterized artery at 24 hours (9.9±4.6% versus 4.1±3.1%, P<0.0001) and recovered by 12 weeks (8.1±4.9% versus 10.1±4.9%, P=0.09). Although the number of CD133+ cells increased 24 hours after catheterization (P=0.02), the numbers of CD34+ cells and endothelial outgrowth colonies were unchanged. Migration of endothelial cells derived from endothelial outgrowth colonies correlated with arterial function before catheterization but was not related to recovery of function following injury. CONCLUSIONS Transradial cardiac catheterization causes endothelial denudation, vascular injury, and vasomotor dysfunction that recover over 12 weeks. Recovery of vascular function does not appear to be dependent on the mobilization or function of endothelial progenitor cells. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT02147119.
Collapse
Affiliation(s)
- Andrew Mitchell
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Takeshi Fujisawa
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Nicholas L Mills
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Mairi Brittan
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - David E Newby
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Nicholas L M Cruden
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| |
Collapse
|
47
|
Yang JG, Wang LL, Ma DC. Effects of vascular endothelial growth factors and their receptors on megakaryocytes and platelets and related diseases. Br J Haematol 2017; 180:321-334. [PMID: 29076133 DOI: 10.1111/bjh.15000] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It is well known that vascular endothelial growth factors (VEGFs) and their receptors (vascular endothelial growth factor receptors, VEGFRs) are expressed in different tissues, and VEGF-VEGFR loops regulate a wide range of responses, including metabolic homeostasis, cell proliferation, migration and tubuleogenesis. As ligands, VEGFs act on three structurally related VEGFRs (VEGFR1, VEGFR2 and VEGFR3 [also termed FLT1, KDR and FLT4, respectively]) that deliver downstream signals. Haematopoietic stem cells (HSCs), megakaryocytic cell lines, cultured megakaryocytes (MKs), primary MKs and abnormal MKs express and secrete VEGFs. During the development from HSCs to MKs, VEGFR1, VEGFR2 and VEGFR3 are expressed at different developmental stages, respectively, and re-expressed, e.g., VEGFR2, and play different roles in commitment, differentiation, proliferation, survival and polyplodization of HSCs/MKs via autocrine, paracrine and/or even intracrine loops. Moreover, VEGFs and their receptors are abnormally expressed in MK-related diseases, including myeloproliferative neoplasms, myelodysplastic syndromes and acute megakaryocytic leukaemia (a rare subtype of acute myeloid leukaemia), and they lead to the disordered proliferation/differentiation of bone marrow cells and angiogenesis, indicating that they are closely related to these diseases. Thus, targeting VEGF-VEGFR loops may be of potential therapeutic value.
Collapse
Affiliation(s)
- Jin-Gang Yang
- Department of Experimental Medicine, General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Li-Li Wang
- Department of Experimental Medicine, General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| | - Dong-Chu Ma
- Department of Experimental Medicine, General Hospital of Shenyang Military Region, Shenyang, Liaoning, China
| |
Collapse
|
48
|
Peristiowati Y. Protective Effects of Catechins Isolate From GMB4 Clone Green Tea Against EPC In Type 2 Diabetes Mellitus. JURNAL NERS 2017. [DOI: 10.20473/jn.v12i2.4685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Introduction : This study aimed to evaluate the role of catechins isolates from GMB4 clone green to in the dynamics of endothelial progenitor cells (EPC) in type 2 diabetes mellitus (DM).Methods: 25 Wistar rats, were divided into 5 groups, including control group, type 2 diabetes mellitus group, treated daily administration of extracts of catechins isolate from GMB4 clones green tea at 20; 40; and 60 mg /kgBB doses for 6 weeks. EPCs analysis was performed by flow cytometry,levels of NO was performed using a spectrophotometer, serum levels of SDF-1a was performed with ELISA technique. Analysis of the expression of SDF-1 and CXCR-4 was performed using immunohistochemistry techniques.Results : The CD34+ and CD133+ count is lower in the DM group compared to the control group (P < 0.05). Of the three doses of catechins, only the highest dose were able to significantly increase CD34+ count compared to rats in the diabetes mellitus group (P < 0.05), Serum NO level is significantly higher in the DM the control group (P < 0.05). All three doses of catechins were able to significantly increase the expression of SDF-1a and CXCR4 in the aorta compared to the DM group or the control group (P < 0.05).Discussion : It can be concluded that high dose cathecin isolate from GMB-4 clone green tea (60 mg/kgBB) may trigger the proliferation and maturation of EPCs in rats with type 2 DM in environment with high level of NO, involving the interacton between SDF-1a and CXCR4 in the aorta.
Collapse
|
49
|
Wang Q, Liu L, Li Y, Wang J, Liu Y, Wu Q, Wang B, Qi B, Qi B. Hypoxic Preconditioning Enhances Biological Function of Endothelial Progenitor Cells via Notch-Jagged1 Signaling Pathway. Med Sci Monit 2017; 23:4665-4667. [PMID: 28959004 PMCID: PMC5633065 DOI: 10.12659/msm.902470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Hypoxic preconditioning may be a key influence on functions of endothelial progenitor cells (EPCs). MATERIAL AND METHODS To investigate the role and mechanism of the Notch-Jagged1 pathway on endothelial progenitor cells in hypoxic preconditioning, endothelial progenitor cells were randomly allocated into 5 groups: 1 Normoxic control group; 2 Hypoxic blank group; 3 Hypoxic+25 μM DAPT group; 4 Hypoxic+50 μM DAPT group; 5 Hypoxic+100 μM DAPT group. After reoxygenation, protein and mRNA levels of Jagged1 were measured by Western blot and quantitative RT-PCR. The MTT test was used to assess proliferation. ELISA was used to measure NO and VEGF secretion. RESULTS Hypoxic preconditioning treatment significantly upregulated both protein and mRNA levels of Jagged1 in endothelial progenitor cells. It also enhanced proliferation ability and elevated secretion of NO and VEGF. Furthermore, after blocking the Notch pathway by using DAPT, Jagged1 expression and EP proliferation, migration, and secretion of NO and VEGF were decreased in a dose-dependent manner. CONCLUSIONS Our results suggest the Notch-Jagged1 pathway enhances EPCs proliferation and secretion ability during hypoxic preconditioning.
Collapse
Affiliation(s)
- Qian Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - LiHua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - YuanYuan Li
- Department of Neurology, Zhumadian Zhongxin Hospital, Zhumadian, Henan, China (mainland)
| | - Jinfeng Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - QinQin Wu
- Department of Geriatrics, The Central Hospital of Wuhan, Wuhan, Hubei, China (mainland)
| | - Bin Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Benming Qi
- Department of Otolaryngology, 1st People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - BenLing Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| |
Collapse
|
50
|
Abstract
Vascular complications contribute significantly to morbidity and mortality of diabetes mellitus. The primary cause of vascular complications in diabetes mellitus is hyperglycaemia, associated with endothelial dysfunction and impaired neovascularization. Circulating endothelial progenitor cells was shown to play important roles in vascular repair and promoting neovascularization. In this review, we will demonstrate the individual effect of high glucose on endothelial progenitor cells. Endothelial progenitor cells isolated from healthy subjects exposed to high glucose conditions or endothelial progenitor cells isolated from diabetic patients exhibit reduced number of endothelial cell colony forming units, impaired abilities of differentiation, proliferation, adhesion and migration, tubulization, secretion, mobilization and homing, whereas enhanced senescence. Increased production of reactive oxygen species by the mitochondria seems to play a crucial role in high glucose-induced endothelial progenitor cells deficit. Later, we will review the agents that might be used to alleviate dysfunction of endothelial progenitor cells induced by high glucose. The conclusions are that the relationship between hyperglycaemia and endothelial progenitor cells dysfunction is only beginning to be recognized, and future studies should pay more attention to the haemodynamic environment of endothelial progenitor cells and ageing factors to discover novel treatment agents.
Collapse
Affiliation(s)
- Hongyan Kang
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xuejiao Ma
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jiajia Liu
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- 2 National Research Center for Rehabilitation Technical Aids, Beijing, China
| | - Xiaoyan Deng
- 1 Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|