1
|
Yang S, Fang Y, Ma Y, Wang F, Wang Y, Jia J, Yang Y, Sun W, Zhou Q, Li Z. Angiogenesis and targeted therapy in the tumour microenvironment: From basic to clinical practice. Clin Transl Med 2025; 15:e70313. [PMID: 40268524 PMCID: PMC12017902 DOI: 10.1002/ctm2.70313] [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: 11/16/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Angiogenesis, as a core marker of cancer survival and growth, is integral to the processes of tumour growth, invasion and metastasis. In recent years, targeted angiogenesis treatment strategies have gradually become an important direction in cancer treatment. Single-cell sequencing technology can provide new insights into targeted angiogenesis by providing a deeper understanding of the heterogeneity of tumour endothelial cells and exploring the interactions between endothelial cells and surrounding cells in the tumour microenvironment. Here, we systematically review the research progress in endothelial cell pathophysiology and its endothelial‒mesenchymal transition and illustrate the heterogeneity of endothelial cells from a single-cell perspective. Finally, we examine the contributions of different cell types within the tumour microenvironment in relation to tumour angiogenesis, as well as the latest progress and strategies in targeted angiogenesis therapy, hoping to provide useful insights into the clinical application of antiangiogenic treatment. Furthermore, a summary of the present progress in the development of potential angiogenesis inhibitors and the ongoing clinical trials for combination therapies is provided. KEY POINTS: Angiogenesis plays a key role in tumour progression, invasion and metastasis, so strategies targeting angiogenesis are gradually becoming an important direction in cancer therapy. Interactions between endothelial cells and stromal cells and immune cells in the tumour microenvironment are significant in angiogenesis. The application of antiangiogenic immunotherapy and nanotechnology in antiangiogenic therapy provides a vital strategy for prolonging the survival of cancer patients.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingshuai Fang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yangcheng Ma
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fuqi Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhang Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiachi Jia
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yabing Yang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Weipeng Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhen Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
2
|
Lutter G, Pommert NS, Zhang X, Seiler J, Saeid Nia M, Meier D, Sellers SL, Gorb SN, Hansen JH, Seoudy H, Müller OJ, Saad M, Haneya A, Frank D, Puehler T, Sathananthan J. Producing and Testing Prototype Tissue-Engineered 3D Tri-Leaflet Valved Stents on Biodegradable Poly-ε-Caprolactone Scaffolds. Int J Mol Sci 2023; 24:17357. [PMID: 38139185 PMCID: PMC10744316 DOI: 10.3390/ijms242417357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Transcatheter pulmonary valve replacement is a minimally-invasive alternative treatment for right ventricular outflow tract dysfunction and has been rapidly evolving over the past years. Heart valve prostheses currently available still have major limitations. Therefore, one of the significant challenges for the future is the roll out of transcatheter tissue engineered pulmonary valve replacement to more patients. In the present study, biodegradable poly-ε-caprolactone (PCL) nanofiber scaffolds in the form of a 3D leaflet matrix were successfully seeded with human endothelial colony-forming cells (ECFCs), human induced pluripotent stem cell-derived MSCs (hMSCs), and porcine MSCs (pMSCs) for three weeks for the generation of 3D tissue-engineered tri-leaflet valved stent grafts. The cell adhesion, proliferation, and distribution of these 3D heart leaflets was analyzed using fluorescence microscopy and scanning electron microscopy (SEM). All cell lineages were able to increase the overgrown leaflet area within the three-week timeframe. While hMSCs showed a consistent growth rate over the course of three weeks, ECFSs showed almost no increase between days 7 and 14 until a growth spurt appeared between days 14 and 21. More than 90% of heart valve leaflets were covered with cells after the full three-week culturing cycle in nearly all leaflet areas, regardless of which cell type was used. This study shows that seeded biodegradable PCL nanofiber scaffolds incorporated in nitinol or biodegradable stents will offer a new therapeutic option in the future.
Collapse
Affiliation(s)
- Georg Lutter
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Nina Sophie Pommert
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Xiling Zhang
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Jette Seiler
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Monireh Saeid Nia
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - David Meier
- Department of Cardiology, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| | - Stephanie L. Sellers
- Centre for Cardiovascular Innovation, St Paul’s and Vancouver General Hospital, Vancouver, BC V6Z 1Y6, Canada; (S.L.S.); (J.S.)
- Cardiovascular Translational Laboratory, Providence Research & Centre for Heart Lung Innovation, Vancouver, BC V6Z 1Y6, Canada
- Centre for Heart Valve Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Stanislav N. Gorb
- Department of Functional Morphology and Biomechanics, Zoological Institute, Christian-Albrecht University of Kiel, 24105 Kiel, Germany
| | - Jan-Hinnerk Hansen
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Hatim Seoudy
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Oliver J. Müller
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Mohammed Saad
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Assad Haneya
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
| | - Derk Frank
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Thomas Puehler
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Janarthanan Sathananthan
- Centre for Cardiovascular Innovation, St Paul’s and Vancouver General Hospital, Vancouver, BC V6Z 1Y6, Canada; (S.L.S.); (J.S.)
- Cardiovascular Translational Laboratory, Providence Research & Centre for Heart Lung Innovation, Vancouver, BC V6Z 1Y6, Canada
- Centre for Heart Valve Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
3
|
Zheng R, Xu T, Wang X, Yang L, Wang J, Huang X. Stem cell therapy in pulmonary hypertension: current practice and future opportunities. Eur Respir Rev 2023; 32:230112. [PMID: 37758272 PMCID: PMC10523152 DOI: 10.1183/16000617.0112-2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/13/2023] [Indexed: 09/30/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease characterised by elevated pulmonary arterial pressure and right-sided heart failure. While conventional drug therapies, including prostacyclin analogues, endothelin receptor antagonists and phosphodiesterase type 5 inhibitors, have been shown to improve the haemodynamic abnormalities of patients with PH, the 5-year mortality rate remains high. Thus, novel therapies are urgently required to prolong the survival of patients with PH. Stem cell therapies, including mesenchymal stem cells, endothelial progenitor cells and induced pluripotent stem cells, have shown therapeutic potential for the treatment of PH and clinical trials on stem cell therapies for PH are ongoing. This review aims to present the latest preclinical achievements of stem cell therapies, focusing on the therapeutic effects of clinical trials and discussing the challenges and future perspectives of large-scale applications.
Collapse
Affiliation(s)
- Ruixuan Zheng
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- These authors contributed equally to this work
| | - Tingting Xu
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- These authors contributed equally to this work
| | - Xinghong Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lehe Yang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Cervantes-Pérez LA, Cervantes-Guevara G, Cervantes-Pérez E, Cervantes-Cardona GA, Nápoles-Echauri A, González-Ojeda A, Fuentes-Orozco C, Cervantes-Pérez G, Reyes-Torres CA, Hernández-Mora FJ, Ron-Magaña AL, Vázquez-Beltrán JC, Hernández-Rivas MI, Ramírez-Ochoa S. Evaluation of the Effects of Atorvastatin and N-Acetyl Cysteine on Platelet Counts in Patients with Primary Immune Thrombocytopenia: An Exploratory Clinical Trial. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1122. [PMID: 37374326 DOI: 10.3390/medicina59061122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Objective: We aimed to evaluate the efficacy of the combination of atorvastatin and N-acetyl cysteine in increasing platelet counts in patients with immune thrombocytopenia who were resistant to steroid therapy or had a relapse after treatment. Material and Methods: The patients included in this study received oral treatment of atorvastatin at a dose of 40 mg daily and N-acetyl cysteine at a dose of 400 mg every 8 h. The desired treatment duration was 12 months, but we included patients who completed at least 1 month of treatment in the analysis. The platelet counts were measured prior to the administration of the study treatment and in the first, third, sixth, and twelfth months of treatment (if available). A p value < 0.05 was considered statistically significant. Results: We included 15 patients who met our inclusion criteria. For the total treatment duration, the global response was 60% (nine patients); eight patients (53.3%) had a complete response and one patient (6.7%) had a partial response. Six patients (40%) were considered as having undergone treatment failure. Of the responder group, five patients maintained a complete response after treatment (55.5%), three patients maintained a partial response (33.3%), and one patient (11.1%) lost their response to the treatment. All of the patients in the responder group had significant increases in their platelet counts after treatment (p < 0.05). Conclusion: This study provides evidence of a possible treatment option for patients with primary immune thrombocytopenia. However, further studies are needed.
Collapse
Affiliation(s)
- Lorena A Cervantes-Pérez
- Department of Hematology, Hospital Civil de Guadalajaras "Fray Antonio Alcalde", Guadalajara 44280,, Mexico
| | - Gabino Cervantes-Guevara
- Department of Welfare and Sustainable Development, Centro Universitario del Norte, Universidad de Guadalajara, Colotlán 46200, Mexico
- Department of Gastroenterology, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Guadalajara 44280, Mexico
| | - Enrique Cervantes-Pérez
- Department of Internal Medicine, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Health Sciences University Center, Universidad de Guadalajara, Guadalajara 44280, Mexico
- Centro Universitario de Tlajomulco, University of Guadalajara, Tlajomulco de Zúñiga 45641, Mexico
| | - Guillermo Alonso Cervantes-Cardona
- Department of Philosophical, Methodological and Instrumental Disciplines, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Adriana Nápoles-Echauri
- Department of Philosophical, Methodological and Instrumental Disciplines, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alejandro González-Ojeda
- Biomedical Research Unit 02, Specialties Hospital of the Western National Medical Center, Mexican Institute of Social Security, Guadalajara 44329, Mexico
| | - Clotilde Fuentes-Orozco
- Biomedical Research Unit 02, Specialties Hospital of the Western National Medical Center, Mexican Institute of Social Security, Guadalajara 44329, Mexico
| | - Gabino Cervantes-Pérez
- Department of Internal Medicine, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Health Sciences University Center, Universidad de Guadalajara, Guadalajara 44280, Mexico
| | - Carlos A Reyes-Torres
- School of Health Sciences, Instituto Tecnológico y de Estudios Superiores de Monterrey, Mexico City 14380, Mexico
| | - Francisco Javier Hernández-Mora
- Human Reproduction, Growth and Child Development Clinic, Health Sciences University Center, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Ana Lucia Ron-Magaña
- Department of Hematology, Hospital Civil de Guadalajaras "Fray Antonio Alcalde", Guadalajara 44280,, Mexico
| | | | - María Isabel Hernández-Rivas
- Departament of Odontology for the Preservation of Health, Health Sciences University Center, Universidad de Guadalajara, Guadalajara 44280, Mexico
| | - Sol Ramírez-Ochoa
- Department of Internal Medicine, Hospital Civil de Guadalajara "Fray Antonio Alcalde", Health Sciences University Center, Universidad de Guadalajara, Guadalajara 44280, Mexico
| |
Collapse
|
5
|
Ramnaraign DJ, Godbout C, Hali K, Hegner C, Bates BD, Desjardins S, Peck J, Schemitsch EH, Nauth A. Endothelial Progenitor Cell Therapy for Fracture Healing: A Dose-Response Study in a Rat Femoral Defect Model. J Tissue Eng Regen Med 2023; 2023:8105599. [PMID: 40226398 PMCID: PMC11918885 DOI: 10.1155/2023/8105599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 04/15/2025]
Abstract
Endothelial progenitor cell (EPC) therapy has been successfully used in orthopaedic preclinical models to heal bone defects. However, no previous studies have investigated the dose-response relationship between EPC therapy and bone healing. This study aimed to assess the effect of different EPC doses on bone healing in a rat model to define an optimal dose. Five-millimeter segmental defects were created in the right femora of Fischer 344 rats, followed by stabilization with a miniplate and screws. Rats were assigned to one of six groups (control, 0.1 M, 0.5 M, 1.0 M, 2.0 M, and 4.0 M; n = 6), receiving 0, 1 × 105, 5 × 105, 1 × 106, 2 × 106, and 4 × 106 EPCs, respectively, delivered into the defect on a gelatin scaffold. Radiographs were taken every two weeks until the animals were euthanized 10 weeks after surgery. The operated femora were then evaluated using micro-computed tomography and biomechanical testing. Overall, the groups that received higher doses of EPCs (0.5 M, 1.0 M, 2.0 M, and 4.0 M) reached better outcomes. At 10 weeks, full radiographic union was observed in 67% of animals in the 0.5 M group, 83% of animals in the 1.0 M group, and 100% of the animals in the 2.0 M and 4.0 M groups, but none in the control and 0.1 M groups. The 2.0 M group also displayed the strongest biomechanical properties, which significantly improved relative to the control and 0.1 M groups. In summary, this study defined a dose-response relationship between EPC therapy and bone healing, with 2 × 106 EPCs being the optimal dose in this model. Our findings emphasize the importance of dosing considerations in the application of cell therapies aimed at tissue regeneration and will help guide future investigations and clinical translation of EPC therapy.
Collapse
Affiliation(s)
- David J. Ramnaraign
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Charles Godbout
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Kalter Hali
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Christian Hegner
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Brent D. Bates
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Sarah Desjardins
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Peck
- Division of Orthopaedic Surgery, Department of Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | | | - Aaron Nauth
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
- Division of Orthopaedic Surgery, Department of Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Regulation of endothelial progenitor cell functions during hyperglycemia: new therapeutic targets in diabetic wound healing. J Mol Med (Berl) 2022; 100:485-498. [PMID: 34997250 DOI: 10.1007/s00109-021-02172-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Diabetes is primarily characterized by hyperglycemia, and its high incidence is often very costly to patients, their families, and national economies. Unsurprisingly, the number and function of endothelial progenitor cells (EPCs) decrease in patients resulting in diabetic wound non-healing. As precursors of endothelial cells (ECs), these cells were discovered in 1997 and found to play an essential role in wound healing. Their function, number, and role in wound healing has been widely investigated. Hitherto, a lot of complex molecular mechanisms have been discovered. In this review, we summarize the mechanisms of how hyperglycemia affects the function and number of EPCs and how the affected cells impact wound healing. We aim to provide a complete summary of the relationship between diabetic hyperglycosemia, EPCs, and wound healing, as well as a better comprehensive platform for subsequent related research.
Collapse
|
7
|
Lutter G, Puehler T, Cyganek L, Seiler J, Rogler A, Herberth T, Knueppel P, Gorb SN, Sathananthan J, Sellers S, Müller OJ, Frank D, Haben I. Biodegradable Poly-ε-Caprolactone Scaffolds with ECFCs and iMSCs for Tissue-Engineered Heart Valves. Int J Mol Sci 2022; 23:527. [PMID: 35008953 PMCID: PMC8745109 DOI: 10.3390/ijms23010527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Clinically used heart valve prostheses, despite their progress, are still associated with limitations. Biodegradable poly-ε-caprolactone (PCL) nanofiber scaffolds, as a matrix, were seeded with human endothelial colony-forming cells (ECFCs) and human induced-pluripotent stem cells-derived MSCs (iMSCs) for the generation of tissue-engineered heart valves. Cell adhesion, proliferation, and distribution, as well as the effects of coating PCL nanofibers, were analyzed by fluorescence microscopy and SEM. Mechanical properties of seeded PCL scaffolds were investigated under uniaxial loading. iPSCs were used to differentiate into iMSCs via mesoderm. The obtained iMSCs exhibited a comparable phenotype and surface marker expression to adult human MSCs and were capable of multilineage differentiation. EFCFs and MSCs showed good adhesion and distribution on PCL fibers, forming a closed cell cover. Coating of the fibers resulted in an increased cell number only at an early time point; from day 7 of colonization, there was no difference between cell numbers on coated and uncoated PCL fibers. The mechanical properties of PCL scaffolds under uniaxial loading were compared with native porcine pulmonary valve leaflets. The Young's modulus and mean elongation at Fmax of unseeded PCL scaffolds were comparable to those of native leaflets (p = ns.). Colonization of PCL scaffolds with human ECFCs or iMSCs did not alter these properties (p = ns.). However, the native heart valves exhibited a maximum tensile stress at a force of 1.2 ± 0.5 N, whereas it was lower in the unseeded PCL scaffolds (0.6 ± 0.0 N, p < 0.05). A closed cell layer on PCL tissues did not change the values of Fmax (ECFCs: 0.6 ± 0.1 N; iMSCs: 0.7 ± 0.1 N). Here, a successful two-phase protocol, based on the timed use of differentiation factors for efficient differentiation of human iPSCs into iMSCs, was developed. Furthermore, we demonstrated the successful colonization of a biodegradable PCL nanofiber matrix with human ECFCs and iMSCs suitable for the generation of tissue-engineered heart valves. A closed cell cover was already evident after 14 days for ECFCs and 21 days for MSCs. The PCL tissue did not show major mechanical differences compared to native heart valves, which was not altered by short-term surface colonization with human cells in the absence of an extracellular matrix.
Collapse
Affiliation(s)
- Georg Lutter
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Thomas Puehler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Jette Seiler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Anita Rogler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Tanja Herberth
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Philipp Knueppel
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Stanislav N. Gorb
- Department of Functional Morphology and Biomechanics, Zoological Institute, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany;
| | - Janarthanan Sathananthan
- Department of Centre for Heart Valve Innovation, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 174, Canada; (J.S.); (S.S.)
| | - Stephanie Sellers
- Department of Centre for Heart Valve Innovation, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 174, Canada; (J.S.); (S.S.)
| | - Oliver J. Müller
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Irma Haben
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| |
Collapse
|
8
|
Chemically Defined Xeno- and Serum-Free Cell Culture Medium to Grow Human Adipose Stem Cells. Cells 2021; 10:cells10020466. [PMID: 33671568 PMCID: PMC7926673 DOI: 10.3390/cells10020466] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is an abundant source of stem cells. However, liposuction cannot yield cell quantities sufficient for direct applications in regenerative medicine. Therefore, the development of GMP-compliant ex vivo expansion protocols is required to ensure the production of a "cell drug" that is safe, reproducible, and cost-effective. Thus, we developed our own basal defined xeno- and serum-free cell culture medium (UrSuppe), specifically formulated to grow human adipose stem cells (hASCs). With this medium, we can directly culture the stromal vascular fraction (SVF) cells in defined cell culture conditions to obtain hASCs. Cells proliferate while remaining undifferentiated, as shown by Flow Cytometry (FACS), Quantitative Reverse Transcription PCR (RT-qPCR) assays, and their secretion products. Using the UrSuppe cell culture medium, maximum cell densities between 0.51 and 0.80 × 105 cells/cm2 (=2.55-4.00 × 105 cells/mL) were obtained. As the expansion of hASCs represents only the first step in a cell therapeutic protocol or further basic research studies, we formulated two chemically defined media to differentiate the expanded hASCs in white or beige/brown adipocytes. These new media could help translate research projects into the clinical application of hASCs and study ex vivo the biology in healthy and dysfunctional states of adipocytes and their precursors. Following the cell culture system developers' practice and obvious reasons related to the formulas' patentability, the defined media's composition will not be disclosed in this study.
Collapse
|
9
|
Pu X, Du L, Hu Y, Fan Y, Xu Q. Stem/Progenitor Cells and Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2020; 41:167-178. [PMID: 33028095 DOI: 10.1161/atvbaha.120.315052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by endothelial dysfunction and vascular remodeling. Despite significant advancement in our understanding of the pathogenesis of PAH in recent years, treatment options for PAH are limited and their prognosis remains poor. PAH is now seen as a severe pulmonary arterial vasculopathy with structural changes driven by excessive vascular proliferation and inflammation. Perturbations of a number of cellular and molecular mechanisms have been described, including pathways involving growth factors, cytokines, metabolic signaling, elastases, and proteases, underscoring the complexity of the disease pathogenesis. Interestingly, emerging evidence suggests that stem/progenitor cells may have an impact on disease development and therapy. In preclinical studies, stem/progenitor cells displayed an ability to promote endothelial repair of dysfunctional arteries and induce neovascularization. The stem cell-based therapy for PAH are now under active investigation. This review article will briefly summarize the updates in the research field, with a special focus on the contribution of stem/progenitor cells to lesion formation via influencing vascular cell functions and highlight the potential clinical application of stem/progenitor cell therapy to PAH.
Collapse
Affiliation(s)
- Xiangyuan Pu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (X.P., L.D., Y.H., Q.X.)
| | - Luping Du
- Department of Cardiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (X.P., L.D., Y.H., Q.X.)
| | - Yanhua Hu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (X.P., L.D., Y.H., Q.X.)
| | - Ye Fan
- Department of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.F.)
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (X.P., L.D., Y.H., Q.X.)
| |
Collapse
|
10
|
Evidence of Accumulated Endothelial Progenitor Cells in the Lungs of Rats with Pulmonary Arterial Hypertension by 89Zr-oxine PET Imaging. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:1108-1117. [PMID: 32490032 PMCID: PMC7256434 DOI: 10.1016/j.omtm.2020.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/27/2020] [Indexed: 12/21/2022]
Abstract
Endothelial progenitor cells (EPCs) play a major role in regulating pulmonary vascular remodeling during pulmonary arterial hypertension (PAH) development. Several preclinical and clinical trials of EPCs transplantation have been performed for the treatment of PAH. However, there is no reliable method to monitor real-time cell trafficking and quantify transplanted EPCs. Here in this paper we isolated EPCs from human peripheral blood, identified their functional integrity, and efficiently labeled the EPCs with 89Zr-oxine and DiO. Labeled EPCs were injected into the tail vein of normal and PAH rats to be tracked in vivo. From the microPET/CT images, we found EPCs were distributed primarily in the lung at 1 h and then migrated to the liver and spleen. We could observe the 3,3′ dioctadecyloxacarbocyanine perchlorate (DiO)-labeled EPCs binding in the pulmonary vasculature by CellVizio confocal. The result of quantitative analysis revealed significantly higher accumulation of EPCs in the lungs of PAH rats than in those of healthy rats. The distribution and higher accumulation of EPCs in the lungs of PAH rats could help to evaluate the safety and provide evidence of effectiveness of EPC therapy.
Collapse
|
11
|
Hao D, Fan Y, Xiao W, Liu R, Pivetti C, Walimbe T, Guo F, Zhang X, Farmer DL, Wang F, Panitch A, Lam KS, Wang A. Rapid endothelialization of small diameter vascular grafts by a bioactive integrin-binding ligand specifically targeting endothelial progenitor cells and endothelial cells. Acta Biomater 2020; 108:178-193. [PMID: 32151698 PMCID: PMC8012081 DOI: 10.1016/j.actbio.2020.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/31/2022]
Abstract
Establishing and maintaining a healthy endothelium on vascular and intravascular devices is crucial for the prevention of thrombosis and stenosis. Generating a biofunctional surface on vascular devices to recruit endothelial progenitor cells (EPCs) and endothelial cells (ECs) has proven efficient in promoting in situ endothelialization. However, molecules conventionally used for EPC/EC capturing generally lack structural stability, capturing specificity, and biological functionalities, which have limited their applications. Discovery of effective, specific, and structurally stable EPC/EC capturing ligands is desperately needed. Using the high-throughput One-Bead One-Compound combinatorial library screening technology, we recently identified a disulfide cyclic octa-peptide LXW7 (cGRGDdvc), which possesses strong binding affinity and functionality to EPCs/ECs, weak binding to platelets, and no binding to inflammatory cells. Because LXW7 is cyclic and 4 out of the 8 amino acids are unnatural D-amino acids, LXW7 is highly proteolytically stable. In this study, we applied LXW7 to modify small diameter vascular grafts using a Click chemistry approach. In vitro studies demonstrated that LXW7-modified grafts significantly improved EPC attachment, proliferation and endothelial differentiation and suppressed platelet attachment. In a rat carotid artery bypass model, LXW7 modification of the small diameter vascular grafts significantly promoted EPC/EC recruitment and rapidly achieved endothelialization. At 6 weeks after implantation, LXW7-modified grafts retained a high patency of 83%, while the untreated grafts had a low patency of 17%. Our results demonstrate that LXW7 is a potent EPC/EC capturing and platelet suppressing ligand and LXW7-modified vascular grafts rapidly generate a healthy and stable endothelial interface between the graft surface and the circulation to reduce thrombosis and improve patency. STATEMENT OF SIGNIFICANCE: In this study, One-Bead One-Compound (OBOC) technology has been applied for the first time in discovering bioactive ligands for tissue regeneration applications. Current molecules used to modify artificial vascular grafts generally lack EPC/EC capturing specificity, biological functionalities and structural stability. Using OBOC technology, we identified LXW7, a constitutionally stable disulfide cyclic octa-peptide with strong binding affinity and biological functionality to EPCs/ECs, very weak binding to platelets and no binding to inflammatory cells. These characteristics are crucial for promoting rapid endothelialization to prevent thrombosis and improve patency of vascular grafts. LXW7 coating technology could be applied to a wide range of vascular and intravascular devices, including grafts, stents, cardiac valves, and catheters, where a "living" endothelium and healthy blood interface are needed.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Yahan Fan
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; Department of Blood Transfusion, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, United States
| | - Christopher Pivetti
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Tanaya Walimbe
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States
| | - Fuzheng Guo
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Xinke Zhang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; School of Pharmaceutical Science, Shandong University, Jinan, Shandong 250012, China
| | - Diana L Farmer
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
| | - Fengshan Wang
- School of Pharmaceutical Science, Shandong University, Jinan, Shandong 250012, China
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, United States; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States; Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, United States.
| |
Collapse
|
12
|
Lopes-Coelho F, Silva F, Gouveia-Fernandes S, Martins C, Lopes N, Domingues G, Brito C, Almeida AM, Pereira SA, Serpa J. Monocytes as Endothelial Progenitor Cells (EPCs), Another Brick in the Wall to Disentangle Tumor Angiogenesis. Cells 2020; 9:cells9010107. [PMID: 31906296 PMCID: PMC7016533 DOI: 10.3390/cells9010107] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bone marrow contains endothelial progenitor cells (EPCs) that, upon pro-angiogenic stimuli, migrate and differentiate into endothelial cells (ECs) and contribute to re-endothelialization and neo-vascularization. There are currently no reliable markers to characterize EPCs, leading to their inaccurate identification. In the past, we showed that, in a panel of tumors, some cells on the vessel wall co-expressed CD14 (monocytic marker) and CD31 (EC marker), indicating a putative differentiation route of monocytes into ECs. Herein, we disclosed monocytes as potential EPCs, using in vitro and in vivo models, and also addressed the cancer context. Monocytes acquired the capacity to express ECs markers and were able to be incorporated into blood vessels, contributing to cancer progression, by being incorporated in tumor neo-vasculature. Reactive oxygen species (ROS) push monocytes to EC differentiation, and this phenotype is reverted by cysteine (a scavenger and precursor of glutathione), which indicates that angiogenesis is controlled by the interplay between the oxidative stress and the scavenging capacity of the tumor microenvironment.
Collapse
Affiliation(s)
- Filipa Lopes-Coelho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Fernanda Silva
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Sofia Gouveia-Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Carmo Martins
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Nuno Lopes
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
| | - Germana Domingues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica, Avenida da República, Estação Agronómica, 2780-157 Oeiras, Portugal; (N.L.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António M Almeida
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Hospital da Luz, Av. Lusíada 100, 1500-650 Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (F.L.-C.); (F.S.); (S.G.-F.); (G.D.); (S.A.P.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto 1099-023 Lisboa, Portugal; (C.M.); (A.M.A.)
- Correspondence: ; Tel.: +350-217-229-800; Fax: +351-217-248-756
| |
Collapse
|
13
|
Kaushik K, Das A. Endothelial progenitor cell therapy for chronic wound tissue regeneration. Cytotherapy 2019; 21:1137-1150. [PMID: 31668487 DOI: 10.1016/j.jcyt.2019.09.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
Despite advancements in wound care, healing of chronic diabetic wounds remains a great challenge for the clinical fraternity because of the intricacies of the healing process. Due to the limitations of existing treatment strategies for chronic wounds, stem/progenitor cell transplantation therapies have been explored as an alternative for tissue regeneration at the wound site. The non-healing phenotype of chronic wounds is directly associated with lack of vascularization. Therefore, endothelial progenitor cell (EPC) transplantation is proving to be a promising approach for the treatment of hypo-vascular chronic wounds. With the existing knowledge in EPC biology, significant efforts have been made to enrich EPCs at the chronic wound site, generating EPCs from somatic cells, induced pluripotent stem cells (iPSCs) using transcription factors, or from adult stem cells using chemicals/drugs for use in transplantation, as well as modulating the endogenous dysfunctional/compromised EPCs under diabetic conditions. This review mainly focuses on the pre-clinical and clinical approaches undertaken to date with EPC-based translational therapy for chronic diabetic as well as non-diabetic wounds to evaluate their vascularity-mediated regeneration potential.
Collapse
Affiliation(s)
- Komal Kaushik
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India.
| |
Collapse
|
14
|
Smith RJ, Yi T, Nasiri B, Breuer CK, Andreadis ST. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response. FASEB J 2019; 33:5089-5100. [PMID: 30629890 DOI: 10.1096/fj.201801856r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recently, our group demonstrated that immobilized VEGF can capture flowing endothelial cells (ECs) from the blood in vitro and promote endothelialization and patency of acellular tissue-engineered vessels (A-TEVs) into the arterial system of an ovine animal model. Here, we demonstrate implantability of submillimeter diameter heparin and VEGF-decorated A-TEVs in a mouse model and discuss the cellular and immunologic response. At 1 mo postimplantation, the graft lumen was fully endothelialized, as shown by expression of EC markers such as CD144, eNOS, CD31, and VEGFR2. Interestingly, the same cells coexpressed leukocyte/macrophage (Mϕ) markers CD14, CD16, VEGFR1, CD38, and EGR2. Notably, there was a stark difference in the cellular makeup between grafts containing VEGF and those containing heparin alone. In VEGF-containing grafts, infiltrating monocytes (MCs) converted into anti-inflammatory M2-Mϕs, and the grafts developed well-demarcated luminal and medial layers resembling those of native arteries. In contrast, in grafts containing only heparin, MCs converted primarily into M1-Mϕs, and the endothelial and smooth muscle layers were not well defined. Our results indicate that VEGF may play an important role in regulating A-TEV patency and regeneration, possibly by regulating the inflammatory response to the implants.-Smith, R. J., Jr., Yi, T., Nasiri, B., Breuer, C. K., Andreadis, S. T. Implantation of VEGF-functionalized cell-free vascular grafts: regenerative and immunological response.
Collapse
Affiliation(s)
- Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, New York, USA
| | - Tai Yi
- Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Bita Nasiri
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, USA; and
| | | | - Stelios T Andreadis
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Amherst, New York, USA.,Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, New York, USA; and.,Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, State University of New York, Amherst, New York, USA
| |
Collapse
|
15
|
Tite T, Popa AC, Balescu LM, Bogdan IM, Pasuk I, Ferreira JMF, Stan GE. Cationic Substitutions in Hydroxyapatite: Current Status of the Derived Biofunctional Effects and Their In Vitro Interrogation Methods. MATERIALS (BASEL, SWITZERLAND) 2018; 11:E2081. [PMID: 30355975 PMCID: PMC6266948 DOI: 10.3390/ma11112081] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/13/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
High-performance bioceramics are required for preventing failure and prolonging the life-time of bone grafting scaffolds and osseous implants. The proper identification and development of materials with extended functionalities addressing socio-economic needs and health problems constitute important and critical steps at the heart of clinical research. Recent findings in the realm of ion-substituted hydroxyapatite (HA) could pave the road towards significant developments in biomedicine, with an emphasis on a new generation of orthopaedic and dentistry applications, since such bioceramics are able to mimic the structural, compositional and mechanical properties of the bone mineral phase. In fact, the fascinating ability of the HA crystalline lattice to allow for the substitution of calcium ions with a plethora of cationic species has been widely explored in the recent period, with consequent modifications of its physical and chemical features, as well as its functional mechanical and in vitro and in vivo biological performance. A comprehensive inventory of the progresses achieved so far is both opportune and of paramount importance, in order to not only gather and summarize information, but to also allow fellow researchers to compare with ease and filter the best solutions for the cation substitution of HA-based materials and enable the development of multi-functional biomedical designs. The review surveys preparation and synthesis methods, pinpoints all the explored cation dopants, and discloses the full application range of substituted HA. Special attention is dedicated to the antimicrobial efficiency spectrum and cytotoxic trade-off concentration values for various cell lines, highlighting new prophylactic routes for the prevention of implant failure. Importantly, the current in vitro biological tests (widely employed to unveil the biological performance of HA-based materials), and their ability to mimic the in vivo biological interactions, are also critically assessed. Future perspectives are discussed, and a series of recommendations are underlined.
Collapse
Affiliation(s)
- Teddy Tite
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| | - Adrian-Claudiu Popa
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
- Army Centre for Medical Research, RO-010195 Bucharest, Romania.
| | | | | | - Iuliana Pasuk
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| | - José M F Ferreira
- Department of Materials and Ceramics Engineering, CICECO, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - George E Stan
- National Institute of Materials Physics, RO-077125 Magurele, Romania.
| |
Collapse
|
16
|
Abstract
Cardiovascular disease is cited as the underlying cause of death in one out of every three deaths within the United States; this burden on the health care system percolates down to affect patients on an individual level. In part, the problem arises from the low regenerative capacity of cardiovascular system cells, for example, cardiac myocytes, and from oxidative stressors to the human body. Endothelial progenitor cells (EPCs) are a type of stem cell, and various clinical conditions including hypertension and renal failure underlie their dysfunction. EPCs are classified as either early or late outgrowth endothelial progenitor cells depending on the time they appear in circulation and at the site of injury after an inciting event. Their function is paracrine through the release of cytokines, growth factors and chemokines such as interleukin-6 and vascular endothelial growth factor, and they are involved in transdifferentiation into vascular smooth muscle cells and potentially cardiac myocytes. They are beneficial to the modification of cardiovascular cell apoptosis, fibrosis, and contractility. In times of stress, the normal function of endothelial progenitor cells is altered; this creates a maladaptive cycle where stress and failed coping mechanisms enhance each other toward the culmination of cardiovascular disease. The development of the cardiovascular system follows gastrulation in the embryonic period, and the cells that form the system are derived from the mesoderm; being mesoderm, the vascular cells exhibit heterogeneity in their origin and function. The need to understand the molecular and cellular regulatory pathways during development can amalgamate efforts of endothelial cell and cardiovascular system pathophysiology for the advancement of patient cardiovascular reserve and function.
Collapse
|
17
|
Isolation and characterization of endothelial colony-forming cells from mononuclear cells of rat bone marrow. Exp Cell Res 2018; 370:116-126. [PMID: 29908162 DOI: 10.1016/j.yexcr.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
Transplantation of bone marrow-derived endothelial progenitor cells (BM-EPCs) has been used as a therapeutic strategy for vascular repair. However, it remains controversial whether BM-EPCs exhibit clonal endothelial colony-forming cell (ECFC) capacity, a characteristic of true EPCs. The aim of this study was to isolate and explore the cellular properties of BM-ECFCs. We isolated BM-ECFCs from rat bone marrow with high purity via an optimized method. This approach involved the removal of selective colonies based on the conventional differential adhesive culture method used to isolate ECFCs from peripheral and umbilical cord blood. Our results indicate that primary colony BM-ECFCs display a panel of surface antigen markers consistent with endothelial cells. These BM-ECFCs coexpress CD34, CD133, and VEGFR2 at high levels, and these levels decrease with passaging. These cells have high potential for proliferation, migration, and formation of capillary-like structures on Matrigel, and these abilities are retained during ex vivo expansion. Furthermore, BM-ECFCs cultured with 10% or 20% fetal bovine serum demonstrated two different patterns of spontaneous capillary-like structure formation. These results provide a foundation for isolation of ECFCs from human bone marrow for autologous cell transplantation and tissue engineering applications in the future.
Collapse
|
18
|
Huertas A, Guignabert C, Barberà JA, Bärtsch P, Bhattacharya J, Bhattacharya S, Bonsignore MR, Dewachter L, Dinh-Xuan AT, Dorfmüller P, Gladwin MT, Humbert M, Kotsimbos T, Vassilakopoulos T, Sanchez O, Savale L, Testa U, Wilkins MR. Pulmonary vascular endothelium: the orchestra conductor in respiratory diseases. Eur Respir J 2018; 51:13993003.00745-2017. [DOI: 10.1183/13993003.00745-2017] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 02/03/2018] [Indexed: 12/15/2022]
Abstract
The European Respiratory Society (ERS) Research Seminar entitled “Pulmonary vascular endothelium: orchestra conductor in respiratory diseases - highlights from basic research to therapy” brought together international experts in dysfunctional pulmonary endothelium, from basic science to translational medicine, to discuss several important aspects in acute and chronic lung diseases. This review will briefly sum up the different topics of discussion from this meeting which was held in Paris, France on October 27–28, 2016. It is important to consider that this paper does not address all aspects of endothelial dysfunction but focuses on specific themes such as: 1) the complex role of the pulmonary endothelium in orchestrating the host response in both health and disease (acute lung injury, chronic obstructive pulmonary disease, high-altitude pulmonary oedema and pulmonary hypertension); and 2) the potential value of dysfunctional pulmonary endothelium as a target for innovative therapies.
Collapse
|
19
|
Evaluation of ex vivo produced endothelial progenitor cells for autologous transplantation in primates. Stem Cell Res Ther 2018; 9:14. [PMID: 29357928 PMCID: PMC5778763 DOI: 10.1186/s13287-018-0769-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 02/05/2023] Open
Abstract
Background Autologous transplantation of endothelial progenitor cells (EPCs) is a promising therapeutic approach in the treatment of various vascular diseases. We previously reported a two-step culture system for scalable generation of human EPCs derived from cord blood CD34+ cells ex vivo. Here, we now apply this culture system to expand and differentiate human and nonhuman primate EPCs from mobilized peripheral blood (PB) CD34+ cells for the therapeutic potential of autologous transplantation. Methods The human and nonhuman primate EPCs from mobilized PB CD34+ cells were cultured according to our previously reported system. The generated adherent cells were then characterized by the morphology, surface markers, nitric oxide (NO)/endothelial NO synthase (eNOS) levels and Dil-acetylated low-density lipoprotein (Dil-Ac-LDL) uptake/fluorescein isothiocyanate (FITC)-lectin binding actives. Furthermore, the efficacy and safety studies were performed by autologous transplantation via hepatic portal vein injection in a nonhuman primate model with acute liver sinusoidal endothelial cell injury. Results The mobilized PB CD34+ cells from both human and nonhuman primate were efficiently expanded and differentiated. Over 2 × 108 adherent cells were generated from 20 mL mobilized primate PB (1.51 × 106 ± 3.39 × 105 CD34+ cells) by 36-day culture and more than 80% of the produced cells were identified as EPCs/endothelial cells (ECs). In the autologous transplant model, the injected EPC/ECs from nonhuman primate PB were scattered in the intercellular spaces of hepatocytes at the hepatic tissues 14 days post-transplantation, indicating successful migration and reconstitution in the liver structure as the functional EPCs/ECs. Conclusions We successfully applied our previous two-step culture system for the generation of primate EPCs from mobilized PB CD34+ cells, evaluated the phenotypes ex vivo, and transplanted autologous EPCs/ECs in a nonhuman primate model. Our study indicates that it may be possible for these ex-vivo high-efficient expanded EPCs to be used in clinical cell therapy.
Collapse
|
20
|
Simara P, Tesarova L, Rehakova D, Farkas S, Salingova B, Kutalkova K, Vavreckova E, Matula P, Matula P, Veverkova L, Koutna I. Reprogramming of Adult Peripheral Blood Cells into Human Induced Pluripotent Stem Cells as a Safe and Accessible Source of Endothelial Cells. Stem Cells Dev 2017; 27:10-22. [PMID: 29117787 PMCID: PMC5756468 DOI: 10.1089/scd.2017.0132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
New approaches in regenerative medicine and vasculogenesis have generated a demand for sufficient numbers of human endothelial cells (ECs). ECs and their progenitors reside on the interior surface of blood and lymphatic vessels or circulate in peripheral blood; however, their numbers are limited, and they are difficult to expand after isolation. Recent advances in human induced pluripotent stem cell (hiPSC) research have opened possible avenues to generate unlimited numbers of ECs from easily accessible cell sources, such as the peripheral blood. In this study, we reprogrammed peripheral blood mononuclear cells, human umbilical vein endothelial cells (HUVECs), and human saphenous vein endothelial cells (HSVECs) into hiPSCs and differentiated them into ECs. The phenotype profiles, functionality, and genome stability of all hiPSC-derived ECs were assessed and compared with HUVECs and HSVECs. hiPSC-derived ECs resembled their natural EC counterparts, as shown by the expression of the endothelial surface markers CD31 and CD144 and the results of the functional analysis. Higher expression of endothelial progenitor markers CD34 and kinase insert domain receptor (KDR) was measured in hiPSC-derived ECs. An analysis of phosphorylated histone H2AX (γH2AX) foci revealed that an increased number of DNA double-strand breaks upon reprogramming into pluripotent cells. However, differentiation into ECs restored a normal number of γH2AX foci. Our hiPSCs retained a normal karyotype, with the exception of the HSVEC-derived hiPSC line, which displayed mosaicism due to a gain of chromosome 1. Peripheral blood from adult donors is a suitable source for the unlimited production of patient-specific ECs through the hiPSC interstage. hiPSC-derived ECs are fully functional and comparable to natural ECs. The protocol is eligible for clinical applications in regenerative medicine, if the genomic stability of the pluripotent cell stage is closely monitored.
Collapse
Affiliation(s)
- Pavel Simara
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Lenka Tesarova
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Daniela Rehakova
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Simon Farkas
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Barbara Salingova
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Katerina Kutalkova
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Eva Vavreckova
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Pavel Matula
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Petr Matula
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
| | - Lenka Veverkova
- I. Surgery Department, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Irena Koutna
- Centre for Biomedical Image Analysis, Faculty of Informatics, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
21
|
Rizo-Roca D, Ríos-Kristjánsson JG, Núñez-Espinosa C, Santos-Alves E, Gonçalves IO, Magalhães J, Ascensão A, Pagès T, Viscor G, Torrella JR. Intermittent hypobaric hypoxia combined with aerobic exercise improves muscle morphofunctional recovery after eccentric exercise to exhaustion in trained rats. J Appl Physiol (1985) 2016; 122:580-592. [PMID: 27765844 DOI: 10.1152/japplphysiol.00501.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/27/2016] [Accepted: 10/13/2016] [Indexed: 01/07/2023] Open
Abstract
Unaccustomed eccentric exercise leads to muscle morphological and functional alterations, including microvasculature damage, the repair of which is modulated by hypoxia. We present the effects of intermittent hypobaric hypoxia and exercise on recovery from eccentric exercise-induced muscle damage (EEIMD). Soleus muscles from trained rats were excised before (CTRL) and 1, 3, 7, and 14 days after a double session of EEIMD protocol. A recovery treatment consisting of one of the following protocols was applied 1 day after the EEIMD: passive normobaric recovery (PNR), a 4-h daily exposure to passive hypobaric hypoxia at 4,000 m (PHR), or hypobaric hypoxia exposure followed by aerobic exercise (AHR). EEIMD produced an increase in the percentage of abnormal fibers compared with CTRL, and it affected the microvasculature by decreasing capillary density (CD, capillaries per mm2) and the capillary-to-fiber ratio (CF). After 14 days, AHR exhibited CD and CF values similar to those of CTRL animals (789 and 3.30 vs. 746 and 3.06) and significantly higher than PNR (575 and 2.62) and PHR (630 and 2.92). Furthermore, VEGF expression showed a significant 43% increase in AHR when compared with PNR. Moreover, after 14 days, the muscle fibers in AHR had a more oxidative phenotype than the other groups, with significantly smaller cross-sectional areas (AHR, 3,745; PNR, 4,502; and PHR, 4,790 µm2), higher citrate synthase activity (AHR, 14.8; PNR, 13.1; and PHR, 12 µmol·min-1·mg-1) and a significant 27% increment in PGC-1α levels compared with PNR. Our data show that hypoxia combined with exercise attenuates or reverses the morphofunctional alterations induced by EEIMD.NEW & NOTEWORTHY Our study provides new insights into the use of intermittent hypobaric hypoxia combined with exercise as a strategy to recover muscle damage induced by eccentric exercise. We analyzed the effects of hypobaric exposure combined with aerobic exercise on histopathological features of muscle damage, fiber morphofunctionality, capillarization, angiogenesis, and the oxidative capacity of damaged soleus muscle. Most of these parameters were improved after a 2-wk protocol of intermittent hypobaric hypoxia combined with aerobic exercise.
Collapse
Affiliation(s)
- D Rizo-Roca
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| | - J G Ríos-Kristjánsson
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| | - C Núñez-Espinosa
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| | - E Santos-Alves
- Research Center in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - I O Gonçalves
- Research Center in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - J Magalhães
- Research Center in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - A Ascensão
- Research Center in Physical Activity, Health and Leisure, Faculty of Sport, University of Porto, Porto, Portugal
| | - T Pagès
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| | - G Viscor
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| | - J R Torrella
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; and
| |
Collapse
|
22
|
Montali M, Barachini S, Panvini FM, Carnicelli V, Fulceri F, Petrini I, Pacini S. Growth Factor Content in Human Sera Affects the Isolation of Mesangiogenic Progenitor Cells (MPCs) from Human Bone Marrow. Front Cell Dev Biol 2016; 4:114. [PMID: 27800477 PMCID: PMC5065953 DOI: 10.3389/fcell.2016.00114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 09/29/2016] [Indexed: 12/31/2022] Open
Abstract
Mesangiogenic Progenitor Cells (MPCs) are human bone marrow-derived multipotent cells, isolated in vitro under selective culture conditions and shown to retain both mesengenic and angiogenic potential. MPCs also co-isolated with multipotent stromal cells (MSCs) when bone marrow primary cultures were set up for clinical applications, using human serum (HS) in place of fetal bovine serum (FBS). MPC culture purity (over 95%) is strictly dependent on HS supplementation with significant batch-to-batch variability. In the present paper we screened different sources of commercially available pooled human AB type serum (PhABS) for their ability to promote MPC production under selective culture conditions. As the majority of "contaminating" cells in MPC cultures were represented by MSC-like cells, we hypothesized a role by differentiating agents present in the sera. Therefore, we tested a number of growth factors (hGF) and found that higher concentrations of FGF-2, EGF, PDGF-AB, and VEGF-A as well as lower concentration of IGF-1 give sub-optimal MPC recovery. Gene expression analysis of hGF receptors was also carried out both in MSCs and MPCs, suggesting that FGF-2, EGF, and PDGF-AB could act promoting MSC proliferation, while VEGF-A contribute to MSC-like cell contamination, triggering MPC differentiation. Here we demonstrated that managing hGF contents, together with applying specific receptors inhibitors (Erlotinib-HCl and Nintedanib), could significantly mitigate the batch-to-batch variability related to serum supplementation. These data represent a fundamental milestone in view of manufacturing MPC-based medicinal products.
Collapse
Affiliation(s)
- Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa Pisa, Italy
| | - Francesca M Panvini
- Department of Clinical and Experimental Medicine, University of Pisa Pisa, Italy
| | - Vittoria Carnicelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa Pisa, Italy
| | - Franca Fulceri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa Pisa, Italy
| | - Iacopo Petrini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa Pisa, Italy
| | - Simone Pacini
- Department of Clinical and Experimental Medicine, University of Pisa Pisa, Italy
| |
Collapse
|
23
|
Castelli G, Parolini I, Cerio AM, D'Angiò A, Pasquini L, Carollo M, Sargiacomo M, Testa U, Pelosi E. Conditioned medium from human umbilical vein endothelial cells markedly improves the proliferation and differentiation of circulating endothelial progenitors. Blood Cells Mol Dis 2016; 61:58-65. [PMID: 27667168 DOI: 10.1016/j.bcmd.2016.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 01/01/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) have been suggested as a precious source for generating functionally competent endothelial cells (ECs), candidate for various clinical applications. However, the paucity of these progenitor cells and the technical difficulties for their in vitro growth represent a main limitation to their use. In the present study we hypothesized that the paracrine effects of human umbilical vein endothelial cells (HUVECs) may improve endothelial cell generation from cord blood (CB) EPCs. In line with this hypothesis we showed that HUVEC conditioned medium (CM) or co-culture with HUVECs markedly improved the proliferation and differentiation and delayed the senescence of CB EPCs. The endothelial-promoting effect of CM seems to be related to smaller vesicles including exosomes (sEV/exo) contained in this medium and transferred to CB CD34(+) EPCs: in fact, purified preparations of sEV/exo isolated from CM mimicked the effect of CM to sustain endothelial formation. These observations provided the interesting indication that mature ECs exert a stimulatory effect on endothelial cell differentiation from CD34(+) cells.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Isabella Parolini
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Anna Maria Cerio
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Agnese D'Angiò
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Luca Pasquini
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Maria Carollo
- Department of Infectious, Parasitic and Immune-mediates Diseases, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Massimo Sargiacomo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 200, 00161 Rome, Italy
| |
Collapse
|
24
|
Testa U, Saulle E, Castelli G, Pelosi E. Endothelial progenitor cells in hematologic malignancies. Stem Cell Investig 2016; 3:26. [PMID: 27583252 DOI: 10.21037/sci.2016.06.07] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/23/2016] [Indexed: 01/09/2023]
Abstract
Studies carried out in the last years have improved the understanding of the cellular and molecular mechanisms controlling angiogenesis during adult life in normal and pathological conditions. Some of these studies have led to the identification of some progenitor cells that sustain angiogenesis through indirect, paracrine mechanisms (hematopoietic angiogenic cells) and through direct mechanisms, i.e., through their capacity to generate a progeny of phenotypically and functionally competent endothelial cells [endothelial colony forming cells (ECFCs)]. The contribution of these progenitors to angiogenetic processes under physiological and pathological conditions is intensively investigated. Angiogenetic mechanisms are stimulated in various hematological malignancies, including chronic myeloid leukemia (CML), acute myeloid leukemia (AML), myelodysplastic syndromes and multiple myeloma, resulting in an increased angiogenesis that contributes to disease progression. In some of these conditions there is preliminary evidence that some endothelial cells could derive from the malignant clone, thus leading to the speculation that the leukemic cell derives from the malignant transformation of a hemangioblastic progenitor, i.e., of a cell capable of differentiation to the hematopoietic and to the endothelial cell lineages. Our understanding of the mechanisms underlying increased angiogenesis in these malignancies not only contributed to a better knowledge of the mechanisms responsible for tumor progression, but also offered the way for the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Ernestina Saulle
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
25
|
Salazar-Martinez E, Rodriguez-Valentin R, Albavera-Hernandez C, Carreon-Rodriguez A, Lazcano-Ponce E. Number of colony-forming unit-Hill colonies among children and teenagers with obesity, dyslipidemia and breastfeeding history. Nutr Metab Cardiovasc Dis 2016; 26:534-540. [PMID: 27113291 DOI: 10.1016/j.numecd.2016.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/23/2016] [Accepted: 03/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS The number of colony-forming unit (CFU)-Hill colonies has been proposed as a biomarker of vascular function and cardiovascular risk in adults but information about its role in children is scarce. This study evaluates the associations between obesity, cardiovascular risk factors and breastfeeding history with the numbers of CFU-Hill colonies in a sample of young people. METHODS AND RESULTS We selected 49 children and teenagers between ages 10 and 17 (65.3% boys) from Mexican Health Care system. Physical activity and Anthropometric measures data were registered. CFU-Hill colonies were cultured from mononuclear cells obtained from venous blood. We detected inverse associations between the formation of CFU-Hill colonies and body mass index (BMI; β = -1.53; 95% confidence interval [CI], -1.92, -1.13), triglycerides (β = -0.26; 95%CI = -0.34, -0.18), total cholesterol (β = -0.13; 95%CI = -0.17, -0.08), Low Density Lipoprotein (LDL) (β = -0.20; 95%CI = -0.31, -0.09) and glucose (β = -0.37; 95%CI = -0.55, -0.18) using multivariate models. Breastfeeding duration showed a 1.46-colony increase for each month of breastfeeding (95%CI = 0.73, 2.18). CONCLUSIONS CFU-Hill colony-forming capacity in children and teenagers was inversely associated with obesity, dyslipidemia and high blood levels of glucose. In contrast a longer breastfeeding duration was directly associated with an increased number of CFU-Hill colonies. However these results must be confirmed with further studies. Our findings support the importance of promoting breastfeeding and monitoring nutritional and metabolic status at an early age to prevent chronic disease development.
Collapse
Affiliation(s)
| | | | | | | | - E Lazcano-Ponce
- National Institute of Public Health, Cuernavaca, Morelos, Mexico
| |
Collapse
|
26
|
Danova M, Comolli G, Manzoni M, Torchio M, Mazzini G. Flow cytometric analysis of circulating endothelial cells and endothelial progenitors for clinical purposes in oncology: A critical evaluation. Mol Clin Oncol 2016; 4:909-917. [PMID: 27284422 DOI: 10.3892/mco.2016.823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/11/2015] [Indexed: 12/19/2022] Open
Abstract
Malignant tumors are characterized by uncontrolled cell growth and metastatic spread, with a pivotal importance of the phenomenon of angiogenesis. For this reason, research has focused on the development of agents targeting the vascular component of the tumor microenvironment and regulating the angiogenic switch. As a result, the therapeutic inhibition of angiogenesis has become an important component of anticancer treatment, however, its utility is partly limited by the lack of an established methodology to assess its efficacy in vivo. Circulating endothelial cells (CECs), which are rare in healthy subjects and significantly increased in different tumor types, represent a promising tool for monitoring the tumor clinical outcome and the treatment response. A cell population circulating into the blood also able to form endothelial colonies in vitro and to promote vasculogenesis is represented by endothelial progenitor cells (EPCs). The number of both of these cell types is extremely low and they cannot be identified using a single marker, therefore, in absence of a definite consensus on their phenotype, require discrimination using combinations of antigens. Multiparameter flow cytometry (FCM) is ideal for rapid processing of high numbers of cells per second and is commonly utilized to quantify CECs and EPCs, however, remains technically challenging since there is as yet no standardized protocol for the identification and enumeration of these rare events. Methodology in studies on CECs and/or EPCs as clinical biomarkers in oncology is heterogeneous and data have been obtained from different studies leading to conflicting conclusions. The present review presented a critical review of the issues that limit the comparability of results of the most significant studies employing FCM for CEC and/or EPC detection in patients with cancer.
Collapse
Affiliation(s)
- Marco Danova
- Internal Medicine and Medical Oncology, Vigevano Hospital, ASST Pavia, I-27029 Vigevano, Italy
| | - Giuditta Comolli
- Microbiology and Virology, Biotechnology Laboratories, IRCCS San Matteo Foundation, I-27100 Pavia, Italy
| | | | - Martina Torchio
- Internal Medicine and Medical Oncology, Vigevano Hospital, ASST Pavia, I-27029 Vigevano, Italy
| | - Giuliano Mazzini
- Molecular Genetics Institute, National Research Council and Biology and Biotechnology Department 'L. Spallanzani', University of Pavia, I-27100 Pavia, Italy
| |
Collapse
|
27
|
Ohnishi H, Mizuno S, Mizuno-Horikawa Y, Kato T. Stromal cell-derived factor-1 (SDF1)-dependent recruitment of bone marrow-derived renal endothelium-like cells in a mouse model of acute kidney injury. J Vet Med Sci 2015; 77:313-9. [PMID: 25833353 PMCID: PMC4383777 DOI: 10.1292/jvms.14-0562] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ischemic acute kidney injury (AKI) is the most key pathological event for accelerating
progression to chronic kidney disease through vascular endothelial injury or dysfunction.
Thus, it is critical to elucidate the molecular mechanism of endothelial protection and
regeneration. Emerging evidence indicates that bone marrow-derived cells (BMCs) contribute
to tissue reconstitution in several types of organs post-injury, but little is known
whether and how BMCs contribute to renal endothelial reconstitution, especially in an
early-stage of AKI. Using a mouse model of ischemic AKI, we provide evidence that
incorporation of BMCs in vascular components (such as endothelial and smooth muscle cells)
becomes evident within four days after renal ischemia and reperfusion, associated with an
increase in stromal cell-derived factor-1 (SDF1) in endothelium and that in
CXCR4/SDF1-receptor in BMCs. Notably, anti-CXCR4 antibody decreased the numbers of
infiltrated BMCs and BMC-derived endothelium-like cells, but not of BMC-derived smooth
muscle cell-like cells. These results suggest that reconstitution of renal endothelium
post-ischemia partially depends on a paracrine loop of SDF1-CXCR4 between resident
endothelium and BMCs. Such a chemokine ligand-receptor system may be attributable for
selecting a cellular lineage (s), required for renal vascular protection, repair and
homeostasis, even in an earlier phase of AKI.
Collapse
Affiliation(s)
- Hiroyuki Ohnishi
- Department of Biochemistry, Osaka University Graduate School of Medicine, 2–2 Yamadaoka, Suita 565–0871; 2. Kinjo Gakuin University College of Pharmacy, 2-1723 Oomori, Moriyama-ku, Nagoya 463-8521, Japan
| | | | | | | |
Collapse
|
28
|
Are Endothelial Progenitor Cells the Real Solution for Cardiovascular Diseases? Focus on Controversies and Perspectives. BIOMED RESEARCH INTERNATIONAL 2015; 2015:835934. [PMID: 26509164 PMCID: PMC4609774 DOI: 10.1155/2015/835934] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/19/2015] [Accepted: 07/15/2015] [Indexed: 12/20/2022]
Abstract
Advanced knowledge in the field of stem cell biology and their ability to provide a cue for counteracting several diseases are leading numerous researchers to focus their attention on “regenerative medicine” as possible solutions for cardiovascular diseases (CVDs). However, the lack of consistent evidence in this arena has hampered the clinical application. The same condition affects the research on endothelial progenitor cells (EPCs), creating more confusion than comprehension. In this review, this aspect is discussed with particular emphasis. In particular, we describe biology and physiology of EPCs, outline their clinical relevance as both new predictive, diagnostic, and prognostic CVD biomarkers and therapeutic agents, discuss advantages, disadvantages, and conflicting data about their use as possible solutions for vascular impairment and clinical applications, and finally underline a very crucial aspect of EPCs “characterization and definition,” which seems to be the real cause of large heterogeneity existing in literature data on this topic.
Collapse
|
29
|
Kachamakova-Trojanowska N, Bukowska-Strakova K, Zukowska M, Dulak J, Jozkowicz A. The real face of endothelial progenitor cells - Circulating angiogenic cells as endothelial prognostic marker? Pharmacol Rep 2015; 67:793-802. [PMID: 26321283 DOI: 10.1016/j.pharep.2015.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 02/08/2023]
Abstract
Endothelial progenitor cells (EPCs) have been extensively studied for almost 19 years now and were considered as a potential marker for endothelial regeneration ability. On the other hand, circulating endothelial cells (CEC) were studied as biomarker for endothelial injury. Yet, in the literature, there is also huge incoherency in regards to terminology and protocols used. This results in misleading conclusions on the role of so called "EPCs", especially in the clinical field. The discrepancies are mainly due to strong phenotypic overlap between EPCs and circulating angiogenic cells (CAC), therefore changes in "EPC" terminology have been suggested. Other factors leading to inconsistent results are varied definitions of the studied populations and the lack of universal data reporting, which could strongly affect data interpretation. The current review is focused on controversies concerning the use of "EPCs"/CAC and CEC as putative endothelial diagnostic markers.
Collapse
Affiliation(s)
- Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Monika Zukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
30
|
Soler E, Grosveld F. Targeting epigenetics to speed up repair. Cell Stem Cell 2014; 14:553-4. [PMID: 24792110 DOI: 10.1016/j.stem.2014.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this issue of Cell Stem Cell, Palii et al. reveal that TAL1 is a master regulator of adhesion and migration networks in human endothelial progenitors and that ex vivo treatment with the histone deacetylase inhibitor TSA enables their faster vascularization after ischemic injury.
Collapse
Affiliation(s)
- Eric Soler
- INSERM UMR967 CEA/DSV/iRCM, 92265 Fontenay-aux-Roses, France.
| | - Frank Grosveld
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
31
|
Affiliation(s)
- Stella Kourembanas
- From the Division of Newborn Medicine, Boston Children's Hospital, Boston, MA (S.K.); and Department of Pediatrics, Harvard Medical School, Boston, MA (S.K.).
| |
Collapse
|