1
|
Senesi G, Guerricchio L, Ghelardoni M, Bertola N, Rebellato S, Grinovero N, Bartolucci M, Costa A, Raimondi A, Grange C, Bolis S, Massa V, Paladini D, Coviello D, Pandolfi A, Bussolati B, Petretto A, Fazio G, Ravera S, Barile L, Balbi C, Bollini S. Extracellular vesicles from II trimester human amniotic fluid as paracrine conveyors counteracting oxidative stress. Redox Biol 2024; 75:103241. [PMID: 38901103 PMCID: PMC11253147 DOI: 10.1016/j.redox.2024.103241] [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: 04/20/2024] [Revised: 06/07/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND We previously demonstrated that the human amniotic fluid (hAF) from II trimester of gestation is a feasible source of stromal progenitors (human amniotic fluid stem cells, hAFSC), with significant paracrine potential for regenerative medicine. Extracellular vesicles (EVs) separated and concentrated from hAFSC secretome can deliver pro-survival, proliferative, anti-fibrotic and cardioprotective effects in preclinical models of skeletal and cardiac muscle injury. While hAFSC-EVs isolation can be significantly influenced by in vitro cell culture, here we profiled EVs directly concentrated from hAF as an alternative option and investigated their paracrine potential against oxidative stress. METHODS II trimester hAF samples were obtained as leftover material from prenatal diagnostic amniocentesis following written informed consent. EVs were separated by size exclusion chromatography and concentrated by ultracentrifugation. hAF-EVs were assessed by nanoparticle tracking analysis, transmission electron microscopy, Western Blot, and flow cytometry; their metabolic activity was evaluated by oximetric and luminometric analyses and their cargo profiled by proteomics and RNA sequencing. hAF-EV paracrine potential was tested in preclinical in vitro models of oxidative stress and dysfunction on murine C2C12 cells and on 3D human cardiac microtissue. RESULTS Our protocol resulted in a yield of 6.31 ± 0.98 × 109 EVs particles per hAF milliliter showing round cup-shaped morphology and 209.63 ± 6.10 nm average size, with relevant expression of CD81, CD63 and CD9 tetraspanin markers. hAF-EVs were enriched in CD133/1, CD326, CD24, CD29, and SSEA4 and able to produce ATP by oxygen consumption. While oxidative stress significantly reduced C2C12 survival, hAF-EV priming resulted in significant rescue of cell viability, with notable recovery of ATP synthesis and concomitant reduction of cell damage and lipid peroxidation activity. 3D human cardiac microtissues treated with hAF-EVs and experiencing H2O2 stress and TGFβ stimulation showed improved survival with a remarkable decrease in the onset of fibrosis. CONCLUSIONS Our results suggest that leftover samples of II trimester human amniotic fluid can represent a feasible source of EVs to counteract oxidative damage on target cells, thus offering a novel candidate therapeutic option to counteract skeletal and cardiac muscle injury.
Collapse
Affiliation(s)
- Giorgia Senesi
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900, Lugano, Switzerland
| | - Laura Guerricchio
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy
| | | | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Stefano Rebellato
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy
| | - Nicole Grinovero
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Ambra Costa
- IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy
| | - Andrea Raimondi
- Institute for Research in Biomedicine, Università della Svizzera Italiana, CH-6500, Bellinzona, Switzerland
| | - Cristina Grange
- VEXTRA Facility and Department of Medical Sciences, University of Turin, 10126, Turin, Italy
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland
| | - Valentina Massa
- Department of Health Sciences, University of Milan, 20146, Milan, Italy
| | - Dario Paladini
- Fetal Medicine and Surgery Unit, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Domenico Coviello
- Human Genetics Laboratory, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara and Center for Advanced Studies and Technology - CAST, 66100, Chieti, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126, Turin, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, 16147, Genova, Italy
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy
| | - Silvia Ravera
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino and Laboratories for Traslational Research Ente Ospedaliero Cantonale, CH-6500, Bellinzona, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, CH-6900, Lugano, Switzerland.
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland.
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, 16132, Genova, Italy; IRCCS Ospedale Policlinico San Martino, 16132, Genova, Italy.
| |
Collapse
|
2
|
Rizzuto AS, Gelpi G, Mangini A, Carugo S, Ruscica M, Macchi C. Exploring the role of epicardial adipose-tissue-derived extracellular vesicles in cardiovascular diseases. iScience 2024; 27:109359. [PMID: 38510143 PMCID: PMC10951984 DOI: 10.1016/j.isci.2024.109359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024] Open
Abstract
Epicardial adipose tissue (EAT) is a fat depot located between the myocardium and the visceral layer of the epicardium, which, owing to its location, can influence surrounding tissues and can act as a local transducer of systemic inflammation. The mechanisms upon which such influence depends on are however unclear. Given the role EAT undoubtedly has in the scheme of cardiovascular diseases (CVDs), understanding the impact of its cellular components is of upmost importance. Extracellular vesicles (EVs) constitute promising candidates to fill the gap in the knowledge concerning the unexplored mechanisms through which EAT promotes onset and progression of CVDs. Owing to their ability of transporting active biomolecules, EAT-derived EVs have been reported to be actively involved in the pathogenesis of ischemia/reperfusion injury, coronary atherosclerosis, heart failure, and atrial fibrillation. Exploring the precise functions EVs exert in this context may aid in connecting the dots between EAT and CVDs.
Collapse
Affiliation(s)
| | - Guido Gelpi
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Mangini
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Carugo
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimiliano Ruscica
- Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, Milan, Italy
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, Milan, Italy
| |
Collapse
|
3
|
Rosner M, Horer S, Feichtinger M, Hengstschläger M. Multipotent fetal stem cells in reproductive biology research. Stem Cell Res Ther 2023; 14:157. [PMID: 37287077 DOI: 10.1186/s13287-023-03379-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Due to the limited accessibility of the in vivo situation, the scarcity of the human tissue, legal constraints, and ethical considerations, the underlying molecular mechanisms of disorders, such as preeclampsia, the pathological consequences of fetomaternal microchimerism, or infertility, are still not fully understood. And although substantial progress has already been made, the therapeutic strategies for reproductive system diseases are still facing limitations. In the recent years, it became more and more evident that stem cells are powerful tools for basic research in human reproduction and stem cell-based approaches moved into the center of endeavors to establish new clinical concepts. Multipotent fetal stem cells derived from the amniotic fluid, amniotic membrane, chorion leave, Wharton´s jelly, or placenta came to the fore because they are easy to acquire, are not associated with ethical concerns or covered by strict legal restrictions, and can be banked for autologous utilization later in life. Compared to adult stem cells, they exhibit a significantly higher differentiation potential and are much easier to propagate in vitro. Compared to pluripotent stem cells, they harbor less mutations, are not tumorigenic, and exhibit low immunogenicity. Studies on multipotent fetal stem cells can be invaluable to gain knowledge on the development of dysfunctional fetal cell types, to characterize the fetal stem cells migrating into the body of a pregnant woman in the context of fetomaternal microchimerism, and to obtain a more comprehensive picture of germ cell development in the course of in vitro differentiation experiments. The in vivo transplantation of fetal stem cells or their paracrine factors can mediate therapeutic effects in preeclampsia and can restore reproductive organ functions. Together with the use of fetal stem cell-derived gametes, such strategies could once help individuals, who do not develop functional gametes, to conceive genetically related children. Although there is still a long way to go, these developments regarding the usage of multipotent fetal stem cells in the clinic should continuously be accompanied by a wide and detailed ethical discussion.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Stefanie Horer
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | | | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
4
|
Wang J, Chen X, Zhang L, Zheng Y, Qian J, Sun N, Ding X, Cui B. Chick early amniotic fluid component improves heart function and protects against inflammation after myocardial infarction in mice. Front Cardiovasc Med 2022; 9:1042852. [DOI: 10.3389/fcvm.2022.1042852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022] Open
Abstract
Myocardial infarction (MI) is the major cause of mortality around the world. We recently demonstrated that chick early amniotic fluid (ceAF) can effectively rescue ischemic heart injury, indicating that it has a therapeutic function in MI. However, its functional components and the underlying mechanisms remain to be clarified. Here, we demonstrated that a fraction of ceAF, peak 8 (P8), had a protective effect on acute MI. P8 significantly decreased cardiomyocyte cross-sectional areas and cardiomyocyte apoptosis in MI mice. Using a human embryonic stem cell-derived cardiomyocyte model, which was subjected to hypoxia and reoxygenation, mimicking MI state, we found that P8 treatment reduced apoptosis and reversed myocardial contractility. Mechanistically, P8 improved cardiac function by inhibiting NF-κB signaling and downregulating inflammatory cytokine expression. Using mass spectrometry, we identified that guanosine and deoxynucleoside were the main functional components of P8 that suppressed the inflammatory response in human embryonic stem cell-derived cardiomyocytes. Collectively, our data suggest that specific components from ceAF are promising therapeutic agents for ischemic heart injury and could be a potential supplement to current medications for MI.
Collapse
|
5
|
Lee YS, Javan H, Reems JA, Li L, Lusty Beech J, Schaaf CI, Pierce J, Phillips JD, Selzman CH. Acellular human amniotic fluid protects the ischemic/reperfused rat myocardium. Am J Physiol Heart Circ Physiol 2022; 322:H406-H416. [DOI: 10.1152/ajpheart.00331.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amniotic products are potent immunomodulators utilized clinically to repair tissue injury. Little information exists regarding the potential of cell-free human amniotic fluid (hAF) to treat cardiovascular disease. Herein, we sought to determine the influence and efficacy of acellular hAF on myocardial ischemia/reperfusion injury. Processed hAF was obtained from volunteer donors at the time of elective caesarean section and manufactured using proprietary methods. Left anterior descending coronary artery ligation was performed on rats for 60 minutes. Thirty minutes after release and reperfusion, either saline or hAF was injected intramyocardially. Serial echocardiography revealed that compared to saline injected rats, hAF animals maintained their ejection fraction and did not adversely remodel through the 4-week period. This preserved ventricular function correlated with decreased infarct size, less fibrosis, and reduced expression of cytokines and infiltrating inflammatory cells. Comparative arrays of different donor hAF lots confirmed the presence of a wide array of immunomodulatory and host-defense proteins. The observed functional cardioprotection was furthermore evident when given intravenously and across multiple hAF donors. In conclusion, our data demonstrate, for the first time, the cardioprotective effect of acellular hAF on myocardial injury. These observations spanned across diverse donors and likely result from the mixture of a plethora of naturally produced cytokines, chemokines, and immune-modulating proteins rather than a single, defined mechanistic culprit. The ubiquitous availability of hAF as a cell-free solution further suggests its potential for widespread adoption as a therapy for myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Young Sook Lee
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Hadi Javan
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jo-Anna Reems
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Ling Li
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jessica Lusty Beech
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Christine I. Schaaf
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jan Pierce
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - John D. Phillips
- Cell Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Craig H. Selzman
- Division of Cardiothoracic Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
6
|
Costa A, Quarto R, Bollini S. Small Extracellular Vesicles from Human Amniotic Fluid Samples as Promising Theranostics. Int J Mol Sci 2022; 23:ijms23020590. [PMID: 35054775 PMCID: PMC8775841 DOI: 10.3390/ijms23020590] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 02/05/2023] Open
Abstract
Since the first evidence that stem cells can provide pro-resolving effects via paracrine secretion of soluble factors, growing interest has been addressed to define the most ideal cell source for clinical translation. Leftover or clinical waste samples of human amniotic fluid obtained following prenatal screening, clinical intervention, or during scheduled caesarean section (C-section) delivery at term have been recently considered an appealing source of mesenchymal progenitors with peculiar regenerative capacity. Human amniotic fluid stem cells (hAFSC) have been demonstrated to support tissue recovery in several preclinical models of disease by exerting paracrine proliferative, anti-inflammatory and regenerative influence. Small extracellular vesicles (EVs) concentrated from the hAFSC secretome (the total soluble trophic factors secreted in the cell-conditioned medium, hAFSC-CM) recapitulate most of the beneficial cell effects. Independent studies in preclinical models of either adult disorders or severe diseases in newborns have suggested a regenerative role of hAFSC-EVs. EVs can be eventually concentrated from amniotic fluid (hAF) to offer useful prenatal information, as recently suggested. In this review, we focus on the most significant aspects of EVs obtained from either hAFSC and hAF and consider the current challenges for their clinical translation, including isolation, characterization and quantification methods.
Collapse
Affiliation(s)
- Ambra Costa
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
| | - Rodolfo Quarto
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sveva Bollini
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Correspondence: ; Tel.: +39-010-555-8394
| |
Collapse
|
7
|
Stem C, Rodman C, Ramamurthy RM, George S, Meares D, Farland A, Atala A, Doering CB, Spencer HT, Porada CD, Almeida-Porada G. Investigating Optimal Autologous Cellular Platforms for Prenatal or Perinatal Factor VIII Delivery to Treat Hemophilia A. Front Cell Dev Biol 2021; 9:678117. [PMID: 34447745 PMCID: PMC8383113 DOI: 10.3389/fcell.2021.678117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
Patients with the severe form of hemophilia A (HA) present with a severe phenotype, and can suffer from life-threatening, spontaneous hemorrhaging. While prophylactic FVIII infusions have revolutionized the clinical management of HA, this treatment is short-lived, expensive, and it is not available to many A patients worldwide. In the present study, we evaluated a panel of readily available cell types for their suitability as cellular vehicles to deliver long-lasting FVIII replacement following transduction with a retroviral vector encoding a B domain-deleted human F8 transgene. Given the immune hurdles that currently plague factor replacement therapy, we focused our investigation on cell types that we deemed to be most relevant to either prenatal or very early postnatal treatment and that could, ideally, be autologously derived. Our findings identify several promising candidates for use as cell-based FVIII delivery vehicles and lay the groundwork for future mechanistic studies to delineate bottlenecks to efficient production and secretion of FVIII following genetic-modification.
Collapse
Affiliation(s)
- Christopher Stem
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher Rodman
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ritu M. Ramamurthy
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Sunil George
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Diane Meares
- Special Hematology Laboratory, Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andrew Farland
- Special Hematology Laboratory, Wake Forest Baptist Medical Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher B. Doering
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, United States
| | - H. Trent Spencer
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Christopher D. Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Fetal Research and Therapy Program, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
8
|
Fanni D, Gerosa C, Loddo C, Castagnola M, Fanos V, Zaffanello M, Faa G. Stem/progenitor cells in fetuses and newborns: overview of immunohistochemical markers. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:22. [PMID: 34219203 PMCID: PMC8255250 DOI: 10.1186/s13619-021-00084-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 04/12/2021] [Indexed: 12/26/2022]
Abstract
Microanatomy of the vast majority of human organs at birth is characterized by marked differences as compared to adult organs, regarding their architecture and the cell types detectable at histology. In preterm neonates, these differences are even more evident, due to the lower level of organ maturation and to ongoing cell differentiation. One of the most remarkable finding in preterm tissues is the presence of huge amounts of stem/progenitor cells in multiple organs, including kidney, brain, heart, adrenals, and lungs. In other organs, such as liver, the completely different burden of cell types in preterm infants is mainly related to the different function of the liver during gestation, mainly focused on hematopoiesis, a function that is taken by bone marrow after birth. Our preliminary studies showed that the antigens expressed by stem/progenitors differ significantly from one organ to the next. Moreover, within each developing human tissue, reactivity for different stem cell markers also changes during gestation, according with the multiple differentiation steps encountered by each progenitor during development. A better knowledge of stem/progenitor cells of preterms will allow neonatologists to boost preterm organ maturation, favoring the differentiation of the multiple cells types that characterize each organ in at term neonates.
Collapse
Affiliation(s)
- D Fanni
- Division of Pathology, University Hospital San Giovanni Di Dio, via Ospedale, 54, Cagliari, Italy.,Department of Biology, College of Science and Technology, Temple University, Phidelphia, USA
| | - C Gerosa
- Division of Pathology, University Hospital San Giovanni Di Dio, via Ospedale, 54, Cagliari, Italy.,Department of Biology, College of Science and Technology, Temple University, Phidelphia, USA
| | - C Loddo
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - M Castagnola
- Laboratory of Biochemistry and Metabolomics, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - V Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - M Zaffanello
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Piazzale Stefani, 1, I-37126, Verona, Italy.
| | - G Faa
- Division of Pathology, University Hospital San Giovanni Di Dio, via Ospedale, 54, Cagliari, Italy.,Department of Biology, College of Science and Technology, Temple University, Phidelphia, USA
| |
Collapse
|
9
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|
10
|
Costa A, Ceresa D, De Palma A, Rossi R, Turturo S, Santamaria S, Balbi C, Villa F, Reverberi D, Cortese K, De Biasio P, Paladini D, Coviello D, Ravera S, Malatesta P, Mauri P, Quarto R, Bollini S. Comprehensive Profiling of Secretome Formulations from Fetal- and Perinatal Human Amniotic Fluid Stem Cells. Int J Mol Sci 2021; 22:ijms22073713. [PMID: 33918297 PMCID: PMC8038201 DOI: 10.3390/ijms22073713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
We previously reported that c-KIT+ human amniotic-fluid derived stem cells obtained from leftover samples of routine II trimester prenatal diagnosis (fetal hAFS) are endowed with regenerative paracrine potential driving pro-survival, anti-fibrotic and proliferative effects. hAFS may also be isolated from III trimester clinical waste samples during scheduled C-sections (perinatal hAFS), thus offering a more easily accessible alternative when compared to fetal hAFS. Nonetheless, little is known about the paracrine profile of perinatal hAFS. Here we provide a detailed characterization of the hAFS total secretome (i.e., the entirety of soluble paracrine factors released by cells in the conditioned medium, hAFS-CM) and the extracellular vesicles (hAFS-EVs) within it, from II trimester fetal- versus III trimester perinatal cells. Fetal- and perinatal hAFS were characterized and subject to hypoxic preconditioning to enhance their paracrine potential. hAFS-CM and hAFS-EV formulations were analyzed for protein and chemokine/cytokine content, and the EV cargo was further investigated by RNA sequencing. The phenotype of fetal- and perinatal hAFS, along with their corresponding secretome formulations, overlapped; yet, fetal hAFS showed immature oxidative phosphorylation activity when compared to perinatal ones. The profiling of their paracrine cargo revealed some differences according to gestational stage and hypoxic preconditioning. Both cell sources provided formulations enriched with neurotrophic, immunomodulatory, anti-fibrotic and endothelial stimulating factors, and the immature fetal hAFS secretome was defined by a more pronounced pro-vasculogenic, regenerative, pro-resolving and anti-aging profile. Small RNA profiling showed microRNA enrichment in both fetal- and perinatal hAFS-EV cargo, with a stably- expressed pro-resolving core as a reference molecular signature. Here we confirm that hAFS represents an appealing source of regenerative paracrine factors; the selection of either fetal or perinatal hAFS secretome formulations for future paracrine therapy should be evaluated considering the specific clinical scenario.
Collapse
Affiliation(s)
- Ambra Costa
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (S.T.); (P.M.)
| | - Davide Ceresa
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Antonella De Palma
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies (ITB-CNR), 20054 Milan, Italy; (A.D.P.); (R.R.); (P.M.)
| | - Rossana Rossi
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies (ITB-CNR), 20054 Milan, Italy; (A.D.P.); (R.R.); (P.M.)
| | - Sara Turturo
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (S.T.); (P.M.)
| | - Sara Santamaria
- Human Anatomy Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.S.); (K.C.); (S.R.)
| | - Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland;
- Center for Molecular Cardiology, University of Zurich, 8952 Zurich, Switzerland
| | - Federico Villa
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS Ospedale Policlinico, San Martino, 16132 Genova, Italy;
| | - Katia Cortese
- Human Anatomy Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.S.); (K.C.); (S.R.)
| | - Pierangela De Biasio
- Prenatal Diagnosis and Perinatal Medicine Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Dario Paladini
- Fetal Medicine and Surgery Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Domenico Coviello
- Laboratory of Human Genetics, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Silvia Ravera
- Human Anatomy Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.S.); (K.C.); (S.R.)
| | - Paolo Malatesta
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (S.T.); (P.M.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Pierluigi Mauri
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies (ITB-CNR), 20054 Milan, Italy; (A.D.P.); (R.R.); (P.M.)
| | - Rodolfo Quarto
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (S.T.); (P.M.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Correspondence: (R.Q.); (S.B.); Tel.: +39-010-5558-257 (S.B.)
| | - Sveva Bollini
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (S.T.); (P.M.)
- Correspondence: (R.Q.); (S.B.); Tel.: +39-010-5558-257 (S.B.)
| |
Collapse
|
11
|
Hu J, Chen X, Li P, Lu X, Yan J, Tan H, Zhang C. Exosomes derived from human amniotic fluid mesenchymal stem cells alleviate cardiac fibrosis via enhancing angiogenesis in vivo and in vitro. Cardiovasc Diagn Ther 2021; 11:348-361. [PMID: 33968614 DOI: 10.21037/cdt-20-1032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Cardiac fibrosis is a pathological process characterized by excess extracellular matrix (ECM) deposition and plays a critical role in nearly all types of heart disease. The mechanism of cardiac fibrosis is still unclear and no effective medication treatment of cardiac fibrosis. Research showed that mesenchymal stem cell (MSC) derived exosomes may play a critical role in cardiac fibrosis. The effect of human amniotic fluid MSC (hAFMSC)-derived exosomes (hAFMSCExos) on cardiac fibrosis has remained unclear. Methods The hAFMSCExos were extracted using a sequential centrifugation approach. The effects of hAFMSCExos on angiogenesis were analyzed both in human umbilical vein endothelial cells (HUVECs) after oxygen and glucose deprivation (OGD) in vitro, and in isoproterenol (ISO) induced-cardiac fibrosis in vivo. Results The hAFMSCExos remarkably up-regulate the motility and migration of HUVECs after OGD compared with phosphate-buffered saline (PBS). Meanwhile, total tube length, total branching points and total loops were significantly raised in HUVECs after OGD treated with hAFMSCExos. The hAFMSCExos alleviated the cardiac fibrosis degree tested by hematoxylin-eosin (H&E) and Masson staining. The protein levels of Collagen I and α-smooth muscle actin (α-SMA) were lower in exosomes group rats than PBS group. Immunofluorescence suggested that hAFMSCExos can promote the expression of CD31 in the rats. Meanwhile, the number of regenerated microvessels was significantly enhanced in rats administrated with exosomes by quantitative analysis of microvessel density. Furthermore, the micro-CT scanning evidenced that hAFMSCExos promote angiogenesis after cardiac fibrosis. The levels of hypoxia-inducible factor 1 α (HIF-1α) and vascular endothelial growth factor (VEGF) expression in the left ventricle accepted HUVECs were higher than PBS treatment at 7 days post-treatment by Western blot analysis. Conclusions The hAFMSCExos have proangiogenic effects on endothelial cells and enhanced angiogenesis in cardiac fibrosis. The hAFMSCExos may be a promising potential treatment strategy for cardiac fibrosis.
Collapse
Affiliation(s)
- Jiajia Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xuliang Chen
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, China
| | - Xiaoxu Lu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianqin Yan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Huiling Tan
- Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Kukumberg M, Phermthai T, Wichitwiengrat S, Wang X, Arjunan S, Chong SY, Fong CY, Wang JW, Rufaihah AJ, Mattar CNZ. Hypoxia-induced amniotic fluid stem cell secretome augments cardiomyocyte proliferation and enhances cardioprotective effects under hypoxic-ischemic conditions. Sci Rep 2021; 11:163. [PMID: 33420256 PMCID: PMC7794288 DOI: 10.1038/s41598-020-80326-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Secretome derived from human amniotic fluid stem cells (AFSC-S) is rich in soluble bioactive factors (SBF) and offers untapped therapeutic potential for regenerative medicine while avoiding putative cell-related complications. Characterization and optimal generation of AFSC-S remains challenging. We hypothesized that modulation of oxygen conditions during AFSC-S generation enriches SBF and confers enhanced regenerative and cardioprotective effects on cardiovascular cells. We collected secretome at 6-hourly intervals up to 30 h following incubation of AFSC in normoxic (21%O2, nAFSC-S) and hypoxic (1%O2, hAFSC-S) conditions. Proliferation of human adult cardiomyocytes (hCM) and umbilical cord endothelial cells (HUVEC) incubated with nAFSC-S or hAFSC-S were examined following culture in normoxia or hypoxia. Lower AFSC counts and richer protein content in AFSC-S were observed in hypoxia. Characterization of AFSC-S by multiplex immunoassay showed higher concentrations of pro-angiogenic and anti-inflammatory SBF. hCM demonstrated highest proliferation with 30h-hAFSC-S in hypoxic culture. The cardioprotective potential of concentrated 30h-hAFSC-S treatment was demonstrated in a myocardial ischemia-reperfusion injury mouse model by infarct size and cell apoptosis reduction and cell proliferation increase when compared to saline treatment controls. Thus, we project that hypoxic-generated AFSC-S, with higher pro-angiogenic and anti-inflammatory SBF, can be harnessed and refined for tailored regenerative applications in ischemic cardiovascular disease.
Collapse
Affiliation(s)
- Marek Kukumberg
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tatsanee Phermthai
- Stem Cell Research and Development for Medical Therapy Unit, Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suparat Wichitwiengrat
- Stem Cell Research and Development for Medical Therapy Unit, Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Subramanian Arjunan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Chui-Yee Fong
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Abdul Jalil Rufaihah
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Citra Nurfarah Zaini Mattar
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Obstetrics and Gynaecology, National University Health System, Singapore, Singapore.
| |
Collapse
|
13
|
Progress and Challenges of Amniotic Fluid Derived Stem Cells in Therapy of Ischemic Heart Disease. Int J Mol Sci 2020; 22:ijms22010102. [PMID: 33374215 PMCID: PMC7794729 DOI: 10.3390/ijms22010102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/13/2020] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular disease is the leading cause of deaths worldwide, claiming an estimated total of 17.9 million lives each year, of which one-third of the people are under the age of 70 years. Since adult cardiomyocytes fail to regenerate, the heart loses the ability to repair itself after an injury, making patients with heart disease suffer from poor prognosis. Pluripotent stem cells have the ability to differentiate into cardiomyocytes in vitro through a well-established process, which is a new advancement in cardiac regeneration therapy. However, pluripotent stem cell-derived cardiomyocytes have certain drawbacks, such as the risk of arrhythmia and immune incompatibility. Thus, amniotic fluid stem cells (AFSCs), a relatively novel source of stem cells, have been exploited for their ability of pluripotent differentiation. In addition, since AFSCs are weakly positive for the major histocompatibility class II molecules, they may have high immune tolerance. In summary, the possibility of development of cardiomyocytes from AFSCs, as well as their transplantation in host cells to produce mechanical contraction, has been discussed. Thus, this review article highlights the progress of AFSC therapy and its application in the treatment of heart diseases in recent years.
Collapse
|
14
|
Singh S, Varshney A, Borkar N, Jindal A, Padhi P, Ahmed I, Srivastava N. Clinical Utility of Stem Cells in Congenital Anomalies: New Horizons in Pediatric Surgery. Indian J Surg 2020. [DOI: 10.1007/s12262-020-02264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
15
|
The Therapeutic Potential of Mesenchymal Stromal Cells in the Treatment of Chemotherapy-Induced Tissue Damage. Stem Cell Rev Rep 2020; 15:356-373. [PMID: 30937640 DOI: 10.1007/s12015-019-09886-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemotherapy constitutes one of the key treatment modalities for solid and hematological malignancies. Albeit being an effective treatment, chemotherapy application is often limited by its damage to healthy tissues, and curative treatment options for chemotherapy-related side effects are largely missing. As mesenchymal stromal cells (MSCs) are known to exhibit regenerative capacity mainly by supporting a beneficial microenvironment for tissue repair, MSC-based therapies may attenuate chemotherapy-induced tissue injuries. An increasing number of animal studies shows favorable effects of MSC-based treatments; however, clinical trials for MSC therapies in the context of chemotherapy-related side effects are rare. In this concise review, we summarize the current knowledge of the effects of MSCs on chemotherapy-induced tissue toxicities. Both preclinical and early clinical trials investigating MSC-based treatments for chemotherapy-related side reactions are presented, and mechanistic explanations about the regenerative effects of MSCs in the context of chemotherapy-induced tissue damage are discussed. Furthermore, challenges of MSC-based treatments are outlined that need closer investigations before these multipotent cells can be safely applied to cancer patients. As any pro-tumorigenicity of MSCs needs to be ruled out prior to clinical utilization of these cells for cancer patients, the pro- and anti-tumorigenic activities of MSCs are discussed in detail.
Collapse
|
16
|
Perrino C, Ferdinandy P, Bøtker HE, Brundel BJJM, Collins P, Davidson SM, den Ruijter HM, Engel FB, Gerdts E, Girao H, Gyöngyösi M, Hausenloy DJ, Lecour S, Madonna R, Marber M, Murphy E, Pesce M, Regitz-Zagrosek V, Sluijter JPG, Steffens S, Gollmann-Tepeköylü C, Van Laake LW, Van Linthout S, Schulz R, Ytrehus K. Improving translational research in sex-specific effects of comorbidities and risk factors in ischaemic heart disease and cardioprotection: position paper and recommendations of the ESC Working Group on Cellular Biology of the Heart. Cardiovasc Res 2020; 117:367-385. [PMID: 32484892 DOI: 10.1093/cvr/cvaa155] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/29/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Ischaemic heart disease (IHD) is a complex disorder and a leading cause of death and morbidity in both men and women. Sex, however, affects several aspects of IHD, including pathophysiology, incidence, clinical presentation, diagnosis as well as treatment and outcome. Several diseases or risk factors frequently associated with IHD can modify cellular signalling cascades, thus affecting ischaemia/reperfusion injury as well as responses to cardioprotective interventions. Importantly, the prevalence and impact of risk factors and several comorbidities differ between males and females, and their effects on IHD development and prognosis might differ according to sex. The cellular and molecular mechanisms underlying these differences are still poorly understood, and their identification might have important translational implications in the prediction or prevention of risk of IHD in men and women. Despite this, most experimental studies on IHD are still undertaken in animal models in the absence of risk factors and comorbidities, and assessment of potential sex-specific differences are largely missing. This ESC WG Position Paper will discuss: (i) the importance of sex as a biological variable in cardiovascular research, (ii) major biological mechanisms underlying sex-related differences relevant to IHD risk factors and comorbidities, (iii) prospects and pitfalls of preclinical models to investigate these associations, and finally (iv) will provide recommendations to guide future research. Although gender differences also affect IHD risk in the clinical setting, they will not be discussed in detail here.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary.,Pharmahungary Group, Hajnoczy str. 6., H-6722 Szeged, Hungary
| | - Hans E Bøtker
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 161, 8200 Aarhus, Denmark
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, Amsterdam, 1108 HV, the Netherlands
| | - Peter Collins
- Imperial College, Faculty of Medicine, National Heart & Lung Institute, South Kensington Campus, London SW7 2AZ, UK.,Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), Schwabachanlage 12, 91054 Erlangen, Germany
| | - Eva Gerdts
- Department for Clinical Science, University of Bergen, PO Box 7804, 5020 Bergen, Norway
| | - Henrique Girao
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Azinhaga Santa Comba, Celas, 3000-548 Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, and Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, 169609, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, 119228, Singapore.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa, Faculty of Health Sciences, Chris Barnard Building, University of Cape Town, Private Bag X3 7935 Observatory, Cape Town, South Africa
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Lungarno Antonio Pacinotti 43, 56126 Pisa, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, 6410 Fannin St #1014, Houston, TX 77030, USA
| | - Michael Marber
- King's College London BHF Centre, The Rayne Institute, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS Via Parea, 4, I-20138 Milan, Italy
| | - Vera Regitz-Zagrosek
- Berlin Institute of Gender in Medicine, Center for Cardiovascular Research, DZHK, partner site Berlin, Geschäftsstelle Potsdamer Str. 58, 10785 Berlin, Germany.,University of Zürich, Rämistrasse 71, 8006 Zürich, Germany
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands.,Circulatory Health Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, the Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Can Gollmann-Tepeköylü
- Department of Cardiac Surgery, Medical University of Innsbruck, Anichstr.35, A - 6020 Innsbruck, Austria
| | - Linda W Van Laake
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, 10178 Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University Giessen, Ludwigstraße 23, 35390 Giessen, Germany
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9037 Tromsø, Norway
| |
Collapse
|
17
|
Maghin E, Garbati P, Quarto R, Piccoli M, Bollini S. Young at Heart: Combining Strategies to Rejuvenate Endogenous Mechanisms of Cardiac Repair. Front Bioeng Biotechnol 2020; 8:447. [PMID: 32478060 PMCID: PMC7237726 DOI: 10.3389/fbioe.2020.00447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
True cardiac regeneration of the injured heart has been broadly described in lower vertebrates by active replacement of lost cardiomyocytes to functionally and structurally restore the myocardial tissue. On the contrary, following severe injury (i.e., myocardial infarction) the adult mammalian heart is endowed with an impaired reparative response by means of meager wound healing program and detrimental remodeling, which can lead over time to cardiomyopathy and heart failure. Lately, a growing body of basic, translational and clinical studies have supported the therapeutic use of stem cells to provide myocardial regeneration, with the working hypothesis that stem cells delivered to the cardiac tissue could result into new cardiovascular cells to replenish the lost ones. Nevertheless, multiple independent evidences have demonstrated that injected stem cells are more likely to modulate the cardiac tissue via beneficial paracrine effects, which can enhance cardiac repair and reinstate the embryonic program and cell cycle activity of endogenous cardiac stromal cells and resident cardiomyocytes. Therefore, increasing interest has been addressed to the therapeutic profiling of the stem cell-derived secretome (namely the total of cell-secreted soluble factors), with specific attention to cell-released extracellular vesicles, including exosomes, carrying cardioprotective and regenerative RNA molecules. In addition, the use of cardiac decellularized extracellular matrix has been recently suggested as promising biomaterial to develop novel therapeutic strategies for myocardial repair, as either source of molecular cues for regeneration, biological scaffold for cardiac tissue engineering or biomaterial platform for the functional release of factors. In this review, we will specifically address the translational relevance of these two approaches with ad hoc interest in their feasibility to rejuvenate endogenous mechanisms of cardiac repair up to functional regeneration.
Collapse
Affiliation(s)
- Edoardo Maghin
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.,Department of Women's and Children Health, University of Padova, Padua, Italy
| | - Patrizia Garbati
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Rodolfo Quarto
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy.,UOC Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Martina Piccoli
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
18
|
Balbi C, Costa A, Barile L, Bollini S. Message in a Bottle: Upgrading Cardiac Repair into Rejuvenation. Cells 2020; 9:cells9030724. [PMID: 32183455 PMCID: PMC7140681 DOI: 10.3390/cells9030724] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
Ischaemic cardiac disease is associated with a loss of cardiomyocytes and an intrinsic lack of myocardial renewal. Recent work has shown that the heart retains limited cardiomyocyte proliferation, which remains inefficient when facing pathological conditions. While broadly active in the neonatal mammalian heart, this mechanism becomes quiescent soon after birth, suggesting loss of regenerative potential with maturation into adulthood. A key question is whether this temporary regenerative window can be enhanced via appropriate stimulation and further extended. Recently the search for novel therapeutic approaches for heart disease has centred on stem cell biology. The “paracrine effect” has been proposed as a promising strategy to boost endogenous reparative and regenerative mechanisms from within the cardiac tissue by exploiting the modulatory potential of soluble stem cell-secreted factors. As such, growing interest has been specifically addressed towards stem/progenitor cell-secreted extracellular vesicles (EVs), which can be easily isolated in vitro from cell-conditioned medium. This review will provide a comprehensive overview of the current paradigm on cardiac repair and regeneration, with a specific focus on the role and mechanism(s) of paracrine action of EVs from cardiac stromal progenitors as compared to exogenous stem cells in order to discuss the optimal choice for future therapy. In addition, the challenges to overcoming translational EV biology from bench to bedside for future cardiac regenerative medicine will be discussed.
Collapse
Affiliation(s)
- Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland;
| | - Ambra Costa
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Correspondence: (L.B.); (S.B.)
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Dept. of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
- Correspondence: (L.B.); (S.B.)
| |
Collapse
|
19
|
Takov K, He Z, Johnston HE, Timms JF, Guillot PV, Yellon DM, Davidson SM. Small extracellular vesicles secreted from human amniotic fluid mesenchymal stromal cells possess cardioprotective and promigratory potential. Basic Res Cardiol 2020; 115:26. [PMID: 32146560 PMCID: PMC7060967 DOI: 10.1007/s00395-020-0785-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) exhibit antiapoptotic and proangiogenic functions in models of myocardial infarction which may be mediated by secreted small extracellular vesicles (sEVs). However, MSCs have frequently been harvested from aged or diseased patients, while the isolated sEVs often contain high levels of impurities. Here, we studied the cardioprotective and proangiogenic activities of size-exclusion chromatography-purified sEVs secreted from human foetal amniotic fluid stem cells (SS-hAFSCs), possessing superior functional potential to that of adult MSCs. We demonstrated for the first time that highly pure (up to 1.7 × 1010 particles/µg protein) and thoroughly characterised SS-hAFSC sEVs protect rat hearts from ischaemia-reperfusion injury in vivo when administered intravenously prior to reperfusion (38 ± 9% infarct size reduction, p < 0.05). SS-hAFSC sEVs did not protect isolated primary cardiomyocytes in models of simulated ischaemia-reperfusion injury in vitro, indicative of indirect cardioprotective effects. SS-hAFSC sEVs were not proangiogenic in vitro, although they markedly stimulated endothelial cell migration. Additionally, sEVs were entirely responsible for the promigratory effects of the medium conditioned by SS-hAFSC. Mechanistically, sEV-induced chemotaxis involved phosphatidylinositol 3-kinase (PI3K) signalling, as its pharmacological inhibition in treated endothelial cells reduced migration by 54 ± 7% (p < 0.001). Together, these data indicate that SS-hAFSC sEVs have multifactorial beneficial effects in a myocardial infarction setting.
Collapse
Affiliation(s)
- Kaloyan Takov
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Harvey E Johnston
- EGA Institute for Women's Health, University College London, London, UK
| | - John F Timms
- EGA Institute for Women's Health, University College London, London, UK
| | - Pascale V Guillot
- EGA Institute for Women's Health, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
20
|
Balbi C, Lodder K, Costa A, Moimas S, Moccia F, van Herwaarden T, Rosti V, Campagnoli F, Palmeri A, De Biasio P, Santini F, Giacca M, Goumans MJ, Barile L, Smits AM, Bollini S. Reactivating endogenous mechanisms of cardiac regeneration via paracrine boosting using the human amniotic fluid stem cell secretome. Int J Cardiol 2019; 287:87-95. [PMID: 30987834 DOI: 10.1016/j.ijcard.2019.04.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND The adult mammalian heart retains residual regenerative capability via endogenous cardiac progenitor cell (CPC) activation and cardiomyocyte proliferation. We previously reported the paracrine cardioprotective capacity of human amniotic fluid-derived stem cells (hAFS) following ischemia or cardiotoxicity. Here we analyse the potential of hAFS secretome fractions for cardiac regeneration and future clinical translation. METHODS hAFS were isolated from amniotic fluid leftover samples from prenatal screening. hAFS conditioned medium (hAFS-CM) was obtained following hypoxic preconditioning. Anti-apoptotic, angiogenic and proliferative effects were evaluated on rodent neonatal cardiomyocytes (r/mNVCM), human endothelial colony forming cells (hECFC) and human CPC. Mice undergoing myocardial infarction (MI) were treated with hAFS-CM, hAFS-extracellular vesicles (hAFS-EV), or EV-depleted hAFS-CM (hAFS-DM) by single intra-myocardial administration and evaluated in the short and long term. RESULTS hAFS-CM improved mNVCM survival under oxidative and hypoxic damage, induced Ca2+-dependent angiogenesis in hECFC and triggered hCPC and rNVCM proliferation. hAFS-CM treatment after MI counteracted scarring, supported cardiac function, angiogenesis and cardiomyocyte cell cycle progression in the long term. hAFS-DM had no effect. hAFS-CM and hAFS-EV equally induced epicardium WT1+ CPC reactivation. Although no CPC cardiovascular differentiation was observed, our data suggests contribution to local angiogenesis by paracrine modulation. hAFS-EV alone were able to recapitulate all the beneficial effects exerted by hAFS-CM, except for stimulation of vessel formation. CONCLUSIONS hAFS-CM and hAFS-EV can improve cardiac repair and trigger cardiac regeneration via paracrine modulation of endogenous mechanisms. While both formulations are effective in sustaining myocardial renewal, hAFS-CM retains higher pro-angiogenic potential, while hAFS-EV particularly enhances cardiac function.
Collapse
Affiliation(s)
- Carolina Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy; Molecular and Cell Cardiology Laboratory, CardioCentro Ticino, Lugano, Switzerland
| | - Kirsten Lodder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ambra Costa
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Silvia Moimas
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Francesco Moccia
- General Physiology Laboratory, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Tessa van Herwaarden
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vittorio Rosti
- Laboratory of Biochemistry, Biotechnology and Advanced Diagnostic, Myelofibrosis Study Centre, IRCCS Ospedale Policlinico San Matteo, Pavia, Italy
| | - Francesca Campagnoli
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Agnese Palmeri
- Dept. of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Pierangela De Biasio
- Dept. of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Santini
- Division of Cardiac Surgery, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lucio Barile
- Molecular and Cell Cardiology Laboratory, CardioCentro Ticino, Lugano, Switzerland
| | - Anke M Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy.
| |
Collapse
|
21
|
The Regenerative Potential of Amniotic Fluid Stem Cell Extracellular Vesicles: Lessons Learned by Comparing Different Isolation Techniques. Sci Rep 2019; 9:1837. [PMID: 30755672 PMCID: PMC6372651 DOI: 10.1038/s41598-018-38320-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/20/2018] [Indexed: 12/30/2022] Open
Abstract
Extracellular vesicles (EVs) derived from amniotic fluid stem cells (AFSCs) mediate anti-apoptotic, pro-angiogenic, and immune-modulatory effects in multiple disease models, such as skeletal muscle atrophy and Alport syndrome. A source of potential variability in EV biological functions is how EV are isolated from parent cells. Currently, a comparative study of different EV isolation strategies using conditioned medium from AFSCs is lacking. Herein, we examined different isolation strategies for AFSC-EVs, using common techniques based on differential sedimentation (ultracentrifugation), solubility (ExoQuick, Total Exosome Isolation Reagent, Exo-PREP), or size-exclusion chromatography (qEV). All techniques isolated AFSC-EVs with typical EV morphology and protein markers. In contrast, AFSC-EV size, protein content, and yield varied depending on the method of isolation. When equal volumes of the different AFSC-EV preparations were used as treatment in a model of lung epithelial injury, we observed a significant variation in how AFSC-EVs were able to protect against cell death. AFSC-EV enhancement of cell survival appeared to be dose dependent, and largely uninfluenced by variation in EV-size distributions, relative EV-purity, or their total protein content. The variation in EV-mediated cell survival obtained with different isolation strategies emphasizes the importance of testing alternative isolation techniques in order to maximize EV regenerative capacity.
Collapse
|
22
|
Hou Y, Li C. Stem/Progenitor Cells and Their Therapeutic Application in Cardiovascular Disease. Front Cell Dev Biol 2018; 6:139. [PMID: 30406100 PMCID: PMC6200850 DOI: 10.3389/fcell.2018.00139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/28/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the world. The stem/progenitor cell-based therapy has emerged as a promising approach for the treatment of a variety of cardiovascular diseases including myocardial infarction, stroke, peripheral arterial disease, and diabetes. An increasing number of evidence has shown that stem/progenitor cell transplantation could replenish damaged cells, improve cardiac and vascular functions, and repair injured tissues in many pre-clinical studies and clinical trials. In this review, we have outlined the major types of stem/progenitor cells, and summarized the studies in applying these cells, especially endothelial stem/progenitor cells and their derivatives, in the treatment of cardiovascular disease. Here the strategies used to improve the stem/progenitor cell-based therapies in cardiovascular disease and the challenges with these therapies in clinical applications are also reviewed.
Collapse
Affiliation(s)
- Yuning Hou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| | - Chunying Li
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
23
|
Vescovo G, Castellani C, Fedrigo M, Virzì GM, Vescovo GM, Tavano R, Pozzobon M, Angelini A. Stem cells transplantation positively modulates the heart-kidney cross talk in cardiorenal syndrome type II. Int J Cardiol 2018; 275:136-144. [PMID: 30509369 DOI: 10.1016/j.ijcard.2018.10.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/04/2018] [Accepted: 10/09/2018] [Indexed: 12/28/2022]
Abstract
INTRODUCTION We investigated the effects of human amniotic fluid stem cells (hAFS) and rat adipose tissue stromal vascular fraction GFP-positive cells (rSVC-GFP) in a model of cardio-renal syndrome type II (CRSII). METHODS AND RESULTS RHF was induced by monocrotaline (MCT) in 28 Sprague-Dawley rats. Three weeks later, four million hAFS or rSVC-GFP cells were injected via tail vein. BNP, sCreatinine, kidney and heart NGAL and MMP9, sCytokines, kidney and heart apoptosis and cells (Cs) engraftment were evaluated. Cell-treated rats showed a significant reduction of serum NGAL and Creatinine compared to CRSII. In both hAFS and rSVC-GFP group, kidney protein expression of NGAL was significantly lower than in CRSII (hAFS p = 0.036 and rSVC-GFP p < 0.0001) and similar to that of controls. In both hAFS and rSVC-GFP treated rats, we observed cell engraftment in the medulla and differentiation into tubular, endothelial and SMCs cells. Apoptosis was significantly decreased in cell-treated rats (hAFS 14.07 ± 1.38 and rSVC-GFP 12.67 ± 2.96 cells/mm2) and similar to controls (9.85 ± 2.1 cell/mm2). TUNEL-positive cells were mainly located in the kidney medulla. Pro-inflammatory cytokines were down regulated in cell-treated groups and similar to controls. In cell-treated rats, kidney and heart tissue NGAL was not complexed with MMP9 as in CRSII group, suggesting inhibition of MMPs activity. CONCLUSION Cell therapy produced improvement in kidney function in rats with CRSII. This was the result of interstitial, vessel and tubular cell engraftment leading to tubular and vessel regeneration, decreased tubular cells apoptosis and mitigated pro-inflammatory milieu. Reduction of NGLA-MMP9 complexes mainly due to decrease MMPs activity prevented further negative heart remodeling.
Collapse
Affiliation(s)
| | - Chiara Castellani
- Dept. Cardiac Thoracic, Vascular Sciences and Public Health, University of Padua, Italy
| | - Marny Fedrigo
- Dept. Cardiac Thoracic, Vascular Sciences and Public Health, University of Padua, Italy
| | - Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplant, San Bortolo Hospital, Vicenza, Italy; IRRIV-International Renal Resarch Institute Vicenza, San Bortolo Hospital, Vicenza, Italy
| | | | - Regina Tavano
- Dept. Biomedical Sciences, University of Padua, Italy
| | - Michela Pozzobon
- Dept. Women and Children Health, University of Padua, Italy; Insitute of Pediatric Research Città della Speranza, Padova, Italy
| | - Annalisa Angelini
- Dept. Cardiac Thoracic, Vascular Sciences and Public Health, University of Padua, Italy.
| |
Collapse
|
24
|
Kunisaki SM. Amniotic Fluid Stem Cells for the Treatment of Surgical Disorders in the Fetus and Neonate. Stem Cells Transl Med 2018; 7:767-773. [PMID: 30085416 PMCID: PMC6216434 DOI: 10.1002/sctm.18-0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/14/2018] [Accepted: 06/08/2018] [Indexed: 12/11/2022] Open
Abstract
Over the past decade, amniotic fluid‐derived stem cells have emerged as a novel experimental approach aimed at improving outcomes in children with congenital anomalies, including spina bifida, heart defects, and diaphragmatic hernia. Interest in these cells for the treatment of prenatally diagnosed diseases has arisen based on numerous studies demonstrating the relative ease of harvesting an abundant quantity of amniocytes from a small aliquot of fluid, the unique properties of amniocytes themselves, and the beneficial effects of amniotic fluid‐derived stem cells in experimental animal models. This report gives a brief overview of the rationale and current status of amniotic fluid stem cell‐based therapies, focusing on its relevance to birth defects affecting the fetus and neonate. The author proposes a roadmap for further study that would be required prior to clinical application of amniotic fluid stem cell technologies. stem cells translational medicine2018;7:767–773
Collapse
Affiliation(s)
- Shaun M Kunisaki
- Department of Surgery, Fetal Diagnosis and Treatment Center and Section of Pediatric Surgery, University of Michigan, C.S. Mott Children's and Von Voigtlander Women's Hospital, Ann Arbor, Michigan, USA
| |
Collapse
|
25
|
Dubé KN, Smart N. Thymosin β4 and the vasculature: multiple roles in development, repair and protection against disease. Expert Opin Biol Ther 2018; 18:131-139. [DOI: 10.1080/14712598.2018.1459558] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Karina N. Dubé
- BHF Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nicola Smart
- BHF Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Chen L, Xia W, Hou M. Mesenchymal stem cells attenuate doxorubicin‑induced cellular senescence through the VEGF/Notch/TGF‑β signaling pathway in H9c2 cardiomyocytes. Int J Mol Med 2018; 42:674-684. [PMID: 29693137 DOI: 10.3892/ijmm.2018.3635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/05/2018] [Indexed: 11/06/2022] Open
Abstract
The clinical use of doxorubicin (Dox) is limited by its cardiotoxicity. The fundamental changes it induces include interstitial myocardial fibrosis and the appearance of senescent cardiomyocytes. Mesenchymal stem cell (MSC)‑based therapies have also been reported to modulate cellular senescence, and have been used effectively to treat age‑related cardiovascular diseases. In the present study, the Transwell system was used to coculture H9c2 cells with MSCs, and their proliferation and viability were assessed. The expression of senescence‑related genes p53 and p16, and telomere length were measured using reverse transcription‑quantitative polymerase chain reaction analysis, and the Jagged‑1/Notch‑1 signaling pathway was detected using western blot analysis. The results revealed that Dox induced the senescence of H9c2 cells, characterized by a low proliferation rate, poor viability, reduced telomere length and impaired telomerase activity, and by marked increases in the expression of p53 and p16. By contrast, when cocultured with MSCs in the presence of Dox, H9c2 cell proliferation and viability increased, whereas the expression levels of p53 and p16 decreased, and telomere length and telomerase activity increased. The mechanism underlying the antisenescence function of MSCs was clarified, which involved the vascular endothelial growth factor (VEGF)/Jagged‑1/Notch‑1/transforming growth factor‑β1 (TGF‑β1) signaling pathway. It was confirmed that inhibiting VEGF, or silencing Jagged‑1 or Notch‑1 with small interfering RNA, or using recombinant TGF‑β1 eliminated the antisenescence effects of MSCs on the Dox‑treated H9c2 cells. The results revealed that MSCs rescued H9c2 cells from Dox‑induced senescence through the release of VEGF, which activated the Jagged‑1/Notch‑1 signaling pathway, leading to the inhibition of TGF‑β1 release. Therefore, treatment with MSCs may have important therapeutic implications on the attenuation of cardiotoxicity in patients with cancer treated with Dox.
Collapse
Affiliation(s)
- Lingli Chen
- Department of Neurology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
27
|
Bollini S, Silini AR, Banerjee A, Wolbank S, Balbi C, Parolini O. Cardiac Restoration Stemming From the Placenta Tree: Insights From Fetal and Perinatal Cell Biology. Front Physiol 2018; 9:385. [PMID: 29695981 PMCID: PMC5904405 DOI: 10.3389/fphys.2018.00385] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Efficient cardiac repair and ultimate regeneration still represents one of the main challenges of modern medicine. Indeed, cardiovascular disease can derive from independent conditions upsetting heart structure and performance: myocardial ischemia and infarction (MI), pharmacological cardiotoxicity, and congenital heart defects, just to name a few. All these disorders have profound consequences on cardiac tissue, inducing the onset of heart failure over time. Since the cure is currently represented by heart transplantation, which is extremely difficult due to the shortage of donors, much effort is being dedicated to developing innovative therapeutic strategies based on stem cell exploitation. Among the broad scenario of stem/progenitor cell subpopulations, fetal and perinatal sources, namely amniotic fluid and term placenta, have gained interest due to their peculiar regenerative capacity, high self-renewal capability, and ease of collection from clinical waste material. In this review, we will provide the state-of-the-art on fetal perinatal stem cells for cardiac repair and regeneration. We will discuss different pathological conditions and the main therapeutic strategies proposed, including cell transplantation, putative paracrine therapy, reprogramming, and tissue engineering approaches.
Collapse
Affiliation(s)
- Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Antonietta R Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Asmita Banerjee
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Carolina Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy.,Institute of Human Anatomy and Cell Biology, "A. Gemelli" Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
28
|
Loukogeorgakis SP, De Coppi P. Concise Review: Amniotic Fluid Stem Cells: The Known, the Unknown, and Potential Regenerative Medicine Applications. Stem Cells 2018; 35:1663-1673. [PMID: 28009066 DOI: 10.1002/stem.2553] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 09/07/2016] [Accepted: 10/01/2016] [Indexed: 12/19/2022]
Abstract
The amniotic fluid has been identified as an untapped source of cells with broad potential, which possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. CD117(c-Kit)+ cells selected from amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumors, making them ideal candidates for regenerative medicine applications. Moreover, their ability to engraft in injured organs and modulate immune and repair responses of host tissues, suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases. Although significant questions remain regarding the origin, heterogeneous phenotype, and expansion potential of amniotic fluid stem cells, evidence to date supports their potential role as a valuable stem cell source for the field of regenerative medicine. Stem Cells 2017;35:1663-1673.
Collapse
Affiliation(s)
- Stavros P Loukogeorgakis
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
29
|
Pei W, Lu T, Wang K, Ji M, Zhang S, Chen F, Li L, Li X, Guan W. Biological characterization and pluripotent identification of ovine amniotic fluid stem cells. Cytotechnology 2018; 70:1009-1021. [PMID: 29502286 DOI: 10.1007/s10616-017-0115-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
Mesenchymal stem cells derived from amniotic fluid have become one of the most potential stem cell source for cell-based therapy for the reason they can be harvested at low cost and without ethical problems. Here, we obtained amniotic fluid stem cells (AFSCs) from ovine amniotic fluid and studied the expansion capacity, cell markers expression, karyotype, and multilineage differentiation ability. In our work, AFSCs were subcultured to passage 62. The cell markers, CD29, CD44, CD73 and OCT4 which analyzed by RT-PCR were positive; CD44, CD73, CD90, CD105, NANOG, OCT4 analyzed by immunofluorescence and flow cytometry were also positive. The growth curves of different passages were all typically sigmoidal. The different passages cells took on a normal karyotype. In addition, AFSCs were successfully induced to differentiate into adipocytes, osteoblasts and chondrocytes. The results suggested that the AFSCs isolated from ovine maintained normal biological characteristics and their multilineage differentiation potential provides many potential applications in cell-based therapies and tissue engineering.
Collapse
Affiliation(s)
- Wenhua Pei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Tengfei Lu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Kunfu Wang
- College of Wildlife Resources, Northeast Forestry University, Harbin, 150040, China
| | - Meng Ji
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China
| | - Shuang Zhang
- Research Center for Sports Scientific Experiment, Harbin Sport University, Harbin, People's Republic of China
| | - Fenghao Chen
- College of Human Movement Science, Harbin Sport University, Harbin, 150040, China
| | - Lu Li
- College of Animal Science and Technology, Agricultural University of Hebei, Baoding, 071000, China
| | - Xiangchen Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China.
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Yuanmingyuan West road, haidian district, Beijing, 100193, China.
| |
Collapse
|
30
|
Guo Y, Li Z, Shi C, Li J, Yao M, Chen X. Trichostatin A attenuates oxidative stress-mediated myocardial injury through the FoxO3a signaling pathway. Int J Mol Med 2017; 40:999-1008. [PMID: 28849190 PMCID: PMC5593460 DOI: 10.3892/ijmm.2017.3101] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
Trichostatin A (TSA), a histone deacetylase inhibitor, is widely used as an anticancer drug. Recently, TSA has been shown to exert a protective effect on ischemia/reperfusion (I/R) injury; however, the underlying mechanisms remain unclear. Forkhead box O3a (FoxO3a), a unique FoxO family member, has been shown to attenuate myocardial injury by increasing resistance to oxidative stress in mice. The present study aimed to investigate whether TSA exerts its cardioprotective effects through the FoxO3a signaling pathway. For this purpose, healthy male Wistar rats were pre-treated with TSA for 5 days before they were subjected to ligation/relaxation of the left anterior descending branch of the coronary artery and to 30 min of ischemia, followed by 24 h of reperfusion. The activities of creatine kinase (CK), lactate dehydrogenase (LDH), aspartate aminotransferase (AST) and superoxide diamutase (SOD), as well as the malondialdehyde (MDA) levels were examined. The H9c2 rat myocardial cell line was cultured in 10% FBS-containing DMEM for 24 h. The cells were incubated with/without TSA (50 nmol/l) for 1 h and then incubated with/without H2O2 (400 µM) for 2 h. Reactive oxygen species (ROS) and mitochondrial membrane potential (Δψm) were measured by probe staining in the H9c2 cells. The expression of FoxO3a, mitochondrial SOD2 and catalase was quantified by western blot analysis. The levels of H3 and H4 acetylation of the FoxO3a promoter region were examined by chromatin immunoprecipitation assay. TSA significantly reduced the myocardial infarct size and the activities of serum LDH, AST and CK in the rats. TSA also decreased the levels of MDA and increased the activities of SOD in the myocardial tissue of the rats. Consistent with the reduced injury to the TSA-treated rats, TSA significantly reduced the H2O2-induced levels of ROS and increased Δψm. In addition, TSA increased the expression of FoxO3a, SOD2 and catalase, which may be related to increasing the level of H4 acetylation of the FoxO3a promoter region. Our results thus revealed that TSA protected the myocardium from oxidative stress-mediated damage by increasing H4 acetylation of the FoxO3a promoter region, and the expression of FoxO3a, SOD2 and catalase.
Collapse
Affiliation(s)
- Yunhui Guo
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhiping Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Canxia Shi
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Meng Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
31
|
Srivastava M, Ahlawat N, Srivastava A. Amniotic Fluid Stem Cells: A New Era in Regenerative Medicine. J Obstet Gynaecol India 2017; 68:15-19. [PMID: 29391670 DOI: 10.1007/s13224-017-1034-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
Regenerative medicine has become an emerging field which focuses on repair, replacement or regeneration of cells, tissues and the entire organs. The regeneration may occur in patient's own body by using their system as a bioreactor, e.g., cell therapy that involves transplantation of stem cells capable of proliferating, differentiating and replacing damaged host cells. As the field of regenerative medicine advances, and sources of stem cells has been intensified. Though embryonic and adult tissues can be used for isolation of pluripotent stem cells, the amniotic fluid (AF) has been proposed as an alternative source of stem cells for tissue regeneration. AF cells could be banked and used for either allogeneic or autologous transplantation.
Collapse
Affiliation(s)
- Mala Srivastava
- 1Institute of Obstetrics and Gynaecology, Sir Ganga Ram Hospital, New Delhi, India
| | - Neha Ahlawat
- 1Institute of Obstetrics and Gynaecology, Sir Ganga Ram Hospital, New Delhi, India
| | - Ankita Srivastava
- Department of Obstetrics and Gynaecology, Lok Nayak Jay Prakash Hospital, New Delhi, India
| |
Collapse
|
32
|
Abstract
For >4 decades, the holy grail in the treatment of acute myocardial infarction has been the mitigation of lethal injury. Despite promising initial results and decades of investigation by the cardiology research community, the only treatment with proven efficacy is early reperfusion of the occluded coronary artery. The remarkable record of failure has led us and others to wonder if cardioprotection is dead. The path to translation, like the ascent to Everest, is certainly littered with corpses. We do, however, highlight a therapeutic principle that provides a glimmer of hope: cellular postconditioning. Administration of cardiosphere-derived cells after reperfusion limits infarct size measured acutely, while providing long-term structural and functional benefits. The recognition that cell therapy may be cardioprotective, and not just regenerative, merits further exploration before we abandon the pursuit entirely.
Collapse
Affiliation(s)
- David J Lefer
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.).
| | - Eduardo Marbán
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.)
| |
Collapse
|
33
|
Antonucci I, Crowley MG, Stuppia L. Amniotic fluid stem cell models: A tool for filling the gaps in knowledge for human genetic diseases. Brain Circ 2017; 3:167-174. [PMID: 30276320 PMCID: PMC6057697 DOI: 10.4103/bc.bc_23_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/06/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022] Open
Abstract
Induced pluripotent stem (iPS) cells have attracted attention in recent years as a model of human genetic diseases. Starting from the diseased somatic cells isolated from an affected patient, iPS cells can be created and subsequently differentiated into various cell types that can be used to gain a better understanding of the disease at a cellular and molecular level. There are limitations of iPS cell generation, however, due to low efficiency, high costs, and lengthy protocols. The use of amniotic fluid stem cells (AFS) presents a worthy alternative as a stem cell source for modeling of human genetic diseases. Prenatal identification of chromosomal or Mendelian diseases may require the collection of amniotic fluid which is not only useful for the sake of diagnosis but also from this, AFS cells can be isolated and cultured. Since AFS cells show some characteristics of pluripotency, having the capacity to differentiate into various cell types derived from all three germ layers in vitro, they are a well-suited model for investigations regarding alterations in the molecular biology of a cell due to a specific genetic disease. This readily accessible source of stem cells can replace the necessity for generating iPS cells. Here, we expand on the applicability and importance of AFS cells as a model for discovery in the field of human genetic disease research. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Ivana Antonucci
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| | - Marci G Crowley
- Center of Excellence for Aging and Brain Repair, University of South Florida, 12901, USA
| | - Liborio Stuppia
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, Annunzio University, Chieti-Pescara, Italy
| |
Collapse
|
34
|
Cancedda R, Bollini S, Descalzi F, Mastrogiacomo M, Tasso R. Learning from Mother Nature: Innovative Tools to Boost Endogenous Repair of Critical or Difficult-to-Heal Large Tissue Defects. Front Bioeng Biotechnol 2017; 5:28. [PMID: 28503549 PMCID: PMC5408079 DOI: 10.3389/fbioe.2017.00028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022] Open
Abstract
For repair of chronic or difficult-to-heal tissue lesions and defects, major constraints exist to a broad application of cell therapy and tissue engineering approaches, i.e., transplantation of “ex vivo” expanded autologous stem/progenitor cells, alone or associated with carrier biomaterials. To enable a large number of patients to benefit, new strategies should be considered. One of the main goals of contemporary regenerative medicine is to develop new regenerative therapies, inspired from Mother Nature. In all injured tissues, when platelets are activated by tissue contact, their released factors promote innate immune cell migration to the wound site. Platelet-derived factors and factors secreted by migrating immune cells create an inflammatory microenvironment, in turn, causing the activation of angiogenesis and vasculogenesis processes. Eventually, repair or regeneration of the injured tissue occurs via paracrine signals activating, mobilizing or recruiting to the wound site cells with healing potential, such as stem cells, progenitors, or undifferentiated cells derived from the reprogramming of tissue differentiated cells. This review, largely based on our studies, discusses the identification of new tools, inspired by cellular and molecular mechanisms overseeing physiological tissue healing, that could reactivate dormant endogenous regeneration mechanisms lost during evolution and ontogenesis.
Collapse
Affiliation(s)
- Ranieri Cancedda
- Biorigen Srl, Genova, Italy.,Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sveva Bollini
- Department of Experimental Medicine, University of Genova, Genova, Italy
| | | | | | - Roberta Tasso
- IRCCS AOU San Martino-IST National Institute of Cancer Research, Genova, Italy
| |
Collapse
|
35
|
Balbi C, Bollini S. Fetal and perinatal stem cells in cardiac regeneration: Moving forward to the paracrine era. Placenta 2017; 59:96-106. [PMID: 28416208 DOI: 10.1016/j.placenta.2017.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/29/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CD) is a major burden for Western society. Regenerative medicine has provided encouraging results, yet it has not addressed the focal defects causing CD and mainly related to the inefficient repair programme of the heart. In this scenario, stem cells have been broadly investigated and their paracrine effect proposed as a possible working strategy to boost endogenous mechanisms of repair and regeneration from within the cardiac tissue. The scientific community is now focusing on identifying the most effective stem cell secretome, as the whole of bioactive factors and extracellular vesicles secreted by stem cells and endowed with regenerative potential. Indeed, the adult stem cell-paracrine potential for cardiac regeneration have been widely analyzed with positive outcome. Nevertheless, low yield, invasive sampling and controversial self-renewal may limit adult stem cell application. On the contrary, fetal and perinatal stem cells, which can be easily isolated from leftover sample via prenatal screening during gestation or as clinical waste material after birth, can offer an ideal alternative. These broadly multipotent immature progenitors share features with both adult and embryonic stem cells, show high self-renewal, but they are not tumorigenic neither cause any ethical concern. While fetal and perinatal stem cells demonstrated to improve cardiac function when injected in the injured heart, the comprehensive characterization of their secretome for future applications is still at its infancy. In this review, we will discuss the paracrine potential of the fetal and perinatal stem cell secretome to provide cardiac repair and resurge the dormant mechanisms of cardiac regeneration for future therapy.
Collapse
Affiliation(s)
- C Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - S Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy.
| |
Collapse
|
36
|
Balbi C, Piccoli M, Barile L, Papait A, Armirotti A, Principi E, Reverberi D, Pascucci L, Becherini P, Varesio L, Mogni M, Coviello D, Bandiera T, Pozzobon M, Cancedda R, Bollini S. First Characterization of Human Amniotic Fluid Stem Cell Extracellular Vesicles as a Powerful Paracrine Tool Endowed with Regenerative Potential. Stem Cells Transl Med 2017; 6:1340-1355. [PMID: 28271621 PMCID: PMC5442724 DOI: 10.1002/sctm.16-0297] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/08/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022] Open
Abstract
Human amniotic fluid stem cells (hAFS) have shown a distinct secretory profile and significant regenerative potential in several preclinical models of disease. Nevertheless, little is known about the detailed characterization of their secretome. Herein we show for the first time that hAFS actively release extracellular vesicles (EV) endowed with significant paracrine potential and regenerative effect. c‐KIT+ hAFS were isolated from leftover samples of amniotic fluid from prenatal screening and stimulated to enhance EV release (24 hours 20% O2 versus 1% O2 preconditioning). The capacity of the c‐KIT+ hAFS‐derived EV (hAFS‐EV) to induce proliferation, survival, immunomodulation, and angiogenesis were investigated in vitro and in vivo. The hAFS‐EV regenerative potential was also assessed in a model of skeletal muscle atrophy (HSA‐Cre, SmnF7/F7 mice), in which mouse AFS transplantation was previously shown to enhance muscle strength and survival. hAFS secreted EV ranged from 50 up to 1,000 nm in size. In vitro analysis defined their role as biological mediators of regenerative, paracrine effects while their modulatory role in decreasing skeletal muscle inflammation in vivo was shown for the first time. Hypoxic preconditioning significantly induced the enrichment of exosomes endowed with regenerative microRNAs within the hAFS‐EV. In conclusion, this is the first study showing that c‐KIT+ hAFS dynamically release EV endowed with remarkable paracrine potential, thus representing an appealing tool for future regenerative therapy. Stem Cells Translational Medicine2017;6:1340–1355
Collapse
Affiliation(s)
- Carolina Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Lucio Barile
- Laboratory of Molecular and Cellular Cardiology, CardioCentro Ticino Foundation_CCT, Lugano Switzerland
| | - Andrea Papait
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Andrea Armirotti
- Drug Discovery and Development Department, IIT-Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Elisa Principi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS AOU San Martino - IST National Institute for Cancer Research, Genova, Italy
| | - Luisa Pascucci
- Veterinary Medicine Department, University of Perugia, Perugia, Italy
| | - Pamela Becherini
- Molecular Biology Laboratory, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Molecular Biology Laboratory, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Massimo Mogni
- Human Genetics Laboratory, E.O. Ospedali Galliera, Genova, Italy
| | | | - Tiziano Bandiera
- Drug Discovery and Development Department, IIT-Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Ranieri Cancedda
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
37
|
Sokol J, Lippert T, Borlongan CV, Stuppia L. Translating amniotic fluid-derived stem cells for transplantation in stroke. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0055-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
38
|
Gholizadeh-Ghaleh Aziz S, Fathi E, Rahmati-Yamchi M, Akbarzadeh A, Fardyazar Z, Pashaiasl M. An update clinical application of amniotic fluid-derived stem cells (AFSCs) in cancer cell therapy and tissue engineering. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:765-774. [DOI: 10.1080/21691401.2016.1216857] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shiva Gholizadeh-Ghaleh Aziz
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Iran
| | | | - Abolfazl Akbarzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Fardyazar
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, School of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Maryam Pashaiasl
- Women?s Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Jiang G, Herron TJ, Di Bernardo J, Walker KA, O'Shea KS, Kunisaki SM. Human Cardiomyocytes Prior to Birth by Integration-Free Reprogramming of Amniotic Fluid Cells. Stem Cells Transl Med 2016; 5:1595-1606. [PMID: 27465073 DOI: 10.5966/sctm.2016-0016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/13/2016] [Indexed: 01/26/2023] Open
Abstract
: The establishment of an abundant source of autologous cardiac progenitor cells would represent a major advance toward eventual clinical translation of regenerative medicine strategies in children with prenatally diagnosed congenital heart disease. In support of this concept, we sought to examine whether functional, transgene-free human cardiomyocytes (CMs) with potential for patient-specific and autologous applications could be reliably generated following routine amniocentesis. Under institutional review board approval, amniotic fluid specimens (8-10 ml) at 20 weeks gestation were expanded and reprogrammed toward pluripotency using nonintegrating Sendai virus (SeV) expressing OCT4, SOX2, cMYC, and KLF4. Following exposure of these induced pluripotent stem cells to cardiogenic differentiation conditions, spontaneously beating amniotic fluid-derived cardiomyocytes (AF-CMs) were successfully generated with high efficiency. After 6 weeks, quantitative gene expression revealed a mixed population of differentiated atrial, ventricular, and nodal AF-CMs, as demonstrated by upregulation of multiple cardiac markers, including MYH6, MYL7, TNNT2, TTN, and HCN4, which were comparable to levels expressed by neonatal dermal fibroblast-derived CM controls. AF-CMs had a normal karyotype and demonstrated loss of NANOG, OCT4, and the SeV transgene. Functional characterization of SIRPA+ AF-CMs showed a higher spontaneous beat frequency in comparison with dermal fibroblast controls but revealed normal calcium transients and appropriate chronotropic responses after β-adrenergic agonist stimulation. Taken together, these data suggest that somatic cells present within human amniotic fluid can be used to generate a highly scalable source of functional, transgene-free, autologous CMs before a child is born. This approach may be ideally suited for patients with prenatally diagnosed cardiac anomalies. SIGNIFICANCE This study presents transgene-free human amniotic fluid-derived cardiomyocytes (AF-CMs) for potential therapy in tissue engineering and regenerative medicine applications. Using 8-10 ml of amniotic fluid harvested at 20 weeks gestation from normal pregnancies, a mixed population of atrial, ventricular, and nodal AF-CMs were reliably generated after Sendai virus reprogramming toward pluripotency. Functional characterization of purified populations of beating AF-CMs revealed normal calcium transients and appropriate chronotropic responses after β-adrenergic agonist stimulation in comparison with dermal fibroblast controls. Because AF-CMs can be generated in fewer than 16 weeks, this approach may be ideally suited for eventual clinical translation at birth in children with prenatally diagnosed cardiac anomalies.
Collapse
Affiliation(s)
- Guihua Jiang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Todd J Herron
- Department of Internal Medicine, Cardiovascular Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Julie Di Bernardo
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kendal A Walker
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - K Sue O'Shea
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shaun M Kunisaki
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Pluripotent Stem Cell Laboratory, University of Michigan Medical School, Ann Arbor, Michigan, USA
- C.S. Mott Children's Hospital and Von Voigtlander Women's Hospital, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
The human amniotic fluid stem cell secretome effectively counteracts doxorubicin-induced cardiotoxicity. Sci Rep 2016; 6:29994. [PMID: 27444332 PMCID: PMC4956770 DOI: 10.1038/srep29994] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/27/2016] [Indexed: 01/16/2023] Open
Abstract
The anthracycline doxorubicin (Dox) is widely used in oncology, but it may cause a cardiomyopathy with bleak prognosis that cannot be effectively prevented. The secretome of human amniotic fluid-derived stem cells (hAFS) has previously been demonstrated to significantly reduce ischemic cardiac damage. Here it is shown that, following hypoxic preconditioning, hAFS conditioned medium (hAFS-CM) antagonizes senescence and apoptosis of cardiomyocytes and cardiac progenitor cells, two major features of Dox cardiotoxicity. Mechanistic studies with mouse neonatal ventricular cardiomyocytes (mNVCM) reveal that hAFS-CM inhibition of Dox-elicited senescence and apoptosis is associated with decreased DNA damage, nuclear translocation of NF-kB, and upregulation of the NF-kB controlled genes, Il6 and Cxcl1, promoting mNVCM survival. Furthermore, hAFS-CM induces expression of the efflux transporter, Abcb1b, and Dox extrusion from mNVCM. The PI3K/Akt signaling cascade, upstream of NF-kB, is potently activated by hAFS-CM and pre-treatment with a PI3K inhibitor abrogates NF-kB accumulation into the nucleus, modulation of Il6, Cxcl1 and Abcb1b, and prevention of Dox-initiated senescence and apoptosis in response to hAFS-CM. These results support the concept that hAFS are a valuable source of cardioprotective factors and lay the foundations for the development of a stem cell-based paracrine treatment of chemotherapy-related cardiotoxicity.
Collapse
|
41
|
Skardal A, Murphy SV, Crowell K, Mack D, Atala A, Soker S. A tunable hydrogel system for long-term release of cell-secreted cytokines and bioprinted in situ wound cell delivery. J Biomed Mater Res B Appl Biomater 2016; 105:1986-2000. [PMID: 27351939 DOI: 10.1002/jbm.b.33736] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
Abstract
For many cellular therapies being evaluated in preclinical and clinical trials, the mechanisms behind their therapeutic effects appear to be the secretion of growth factors and cytokines, also known as paracrine activity. Often, delivered cells are transient, and half-lives of the growth factors that they secrete are short, limiting their long-term effectiveness. The goal of this study was to optimize a hydrogel system capable of in situ cell delivery that could sequester and release growth factors secreted from those cells after the cells were no longer present. Here, we demonstrate the use of a fast photocross-linkable heparin-conjugated hyaluronic acid (HA-HP) hydrogel as a cell delivery vehicle for sustained growth factor release, which extends paracrine activity. The hydrogel could be modulated through cross-linking geometries and heparinization to support sustained release proteins and heparin-binding growth factors. To test the hydrogel in vivo, we used it to deliver amniotic fluid-derived stem (AFS) cells, which are known to secrete cytokines and growth factors, in full thickness skin wounds in a nu/nu murine model. Despite transience of the AFS cells in vivo, the HA-HP hydrogel with AFS cells improved wound closure and reepithelialization and increased vascularization and production of extracellular matrix in vivo. These results suggest that HA-HP hydrogel has the potential to prolong the paracrine activity of cells, thereby increasing their therapeutic effectiveness in wound healing. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1986-2000, 2017.
Collapse
Affiliation(s)
- Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Kathryn Crowell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - David Mack
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Bioengineering, University of Washington, Seattle, Washington
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| |
Collapse
|
42
|
Amniotic Fluid Stem Cells: A Novel Source for Modeling of Human Genetic Diseases. Int J Mol Sci 2016; 17:ijms17040607. [PMID: 27110774 PMCID: PMC4849058 DOI: 10.3390/ijms17040607] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/01/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
In recent years, great interest has been devoted to the use of Induced Pluripotent Stem cells (iPS) for modeling of human genetic diseases, due to the possibility of reprogramming somatic cells of affected patients into pluripotent cells, enabling differentiation into several cell types, and allowing investigations into the molecular mechanisms of the disease. However, the protocol of iPS generation still suffers from technical limitations, showing low efficiency, being expensive and time consuming. Amniotic Fluid Stem cells (AFS) represent a potential alternative novel source of stem cells for modeling of human genetic diseases. In fact, by means of prenatal diagnosis, a number of fetuses affected by chromosomal or Mendelian diseases can be identified, and the amniotic fluid collected for genetic testing can be used, after diagnosis, for the isolation, culture and differentiation of AFS cells. This can provide a useful stem cell model for the investigation of the molecular basis of the diagnosed disease without the necessity of producing iPS, since AFS cells show some features of pluripotency and are able to differentiate in cells derived from all three germ layers “in vitro”. In this article, we describe the potential benefits provided by using AFS cells in the modeling of human genetic diseases.
Collapse
|
43
|
Dziadosz M, Basch RS, Young BK. Human amniotic fluid: a source of stem cells for possible therapeutic use. Am J Obstet Gynecol 2016; 214:321-7. [PMID: 26767797 DOI: 10.1016/j.ajog.2015.12.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/22/2015] [Accepted: 12/31/2015] [Indexed: 12/23/2022]
Abstract
Stem cells are undifferentiated cells with the capacity for differentiation. Amniotic fluid cells have emerged only recently as a possible source of stem cells for clinical purposes. There are no ethical or sampling constraints for the use of amniocentesis as a standard clinical procedure for obtaining an abundant supply of amniotic fluid cells. Amniotic fluid cells of human origin proliferate rapidly and are multipotent with the potential for expansion in vitro to multiple cell lines. Tissue engineering technologies that use amniotic fluid cells are being explored. Amniotic fluid cells may be of clinical benefit for fetal therapies, degenerative disease, and regenerative medicine applications. We present a comprehensive review of the evolution of human amniotic fluid cells as a possible modality for therapeutic use.
Collapse
Affiliation(s)
- Margaret Dziadosz
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY
| | - Ross S Basch
- Department of Pathology, New York University Langone Medical Center, New York, NY
| | - Bruce K Young
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, New York University Langone Medical Center, New York, NY.
| |
Collapse
|
44
|
Tian Y, Tao L, Zhao S, Tai D, Liu D, Liu P. Isolation and morphological characterization of ovine amniotic fluid mesenchymal stem cells. Exp Anim 2015; 65:125-34. [PMID: 26616638 PMCID: PMC4873481 DOI: 10.1538/expanim.15-0031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the most promising cell populations for tissue engineering and regenerative medicine. Of utmost importance to MSC research is identification of MSC sources that are easily obtainable and stable. Several studies have shown that MSCs can be isolated from amniotic fluid. The sheep is one of the main types of farm animal, and it has many biophysical and biochemical similarities to humans. Here, we obtained MSCs from ovine amniotic fluid and determined the expansion capacity, surface and intracellular marker expression, karyotype, and multilineage differentiation ability of these ovine amniotic fluid mesenchymal stem cells (oAF-MSCs). Moreover, expression levels of differentiation markers were measured using reverse transcription-qPCR (RT-qPCR). Our phenotypic analysis shows that the isolated oAF-MSCs are indeed MSCs.
Collapse
Affiliation(s)
- Yunyun Tian
- College of Life Sciences, Inner Mongolia University, Inner Mongolia, Hohhot 010021, P.R.China
| | | | | | | | | | | |
Collapse
|
45
|
Murphy SV, Hale A, Reid T, Olson J, Kidiyoor A, Tan J, Zhou Z, Jackson J, Atala A. Use of trimetasphere metallofullerene MRI contrast agent for the non-invasive longitudinal tracking of stem cells in the lung. Methods 2015; 99:99-111. [PMID: 26546729 DOI: 10.1016/j.ymeth.2015.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Revised: 10/18/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
Magnetic Resonance Imaging (MRI) is a commonly used, non-invasive imaging technique that provides visualization of soft tissues with high spatial resolution. In both a research and clinical setting, the major challenge has been identifying a non-invasive and safe method for longitudinal tracking of delivered cells in vivo. The labeling and tracking of contrast agent labeled cells using MRI has the potential to fulfill this need. Contrast agents are often used to enhance the image contrast between the tissue of interest and surrounding tissues with MRI. The most commonly used MRI contrast agents contain Gd(III) ions. However, Gd(III) ions are highly toxic in their ionic form, as they tend to accumulate in the liver, spleen, kidney and bones and block calcium channels. Endohedral metallofullerenes such as trimetallic nitride endohedral metallofullerenes (Trimetasphere®) are one unique class of fullerene molecules where a Gd3N cluster is encapsulated inside a C80 carbon cage referred to as Gd3N@C80. These endohedral metallofullerenes have several advantages over small chelated Gd(III) complexes such as increased stability of the Gd(III) ion, minimal toxic effects, high solubility in water and high proton relativity. In this study, we describe the evaluation of gadolinium-based Trimetasphere® positive contrast agent for the in vitro labeling and in vivo tracking of human amniotic fluid-derived stem cells within lung tissue. In addition, we conducted a 'proof-of-concept' experiment demonstrating that this methodology can be used to track the homing of stem cells to injured lung tissue and provide longitudinal analysis of cell localization over an extended time course.
Collapse
Affiliation(s)
- Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Austin Hale
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Tanya Reid
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - John Olson
- Center for Biomolecular Imaging, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| | - Amritha Kidiyoor
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Josh Tan
- Center for Biomolecular Imaging, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| | - Zhiguo Zhou
- Luna nanoWorks Division, Luna Innovations, Incorporated, Danville, VA 24541, USA.
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
46
|
Schiavo AA, Franzin C, Albiero M, Piccoli M, Spiro G, Bertin E, Urbani L, Visentin S, Cosmi E, Fadini GP, De Coppi P, Pozzobon M. Endothelial properties of third-trimester amniotic fluid stem cells cultured in hypoxia. Stem Cell Res Ther 2015; 6:209. [PMID: 26519360 PMCID: PMC4628318 DOI: 10.1186/s13287-015-0204-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/02/2015] [Accepted: 10/15/2015] [Indexed: 12/22/2022] Open
Abstract
Introduction Endothelial dysfunction is found in different pathologies such as diabetes and renal and heart diseases, representing one of the major health problems. The reduced vasodilation of impaired endothelium starts a prothrombotic state associated with irregular blood flow. We aimed to explore the potential of amniotic fluid stem (AFS) cells as a source for regenerative medicine in this field; for the first time, we focused on third-trimester amniotic fluid AFS cells and compared them with the already-described AFS cells from the second trimester. Methods Cells from the two trimesters were cultured, selected and expanded in normoxia (20 % oxygen) and hypoxia (5 % oxygen). Cells were analysed to compare markers, proliferation rate and differentiation abilities. Endothelial potential was assessed not only in vitro—Matrigel tube formation assay, acetylated human low-density lipoprotein (AcLDL) uptake—but also in vivo (Matrigel plug with cell injection and two animal models). Specifically, for the latter, we used established protocols to assess the involvement of AFS cells in two different mouse models of endothelial dysfunction: (1) a chronic ischemia model with local injection of cells and (2) an electric carotid damage where cells were systemically injected. Results We isolated and expanded AFS cells from third-trimester amniotic fluid samples by using CD117 as a selection marker. Hypoxia enhanced the proliferation rate, the surface protein pattern was conserved between the trimesters and comparable differentiation was achieved after culture in both normoxia and hypoxia. Notably, the expression of early endothelial transcription factors and AngiomiRs was detected before and after induction. When in vivo, AFS cells from both trimesters expanded in hypoxia were able to rescue the surface blood flow when locally injected in mice after chronic ischemia damage, and importantly AFS cells at term of gestation possessed enhanced ability to fix carotid artery electric damage compared with AFS cells from the second trimester. Conclusions To the best of our knowledge, this is the first research work that fully characterizes AFS cells from the third trimester for regenerative medicine purposes. The results highlight how AFS cells, in particular at term of gestation and cultured in hypoxia, can be considered a promising source of stem cells possessing significant endothelial regenerative potential. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0204-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Alex Schiavo
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy. .,Department of Woman and Children Health, University of Padova, via Giustinani 2, 35100, Padova, Italy.
| | - Chiara Franzin
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Mattia Albiero
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy. .,Medicine Department (DIMED), University of Padova, via Giustiniani 2, 35100, Padova, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Giovanna Spiro
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy. .,Medicine Department (DIMED), University of Padova, via Giustiniani 2, 35100, Padova, Italy.
| | - Enrica Bertin
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| | - Luca Urbani
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy. .,Stem Cells and Regenerative Medicine Section, Developmental biology and Cancer Program, Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK.
| | - Silvia Visentin
- Department of Woman and Children Health, University of Padova, via Giustinani 2, 35100, Padova, Italy.
| | - Erich Cosmi
- Department of Woman and Children Health, University of Padova, via Giustinani 2, 35100, Padova, Italy.
| | - Gian Paolo Fadini
- Venetian Institute of Molecular Medicine, via Orus 2, 35129, Padova, Italy. .,Medicine Department (DIMED), University of Padova, via Giustiniani 2, 35100, Padova, Italy.
| | - Paolo De Coppi
- Stem Cells and Regenerative Medicine Section, Developmental biology and Cancer Program, Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK.
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research Città della Speranza, Corso Stati Uniti 4, 35127, Padova, Italy.
| |
Collapse
|
47
|
Stem cells from amniotic fluid--Potential for regenerative medicine. Best Pract Res Clin Obstet Gynaecol 2015; 31:45-57. [PMID: 26542929 DOI: 10.1016/j.bpobgyn.2015.08.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Abstract
Regenerative medicine has recently been established as an emerging field focussing on repair, replacement or regeneration of cells, tissues and whole organs. The significant recent advances in the field have intensified the search for novel sources of stem cells with potential for therapy. Recently, researchers have identified the amniotic fluid as an untapped source of stem cells that are multipotent, possess immunomodulatory properties and do not have the ethical and legal limitations of embryonic stem cells. Stem cells from the amniotic fluid have been shown to differentiate into cell lineages representing all three embryonic germ layers without generating tumours, which make them an ideal candidate for tissue engineering applications. In addition, their ability to engraft in injured organs and modulate immune and repair responses of host tissues suggest that transplantation of such cells may be useful for the treatment of various degenerative and inflammatory diseases affecting major tissues/organs. This review summarises the evidence on amniotic fluid cells over the past 15 years and explores the potential therapeutic applications of amniotic fluid stem cells and amniotic fluid mesenchymal stem cells.
Collapse
|
48
|
Elias M, Hoover J, Nguyen H, Reyes S, Lawton C, Borlongan CV. Stroke therapy: the potential of amniotic fluid-derived stem cells. FUTURE NEUROLOGY 2015; 10:321-326. [PMID: 26401122 DOI: 10.2217/fnl.15.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maya Elias
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Jaclyn Hoover
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Hung Nguyen
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Stephanny Reyes
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Christopher Lawton
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA
| | - Cesar V Borlongan
- Department of Neurosurgery & Brain Repair, University of South Florida College of Medicine, 12901 Bruce B Downs Blvd, FL 33612, USA ; Center of Excellence for Aging & Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
49
|
Peng SY, Chou CJ, Cheng PJ, Tseng TY, Cheng WTK, Shaw SWS, Wu SC. Intramuscular Transplantation of Pig Amniotic Fluid-Derived Progenitor Cells Has Therapeutic Potential in a Mouse Model of Myocardial Infarction. Cell Transplant 2015; 24:1003-12. [DOI: 10.3727/096368914x680109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Acute myocardial infarction (MI) is a fatal event that causes a large number of deaths worldwide. MI results in pathological remodeling and decreased cardiac function, which could lead to heart failure and fatal arrhythmia. Cell therapy is a potential strategy to repair the damage through enhanced angiogenesis or by modulation of the inflammatory process via paracrine signaling. Amniotic fluid-derived progenitor cells (AFPCs) have been reported to differentiate into several lineages and can be used without ethical concerns or risk of teratoma formation. Since pigs are anatomically, physiologically, and genetically similar to humans, and pregnant pigs can be an abundant source of AFPCs, we used porcine AFPCs (pAFPCs) as our target cells. Intramyocardial injection of AFPCs has been shown to cure MI in animal models. However, intramuscular transplantation of cells has not been extensively investigated. In this study, we investigated the therapeutic potential of intramuscular injection of pAFPCs on acute MI. MI mice were divided into 1) PBS control, 2) medium cell dose (1 × 106 cells per leg; cell-M), and 3) high cell dose (4 × 106 cells per leg; cell-H) groups. Cells or PBS were directly injected into the hamstring muscle 20 min after MI surgery. Four weeks after MI surgery, the cell-M and cell-H groups exhibited significantly better ejection fraction, significantly greater wall thickness, smaller infarct scar sizes, and lower LV expansion index compared to the PBS group. Using in vivo imaging, we showed that the hamstring muscles from animals in the cell-M and cell-H groups had RFP-positive signals. In summary, intramuscular injection of porcine AFPCs reduced scar size, reduced pathological remodeling, and preserved heart function after MI.
Collapse
Affiliation(s)
- Shao-Yu Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chih-Jen Chou
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Po-Jen Cheng
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - Tse-Yang Tseng
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Winston Teng-Kui Cheng
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan
| | - S. W. Steven Shaw
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, College of Medicine, Taoyuan, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
- Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
50
|
Pipino C, Pandolfi A. Osteogenic differentiation of amniotic fluid mesenchymal stromal cells and their bone regeneration potential. World J Stem Cells 2015; 7:681-690. [PMID: 26029340 PMCID: PMC4444609 DOI: 10.4252/wjsc.v7.i4.681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/16/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
Abstract
In orthopedics, tissue engineering approach using stem cells is a valid line of treatment for patients with bone defects. In this context, mesenchymal stromal cells of various origins have been extensively studied and continue to be a matter of debate. Although mesenchymal stromal cells from bone marrow are already clinically applied, recent evidence suggests that one may use mesenchymal stromal cells from extra-embryonic tissues, such as amniotic fluid, as an innovative and advantageous resource for bone regeneration. The use of cells from amniotic fluid does not raise ethical problems and provides a sufficient number of cells without invasive procedures. Furthermore, they do not develop into teratomas when transplanted, a consequence observed with pluripotent stem cells. In addition, their multipotent differentiation ability, low immunogenicity, and anti-inflammatory properties make them ideal candidates for bone regenerative medicine. We here present an overview of the features of amniotic fluid mesenchymal stromal cells and their potential in the osteogenic differentiation process. We have examined the papers actually available on this regard, with particular interest in the strategies applied to improve in vitro osteogenesis. Importantly, a detailed understanding of the behavior of amniotic fluid mesenchymal stromal cells and their osteogenic ability is desirable considering a feasible application in bone regenerative medicine.
Collapse
|