1
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
2
|
Park G, Hwang DY, Kim DY, Han JY, Lee E, Hwang H, Park JS, Kim DW, Hong S, Yim SV, Hong HS, Son Y. Identification of CD141 +vasculogenic precursor cells from human bone marrow and their endothelial engagement in the arteriogenesis by co-transplantation with mesenchymal stem cells. Stem Cell Res Ther 2024; 15:388. [PMID: 39482744 PMCID: PMC11526567 DOI: 10.1186/s13287-024-03994-9] [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: 07/09/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is a condition characterized by insufficient blood flow to the lower limbs, resulting in severe ischemia and potentially leading to amputation. This study aims to identify novel vasculogenic precursor cells (VPCs) in human bone marrow and evaluate their efficacy in combination with bone marrow-derived mesenchymal stem cells (BM-MSCs) for the treatment of CLI. METHODS Ex vivo cultured VPCs and BM-MSCs from bone marrow were characterized and their effects on neovascularization and long-term tissue regeneration were tested in a mouse CLI model. RESULTS VPCs, expressing high levels of hepatocyte growth factor and c-MET, were identified from human bone marrow aspirates. These cells exhibited strong vasculogenic capacity in vitro but possessed a cellular phenotype distinct from those of previously reported endothelial precursor cells in circulation or cord blood. They also expressed most surface markers of BM-MSCs and demonstrated multipotent differentiation ability. Screening of 376 surface markers revealed that VPCs uniquely display CD141 (thrombomodulin). CD141+VPCs are present in BM aspirates as a rare population and can be expanded ex vivo with a population doubling time of approximately 20 h, generating an elaborate vascular network even under angiogenic factor-deficient conditions and recruiting BM-MSCs to the network as pericyte-like cells. Intramuscular transplantation of a combination of human CD141+VPCs and BM-MSCs at a ratio of 2:1 resulted in limb salvage, blood flow recovery, and regeneration of large vessels in the femoral artery-removed CLI model, with an efficacy superior to that of singular transplantation. Importantly, large arteries and arterioles in dual cell transplantation expressed human CD31 in the intima and human α-smooth muscle actin in media layer at 4 and 12 weeks, likely indicating their lineage commitment to endothelial cells and vascular smooth muscle, respectively, in vivo. CONCLUSION Dual-cell therapy using BM-derived CD141+ VPCs and BM-MSCs holds potential for further development in clinical trials to treat peripheral artery disease and diabetic ulcers.
Collapse
Affiliation(s)
- Gabee Park
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Dae Yeon Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Ji Young Han
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Euiseon Lee
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Hwakyung Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Dae Wook Kim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Seonmin Hong
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Sung Vin Yim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea.
- East-West Medical Research Institute, Kyung Hee University, Seoul, Korea.
| | - Youngsook Son
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea.
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea.
| |
Collapse
|
3
|
Chen DX, Lu CH, Na N, Yin RX, Huang F. Endothelial progenitor cell-derived extracellular vesicles: the world of potential prospects for the treatment of cardiovascular diseases. Cell Biosci 2024; 14:72. [PMID: 38840175 DOI: 10.1186/s13578-024-01255-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
Cardiovascular diseases (CVDs) have emerged as a predominant threat to human health, surpassing the incidence and mortality rates of neoplastic diseases. Extracellular vesicles (EVs) serve as vital mediators in intercellular communication and material exchange. Endothelial progenitor cells (EPCs), recognized as precursors of vascular endothelial cells (ECs), have garnered considerable attention in recent years due to the potential therapeutic value of their derived extracellular vesicles (EPC-EVs) in the context of CVDs. This comprehensive review systematically explores the origins, characteristics, and functions of EPCs, alongside the classification, properties, biogenesis, and extraction techniques of EVs, with particular emphasis on their protective roles in CVDs. Additionally, we delve into the essential bioactive components of EPC-EVs, including microRNAs, long non-coding RNAs, and proteins, analyzing their beneficial effects in promoting angiogenesis, anti-inflammatory and anti-oxidant activities, anti-fibrosis, anti-apoptosis, and myocardial regeneration. Furthermore, this review comprehensively investigates the therapeutic potential of EPC-EVs across various CVDs, encompassing acute myocardial infarction, myocardial ischemia-reperfusion injury, atherosclerosis, non-ischemic cardiomyopathies, and diabetic cardiovascular disease. Lastly, we summarize the potential challenges associated with the clinical application of EPC-EVs and outline future directions, aiming to offer a valuable resource for both theoretical insights and practical applications of EPC-EVs in managing CVDs.
Collapse
Affiliation(s)
- De-Xin Chen
- Department of Cardiology & Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Chuang-Hong Lu
- Department of Cardiology & Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Na Na
- Department of Neuroscience, Scripps Research Institute, No.10550 North Torrey Pines Road, La Jolla, San Diego, CA, 92037, USA
| | - Rui-Xing Yin
- Department of Cardiology & Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Feng Huang
- Department of Cardiology & Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
Xining Z, Sai L. The Evolving Function of Vasculature and Pro-angiogenic Therapy in Fat Grafting. Cell Transplant 2024; 33:9636897241264976. [PMID: 39056562 PMCID: PMC11282510 DOI: 10.1177/09636897241264976] [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: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous fat grating is a widely-accepted method to correct soft tissue deficiency. Although fat transplantation shows excellent biocompatibility and simple applicability, the relatively low retention rate caused by fat necrosis is still a challenge. The vasculature is integral after fat grafting, serving multiple crucial functions. Rapid and effective angiogenesis within grafts is essential for supplying oxygen necessary for adipocytes' survival. It facilitates the influx of inflammatory cells to remove necrotic adipocytes and aids in the delivery of regenerative cells for adipose tissue regeneration in fat grafts. The vasculature also provides a niche for interaction between adipose progenitor cells and vascular progenitor cells, enhancing angiogenesis and adipogenesis in grafts. Various methods, such as enriching grafts with diverse pro-angiogenic cells or utilizing cell-free approaches, have been employed to enhance angiogenesis. Beige and dedifferentiated adipocytes in grafts could increase vessel density. This review aims to outline the function of vasculature in fat grafting and discuss different cell or cell-free approaches that can enhance angiogenesis following fat grafting.
Collapse
Affiliation(s)
- Zhang Xining
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luo Sai
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Zhao L, Lee AS, Sasagawa K, Sokol J, Wang Y, Ransom RC, Zhao X, Ma C, Steininger HM, Koepke LS, Borrelli MR, Brewer RE, Lee LL, Huang X, Ambrosi TH, Sinha R, Hoover MY, Seita J, Weissman IL, Wu JC, Wan DC, Xiao J, Longaker MT, Nguyen PK, Chan CK. A Combination of Distinct Vascular Stem/Progenitor Cells for Neovascularization and Ischemic Rescue. Arterioscler Thromb Vasc Biol 2023; 43:1262-1277. [PMID: 37051932 PMCID: PMC10281192 DOI: 10.1161/atvbaha.122.317943] [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: 12/10/2021] [Revised: 03/09/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Peripheral vascular disease remains a leading cause of vascular morbidity and mortality worldwide despite advances in medical and surgical therapy. Besides traditional approaches, which can only restore blood flow to native arteries, an alternative approach is to enhance the growth of new vessels, thereby facilitating the physiological response to ischemia. METHODS The ActinCreER/R26VT2/GK3 Rainbow reporter mouse was used for unbiased in vivo survey of injury-responsive vasculogenic clonal formation. Prospective isolation and transplantation were used to determine vessel-forming capacity of different populations. Single-cell RNA-sequencing was used to characterize distinct vessel-forming populations and their interactions. RESULTS Two populations of distinct vascular stem/progenitor cells (VSPCs) were identified from adipose-derived mesenchymal stromal cells: VSPC1 is CD45-Ter119-Tie2+PDGFRa-CD31+CD105highSca1low, which gives rise to stunted vessels (incomplete tubular structures) in a transplant setting, and VSPC2 which is CD45-Ter119-Tie2+PDGFRa+CD31-CD105lowSca1high and forms stunted vessels and fat. Interestingly, cotransplantation of VSPC1 and VSPC2 is required to form functional vessels that improve perfusion in the mouse hindlimb ischemia model. Similarly, VSPC1 and VSPC2 populations isolated from human adipose tissue could rescue the ischemic condition in mice. CONCLUSIONS These findings suggest that autologous cotransplantation of synergistic VSPCs from nonessential adipose tissue can promote neovascularization and represents a promising treatment for ischemic disease.
Collapse
Affiliation(s)
- Liming Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Andrew S. Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
| | - Koki Sasagawa
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Jan Sokol
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Ryan C. Ransom
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xin Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Chao Ma
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Holly M. Steininger
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lauren S. Koepke
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Mimi R. Borrelli
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rachel E. Brewer
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lorene L.Y. Lee
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xianxi Huang
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Thomas H. Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Malachia Y. Hoover
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| | - Joseph C. Wu
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Michael T. Longaker
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Charles K.F. Chan
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| |
Collapse
|
6
|
Marshall AJ, Gaubert A, Kapoor A, Tan A, McIntosh E, Jang JY, Yew B, Ho JK, Blanken AE, Dutt S, Sible IJ, Li Y, Rodgers K, Nation DA. Blood-Derived Progenitor Cells Are Depleted in Older Adults with Cognitive Impairment: A Role for Vascular Resilience? J Alzheimers Dis 2023; 93:1041-1050. [PMID: 37154177 PMCID: PMC10258882 DOI: 10.3233/jad-220269] [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] [Accepted: 03/23/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Depletion of blood-derived progenitor cells, including so called "early endothelial progenitor cells", has been observed in individuals with early stage Alzheimer's disease relative to matched older control subjects. These findings could implicate the loss of angiogenic support from hematopoietic progenitors or endothelial progenitors in cognitive dysfunction. OBJECTIVE To investigate links between progenitor cell proliferation and mild levels of cognitive dysfunction. METHODS We conducted in vitro studies of blood-derived progenitor cells using blood samples from sixty-five older adults who were free of stroke or dementia. Peripheral blood mononuclear cells from venous blood samples were cultured in CFU-Hill media and the number of colony forming units were counted after 5 days in vitro. Neuropsychological testing was administered to all participants. RESULTS Fewer colony forming units were observed in samples from older adults with a Clinical Dementia Rating global score of 0.5 versus 0. Older adults whose samples developed fewer colony forming units exhibited worse performance on neuropsychological measures of memory, executive functioning, and language ability. CONCLUSION These data suggest blood progenitors may represent a vascular resilience marker related to cognitive dysfunction in older adults.
Collapse
Affiliation(s)
- Anisa J. Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Aimee Gaubert
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Alick Tan
- Department of Clinical Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Elissa McIntosh
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Jung Yun Jang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Belinda Yew
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean K. Ho
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Anna E. Blanken
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Isabel J. Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Yanrong Li
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Daniel A. Nation
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
7
|
Yu QC, Geng A, Preusch CB, Chen Y, Peng G, Xu Y, Jia Y, Miao Y, Xue H, Gao D, Bao L, Pan W, Chen J, Garcia KC, Cheung TH, Zeng YA. Activation of Wnt/β-catenin signaling by Zeb1 in endothelial progenitors induces vascular quiescence entry. Cell Rep 2022; 41:111694. [DOI: 10.1016/j.celrep.2022.111694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 08/05/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
|
8
|
Steens J, Klein D. HOX genes in stem cells: Maintaining cellular identity and regulation of differentiation. Front Cell Dev Biol 2022; 10:1002909. [PMID: 36176275 PMCID: PMC9514042 DOI: 10.3389/fcell.2022.1002909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Stem cells display a unique cell type within the body that has the capacity to self-renew and differentiate into specialized cell types. Compared to pluripotent stem cells, adult stem cells (ASC) such as mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) exhibit restricted differentiation capabilities that are limited to cell types typically found in the tissue of origin, which implicates that there must be a certain code or priming determined by the tissue of origin. HOX genes, a subset of homeobox genes encoding transcription factors that are generally repressed in undifferentiated pluripotent stem cells, emerged here as master regulators of cell identity and cell fate during embryogenesis, and in maintaining this positional identity throughout life as well as specifying various regional properties of respective tissues. Concurrently, intricate molecular circuits regulated by diverse stem cell-typical signaling pathways, balance stem cell maintenance, proliferation and differentiation. However, it still needs to be unraveled how stem cell-related signaling pathways establish and regulate ASC-specific HOX expression pattern with different temporal-spatial topography, known as the HOX code. This comprehensive review therefore summarizes the current knowledge of specific ASC-related HOX expression patterns and how these were integrated into stem cell-related signaling pathways. Understanding the mechanism of HOX gene regulation in stem cells may provide new ways to manipulate stem cell fate and function leading to improved and new approaches in the field of regenerative medicine.
Collapse
|
9
|
Chesnais F, Hue J, Roy E, Branco M, Stokes R, Pellon A, Le Caillec J, Elbahtety E, Battilocchi M, Danovi D, Veschini L. High content Image Analysis to study phenotypic heterogeneity in endothelial cell monolayers. J Cell Sci 2022; 135:273879. [PMID: 34982151 DOI: 10.1242/jcs.259104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
Endothelial cells (EC) are heterogeneous across and within tissues, reflecting distinct, specialised functions. EC heterogeneity has been proposed to underpin EC plasticity independently from vessel microenvironments. However, heterogeneity driven by contact-dependent or short-range cell-cell crosstalk cannot be evaluated with single cell transcriptomic approaches as spatial and contextual information is lost. Nonetheless, quantification of EC heterogeneity and understanding of its molecular drivers is key to developing novel therapeutics for cancer, cardiovascular diseases and for revascularisation in regenerative medicine. Here, we developed an EC profiling tool (ECPT) to examine individual cells within intact monolayers. We used ECPT to characterise different phenotypes in arterial, venous and microvascular EC populations. In line with other studies, we measured heterogeneity in terms of cell cycle, proliferation, and junction organisation. ECPT uncovered a previously under-appreciated single-cell heterogeneity in NOTCH activation. We correlated cell proliferation with different NOTCH activation states at the single cell and population levels. The positional and relational information extracted with our novel approach is key to elucidating the molecular mechanisms underpinning EC heterogeneity.
Collapse
Affiliation(s)
- Francois Chesnais
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Jonas Hue
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Errin Roy
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Marco Branco
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Ruby Stokes
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Aize Pellon
- Centre for host-microbiome interactions, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Juliette Le Caillec
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Eyad Elbahtety
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Matteo Battilocchi
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Davide Danovi
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK.,bit.bio, Babraham Research Campus, The Dorothy Hodgkin Building, Cambridge CB22 3FH, UK
| | - Lorenzo Veschini
- Academic centre of reconstructive science, Faculty of Dentistry Oral & Craniofacial Sciences, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
10
|
Moraes de Lima Perini M, Valuch CR, Dadwal UC, Awosanya OD, Mostardo SL, Blosser RJ, Knox AM, McGuire AC, Battina HL, Nazzal M, Kacena MA, Li J. Characterization and assessment of lung and bone marrow derived endothelial cells and their bone regenerative potential. Front Endocrinol (Lausanne) 2022; 13:935391. [PMID: 36120459 PMCID: PMC9470942 DOI: 10.3389/fendo.2022.935391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is important for successful fracture repair. Aging negatively affects the number and activity of endothelial cells (ECs) and subsequently leads to impaired bone healing. We previously showed that implantation of lung-derived endothelial cells (LECs) improved fracture healing in rats. In this study, we characterized and compared neonatal lung and bone marrow-derived endothelial cells (neonatal LECs and neonatal BMECs) and further asses3sed if implantation of neonatal BMECs could enhance bone healing in both young and aged mice. We assessed neonatal EC tube formation, proliferation, and wound migration ability in vitro in ECs isolated from the bone marrow and lungs of neonatal mice. The in vitro studies demonstrated that both neonatal LECs and neonatal BMECs exhibited EC traits. To test the function of neonatal ECs in vivo, we created a femoral fracture in young and aged mice and implanted a collagen sponge to deliver neonatal BMECs at the fracture site. In the mouse fracture model, endochondral ossification was delayed in aged control mice compared to young controls. Neonatal BMECs significantly improved endochondral bone formation only in aged mice. These data suggest BMECs have potential to enhance aged bone healing. Compared to LECs, BMECs are more feasible for translational cell therapy and clinical applications in bone repair. Future studies are needed to examine the fate and function of BMECs implanted into the fracture sites.
Collapse
Affiliation(s)
| | - Conner R. Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
| | - Ushashi C. Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Olatundun D. Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sarah L. Mostardo
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rachel J. Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Adam M. Knox
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anthony C. McGuire
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hanisha L. Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Murad Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Richard L. Roudebush Veterans Affairs (VA) Medical Center, Indianapolis, IN, United States
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, United States
- *Correspondence: Jiliang Li,
| |
Collapse
|
11
|
Dight J, Zhao J, Styke C, Khosrotehrani K, Patel J. Resident vascular endothelial progenitor definition and function: the age of reckoning. Angiogenesis 2021; 25:15-33. [PMID: 34499264 PMCID: PMC8813834 DOI: 10.1007/s10456-021-09817-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/05/2021] [Indexed: 02/07/2023]
Abstract
The cardiovascular system is composed around the central function of the endothelium that lines the inner surfaces of its vessels. In recent years, the existence of a progenitor population within the endothelium has been validated through the study of endothelial colony-forming cells (ECFCs) in human peripheral blood and certain vascular beds. However, our knowledge on endothelial populations in vivo that can give rise to ECFCs in culture has been limited. In this review we report and analyse recent attempts at describing progenitor populations in vivo from murine studies that reflect the self-renewal and stemness capacity observed in ECFCs. We pinpoint seminal discoveries within the field, which have phenotypically defined, and functionally scrutinised these endothelial progenitors. Furthermore, we review recent publications utilising single-cell sequencing technologies to better understand the endothelium in homeostasis and pathology.
Collapse
Affiliation(s)
- James Dight
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Jilai Zhao
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Cassandra Styke
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| | - Jatin Patel
- The University of Queensland Diamantina Institute, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia. .,Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
| |
Collapse
|
12
|
Dierick F, Solinc J, Bignard J, Soubrier F, Nadaud S. Progenitor/Stem Cells in Vascular Remodeling during Pulmonary Arterial Hypertension. Cells 2021; 10:cells10061338. [PMID: 34071347 PMCID: PMC8226806 DOI: 10.3390/cells10061338] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by an important occlusive vascular remodeling with the production of new endothelial cells, smooth muscle cells, myofibroblasts, and fibroblasts. Identifying the cellular processes leading to vascular proliferation and dysfunction is a major goal in order to decipher the mechanisms leading to PAH development. In addition to in situ proliferation of vascular cells, studies from the past 20 years have unveiled the role of circulating and resident vascular in pulmonary vascular remodeling. This review aims at summarizing the current knowledge on the different progenitor and stem cells that have been shown to participate in pulmonary vascular lesions and on the pathways regulating their recruitment during PAH. Finally, this review also addresses the therapeutic potential of circulating endothelial progenitor cells and mesenchymal stem cells.
Collapse
Affiliation(s)
- France Dierick
- Lady Davis Institute for Medical Research, McGill University, Montréal, QC H3T 1E2, Canada;
| | - Julien Solinc
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Juliette Bignard
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Florent Soubrier
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
| | - Sophie Nadaud
- UMR_S 1166, Faculté de Médecine Pitié-Salpêtrière, INSERM, Sorbonne Université, 75013 Paris, France; (J.S.); (J.B.); (F.S.)
- Correspondence:
| |
Collapse
|
13
|
Vašíček J, Baláži A, Bauer M, Svoradová A, Tirpáková M, Tomka M, Chrenek P. Molecular Profiling and Gene Banking of Rabbit EPCs Derived from Two Biological Sources. Genes (Basel) 2021; 12:genes12030366. [PMID: 33806502 PMCID: PMC7998175 DOI: 10.3390/genes12030366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been broadly studied for several years due to their outstanding regenerative potential. Moreover, these cells might be a valuable source of genetic information for the preservation of endangered animal species. However, a controversy regarding their characterization still exists. The aim of this study was to isolate and compare the rabbit peripheral blood- and bone marrow-derived EPCs with human umbilical vein endothelial cells (HUVECs) in terms of their phenotype and morphology that could be affected by the passage number or cryopreservation as well as to assess their possible neuro-differentiation potential. Briefly, cells were isolated and cultured under standard endothelial conditions until passage 3. The morphological changes during the culture were monitored and each passage was analyzed for the typical phenotype using flow cytometry, quantitative real–time polymerase chain reaction (qPCR) and novel digital droplet PCR (ddPCR), and compared to HUVECs. The neurogenic differentiation was induced using a commercial kit. Rabbit cells were also cryopreserved for at least 3 months and then analyzed after thawing. According to the obtained results, both rabbit EPCs exhibit a spindle-shaped morphology and high proliferation rate. The both cell lines possess same stable phenotype: CD14−CD29+CD31−CD34−CD44+CD45−CD49f+CD73+CD90+CD105+CD133−CD146−CD166+VE-cadherin+VEGFR-2+SSEA-4+MSCA-1−vWF+eNOS+AcLDL+ALDH+vimentin+desmin+α-SMA+, slightly different from HUVECs. Moreover, both induced rabbit EPCs exhibit neuron-like morphological changes and expression of neuronal markers ENO2 and MAP2. In addition, cryopreserved rabbit cells maintained high viability (>85%) and endothelial phenotype after thawing. In conclusion, our findings suggest that cells expanded from the rabbit peripheral blood and bone marrow are of the endothelial origin with a stable marker expression and interesting proliferation and differentiation capacity.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| | - Andrej Baláži
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Miroslav Bauer
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Botany and Genetics, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nábrežie mládeže 91, 949 74 Nitra, Slovakia
| | - Andrea Svoradová
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Mária Tirpáková
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- AgroBioTech Research Center, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Marián Tomka
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Peter Chrenek
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| |
Collapse
|
14
|
Hassanpour M, Aghamohamadzade N, Cheraghi O, Heidarzadeh M, Nouri M. Current status of cardiac regenerative medicine; An update on point of view to cell therapy application. J Cardiovasc Thorac Res 2021; 12:256-268. [PMID: 33510874 PMCID: PMC7828760 DOI: 10.34172/jcvtr.2020.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 09/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Because of the economic and social burden of acute myocardial infarction and its chronic consequences in surviving patients, understanding the pathophysiology of myocardial infarction injury is a major priority for cardiovascular research. MI is defined as cardiomyocytes death caused by an ischemic that resulted from the apoptosis, necrosis, necroptosis, and autophagy. The phases of normal repair following MI including inflammatory, proliferation, and maturation. Normal repair is slow and inefficient generally so that other treatments are required. Because of difficulties, outcomes, and backwashes of traditional therapies including coronary artery bypass grafting, balloon angioplasty, heart transplantation, and artificial heart operations, the novel strategy in the treatment of MI, cell therapy, was newly emerged. In cell therapy, a new population of cells has created that substitute with damaged cells. Different types of stem cell and progenitor cells have been shown to improve cardiac function through various mechanisms, including the formation of new myocytes, endothelial cells, and vascular smooth muscle cells. Bone marrow- and/or adipose tissue-derived mesenchymal stem cells, embryonic stem cells, autologous skeletal myoblasts, induced pluripotent stem cells, endothelial progenitor cells, cardiac progenitor cells and cardiac pericytes considered as a source for cell therapy. In this study, we focused on the point of view of the cell sources.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Omid Cheraghi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Abstract
Blood vessel formation is a key feature in physiologic and pathologic processes. Once considered a homogeneous cell population that functions as a passive physical barrier between blood and tissue, endothelial cells (ECs) are now recognized to be quite "heterogeneous." While numerous attempts to enhance endothelial repair and replacement have been attempted using so called "endothelial progenitor cells" it is now clear that a better understanding of the origin, location, and activation of stem and progenitor cells of the resident vascular endothelium is required before attempting exogenous cell therapy approaches. This chapter provides an overview for performance of single-cell clonogenic studies of human umbilical cord blood circulating endothelial colony-forming cells (ECFC) that represent distinct precursors for the endothelial lineage with vessel forming potential.
Collapse
|
16
|
Shi X, He L, Zhang SM, Luo J. Human iPS Cell-derived Tissue Engineered Vascular Graft: Recent Advances and Future Directions. Stem Cell Rev Rep 2020; 17:862-877. [PMID: 33230612 DOI: 10.1007/s12015-020-10091-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 12/19/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) generated from human primary cells represent a promising vascular interventional therapy. However, generation and application of these TEVGs may be significantly hindered by the limited accessibility, finite expandability, donor-donor functional variation and immune-incompatibility of primary seed cells from donors. Alternatively, human induced pluripotent stem cells (hiPSCs) offer an infinite source to obtain functional vascular cells in large quantity and comparable quality for TEVG construction. To date, TEVGs (hiPSC-TEVGs) with significant mechanical strength and implantability have been generated using hiPSC-derived seed cells. Despite being in its incipient stage, this emerging field of hiPSC-TEVG research has achieved significant progress and presented promising future potential. Meanwhile, a series of challenges pertaining hiPSC differentiation, vascular tissue engineering technologies and future production and application await to be addressed. Herein, we have composed this review to introduce progress in TEVG generation using hiPSCs, summarize the current major challenges, and encapsulate the future directions of research on hiPSC-based TEVGs. Graphical abstract.
Collapse
Affiliation(s)
- Xiangyu Shi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA
| | - Lile He
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, 06520, New Haven, CT, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine , Yale School of Medicine, 300 George Street, Room 752, New Haven, CT, 06511, USA. .,Yale Stem Cell Center, 06520, New Haven, CT, USA.
| |
Collapse
|
17
|
de Cavanagh EMV, González SA, Inserra F, Forcada P, Castellaro C, Chiabaut-Svane J, Obregón S, Casarini MJ, Kempny P, Kotliar C. Blood pressure control is not enough to normalize endothelial repair by progenitor cells. Am J Physiol Heart Circ Physiol 2020; 319:H744-H752. [PMID: 32795193 DOI: 10.1152/ajpheart.00333.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Patients presenting with classical cardiovascular risk factors within acceptable or average value ranges often develop cardiovascular disease, suggesting that other risk factors need to be considered. Considering that endothelial progenitor cells (EPCs) contribute to endothelial repair, we investigated whether EPCs might be such a factor. We compared the ability of peripheral blood EPCs to attach to extracellular matrix proteins and to grow and function in culture, between controlled hypertensive patients exhibiting a Framingham score (FS) of <10% while showing severe vascular impairment (intima-media thickness/diameter, carotid-femoral pulse wave velocity, brachial artery flow-mediated dilation, carotid and femoral atherosclerotic plaque presence; vulnerable group, N = 30) and those with an FS of ≥10% and scarce vascular changes (protected group, N = 30). When compared with vulnerable patients, protected patients had significantly higher early and late-EPC and early and late-tunneling nanotube (TNT) numbers. Significant negative associations were found between vascular damage severity and early EPC, late-EPC, or late-TNT numbers, whereas EPC or TNT numbers and patient characteristics or cardiovascular risk factors were not associated. Except for protected patients, in all controlled hypertensive patients, early and late-EPC and early and late-TNT counts were significantly lower than those in the normotensive subjects studied (N = 30). We found that the disparity in vascular status between patients presenting with both an FS of ≥10% and scarce vascular changes and those presenting with both an FS of <10% and severe vascular impairment is related to differences in peripheral blood EPC and TNT numbers. These observations support the role of EPCs as contributors to vascular injury repair and suggest that EPC numbers may be a potential cardiovascular risk factor to be included in the FS calculation.NEW & NOTEWORTHY As individuals who present with risk factors within acceptable or average value ranges often develop cardiovascular (CV) disease, it has been suggested that other CV risk factors need to be considered in addition to those that are commonly combined in the Framingham score (FS) to estimate the risk of general CV disease. We investigated whether peripheral endothelial progenitor cells (EPCs) and tunneling nanotubes (TNTs) deserve to be considered. Here we report that EPCs and TNTs are significantly lower in controlled hypertensive patients versus normotensive subjects and that the disparity in vascular status between patients presenting with an FS of ≥10% with scarce vascular changes and those presenting with an FS of <10% with severe vascular impairment is related to differences in EPC and TNT numbers. These data point to EPC and TNT numbers as potential CV risk factors to be included in the FS calculation.
Collapse
Affiliation(s)
| | - Sergio A González
- Cardiometabolic Unit, Cardiology Department, Austral University Hospital, Pilar, Argentina
| | | | | | - Carlos Castellaro
- Cardiometabolic Unit, Cardiology Department, Austral University Hospital, Pilar, Argentina.,Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno," Buenos Aires, Argentina
| | - Jorge Chiabaut-Svane
- Cardiometabolic Unit, Cardiology Department, Austral University Hospital, Pilar, Argentina
| | - Sebastián Obregón
- Arterial Hypertension Center, Department of Cardiology, Austral University Hospital, Buenos Aires, Argentina
| | | | - Pablo Kempny
- Cardiometabolic Unit, Cardiology Department, Austral University Hospital, Pilar, Argentina
| | - Carol Kotliar
- Arterial Hypertension Center, Department of Cardiology, Austral University Hospital, Buenos Aires, Argentina
| |
Collapse
|
18
|
Komici K, Faris P, Negri S, Rosti V, García-Carrasco M, Mendoza-Pinto C, Berra-Romani R, Cervera R, Guerra G, Moccia F. Systemic lupus erythematosus, endothelial progenitor cells and intracellular Ca2+ signaling: A novel approach for an old disease. J Autoimmun 2020; 112:102486. [DOI: 10.1016/j.jaut.2020.102486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 02/07/2023]
|
19
|
Czapla J, Cichoń T, Pilny E, Jarosz-Biej M, Matuszczak S, Drzyzga A, Krakowczyk Ł, Smolarczyk R. Adipose tissue-derived stromal cells stimulated macrophages-endothelial cells interactions promote effective ischemic muscle neovascularization. Eur J Pharmacol 2020; 883:173354. [PMID: 32663541 DOI: 10.1016/j.ejphar.2020.173354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/31/2022]
Abstract
Neovascularization, the process of new blood vessels formation in response to hypoxia induced signals, is an essential step during wound healing or ischemia repair. It follows as a cascade of consecutive events leading to new blood vessels formation and their subsequent remodeling to a mature and functional state, enabling tissue regeneration. Any disruption in consecutive stages of neovascularization can lead to chronic wounds or impairment of tissue repair. In the study we try to explain the biological basis of accelerated blood vessels formation in ischemic tissue after adipose tissue-derived stromal cells (ADSCs) administration. Experiments were performed on mouse models of hindlimb ischemia. We have evaluated the level of immune cells (neutrophils, macrophages) infiltration. The novelty of our work was the assessment of bone marrow-derived stem/progenitor cells (BMDCs) infiltration and their contribution to the neovascularization process in ischemic tissue. We have noticed that ADSCs regulated immune response and affected the kinetics and ratio of macrophages population infiltrating ischemic tissue. Our research revealed that ADSCs promoted changes in the morphology of infiltrating macrophages and their tight association with forming blood vessels. We assume that recruited macrophages may take over the role of pericytes and stabilize the new blood vessel or even differentiate into endothelial cells, which in consequence can accelerate vascular formation upon ADSCs administration. Our findings indicate that administration of ADSCs into ischemic muscle influence spatio-temporal distribution of infiltrating cells (macrophages, neutrophils and BMDCs), which are involved in each step of vascular formation, promoting effective ischemic tissue neovascularization.
Collapse
Affiliation(s)
- Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland.
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Ewelina Pilny
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland; Department of Organic Chemistry, Biochemistry and Biotechnology, Silesian University of Technology, Księdza Marcina Strzody 9 Street, 44-100, Gliwice, Poland
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Alina Drzyzga
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| | - Łukasz Krakowczyk
- Department of Oncologic and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15 Street, 44-101, Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-101, Gliwice, Poland
| |
Collapse
|
20
|
Negri S, Faris P, Rosti V, Antognazza MR, Lodola F, Moccia F. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis. Cells 2020; 9:cells9061341. [PMID: 32471282 PMCID: PMC7349285 DOI: 10.3390/cells9061341] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+ signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+ concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Lodola
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
- Correspondence:
| |
Collapse
|
21
|
Valverde-Villegas JM, Durand M, Bedin AS, Rutagwera D, Kankasa C, Tuaillon E, Nagot N, Vande Perre P, Molès JP. Large Stem/Progenitor-Like Cell Subsets can Also be Identified in the CD45 - and CD45 +/High Populations in Early Human Milk. J Hum Lact 2020; 36:303-309. [PMID: 31815589 DOI: 10.1177/0890334419885315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Stem/progenitor cells have been identified in human milk. However, characterization and percentages of cell subsets in human milk using hematopoietic stem and progenitor cell markers according to the differential expression of CD45, i.e., as CD45dim/+ (mainly hematopoietic stem/progenitor cells) and CD45- (mainly non-hematopoietic stem/progenitor cells), have not been assessed to date. RESEARCH AIM To characterize stem/progenitor-like cell phenotypes in human milk and to report the percentages of these cells at two different lactation stages compared to peripheral blood. METHODS Human milk samples paired with peripheral blood samples (N = 10) were analyzed by flow cytometry using CD45, CD34, CD133, CD38, and lineage-negative markers. The percentage of cell subsets was analyzed in colostrum (Day 3 postpartum) and transitional milk (Day 5/6 postpartum) and compared with the peripheral blood counterpart. RESULTS The percentage of CD45-CD34+ cells was predominant in both colostrum and transitional milk. The percentage of CD45+/highCD133+ cells was high in colostrum while the percentage of CD45-CD133+ cells was high in transitional milk. Furthermore, the median percentages of the CD45-CD34+, CD45-CD133+, and CD45dimCD133+ cell subsets were higher in colostrum than its peripheral blood counterpart (0.11% vs. 0.002%; 0.17% vs. 0.0005%; 0.09% vs. 0.05%, p = .04, respectively); also CD45-CD34-CD133+ and CD45dimCD34-CD133+ cell subsets were higher in colostrum than peripheral blood (1.32% vs. 0.0% and 2.4% vs. 0.06%, p = .04), respectively). CONCLUSION Early human milk is an abundant reservoir of hematopoietic stem/progenitor-like cells in the CD45+/high population and non-hematopoietic stem/progenitor-like cells in the CD45- population.
Collapse
Affiliation(s)
| | - Mélusine Durand
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France
| | - Anne-Sophie Bedin
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France
| | - David Rutagwera
- 119183 Department of Paediatrics and Child Health, University Teaching Hospital, School of Medicine University of Zambia, Lusaka, Zambia
| | - Chipepo Kankasa
- 119183 Department of Paediatrics and Child Health, University Teaching Hospital, School of Medicine University of Zambia, Lusaka, Zambia
| | - Edouard Tuaillon
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France.,27037 CHU Montpellier, Department of Bacteriology-Virology and Department of Medical Information, Montpellier, France
| | - Nicolas Nagot
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France.,27037 CHU Montpellier, Department of Bacteriology-Virology and Department of Medical Information, Montpellier, France
| | - Philippe Vande Perre
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France.,27037 CHU Montpellier, Department of Bacteriology-Virology and Department of Medical Information, Montpellier, France
| | - Jean-Pierre Molès
- 27102 Pathogenesis and Control of Chronic Infections (PCCI), INSERM, University of Montpellier, EFS, Montpellier, France
| |
Collapse
|
22
|
He D, Mao A, Zheng CB, Kan H, Zhang K, Zhang Z, Feng L, Ma X. Aortic heterogeneity across segments and under high fat/salt/glucose conditions at the single-cell level. Natl Sci Rev 2020; 7:881-896. [PMID: 34692110 PMCID: PMC8289085 DOI: 10.1093/nsr/nwaa038] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 12/24/2022] Open
Abstract
The aorta, with ascending, arch, thoracic and abdominal segments, responds to the heartbeat, senses metabolites and distributes blood to all parts of the body. However, the heterogeneity across aortic segments and how metabolic pathologies change it are not known. Here, a total of 216 612 individual cells from the ascending aorta, aortic arch, and thoracic and abdominal segments of mouse aortas under normal conditions or with high blood glucose levels, high dietary salt, or high fat intake were profiled using single-cell RNA sequencing. We generated a compendium of 10 distinct cell types, mainly endothelial (EC), smooth muscle (SMC), stromal and immune cells. The distributions of the different cells and their intercommunication were influenced by the hemodynamic microenvironment across anatomical segments, and the spatial heterogeneity of ECs and SMCs may contribute to differential vascular dilation and constriction that were measured by wire myography. Importantly, the composition of aortic cells, their gene expression profiles and their regulatory intercellular networks broadly changed in response to high fat/salt/glucose conditions. Notably, the abdominal aorta showed the most dramatic changes in cellular composition, particularly involving ECs, fibroblasts and myeloid cells with cardiovascular risk factor-related regulons and gene expression networks. Our study elucidates the nature and range of aortic cell diversity, with implications for the treatment of metabolic pathologies.
Collapse
Affiliation(s)
- Dongxu He
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Aiqin Mao
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Hao Kan
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ka Zhang
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhiming Zhang
- School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Lei Feng
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Ma
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
23
|
Ren R, Guo J, Shi J, Tian Y, Li M, Kang H. PKM2 regulates angiogenesis of VR-EPCs through modulating glycolysis, mitochondrial fission, and fusion. J Cell Physiol 2020; 235:6204-6217. [PMID: 32017072 DOI: 10.1002/jcp.29549] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
Abstract
Vascular resident endothelial progenitor cells (VR-EPCs) have a certain ability to differentiate into endothelial cells (ECs) and participate in the process of angiogenesis. Glycolysis and mitochondrial fission and fusion play a pivotal role in angiogenesis. Pyruvate kinase muscle isoenzyme 2 (PKM2), which mediates energy metabolism and mitochondrial morphology, is regarded as the focus of VR-EPCs angiogenesis in our study. VR-EPCs were isolated from the hearts of 12-weeks-old Sprague-Dawley rats. The role of PKM2 on angiogenesis was evaluated by tube formation assay, wound healing assay, transwell assay, and chick chorioallantoic membrane assay. Western blot analysis, flow cytometry, mitochondrial membrane potential detection, reactive oxygen species (ROS) detection, immunofluorescence staining, and quantitative real-time polymerase chain reaction were used to investigate the potential mechanism of PKM2 for regulating VR-EPCs angiogenesis. We explored the function of PKM2 on the angiogenesis of VR-EPCs. DASA-58 (the activator of PKM2) promoted VR-EPCs proliferation and PKM2 activity, it also could promote angiogenic differentiation. At the same time, DASA-58 significantly enhanced glycolysis, mitochondrial fusion, slightly increased mitochondrial membrane potential, and maintained ROS at a low level. C3k, an inhibitor of PKM2, inhibited PKM2 activity, expression of angiogenesis-related genes and tube formation. Besides, C3k drastically reduced glycolysis and mitochondrial membrane potential while significantly promoting mitochondrial fission and ROS level. Activation of PKM2 could promote VR-EPCs angiogenesis through modulating glycolysis, mitochondrial fission and fusion. By contrast, PKM2 inhibitor has opposite effects.
Collapse
Affiliation(s)
- Ranyue Ren
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiachao Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Shi
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Tian
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengwei Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Kang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Steffen E, Mayer von Wittgenstein WBE, Hennig M, Niepmann ST, Zietzer A, Werner N, Rassaf T, Nickenig G, Wassmann S, Zimmer S, Steinmetz M. Murine sca1/flk1-positive cells are not endothelial progenitor cells, but B2 lymphocytes. Basic Res Cardiol 2020; 115:18. [PMID: 31980946 PMCID: PMC6981106 DOI: 10.1007/s00395-020-0774-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Abstract
Circulating sca1+/flk1+ cells are hypothesized to be endothelial progenitor cells (EPCs) in mice that contribute to atheroprotection by replacing dysfunctional endothelial cells. Decreased numbers of circulating sca1+/flk1+ cells correlate with increased atherosclerotic lesions and impaired reendothelialization upon electric injury of the common carotid artery. However, legitimate doubts remain about the identity of the putative EPCs and their contribution to endothelial restoration. Hence, our study aimed to establish a phenotype for sca1+/flk1+ cells to gain a better understanding of their role in atherosclerotic disease. In wild-type mice, sca1+/flk1+ cells were mobilized into the peripheral circulation by granulocyte-colony stimulating factor (G-CSF) treatment and this movement correlated with improved endothelial regeneration upon carotid artery injury. Multicolor flow cytometry analysis revealed that sca1+/flk1+ cells predominantly co-expressed surface markers of conventional B cells (B2 cells). In RAG2-deficient mice and upon B2 cell depletion, sca1+/flk1+ cells were fully depleted. In the absence of monocytes, sca1+/flk1+ cell levels were unchanged. A PCR array focused on cell surface markers and next-generation sequencing (NGS) of purified sca1+/flk1+ cells confirmed their phenotype to be predominantly that of B cells. Finally, the depletion of B2 cells, including sca1+/flk1+ cells, in G-CSF-treated wild-type mice partly abolished the endothelial regenerating effect of G-CSF, indicating an atheroprotective role for sca1+/flk1+ B2 cells. In summary, we characterized sca1+/flk1+ cells as a subset of predominantly B2 cells, which are apparently involved in endothelial regeneration.
Collapse
Affiliation(s)
- Eva Steffen
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| | | | - Marie Hennig
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sven Thomas Niepmann
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Andreas Zietzer
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Nikos Werner
- Krankenhaus der Barmherzigen Brüder, Innere Medizin III, Trier, Germany
| | - Tienush Rassaf
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Essen, Germany
| | - Georg Nickenig
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sven Wassmann
- Cardiology Pasing, Munich, Germany.,University of the Saarland, Homburg, Saar, Germany
| | - Sebastian Zimmer
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Martin Steinmetz
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Essen, Germany
| |
Collapse
|
25
|
Baruah J, Wary KK. Exosomes in the Regulation of Vascular Endothelial Cell Regeneration. Front Cell Dev Biol 2020; 7:353. [PMID: 31998716 PMCID: PMC6962177 DOI: 10.3389/fcell.2019.00353] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Exosomes have been described as nanoscale membranous extracellular vesicles that emerge from a variety of cells and tissues and are enriched with biologically active genomic and non-genomic biomolecules capable of transducing cell to cell communication. Exosome release, and exosome mediated signaling and cross-talks have been reported in several pathophysiological states. Therefore, exosomes have the potential to become suitable for the diagnosis, prognosis and treatment of specific diseases, including endothelial cell (EC) dysfunction and regeneration. The role of EC-derived exosomes in the mechanisms of cardiovascular tissue regenerative processes represents currently an area of intense research activity. Recent studies have described the potential of exosomes to influence the pathophysiology of immune signaling, tumor metastasis, and angiogenesis. In this review, we briefly discuss progress made in our understanding of the composition and the roles of exosomes in relation to EC regeneration as well as revascularization of ischemic tissues.
Collapse
Affiliation(s)
- Jugajyoti Baruah
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA, United States
| | - Kishore K Wary
- Department of Pharmacology, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Afra S, Matin MM. Potential of mesenchymal stem cells for bioengineered blood vessels in comparison with other eligible cell sources. Cell Tissue Res 2020; 380:1-13. [PMID: 31897835 DOI: 10.1007/s00441-019-03161-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022]
Abstract
Application of stem cells in tissue engineering has proved to be effective in many cases due to great proliferation and differentiation potentials as well as possible paracrine effects of these cells. Human mesenchymal stem cells (MSCs) are recognized as a valuable source for vascular tissue engineering, which requires endothelial and perivascular cells. The goal of this review is to survey the potential of MSCs for engineering functional blood vessels in comparison with other cell types including bone marrow mononuclear cells, endothelial precursor cells, differentiated adult autologous smooth muscle cells, autologous endothelial cells, embryonic stem cells, and induced pluripotent stem cells. In conclusion, MSCs represent a preference in making autologous tissue-engineered vascular grafts (TEVGs) as well as off-the-shelf TEVGs for emergency vascular surgery cases.
Collapse
Affiliation(s)
- Simindokht Afra
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
27
|
Cong X, Zhang SM, Batty L, Luo J. Application of Human Induced Pluripotent Stem Cells in Generating Tissue-Engineered Blood Vessels as Vascular Grafts. Stem Cells Dev 2019; 28:1581-1594. [PMID: 31663439 DOI: 10.1089/scd.2019.0234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In pace with the advancement of tissue engineering during recent decades, tissue-engineered blood vessels (TEBVs) have been generated using primary seed cells, and their impressive success in clinical trials have demonstrated the great potential of these TEBVs as implantable vascular grafts in human regenerative medicine. However, the production, therapeutic efficacy, and readiness in emergencies of current TEBVs could be hindered by the accessibility, expandability, and donor-donor variation of patient-specific primary seed cells. Alternatively, using human induced pluripotent stem cells (hiPSCs) to derive seed vascular cells for vascular tissue engineering could fundamentally address this current dilemma in TEBV production. As an emerging research field with a promising future, the generation of hiPSC-based TEBVs has been reported recently with significant progress. Simultaneously, to further promote hiPSC-based TEBVs into vascular grafts for clinical use, several challenges related to the safety, readiness, and structural integrity of vascular tissue need to be addressed. Herein, this review will focus on the evolution and role of hiPSCs in vascular tissue engineering technology and summarize the current progress, challenges, and future directions of research on hiPSC-based TEBVs.
Collapse
Affiliation(s)
- Xiaoqiang Cong
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cardiology, Bethune First Hospital of Jilin University, ChangChun, China
| | - Shang-Min Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Luke Batty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Yale Stem Cell Center, School of Medicine, Yale University, New Haven, Connecticut
| |
Collapse
|
28
|
Endothelial Ca 2+ Signaling, Angiogenesis and Vasculogenesis: just What It Takes to Make a Blood Vessel. Int J Mol Sci 2019; 20:ijms20163962. [PMID: 31416282 PMCID: PMC6721072 DOI: 10.3390/ijms20163962] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
It has long been known that endothelial Ca2+ signals drive angiogenesis by recruiting multiple Ca2+-sensitive decoders in response to pro-angiogenic cues, such as vascular endothelial growth factor, basic fibroblast growth factor, stromal derived factor-1α and angiopoietins. Recently, it was shown that intracellular Ca2+ signaling also drives vasculogenesis by stimulation proliferation, tube formation and neovessel formation in endothelial progenitor cells. Herein, we survey how growth factors, chemokines and angiogenic modulators use endothelial Ca2+ signaling to regulate angiogenesis and vasculogenesis. The endothelial Ca2+ response to pro-angiogenic cues may adopt different waveforms, ranging from Ca2+ transients or biphasic Ca2+ signals to repetitive Ca2+ oscillations, and is mainly driven by endogenous Ca2+ release through inositol-1,4,5-trisphosphate receptors and by store-operated Ca2+ entry through Orai1 channels. Lysosomal Ca2+ release through nicotinic acid adenine dinucleotide phosphate-gated two-pore channels is, however, emerging as a crucial pro-angiogenic pathway, which sustains intracellular Ca2+ mobilization. Understanding how endothelial Ca2+ signaling regulates angiogenesis and vasculogenesis could shed light on alternative strategies to induce therapeutic angiogenesis or interfere with the aberrant vascularization featuring cancer and intraocular disorders.
Collapse
|
29
|
Somani A, Nair SL, Milbauer LC, Zhu G, Sajja S, Solovey A, Chen Y, Hebbel RP. Blood outgrowth endothelial cells overexpressing eNOS mitigate pulmonary hypertension in rats: a unique carrier cell enabling autologous cell-based gene therapy. Transl Res 2019; 210:1-7. [PMID: 31082372 PMCID: PMC6741773 DOI: 10.1016/j.trsl.2019.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
We have investigated a unique cell type, blood outgrowth endothelial cells (BOEC), as a cell-based gene therapy approach to pulmonary hypertension. BOEC are bona fide endothelial cells, obtained from peripheral blood, that can be expanded to vast numbers, and are amenable to both cryopreservation and genetic modification. We established primary cultures of rat BOEC and genetically altered them to over-express human eNOS plus green fluorescent protein (rBOEC/eNOS) or to express GFP only (rBOEC/GFP). We gave monocrotaline to rats on day 0, and they developed severe pulmonary hypertension. As a Prevention model, we infused saline or rBOEC/GFP or rBOEC/eNOS on day 3, and then examined endpoints on day 24. The rBOEC/eNOS recipients developed elevated NOx (serum and lung) and less severe: elevation of right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and pulmonary arteriolar muscularization and loss of alveolar density. As an Intervention model, we waited until day 21 to give the test infusions, and we examined endpoints on day 35. The rBOEC/eNOS recipients again developed elevated NOx and manifested the same improvements. Indeed, rBOEC/eNOS infusion not only prevented worsening of RVSP but also partially reversed established arteriolar muscularization. These data suggest that BOEC may be useful as a carrier cell for genetic strategies targeting pulmonary hypertension. Their properties render BOEC amenable to preclinical and scale-up studies, available for autologous therapies, and tolerant of modification and storage for potential future use in patients at risk for PAH, eg, as defined by genetics or medical condition.
Collapse
Affiliation(s)
- Arif Somani
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota.
| | - Sethu L Nair
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Liming C Milbauer
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Suchitra Sajja
- Pediatric Critical Care Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Anna Solovey
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Yingjie Chen
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Robert P Hebbel
- Division of Hematology-Oncology-Transplantation, Department of Medicine; and Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
30
|
|
31
|
Brown N, Khan F, Alshaikh B, Berka N, Liacini A, Alawad E, Yusuf K. CD-34 + and VE-cadherin + endothelial progenitor cells in preeclampsia and normotensive pregnancies. Pregnancy Hypertens 2019; 16:42-47. [PMID: 31056159 DOI: 10.1016/j.preghy.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/12/2019] [Accepted: 02/22/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The objective of our study was to determine levels of endothelial progenitor cells (EPCs) in preeclampsia and normotensive pregnant women. STUDY DESIGN Prospective cohort study of women with preeclampsia and normotensive pregnancies. EPCs were estimated by flow cytometry. Multiple linear regression was used to assess the association of EPCs with preeclampsia adjusting for maternal age, body mass index (BMI), gestation and ethnicity. MAIN OUTCOME MEASURE Levels of EPCs in preeclampsia and normotensive pregnancies, with CD-34 and vascular endothelial (VE)-cadherin as markers of EPCs. VE-cadherin is an endothelial cell adhesion molecule used to delineate endothelial lineage of EPCs. RESULTS There were thirty women in the preeclampsia group and thirty-three in the normotensive group. The two groups were similar except for the BMI and blood pressures, which were higher in preeclampsia. On multiple linear regression, EPCs numbers were significantly higher by 29 (95% confidence interval 11.7-46.6, p = 0.001) in preeclampsia compared to the normotensive group. There was significant positive correlation between EPCs and systolic blood pressure in preeclampsia (Spearman correlation coefficient 0.39, p = 0.03). CONCLUSION Although widely used in cardiovascular disease other than preeclampsia, this is the first study using VE-cadherin as a marker of endothelial lineage to define EPCs in preeclampsia. Our results suggest the higher number of EPCs in preeclampsia may be a response of the bone marrow to endothelial injury.
Collapse
Affiliation(s)
- Nicole Brown
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Faisal Khan
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Belal Alshaikh
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Noureddine Berka
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Abdelhamid Liacini
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Essa Alawad
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Kamran Yusuf
- Section of Neonatology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta, Canada.
| |
Collapse
|
32
|
Landers-Ramos RQ, Sapp RM, Shill DD, Hagberg JM, Prior SJ. Exercise and Cardiovascular Progenitor Cells. Compr Physiol 2019; 9:767-797. [PMID: 30892694 DOI: 10.1002/cphy.c180030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autologous stem/progenitor cell-based methods to restore blood flow and function to ischemic tissues are clinically appealing for the substantial proportion of the population with cardiovascular diseases. Early preclinical and case studies established the therapeutic potential of autologous cell therapies for neovascularization in ischemic tissues. However, trials over the past ∼15 years reveal the benefits of such therapies to be much smaller than originally estimated and a definitive clinical benefit is yet to be established. Recently, there has been an emphasis on improving the number and function of cells [herein generally referred to as circulating angiogenic cells (CACs)] used for autologous cell therapies. CACs include of several subsets of circulating cells, including endothelial progenitor cells, with proangiogenic potential that is largely exerted through paracrine functions. As exercise is known to improve CV outcomes such as angiogenesis and endothelial function, much attention is being given to exercise to improve the number and function of CACs. Accordingly, there is a growing body of evidence that acute, short-term, and chronic exercise have beneficial effects on the number and function of different subsets of CACs. In particular, recent studies show that aerobic exercise training can increase the number of CACs in circulation and enhance the function of isolated CACs as assessed in ex vivo assays. This review summarizes the roles of different subsets of CACs and the effects of acute and chronic exercise on CAC number and function, with a focus on the number and paracrine function of circulating CD34+ cells, CD31+ cells, and CD62E+ cells. © 2019 American Physiological Society. Compr Physiol 9:767-797, 2019.
Collapse
Affiliation(s)
- Rian Q Landers-Ramos
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA.,Education and Clinical Center, Baltimore Veterans Affairs Geriatric Research, Baltimore, Maryland, USA.,University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| | - Ryan M Sapp
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - Daniel D Shill
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - James M Hagberg
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA
| | - Steven J Prior
- University of Maryland School of Public Health, Department of Kinesiology, College Park, Maryland, USA.,Education and Clinical Center, Baltimore Veterans Affairs Geriatric Research, Baltimore, Maryland, USA.,University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Diabetic endothelial colony forming cells have the potential for restoration with glycomimetics. Sci Rep 2019; 9:2309. [PMID: 30783159 PMCID: PMC6381138 DOI: 10.1038/s41598-019-38921-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Endothelial colony forming progenitor cell (ECFC) function is compromised in diabetes, leading to poor vascular endothelial repair, which contributes to impaired diabetic foot ulcer healing. We have generated novel glycomimetic drugs with protective effects against endothelial dysfunction. We investigated the effect of glycomimetic C3 on the functional capacity of diabetic ECFCs. ECFCs were isolated from healthy controls and patients with diabetes with neuroischaemic (NI) or neuropathic (NP) foot ulcers. Functionally, diabetic ECFCs demonstrated delayed colony formation (p < 0.02), differential proliferative capacity (p < 0.001) and reduced NO bioavailability (NI ECFCs; p < 0.05). Chemokinetic migration and angiogenesis were also reduced in diabetic ECFCs (p < 0.01 and p < 0.001), and defects in wound closure and tube formation were apparent in NP ECFCs (p < 0.01). Differential patterns in mitochondrial activity were pronounced, with raised activity in NI and depressed activity in NP cells (p < 0.05). The application of glycomimetic improved scratch wound closure in vitro in patient ECFCs (p < 0.01), most significantly in NI cells (p < 0.001), where tube formation (p < 0.05) was also improved. We demonstrate restoration of the deficits in NI cells but not NP cells, using a novel glycomimetic agent, which may be advantageous for therapeutic cell transplantation or as a localised treatment for NI but not NP patients.
Collapse
|
34
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
35
|
Argentati C, Morena F, Bazzucchi M, Armentano I, Emiliani C, Martino S. Adipose Stem Cell Translational Applications: From Bench-to-Bedside. Int J Mol Sci 2018; 19:E3475. [PMID: 30400641 PMCID: PMC6275042 DOI: 10.3390/ijms19113475] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023] Open
Abstract
During the last five years, there has been a significantly increasing interest in adult adipose stem cells (ASCs) as a suitable tool for translational medicine applications. The abundant and renewable source of ASCs and the relatively simple procedure for cell isolation are only some of the reasons for this success. Here, we document the advances in the biology and in the innovative biotechnological applications of ASCs. We discuss how the multipotential property boosts ASCs toward mesenchymal and non-mesenchymal differentiation cell lineages and how their character is maintained even if they are combined with gene delivery systems and/or biomaterials, both in vitro and in vivo.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Ilaria Armentano
- Department of Ecological and Biological Sciences, Tuscia University Largo dell'Università, snc, 01100 Viterbo, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
36
|
Abstract
Vascular, resident stem cells are present in all 3 layers of the vessel wall; they play a role in vascular formation under physiological conditions and in remodeling in pathological situations. Throughout development and adult early life, resident stem cells participate in vessel formation through vasculogenesis and angiogenesis. In adults, the vascular stem cells are mostly quiescent in their niches but can be activated in response to injury and participate in endothelial repair and smooth muscle cell accumulation to form neointima. However, delineation of the characteristics and of the migration and differentiation behaviors of these stem cells is an area of ongoing investigation. A set of genetic mouse models for cell lineage tracing has been developed to specifically address the nature of these cells and both migration and differentiation processes during physiological angiogenesis and in vascular diseases. This review summarizes the current knowledge on resident stem cells, which has become more defined and refined in vascular biology research, thus contributing to the development of new potential therapeutic strategies to promote endothelial regeneration and ameliorate vascular disease development.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| | - Ting Chen
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences (B.Z.)
| | - Qingbo Xu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| |
Collapse
|