151
|
Liu J, Zhang L, Liu M. Mechanisms supporting potential use of bone marrow-derived mesenchymal stem cells in psychocardiology. Am J Transl Res 2019; 11:6717-6738. [PMID: 31814884 PMCID: PMC6895510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Despite great efforts made in recent years, globally cardiovascular disease (CVD) remains the most common and devastating disease. Pharmacological, interventional and surgical treatments have proved to be only partly satisfactory for the majority of patients. A major underlying cause of poor prognosis is a high comorbidity rate between CVD and mental illness, which calls for the approaches of psychocardiology. As psychiatric disorders and CVD can influence each other bidirectionally, it is necessary to develop novel therapies targeting both systems simultaneously. Therefore, innovative stem cell (SC) therapy has become the most promising treatment strategy in psychocardiology. Bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs), among all different types of SCs, have drawn the most attention due to unique advantages in terms of ethical considerations, low immunogenicity and simplicity of preparation. In this review, we survey recent publications and clinical trials to summarize the knowledge and progress gained so far. Moreover, we discuss the feasibility of the clinical application of BM-MSCs in the area of psychocardiology.
Collapse
Affiliation(s)
- Jianyang Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Lijun Zhang
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| | - Meiyan Liu
- Department of Cardiology, Beijing Anzhen Hospital Affiliated to Capital Medical University Beijing 100029, China
| |
Collapse
|
152
|
In Vivo MRI Tracking of Mesenchymal Stromal Cells Labeled with Ultrasmall Paramagnetic Iron Oxide Particles after Intramyocardial Transplantation in Patients with Chronic Ischemic Heart Disease. Stem Cells Int 2019; 2019:2754927. [PMID: 31814830 PMCID: PMC6877937 DOI: 10.1155/2019/2754927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 09/28/2019] [Indexed: 01/17/2023] Open
Abstract
Background While regenerative stem cell therapy for ischemic heart disease has moved into phase 3 studies, little is still known about retention and migration of cell posttransplantation. In human studies, the ability to track transplanted cells has been limited to labeling with radioisotopes and tracking using nuclear imaging. This method is limited by low resolution and short half-lives of available radioisotopes. Longitudinal tracking using magnetic resonance imaging (MRI) of myocardial injected cells labeled with iron oxide nanoparticles has shown promising results in numerous preclinical studies but has yet to be evaluated in human studies. We aimed to evaluate MRI tracking of mesenchymal stromal cells (MSCs) labeled with ultrasmall paramagnetic iron oxide (USPIO) nanoparticles after intramyocardial transplantation in patients with ischemic heart disease (IHD). Methods Five no-option patients with chronic symptomatic IHD underwent NOGA-guided intramyocardial transplantation of USPIO-labeled MSCs. Serial MRI scans were performed to track labeled cells both visually and using semiautomated T2∗ relaxation time analysis. For safety, we followed symptoms, quality of life, and myocardial function for 6 months. Results USPIO-labeled MSCs were tracked for up to 14 days after transplantation at injection sites both visually and using semiautomated regional T2∗ relaxation time analysis. Labeling of MSCs did not impair long-term safety of treatment. Conclusion This was a first-in-man clinical experience aimed at evaluating the utility of MRI tracking of USPIO-labeled bone marrow-derived autologous MSCs after intramyocardial injection in patients with chronic IHD. The treatment was safe, and cells were detectable at injection sites up to 14 days after transplantation. Further studies are needed to clarify if MSCs migrate out of the injection area into other areas of the myocardium or if injected cells are washed out into the peripheral circulation. The trial is registered with ClinicalTrials.gov NCT03651791.
Collapse
|
153
|
Kot M, Baj-Krzyworzeka M, Szatanek R, Musiał-Wysocka A, Suda-Szczurek M, Majka M. The Importance of HLA Assessment in "Off-the-Shelf" Allogeneic Mesenchymal Stem Cells Based-Therapies. Int J Mol Sci 2019; 20:E5680. [PMID: 31766164 PMCID: PMC6888380 DOI: 10.3390/ijms20225680] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
The need for more effective therapies of chronic and acute diseases has led to the attempts of developing more adequate and less invasive treatment methods. Regenerative medicine relies mainly on the therapeutic potential of stem cells. Mesenchymal stem cells (MSCs), due to their immunosuppressive properties and tissue repair abilities, seem to be an ideal tool for cell-based therapies. Taking into account all available sources of MSCs, perinatal tissues become an attractive source of allogeneic MSCs. The allogeneic MSCs provide "off-the-shelf" cellular therapy, however, their allogenicity may be viewed as a limitation for their use. Moreover, some evidence suggests that MSCs are not as immune-privileged as it was previously reported. Therefore, understanding their interactions with the recipient's immune system is crucial for their successful clinical application. In this review, we discuss both autologous and allogeneic application of MSCs, focusing on current approaches to allogeneic MSCs therapies, with a particular interest in the role of human leukocyte antigens (HLA) and HLA-matching in allogeneic MSCs transplantation. Importantly, the evidence from the currently completed and ongoing clinical trials demonstrates that allogeneic MSCs transplantation is safe and seems to cause no major side-effects to the patient. These findings strongly support the case for MSCs efficacy in treatment of a variety of diseases and their use as an "off-the-shelf" medical product.
Collapse
Affiliation(s)
- Marta Kot
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Rafał Szatanek
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Aleksandra Musiał-Wysocka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Magdalena Suda-Szczurek
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Marcin Majka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| |
Collapse
|
154
|
Haenel A, Ghosn M, Karimi T, Vykoukal J, Shah D, Valderrabano M, Schulz DG, Raizner A, Schmitz C, Alt EU. Unmodified autologous stem cells at point of care for chronic myocardial infarction. World J Stem Cells 2019; 11:831-858. [PMID: 31692971 PMCID: PMC6828597 DOI: 10.4252/wjsc.v11.i10.831] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/03/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Numerous studies investigated cell-based therapies for myocardial infarction (MI). The conflicting results of these studies have established the need for developing innovative approaches for applying cell-based therapy for MI. Experimental studies on animal models demonstrated the potential of fresh, uncultured, unmodified, autologous adipose-derived regenerative cells (UA-ADRCs) for treating acute MI. In contrast, studies on the treatment of chronic MI (CMI; > 4 wk post-MI) with UA-ADRCs have not been published so far. Among several methods for delivering cells to the myocardium, retrograde delivery into a temporarily blocked coronary vein has recently been demonstrated as an effective option.
AIM To test the hypothesis that in experimentally-induced chronic myocardial infarction (CMI; > 4 wk post-MI) in pigs, retrograde delivery of fresh, uncultured, unmodified, autologous adipose-derived regenerative cells (UA-ADRCs) into a temporarily blocked coronary vein improves cardiac function and structure.
METHODS The left anterior descending (LAD) coronary artery of pigs was blocked for 180 min at time point T0. Then, either 18 × 106 UA-ADRCs prepared at “point of care” or saline as control were retrogradely delivered via an over-the-wire balloon catheter placed in the temporarily blocked LAD vein 4 wk after T0 (T1). Effects of cells or saline were assessed by cardiac magnetic resonance (CMR) imaging, late gadolinium enhancement CMR imaging, and post mortem histologic analysis 10 wk after T0 (T2).
RESULTS Unlike the delivery of saline, delivery of UA-ADRCs demonstrated statistically significant improvements in cardiac function and structure at T2 compared to T1 (all values given as mean ± SE): Increased mean LVEF (UA-ADRCs group: 34.3% ± 2.9% at T1 vs 40.4 ± 2.6% at T2, P = 0.037; saline group: 37.8% ± 2.6% at T1 vs 36.2% ± 2.4% at T2, P > 0.999), increased mean cardiac output (UA-ADRCs group: 2.7 ± 0.2 L/min at T1 vs 3.8 ± 0.2 L/min at T2, P = 0.002; saline group: 3.4 ± 0.3 L/min at T1 vs 3.6 ± 0.3 L/min at T2, P = 0.798), increased mean mass of the left ventricle (UA-ADRCs group: 55.3 ± 5.0 g at T1 vs 71.3 ± 4.5 g at T2, P < 0.001; saline group: 63.2 ± 3.4 g at T1 vs 68.4 ± 4.0 g at T2, P = 0.321) and reduced mean relative amount of scar volume of the left ventricular wall (UA-ADRCs group: 20.9% ± 2.3% at T1 vs 16.6% ± 1.2% at T2, P = 0.042; saline group: 17.6% ± 1.4% at T1 vs 22.7% ± 1.8% at T2, P = 0.022).
CONCLUSION Retrograde cell delivery of UA-ADRCs in a porcine model for the study of CMI significantly improved myocardial function, increased myocardial mass and reduced the formation of scar tissue.
Collapse
Affiliation(s)
- Alexander Haenel
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, United States
- The Methodist Hospital Research Institute, Houston, TX 77030, United States
- Department of Radiology and Nuclear Medicine, University Hospital Schleswig-Holstein, Lübeck D-23562, Germany
| | - Mohamad Ghosn
- Houston Methodist DeBakey Heart and Vascular Center, Houston, TX 77030, United States
| | - Tahereh Karimi
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, United States
| | - Jody Vykoukal
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, United States
| | - Dipan Shah
- Houston Methodist DeBakey Heart and Vascular Center, Houston, TX 77030, United States
| | - Miguel Valderrabano
- Houston Methodist DeBakey Heart and Vascular Center, Houston, TX 77030, United States
| | - Daryl G Schulz
- The Methodist Hospital Research Institute, Houston, TX 77030, United States
| | - Albert Raizner
- Houston Methodist DeBakey Heart and Vascular Center, Houston, TX 77030, United States
| | - Christoph Schmitz
- Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich D-80336, Germany
| | - Eckhard U Alt
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, United States
- The Methodist Hospital Research Institute, Houston, TX 77030, United States
- Isar Klinikum Munich, Munich D-80331, Germany
| |
Collapse
|
155
|
Rieger AC, Myerburg RJ, Florea V, Tompkins BA, Natsumeda M, Premer C, Khan A, Schulman IH, Vidro-Casiano M, DiFede DL, Heldman AW, Mitrani R, Hare JM. Genetic determinants of responsiveness to mesenchymal stem cell injections in non-ischemic dilated cardiomyopathy. EBioMedicine 2019; 48:377-385. [PMID: 31648988 PMCID: PMC6838383 DOI: 10.1016/j.ebiom.2019.09.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Non-ischemic dilated cardiomyopathy (NIDCM) responds variably to intramyocardial injection of mesenchymal stem cells (MSCs). We hypothesized that NIDCM genotype may influence responsiveness to MSC therapy and performed genotyping on all patients in the POSEIDON-DCM trial. METHODS POSEIDON-DCM patients (n = 34) underwent genetic sequence analysis and deletion/duplication testing. The results were classified as positive for pathological variants (PV+; n = 8), negative for any variants (V-; n = 6), or as variants of uncertain significance (VUS; n = 20). All outcomes of therapy were analysed for each category of genetic results. FINDINGS The 3 groups were indistinguishable at baseline with regard to ejection fraction (EF), demographics, medication use, or functional parameters. V- patients had an increase in EF at 12 months: +13.6% (IQR = +7.8%; +20.5%; p = 0.002), compared with VUS (+6.5%; IQR = +0.9%, +11.1%; p = 0.005) and PV+(-5.9%; IQR = -12.7%, +1.0; p = 0.2; p = 0.01 between groups). Six-minute walk distance improved in V- patients, but not in VUS and PV+. V- patients improved MLHFQ, compared to the other 2 groups, which did not improve over time. EPCCFUs increased by 9.7 ± 1.9 in V- (p = 0.009) compared to VUS and PV+ patients. V- patients had one-year survival (100%) compared with VUS (85%) and PV+ (40%; p = 0.015 log-rank). Similarly, MACE rates were lower in V- (0%) than PV+ (61.9%) or VUS (42.2%; p = 0.021 log-rank). INTERPRETATION Our findings support the concept that the genetic profile of NIDCM patients plays a role in responsiveness to MSC therapy, with V- patients more likely to benefit and the converse for PV+. This observation emphasizes the need for further genetic studies, because of important implications for the management of NIDCM syndromes.
Collapse
Affiliation(s)
- Angela C Rieger
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert J Myerburg
- Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Victoria Florea
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Bryon A Tompkins
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Makoto Natsumeda
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ivonne H Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States; Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mayra Vidro-Casiano
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Darcy L DiFede
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alan W Heldman
- Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Raul Mitrani
- Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States; Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL, United States.
| |
Collapse
|
156
|
Schulman IH, Khan A, Hare JM. Interdisciplinary Stem Cell Institute at the University of Miami Miller School of Medicine. Circ Res 2019; 123:1030-1032. [PMID: 30355165 DOI: 10.1161/circresaha.118.310426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - Aisha Khan
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, FL
| |
Collapse
|
157
|
Fernandes GC, Fernandes ADF, Rivera M, Khan A, Schulman IH, Lambrakos LK, Myerburg RJ, Goldberger JJ, Hare JM, Mitrani RD. A meta‐analysis of arrhythmia endpoints in randomized controlled trials of transendocardial stem cell injections for chronic ischemic heart disease. J Cardiovasc Electrophysiol 2019; 30:2492-2500. [DOI: 10.1111/jce.14185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/02/2019] [Accepted: 09/11/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Gilson C. Fernandes
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Manuel Rivera
- Division of Cardiology and Cardiovascular Washington University in Saint Louis Saint Louis Missouri
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Ivonne H. Schulman
- Department of Medicine University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Litsa K. Lambrakos
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | - Robert J. Myerburg
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Joshua M. Hare
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Raul D. Mitrani
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
158
|
Fernández-Avilés F, Sanz-Ruiz R, Bogaert J, Casado Plasencia A, Gilaberte I, Belmans A, Fernández-Santos ME, Charron D, Mulet M, Yotti R, Palacios I, Luque M, Sádaba R, San Román JA, Larman M, Sánchez PL, Sanchís J, Jiménez MF, Claus P, Al-Daccak R, Lombardo E, Abad JL, DelaRosa O, Corcóstegui L, Bermejo J, Janssens S. Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With ST-Segment Elevation Myocardial Infarction and Left Ventricular Dysfunction. Circ Res 2019; 123:579-589. [PMID: 29921651 DOI: 10.1161/circresaha.118.312823] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
RATIONALE Allogeneic cardiac stem cells (AlloCSC-01) have shown protective, immunoregulatory, and regenerative properties with a robust safety profile in large animal models of heart disease. OBJECTIVE To investigate the safety and feasibility of early administration of AlloCSC-01 in patients with ST-segment-elevation myocardial infarction. METHODS AND RESULTS CAREMI (Safety and Efficacy of Intracoronary Infusion of Allogeneic Human Cardiac Stem Cells in Patients With STEMI and Left Ventricular Dysfunction) was a phase I/II multicenter, randomized, double-blind, placebo-controlled trial in patients with ST-segment-elevation myocardial infarction, left ventricular ejection fraction ≤45%, and infarct size ≥25% of left ventricular mass by cardiac magnetic resonance, who were randomized (2:1) to receive AlloCSC-01 or placebo through the intracoronary route at days 5 to 7. The primary end point was safety and included all-cause death and major adverse cardiac events at 30 days (all-cause death, reinfarction, hospitalization because of heart failure, sustained ventricular tachycardia, ventricular fibrillation, and stroke). Secondary safety end points included major adverse cardiac events at 6 and 12 months, adverse events, and immunologic surveillance. Secondary exploratory efficacy end points were changes in infarct size (percentage of left ventricular mass) and indices of ventricular remodeling by magnetic resonance at 12 months. Forty-nine patients were included (92% male, 55±11 years), 33 randomized to AlloCSC-01 and 16 to placebo. No deaths or major adverse cardiac events were reported at 12 months. One severe adverse events in each group was considered possibly related to study treatment (allergic dermatitis and rash). AlloCSC-01 elicited low levels of donor-specific antibodies in 2 patients. No immune-related adverse events were found, and no differences between groups were observed in magnetic resonance-based efficacy parameters at 12 months. The estimated treatment effect of AlloCSC-01 on the absolute change from baseline in infarct size was -2.3% (95% confidence interval, -6.5% to 1.9%). CONCLUSIONS AlloCSC-01 can be safely administered in ST-segment-elevation myocardial infarction patients with left ventricular dysfunction early after revascularization. Low immunogenicity and absence of immune-mediated events will facilitate adequately powered studies to demonstrate their clinical efficacy in this setting. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov . Unique identifier: NCT02439398.
Collapse
Affiliation(s)
- Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Ricardo Sanz-Ruiz
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | | | - Ana Casado Plasencia
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Inmaculada Gilaberte
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Ann Belmans
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Maria Eugenia Fernández-Santos
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Dominique Charron
- HLA et Medicine (HLA-MED), Hôpital Saint-Louis, Paris, France (D.C., R.A.-D.)
| | - Miguel Mulet
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Raquel Yotti
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.)
| | - Itziar Palacios
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Manuel Luque
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Rafael Sádaba
- Department of Cardiac Surgery, Complejo Hospitalario de Navarra, Pamplona, Spain (R.S.)
| | - J Alberto San Román
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Instituto de Ciencias del Corazón (ICICOR), Valladolid, Spain (J.A.S.R.)
| | - Mariano Larman
- Department of Cardiology, Policlínia Guipuzcoa, San Sebastián, Spain (M.L.)
| | - Pedro L Sánchez
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Hospital Clínico Universitario, Salamanca, Spain (P.L.S.)
| | - Juan Sanchís
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, Hospital Clínico Universitario, Valencia, Spain (J.S.)
| | - Manuel F Jiménez
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiology, IBIMA, UMA, UGC Corazón Hospital Clínico Virgen de la Victoria, Málaga, Spain (M.F.J.)
| | - Piet Claus
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Reem Al-Daccak
- HLA et Medicine (HLA-MED), Hôpital Saint-Louis, Paris, France (D.C., R.A.-D.)
| | - Eleuterio Lombardo
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - José Luis Abad
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Olga DelaRosa
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Lucia Corcóstegui
- Coretherapix S.L.U./Tigenix Group Madrid, Spain (I.G., M.M., I.P., M.L., E.L., J.L.A., O.D., L.C.)
| | - Javier Bermejo
- From the Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, and Facultad de Medicina, Universidad Complutense, Madrid, Spain (R.S.-R., A.C.P., M.E.F.-S., R.Y., J.B.).,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (F.F.-A., R.S.-R., A.C.P., M.E.F.-S., R.Y., J.A.S.R., P.L.S., J.S., M.F.J., J.B.).,Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| | - Stefan Janssens
- Department of Cardiovascular Medicine, University Hospitals and KU Leuven, Belgium (J.B., A.B., P.C., S.J.)
| |
Collapse
|
159
|
Bittle GJ, Morales D, Deatrick KB, Parchment N, Saha P, Mishra R, Sharma S, Pietris N, Vasilenko A, Bor C, Ambastha C, Gunasekaran M, Li D, Kaushal S. Stem Cell Therapy for Hypoplastic Left Heart Syndrome: Mechanism, Clinical Application, and Future Directions. Circ Res 2019; 123:288-300. [PMID: 29976693 DOI: 10.1161/circresaha.117.311206] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypoplastic left heart syndrome is a type of congenital heart disease characterized by underdevelopment of the left ventricle, outflow tract, and aorta. The condition is fatal if aggressive palliative operations are not undertaken, but even after the complete 3-staged surgical palliation, there is significant morbidity because of progressive and ultimately intractable right ventricular failure. For this reason, there is interest in developing novel therapies for the management of right ventricular dysfunction in patients with hypoplastic left heart syndrome. Stem cell therapy may represent one such innovative approach. The field has identified numerous stem cell populations from different tissues (cardiac or bone marrow or umbilical cord blood), different age groups (adult versus neonate-derived), and different donors (autologous versus allogeneic), with preclinical and clinical experience demonstrating the potential utility of each cell type. Preclinical trials in small and large animal models have elucidated several mechanisms by which stem cells affect the injured myocardium. Our current understanding of stem cell activity is undergoing a shift from a paradigm based on cellular engraftment and differentiation to one recognizing a primarily paracrine effect. Recent studies have comprehensively evaluated the individual components of the stem cells' secretomes, shedding new light on the intracellular and extracellular pathways at the center of their therapeutic effects. This research has laid the groundwork for clinical application, and there are now several trials of stem cell therapies in pediatric populations that will provide important insights into the value of this therapeutic strategy in the management of hypoplastic left heart syndrome and other forms of congenital heart disease. This article reviews the many stem cell types applied to congenital heart disease, their preclinical investigation and the mechanisms by which they might affect right ventricular dysfunction in patients with hypoplastic left heart syndrome, and finally, the completed and ongoing clinical trials of stem cell therapy in patients with congenital heart disease.
Collapse
Affiliation(s)
- Gregory J Bittle
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - David Morales
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Kristopher B Deatrick
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Nathaniel Parchment
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Progyaparamita Saha
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Rachana Mishra
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Sudhish Sharma
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Nicholas Pietris
- Division of Cardiology (N. Pietris), University of Maryland School of Medicine, Baltimore
| | - Alexander Vasilenko
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Casey Bor
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Chetan Ambastha
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Muthukumar Gunasekaran
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Deqiang Li
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| | - Sunjay Kaushal
- From the Division of Cardiac Surgery (G.J.B., D.M., K.B.D., N. Parchment, P.S., R.M., S.S., A.V., C.B., C.A., M.G., D.L., S.K.)
| |
Collapse
|
160
|
Affiliation(s)
- Eugene Braunwald
- From the TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
161
|
Banerjee MN, Bolli R, Hare JM. Clinical Studies of Cell Therapy in Cardiovascular Medicine: Recent Developments and Future Directions. Circ Res 2019; 123:266-287. [PMID: 29976692 DOI: 10.1161/circresaha.118.311217] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the rising prevalence of cardiovascular disease worldwide and the limited therapeutic options for severe heart failure, novel technologies that harness the regenerative capacity of the heart are sorely needed. The therapeutic use of stem cells has the potential to reverse myocardial injury and improve cardiac function, in contrast to most current medical therapies that only mitigate heart failure symptoms. Nearly 2 decades and >200 trials for cardiovascular disease have revealed that most cell types are safe; however, their efficacy remains controversial, limiting the transition of this therapy from investigation to practice. Lessons learned from these initial studies are driving the design of new clinical trials; higher fidelity of cell isolation techniques, standardization of conditions, more consistent use of state of the art measurement techniques, and assessment of multiple end points to garner insights into the efficacy of stem cells. Translation to clinical trials has almost outpaced our mechanistic understanding, and individual patient factors likely play a large role in stem cell efficacy. Therefore, careful analysis of dosing, delivery methods, and the ideal patient populations is necessary to translate cell therapy from research to practice. We are at a pivotal stage in the field in which information from many relatively small clinical trials must guide carefully executed efficacy trials. Larger efficacy trials are being launched to answer questions about older, first-generation stem cell therapeutics, while novel, second-generation products are being introduced into the clinical realm. This review critically examines the current state of clinical research on cell-based therapies for cardiovascular disease, highlighting the controversies in the field, improvements in clinical trial design, and the application of exciting new cell products.
Collapse
Affiliation(s)
- Monisha N Banerjee
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.).,Department of Surgery (M.N.B)
| | - Roberto Bolli
- University of Miami Miller School of Medicine, FL; and Institute of Molecular Cardiology, University of Louisville, KY (R.B.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.) .,Department of Medicine (J.M.H.)
| |
Collapse
|
162
|
Premer C, Wanschel A, Porras V, Balkan W, Legendre-Hyldig T, Saltzman RG, Dong C, Schulman IH, Hare JM. Mesenchymal Stem Cell Secretion of SDF-1α Modulates Endothelial Function in Dilated Cardiomyopathy. Front Physiol 2019; 10:1182. [PMID: 31616309 PMCID: PMC6769040 DOI: 10.3389/fphys.2019.01182] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/02/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Endothelial dysfunction contributes to the pathophysiology of dilated cardiomyopathy (DCM). Allogeneic but not autologous mesenchymal stem cells (MSCs) improve endothelial function in DCM patients. We hypothesized that these effects are modulated by release of stromal derived factor-1α (SDF-1α). METHODS Plasma TNFα and endothelial progenitor cell-colony forming units (EPC-CFUs) were assessed at baseline and 3-months post-injection in a subset of POSEIDON-DCM patients that received autologous (n = 11) or allogeneic (n = 10) MSCs. SDF-1α secretion by MSCs, endothelial cell (EC) TNFα mRNA expression, and levels of reactive oxygen species (ROS) in response to SDF-1α were measured in vitro. RESULTS As previously shown, DCM patients (n = 21) had reduced EPC-CFUs at baseline (3 ± 3), which were restored to normal by allogeneic MSCs 3-months post-treatment (Δ10 ± 4). DCM patients had elevated baseline plasma TNFα (n = 15, 22 ± 9.4 pg/mL). Allogeneic MSCs (n = 8) decreased, and autologous MSCs (n = 7) increased, plasma TNFα (-7.1 ± 3.1 vs. 22.2 ± 17.1 pg/mL, respectively; P = 0.0005). In culture, autologous MSCs (n = 11) secreted higher levels of SDF-1α than allogeneic MSCs (n = 6) [76.0 (63.7, 100.9) vs. 22.8 (7.2, 43.5) pg/mL, P = 0.0002]. SDF-1α and plasma TNFα negatively correlated with EPC-CFUs in both treatment groups (R = -0.7, P = 0.0004). ECs treated with 20 ng SDF-1α expressed lower levels of TNFα mRNA than cells treated with 100 ng (0.7 ± 0.2 vs. 2.1 ± 0.3, P = 0.0008). SDF-1α at low but not high concentration inhibited the generation of ROS. CONCLUSION MSC secretion of SDF-1α inversely correlates with EPC-CFU production in DCM patients and therefore may be a modulator of MSC therapeutic effect in this clinical setting. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT01392625, identifier NCT01392625.
Collapse
Affiliation(s)
- Courtney Premer
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Amarylis Wanschel
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Valeria Porras
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Tatiana Legendre-Hyldig
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Russell G. Saltzman
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Chunming Dong
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
- Katz Family Division of Nephrology and Hypertension, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
163
|
Diaz-Navarro R, Urrútia G, Cleland JGF, Poloni D, Villagran F, Bangdiwala S, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Hippokratia 2019. [DOI: 10.1002/14651858.cd013433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Rienzi Diaz-Navarro
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Gerard Urrútia
- CIBER Epidemiología y Salud Pública (CIBERESP); Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau); Sant Antoni Maria Claret, 167 Pavilion 18 (D-53) Barcelona Catalonia Spain 08025
| | - John GF Cleland
- Imperial College London; National Heart and Lung Institute; London UK
| | - Daniel Poloni
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Francisco Villagran
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Shrikant Bangdiwala
- Collaborative Studies Coordinating Center; Department of Biostatistics, Gillings School of Global Public Health; Suite 203, Bank of America Center 137 E. Franklin Street Chapel Hill North Carolina USA 27514-4145
| | - Gabriel Rada
- Pontificia Universidad Católica de Chile; Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine; Lira 44, Decanato Primer piso Santiago Chile
| | - Eva Madrid
- Cochrane Centre School of Medicine Universidad de Valparaiso; Interdisciplinary Centre for Health Studies CIESAL; Viña del Mar Chile
- Universidad de Valparaiso; Chilean Cochrane Centre; Valparaiso Chile
| |
Collapse
|
164
|
Reparative and Regenerative Effects of Mesenchymal Stromal Cells-Promising Potential for Kidney Transplantation? Int J Mol Sci 2019; 20:ijms20184614. [PMID: 31540361 PMCID: PMC6770554 DOI: 10.3390/ijms20184614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) possess reparative, regenerative and immunomodulatory properties. The current literature suggests that MSCs could improve kidney transplant outcome via immunomodulation. In many clinical domains, research has also focussed on the regenerative and reparative effects of therapies with MSCs. However, in the field of transplantation, data on this subject remain scarce. This review provides an overview of what is known about the regenerative and reparative effects of MSCs in various fields ranging from wound care to fracture healing and also examines the potential of these promising MSC properties to improve the outcome of kidney transplantations.
Collapse
|
165
|
Jayaraj JS, Janapala RN, Qaseem A, Usman N, Fathima N, Kashif T, Reddy VK, Bakshi S. Efficacy and Safety of Stem Cell Therapy in Advanced Heart Failure Patients: A Systematic Review with a Meta-analysis of Recent Trials Between 2017 and 2019. Cureus 2019; 11:e5585. [PMID: 31696004 PMCID: PMC6820892 DOI: 10.7759/cureus.5585] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective The effects of stem cell therapy in patients with advanced heart failure is an ongoing debate. This study aimed to assess the effectiveness and safety of stem cell therapy plus the standard of care as compared to the placebo plus the standard of care in advanced heart failure patients. Methods A comprehensive keyword search of PubMed between 2017 and 2019 was performed to extract trials conducted with stem cell therapy controlled with placebo in advanced heart failure. We included randomized controlled trials (RCTs) with data on safety and efficacy in patients with advanced heart failure after stem cell transplantation. Results Six RCTs, consisting of 569 patients, were selected. Three-hundred sixty-seven (367) out of 369 participants from the eligible four out of six RCTs were included for efficacy analysis, as we lost two patients from the final analysis due to early death. Five-hundred twenty-six (526) out of 527 participants from the eligible five out of six RCTs were included for safety analysis, as we lost one patient from the final analysis for not being able to receive the intervention. Stem cell transplantation significantly improved left ventricular ejection fraction (LVEF) by 4.58% (95% CI: 3.73-5.43%; p = 0.00001), improved left ventricular end-systolic volume (LVESV) by -5.18 ml (95% CI: -9.74 to -0.63 ml; p =0.03), and there was no difference in the risk of all-cause mortality (OR 0.97; 95% CI: 0.52 to 1.78%; p = 0.91). The above results correlate with the previous meta-analysis data conducted in 2016. Conclusions This meta-analysis provided the cumulative efficacy and safety results of stem cell transplantation in advanced heart failure based on recent RCTs. The above results suggest that stem cell therapy was associated with a moderate improvement in LVEF, and the safety analysis indicates no increased risk of mortality in patients with advanced heart failure. This meta-analysis recommends conducting more RCTs comparing stem cell transplantation and placebo with a larger patient population and longer follow-up.
Collapse
Affiliation(s)
- Joseph S Jayaraj
- Internal Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, USA
| | - Rajesh Naidu Janapala
- Internal Medicine, Icahn School of Medicine at Mount Sinai, Queens Hospital Center, New York, USA
| | - Aisha Qaseem
- Internal Medicine, Emory Johns Creek Hospital, Georgia, USA
| | - Norina Usman
- Internal Medicine, Veterans Affairs Palo Alto Health Care System - Stanford University School of Medicine, Palo Alto, USA
| | - Nida Fathima
- Internal Medicine, Sri Siddhartha Medical College, Tumkur, IND
| | - Tooba Kashif
- Cardiology, Heart and Vascular Institute, Dearborn, USA
| | - Vineeth K Reddy
- Internal Medicine, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | - Sanjiv Bakshi
- Cardiology, Icahn School of Medicine at Mount Sinai , Queens Hospital Center, New York, USA
| |
Collapse
|
166
|
Pooria A, Pourya A, Gheini A. Animal- and human-based evidence for the protective effects of stem cell therapy against cardiovascular disorders. J Cell Physiol 2019; 234:14927-14940. [PMID: 30811030 DOI: 10.1002/jcp.28330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 01/24/2023]
Abstract
The increasing rate of mortality and morbidity because of cardiac diseases has called for efficient therapeutic needs. With the advancement in cell-based therapies, stem cells are abundantly studied in this area. Nearly, all sources of stem cells are experimented to treat cardiac injuries. Tissue engineering has also backed this technique by providing an advantageous platform to improve stem cell therapy. After in vitro studies, primary treatment-based research studies comprise small and large animal studies. Furthermore, these studies are implemented in human models in the form of clinical trials. Purpose of this review is to highlight the animal- and human-based studies, exploiting various stem cell sources, to treat cardiovascular disorders.
Collapse
Affiliation(s)
- Ali Pooria
- Department of Cardiology, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afsoun Pourya
- Student of Research committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Gheini
- Department of Cardiology, Lorestan University of Medical Sciences, Khoramabad, Iran
| |
Collapse
|
167
|
Aghabozorgi AS, Ahangari N, Eftekhaari TE, Torbati PN, Bahiraee A, Ebrahimi R, Pasdar A. Circulating exosomal miRNAs in cardiovascular disease pathogenesis: New emerging hopes. J Cell Physiol 2019; 234:21796-21809. [PMID: 31273798 DOI: 10.1002/jcp.28942] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVDs) are one of the leading causes of morbidity and mortality. Standard therapies have failed to significantly increase patients' survival. Moreover, the majority of conventional screening procedures are ineffective for the diagnosis of CVDs at early stages. Accumulating evidence suggests that numerous cell types release a class of nano-sized vesicles named exosomes into the extracellular space. Exosomes are widely distributed in various body fluids and contain a number of diverse biomolecules such as proteins, lipids, and both mRNA and noncoding RNAs which reflect host-cell molecular architecture. MicroRNAs (miRNAs), which can be found in exosomes, could be taken up by both neighboring and distal cells. Not only has recent evidence indicated the regulatory role of exosomal miRNAs in the pathogenesis of CVD, but it has also been shown that differential expression of exosomal miRNAs in CVDs has made them promising biomarkers for early detection of CVDs. Owing to these remarkable features, exosomal miRNAs have emerged as hot spots in research. This review summarizes the role of exosomal miRNAs in the pathogenesis of CVDs and discusses their potential application in the clinical setting as both therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Amirsaeed S Aghabozorgi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Najmeh Ahangari
- Department of Modern Sciences & Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tasnim E Eftekhaari
- Molecular Medicine Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Paria N Torbati
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Pasdar
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, UK
| |
Collapse
|
168
|
Attenuation of frailty in older adults with mesenchymal stem cells. Mech Ageing Dev 2019; 181:47-58. [DOI: 10.1016/j.mad.2019.111120] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/20/2019] [Accepted: 05/29/2019] [Indexed: 01/13/2023]
|
169
|
Yaron JR, Kwiecien JM, Zhang L, Ambadapadi S, Wakefield DN, Clapp WL, Dabrowski W, Burgin M, Munk BH, McFadden G, Chen H, Lucas AR. Modifying the Organ Matrix Pre-engraftment: A New Transplant Paradigm? Trends Mol Med 2019; 25:626-639. [DOI: 10.1016/j.molmed.2019.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
|
170
|
Yagyu T, Yasuda S, Nagaya N, Doi K, Nakatani T, Satomi K, Shimizu W, Kusano K, Anzai T, Noguchi T, Ohgushi H, Kitamura S, Kangawa K, Ogawa H. Long-Term Results of Intracardiac Mesenchymal Stem Cell Transplantation in Patients With Cardiomyopathy. Circ J 2019; 83:1590-1599. [PMID: 31105128 DOI: 10.1253/circj.cj-18-1179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), which have the potential to differentiate into cardiomyocytes or vascular endothelial cells, have been used clinically as therapy for cardiomyopathy. In this study, we aimed to evaluate the long-term follow-up results. METHODS AND RESULTS We studied 8 patients with symptomatic heart failure (HF) on guideline-directed therapy (ischemic cardiomyopathy, n=3; nonischemic cardiomyopathy, n=5) who underwent intracardiac MSC transplantation using a catheter-based injection method between May 2004 and April 2006. Major adverse events and hospitalizations were investigated up to 10 years afterward. Compared with baseline, there were no significant differences in B-type natriuretic peptide (BNP) (from 211 to 173 pg/mL), left ventricular ejection fraction (LVEF) (from 24% to 26%), and peak oxygen uptake (from 16.5 to 19.2 mL/min/kg) at 2 months. During the follow-up period, no patients experienced serious adverse events such as arrhythmias. Three patients died of pneumonia in the 1st year, liver cancer in the 6th year, and HF in the 7th year. Of the remaining 5 patients, 3 patients were hospitalized for exacerbated HF, 1 of whom required heart transplantation in the 2nd year; 2 patients survived for 10 years without worsening HF. CONCLUSIONS The results of this exploratory study of intracardiac MSCs administration suggest further research regarding the feasibility and efficacy is warranted.
Collapse
Affiliation(s)
- Takeshi Yagyu
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences
| | | | - Kaori Doi
- Research Institute, National Cerebral and Cardiovascular Center
| | - Takeshi Nakatani
- Department of Transplantation, National Cerebral and Cardiovascular Center
| | - Kazuhiro Satomi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiology, Tokyo Medical University
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Hajime Ohgushi
- National Institute of Advanced Industrial Science and Technology
| | - Soichiro Kitamura
- Department of Transplantation, National Cerebral and Cardiovascular Center
| | - Kenji Kangawa
- Research Institute, National Cerebral and Cardiovascular Center
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
- Research Institute, National Cerebral and Cardiovascular Center
| |
Collapse
|
171
|
Avivar-Valderas A, Martín-Martín C, Ramírez C, Del Río B, Menta R, Mancheño-Corvo P, Ortiz-Virumbrales M, Herrero-Méndez Á, Panés J, García-Olmo D, Castañer JL, Palacios I, Lombardo E, Dalemans W, DelaRosa O. Dissecting Allo-Sensitization After Local Administration of Human Allogeneic Adipose Mesenchymal Stem Cells in Perianal Fistulas of Crohn's Disease Patients. Front Immunol 2019; 10:1244. [PMID: 31258526 PMCID: PMC6587893 DOI: 10.3389/fimmu.2019.01244] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022] Open
Abstract
Adipose mesenchymal stem cells (ASC) are considered minimally immunogenic. This is due to the low expression of human leukocyte antigens I (HLA-I), lack of HLA-II expression and low expression of co-stimulatory molecules such as CD40 and CD80. The low rate of observed immunological rejection as well as the immunomodulatory qualities, position ASC as a promising cell-based therapy for the treatment of a variety of inflammatory indications. Yet, few studies have addressed relevant aspects of immunogenicity such as ASC donor-to-patient HLA histocompatibility or assessment of immune response triggered by ASC administration, particularly in the cases of presensitization. The present study aims to assess allo-immune responses in a cohort of Crohn's disease patients administered with allogeneic ASC (darvadstrocel formerly Cx601) for the treatment of complex perianal fistulas. We identified donor-specific antibodies (DSA) generation in a proportion of patients and observed that patients showing preexisting immunity were prone to generating DSA after allogeneic therapy. Noteworthy, naïve patients generating DSA at week 12 (W12) showed a significant reduction in DSA titer at week 52 (W52), whereas DSA titer was reduced in pre-sensitized patients only with no specificities against the donor administered. Remarkably, we did not observe any correlation of DSA generation with ASC therapeutic efficacy. In vitro complement-dependent cytotoxicity (CDC) studies have revealed limited cytotoxic levels based upon HLA-I expression and binding capacity even in pro-inflammatory conditions. We sought to identify CDC coping mechanisms contributing to the limited cytotoxic killing observed in ASC in vitro. We found that ASC express membrane-bound complement regulatory proteins (mCRPs) CD55, CD46, and CD59 at basal levels, with CD46 more actively expressed in pro-inflammatory conditions. We demonstrated that CD46 is a main driver of CDC signaling; its depletion significantly enhances sensitivity of ASC to CDC. In summary, despite relatively high clearance, DSA generation may represent a major challenge for allogeneic cell therapy management. Sensitization may be a significant concern when evaluating re-treatment or multi-donor trials. It is still unknown whether DSA generation could potentially be the consequence of donor-to-patient interaction and, therefore, subsequently link to efficacy or biological activity. Lastly, we propose that CDC modulators such as CD46 could be used to ultimately link CDC specificity with allogeneic cell therapy efficacy.
Collapse
Affiliation(s)
| | | | - Cristina Ramírez
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Borja Del Río
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Ramón Menta
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | | | | | | | - Julián Panés
- Department of Gastroenterology, Hospital Clínic, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Damián García-Olmo
- Department of Surgery, Hospital U. Fundación Jiménez Díaz, Madrid, Spain
| | - José Luís Castañer
- Department of Immunology, University Hospital Ramon y Cajal, Madrid, Spain
| | - Itziar Palacios
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | - Eleuterio Lombardo
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| | | | - Olga DelaRosa
- Takeda Madrid, Cell Therapy Technology Center-Cell Therapies, Madrid, Spain
| |
Collapse
|
172
|
Stem cell therapy in heart failure: Where do we stand today? Biochim Biophys Acta Mol Basis Dis 2019; 1866:165489. [PMID: 31199998 DOI: 10.1016/j.bbadis.2019.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023]
Abstract
Heart failure is a global epidemic that drastically cuts short longevity and compromises quality of life. Approximately 6 million Americans ≥20 years of age carry a diagnosis of heart failure. Worldwide, about 40 million adults are affected. The treatment of HF depends on the etiology. If left untreated it rapidly progresses and compromises quality of life. One of the newer technologies still in its infancy is stem cell therapy for heart failure. This review attempts to highlight the clinical studies done in ischemic cardiomyopathy, dilated cardiomyopathy and restrictive cardiomyopathy. A combined approach of simultaneous revascularization and stem cell therapy appears to produce maximum benefit in ischemic cardiomyopathy. Treatment of dilated cardiomyopathy with stem cells also holds promise but needs more definition with regards to timing, route of cell delivery and type of cell used to achieve reproducible results. The variability noted in response to stem cell therapy in patients could also be secondary to their co-morbidities. Abnormalities of glucose metabolism and diabetes in particular impair stem cell and angiogenic cell mobilization. This opens up a whole new area of investigation into exploring the biochemical microenvironment which could influence the efficacy of stem cell therapy. This article is part of a Special Issue entitled: Stem Cells and Their Applications to Human Diseases edited by Hemachandra Reddy.
Collapse
|
173
|
Burkhart HM, Qureshi MY, Rossano JW, Cantero Peral S, O'Leary PW, Hathcock M, Kremers W, Nelson TJ. Autologous stem cell therapy for hypoplastic left heart syndrome: Safety and feasibility of intraoperative intramyocardial injections. J Thorac Cardiovasc Surg 2019; 158:1614-1623. [PMID: 31345560 DOI: 10.1016/j.jtcvs.2019.06.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Staged surgical palliation for hypoplastic left heart syndrome results in an increased workload on the right ventricle serving as the systemic ventricle. Concerns for cardiac dysfunction and long-term heart failure have generated interest in first-in-infant, cell-based therapies as an additional surgical treatment modality. METHODS A phase 1 clinical trial was conducted to evaluate the safety and feasibility of direct intramyocardial injection of autologous umbilical cord blood-derived mononuclear cells in 10 infants with hypoplastic left heart syndrome at the time of stage II palliation. RESULTS All 10 patients underwent successful stage II palliation and intramyocardial injection of umbilical cord blood-derived mononuclear cells. Operative mortality was 0%. There was a single adverse event related to cell delivery: An injection site epicardial bleed that required simple oversew. The cohort did not demonstrate any significant safety concerns over 6 months. Additionally, the treatment group did not demonstrate any reduction in cardiac function in the context of the study related intramyocardial injections of autologous cells. CONCLUSIONS This phase 1 clinical trial showed that delivering autologous umbilical cord blood-derived mononuclear cells directly into the right ventricular myocardium during planned stage II surgical palliation for hypoplastic left heart syndrome was safe and feasible. Secondary findings of preservation of baseline right ventricular function throughout follow-up and normalized growth rates support the design of a phase 2b follow-up trial.
Collapse
Affiliation(s)
- Harold M Burkhart
- Division of Cardiovascular and Thoracic Surgery, University of Oklahoma, Oklahoma City, Okla.
| | | | - Joseph W Rossano
- Cardiac Center, Children's Hospital of Philadelphia, Philadelphia, Pa
| | | | | | - Matthew Hathcock
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minn
| | - Walter Kremers
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minn
| | - Timothy J Nelson
- Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minn; Division of General Internal Medicine, Mayo Clinic, Rochester, Minn; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minn; Center for Regenerative Medicine, Mayo Clinic, Rochester, Minn
| |
Collapse
|
174
|
Verdonschot JAJ, Hazebroek MR, Ware JS, Prasad SK, Heymans SRB. Role of Targeted Therapy in Dilated Cardiomyopathy: The Challenging Road Toward a Personalized Approach. J Am Heart Assoc 2019; 8:e012514. [PMID: 31433726 PMCID: PMC6585365 DOI: 10.1161/jaha.119.012514] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Job A. J. Verdonschot
- Department of CardiologyCARIMMaastricht University Medical CentreMaastrichtThe Netherlands
- Department of Clinical GeneticsMaastricht University Medical CentreMaastrichtThe Netherlands
| | - Mark R. Hazebroek
- Department of CardiologyCARIMMaastricht University Medical CentreMaastrichtThe Netherlands
| | - James S. Ware
- Cardiovascular Research CentreRoyal Brompton & Harefield Hospitals NHS TrustLondonUnited Kingdom
- National Heart and Lung InstituteImperial College LondonLondonUnited Kingdom
- London Institute of Medical SciencesImperial College LondonLondonUnited Kingdom
| | - Sanjay K. Prasad
- Cardiovascular Research CentreRoyal Brompton & Harefield Hospitals NHS TrustLondonUnited Kingdom
- National Heart and Lung InstituteImperial College LondonLondonUnited Kingdom
| | - Stephane R. B. Heymans
- Department of CardiologyCARIMMaastricht University Medical CentreMaastrichtThe Netherlands
- Netherlands Heart InstituteUtrechtthe Netherlands
- Department of Cardiovascular ResearchUniversity of LeuvenBelgium
| |
Collapse
|
175
|
Fan M, Huang Y, Chen Z, Xia Y, Chen A, Lu D, Wu Y, Zhang N, Qian J. Efficacy of mesenchymal stem cell therapy in systolic heart failure: a systematic review and meta-analysis. Stem Cell Res Ther 2019; 10:150. [PMID: 31151406 PMCID: PMC6544951 DOI: 10.1186/s13287-019-1258-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/28/2022] Open
Abstract
Background Heart failure (HF) is the end stage of most heart disease. Mesenchymal stem cells (MSCs), with their specific biological effects, have been applied in several clinical trials to evaluate the efficacy in HF therapy. We performed this meta-analysis to review the clinical evidence of their therapeutic effect on HF. Methods Three databases were searched. The outcomes of interest were death, readmission, the 6-min walk test (6MWT), New York Heart Association (NYHA) class and left ventricular ejection fraction (LVEF). The relative risk (RR) and weighted mean difference (WMD) were calculated to evaluate the effects of MSCs on HF compared to placebo. Results A total of nine studies were included, involving 612 patients who underwent MSCs or placebo treatment. The overall rate of death showed a trend of reduction of 36% (RR [CI] = 0.64 [0.35, 1.16], p = 0.143) in the MSC treatment group. The incidence of readmission was reduced by 34% (RR [CI] = 0.66 [0.51, 0.85], p = 0.001). The patients in the MSC treatment group realised an average of 40.44 m (WMD [95% CI] = 40.44 m [19.07, 61.82], p < 0.0001) improvement in 6MWT. The NYHA class was reduced obviously in the MSC group (WMD [95% CI] = − 0.42 [− 0.64, − 0.20], p < 0.0001). The changes of LVEF from baseline were significantly more than 5.25% (WMD [95% CI] = 5.25 [3.58, 6.92], p < 0.0001) in the MSCs group, unlike in the placebo group. Conclusions Our results suggested that MSC treatment is an effective therapy for HF by improving the prognosis and exercise capacity. SCs derived from allosomes have superior therapeutic effects, and intracoronary injection is the optimum MSC delivery approach. Short-term cryopreservation is feasible in MSCs storage or transport.
Collapse
Affiliation(s)
- Mengkang Fan
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yin Huang
- Department of Geriatric Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhangwei Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yan Xia
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ao Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Danbo Lu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuan Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ning Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Juying Qian
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
176
|
Wang C, Li J, Zhang B, Li Y. Safety and efficacy of bone marrow-derived cells therapy on cardiomyopathy: a meta-analysis. Stem Cell Res Ther 2019; 10:137. [PMID: 31109372 PMCID: PMC6528271 DOI: 10.1186/s13287-019-1238-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Controversial results still existed on the clinical utility of bone marrow-derived cells (BMCs) for cardiomyopathy (CMP). This study aims to reveal the true power of this promising approach by synthesizing all the available data on this subject matter. METHODS Twenty studies including 1418 patients were identified from systematic search. Weighted mean differences for changes in left ventricular ejection fraction (LVEF), left ventricular end-diastolic volume (LVEDV), left ventricular end-systolic volume (LVESV), 6-min walk distance, and NYHA functional class were estimated with a random-effects model. Major adverse cardiovascular event (MACE), rehospitalization, all-cause mortality, and patients' quality of life were also calculated. RESULTS Compared with the control group, BMC therapy resulted in greater LVEF (3.72%, 95% CI 2.31 to 5.13, P < 0.0001), 6-min walk distance (53.16, 95% CI 25.17 to 81.10, P = 0.0002), NYHA functional class (- 0.48, 95% CI - 0.65 to - 0.31, P < 0.0001), and smaller LVESV (- 16.79, 95% CI - 27.21 to - 6.38, P = 0.002). BMC treatment significantly reduced the mortality rate and improved patients' quality of life. No significant difference was found between the BMCs and control group in LVEDV, MACE, and rehospitalization rate. However, the outcomes showed a clear trend in favor of the BMC group. Subgroup analysis showed that LVEF improved greater in a subgroup of intracoronary infusion, BMSC, or higher cell dose. CONCLUSION The results of the current meta-analysis suggest that BMC treatment for CMP is safe and feasible. This therapy was associated with persistent improvements in LV function, LV remodeling, functional class, patients' survival, and quality of life. Intracoronary infusion of high-dose (> 108) BMSC might be a better therapeutic option for CMP patients. Further evidences are needed to verify our results.
Collapse
Affiliation(s)
- Chao Wang
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China.
| | - Jingzhao Li
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| | - Boya Zhang
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| | - Yongjian Li
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| |
Collapse
|
177
|
Cell Spray Transplantation of Adipose-derived Mesenchymal Stem Cell Recovers Ischemic Cardiomyopathy in a Porcine Model. Transplantation 2019; 102:2012-2024. [PMID: 30048399 DOI: 10.1097/tp.0000000000002385] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Allogeneic adipose-derived mesenchymal stem cells (ADSC) are promising cell sources for cell therapy to treat ischemic cardiomyopathy (ICM). We hypothesized that ADSC transplantation via the new cell spray method may be a feasible, safe, and effective treatment for ICM. METHODS Human ADSCs were acquired from white adipose tissue. Porcine ICM models were established by constriction of the left anterior descending coronary artery. Adipose-derived mesenchymal stem cells were spread over the surface of the heart via cell spray in fibrinogen and thrombin solutions. The cardiac function was compared with that of the control group. RESULTS Adipose-derived mesenchymal stem cells were successfully transplanted forming a graft-like gel film covering the infarct myocardium. Premature ventricular contractions were rarely detected in the first 3 days after transplantation. Echocardiography and magnetic resonance imaging revealed improved cardiac performance of the ADSC group at 4 and 8 weeks after transplantation. Systolic and diastolic parameters were significantly greater in the ADSC group at 8 weeks after transplantation. Histological examination showed significantly attenuated left ventricular remodeling and a greater vascular density in the infarct border area in the ADSC group. Moreover, the coronary flow reserve was maintained, and expression levels of angiogenesis-related factors in the infarct border and remote areas were significantly increased. CONCLUSIONS Spray method implantation of allogenic ADSCs can improve recovery of cardiac function in a porcine infarction model. This new allogenic cell delivery system may help to resolve current limitations of invasiveness and cost in stem cell therapy.
Collapse
|
178
|
Poglajen G, Zemljič G, Cerar A, Frljak S, Jaklič M, Androcec V, Vrtovec B. Transendocardial CD34+ Cell Therapy does not Increase the Risk of Ventricular Arrhythmias in Patients with Chronic Heart Failure. Cell Transplant 2019; 28:856-863. [PMID: 31046425 PMCID: PMC6719496 DOI: 10.1177/0963689719840351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ventricular arrhythmias (VA) are of major concern in the field of cell therapy, potentially limiting its safety and efficacy. We sought to investigate the effects of CD34+ cell therapy on VA burden in patients with chronic heart failure (CHF). We performed registry data analysis of patients with CHF and implanted ICD/CRT devices treated with transendocardial CD 34+ cell therapy. Demographic, echocardiographic, and biochemical parameters were analyzed. Device records were reviewed and the number and type of VA 1 year prior to and 1 year after cell therapy were analyzed. All patients underwent electroanatomical mapping, and myocardial scar was defined as unipolar voltage (UV) <8.3 mV and linear local shortening (LLS) <6%. Of 209 patients screened, 48 met inclusion criteria. The mean age of the patients was 52 years and 88% were male. Nonischemic and ischemic cardiomyopathy were present in 55% and 45% of patients. The average serum creatinine was 91±26 µmol/L, serum bilirubin 18±9 µmol/L, NT-proBNP 1767 (468, 2446) pg/mL, LVEF 27±9% and 6’ walk test 442±123 m. The average scar burden in patients with nonischemic and ischemic DCM was 58±15% and 51±25% (P=0.48). No significant difference in VA burden was observed before and after cell therapy (48% vs. 44%; P=0.68). ICD activation occurred in 19% and 27% of patients before and after cell therapy (P=0.33). According to our results, transendocardial CD34+ cell therapy does not appear to increase the risk of VA in chronic heart failure patients.
Collapse
Affiliation(s)
- Gregor Poglajen
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia.,2 Faculy of Medicine, Ljubljana, Slovenia
| | - Gregor Zemljič
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Andraž Cerar
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Sabina Frljak
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Martina Jaklič
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Vesna Androcec
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia
| | - Bojan Vrtovec
- 1 Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Slovenia.,2 Faculy of Medicine, Ljubljana, Slovenia
| |
Collapse
|
179
|
Janssens SP. Mesenchymal Cell Therapy for Dilated Cardiomyopathy: Time to Test the Water. J Am Coll Cardiol 2019; 69:538-540. [PMID: 28153109 DOI: 10.1016/j.jacc.2016.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Stefan P Janssens
- Department of Cardiovascular Diseases, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
180
|
Regmi S, Pathak S, Kim JO, Yong CS, Jeong JH. Mesenchymal stem cell therapy for the treatment of inflammatory diseases: Challenges, opportunities, and future perspectives. Eur J Cell Biol 2019; 98:151041. [PMID: 31023504 DOI: 10.1016/j.ejcb.2019.04.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/01/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising alternative agents for the treatment of inflammatory disorders due to their immunomodulatory functions, and several clinical trials on MSC-based products are currently being conducted. In this review, we discuss recent progress made on the use of MSCs as immunomodulatory agents, developmental challenges posed by MSC-based therapy, and the strategies being used to overcome these challenges. In this context, current understanding of the mechanisms responsible for MSC interactions with the immune system and the molecular responses of MSCs to inflammatory signals are discussed. The immunosuppressive activities of MSCs are initiated by cell-to-cell contact and the release of immuno-regulatory molecules. By doing so, MSCs can inhibit the proliferation and function of T cells, natural killer cells, B cells, and dendritic cells, and can also increase the proliferation of regulatory T cells. However, various problems, such as low transplanted cell viability, poor homing and engraftment into injured tissues, MSC heterogeneity, and lack of adequate information on optimum MSC doses impede clinical applications. On the other hand, it has been shown that the immunomodulatory activities and viabilities of MSCs might be enhanced by 3D-cultured systems, genetic modifications, preconditioning, and targeted-delivery.
Collapse
Affiliation(s)
- Shobha Regmi
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Shiva Pathak
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
181
|
Cardioprotective microRNAs: Lessons from stem cell-derived exosomal microRNAs to treat cardiovascular disease. Atherosclerosis 2019; 285:1-9. [PMID: 30939341 DOI: 10.1016/j.atherosclerosis.2019.03.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/28/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
The stem cell-based therapy has emerged as a promising therapeutic strategy for treating cardiovascular ischemic diseases (CVIDs), such as myocardial infarction (MI). However, some important functional shortcomings of stem cell transplantation, such as immune rejection, tumorigenicity and infusional toxicity, have overshadowed stem cell therapy in the setting of cardiovascular diseases (CVDs). Accumulating evidence suggests that the therapeutic effects of transplanted stem cells are predominately mediated by secreting paracrine factors, importantly, microRNAs (miRs) present in the secreted exosomes. Therefore, novel cell-free therapy based on the stem cell-secreted exosomal miRs can be considered as a safe and effective alternative tool to stem cell therapy for the treatment of CVDs. Stem cell-derived miRs have recently been found to transfer, via exosomes, from a transplanted stem cell into a recipient cardiac cell, where they regulate various cellular process, such as proliferation, apoptosis, stress responses, as well as differentiation and angiogenesis. The present review aimed to summarize cardioprotective exosomal miRs secreted by transplanted stem cells from various sources, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and cardiac stem/progenitor cells, which showed beneficial modulatory effects on the myocardial infracted heart. In summary, stem cell-exosomal miRs, including miR-19a, mirR-21, miR-21-5p, miR-21-a5p, miR-22 miR-24, miR-26a, miR-29, miR-125b-5p, miR-126, miR-201, miR-210, and miR-294, have been shown to have cardioprotective effects by enhancing cardiomyocyte survival and function and attenuating cardiac fibrosis. Additionally, MCS-exosomal miRs, including miR-126, miR-210, miR-21, miR-23a-3p and miR-130a-3p, are found to exert cardioprotective effects through induction of angiogenesis in ischemic heart after MI.
Collapse
|
182
|
Mostafavian Z, Vakilian F, Torkmanzade L, Moghiman T. Effect of Stem Cell Therapy on Patients' Quality of Life in Heart Failure with Reduced Ejection Fraction. J Med Life 2019; 11:359-364. [PMID: 30894895 PMCID: PMC6418329 DOI: 10.25122/jml-2018-0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: Heart failure with reduced ejection fraction (HFrEF) is a debilitating disease in which Left Ventricular Ejection Fraction (LVEF) is ≤ 40%, and it involves various organs. Regarding the novelty of stem cell therapy in HF, we aimed at studying the effect of stem cell therapy on the QoL of patients with HFrEF. Materials and Methods: In a prospective study, 30 patients diagnosed with HFrEF who had undergone stem cell injection (study group) and 30 patients with HFrEF receiving guideline-directed medical therapy (control group) were recruited by convenience sampling during 2016 in Mashhad, Iran. Patients' quality of life, left ventricular ejection fraction and their disability degree were studied twice with a 3-month interval. For data analysis, paired t-test, chi2 and multivariate linear regression were used. Results: The mean age of study and control groups was 61.3±10.24 and 60.93±7.88 years respectively. Ninety percent of the cases and 56.7% of the controls were male (P=0.003). A significant difference in QoL was observed before and after treatment in each group (P<0.05). However, the QoL score showed no statistical difference between the two groups following treatment (P=0.13). The same result was achieved for LVEF (P=0.18); whereas the NYHA function class showed a significant difference between the two groups following treatment (P=0.017). Conclusions: According to the results, it seems that the treatment of HFrEF patients with stem cells is as effective as conventional therapies in improving the LVEF and QoL and more efficient than conventional treatments in increasing the patients' general satisfaction with life.
Collapse
Affiliation(s)
- Zahra Mostafavian
- Department of Community Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Farveh Vakilian
- Cardiothoracic Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Torkmanzade
- Faculty of Medicine, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | | |
Collapse
|
183
|
Sun XL, Hao QK, Tang RJ, Xiao C, Ge ML, Dong BR. Frailty and Rejuvenation with Stem Cells: Therapeutic Opportunities and Clinical Challenges. Rejuvenation Res 2019; 22:484-497. [PMID: 30693831 PMCID: PMC6919243 DOI: 10.1089/rej.2017.2048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Frailty, one appealing target for improving successful aging of the elderly population, is a common clinical syndrome based on the accumulation of multisystemic function declines and the increase in susceptibility to stressors during biological aging. The age-dependent senescence, the frailty-related stem cell depletion, chronic inflammation, imbalance of immune homeostasis, and the reduction of multipotent stem cells collectively suggest the rational hypothesis that it is possible to (partially) cure frailty with stem cells. This systematic review has included all of the human trials of stem cell therapy for frailty from the main electronic databases and printed materials and screened the closely related reviews themed on the mechanisms of aging, frailty, and stem cells, to provide more insights in stem cell strategies for frailty, one promising method to recover health from a frail status. To date, a total of four trials about this subject have been registered on clinicaltrials.gov. The use of mesenchymal stem cells (MSCs), doses of 100 million cells, single peripheral intravenous infusion, follow-up periods of 6–12 months, and a focus primarily on safety and secondarily on efficacy are common characteristics of these studies. We conclude that intravenous infusion of allogenic MSCs is safe, well tolerated, and preliminarily effective clinically. More preclinical experiments and clinical trials are warranted to precisely elucidate the mechanism, safety, and efficacy of frailty stem cell therapy.
Collapse
Affiliation(s)
- Xue-Lian Sun
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| | - Qiu-Kui Hao
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ren-Jie Tang
- Department of Urinary Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Urology, Chengdu Sixth People's Hospital, Chengdu, China
| | - Chun Xiao
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Sichuan Engineering Research Institute of Chi Ding Sheng Tong, Chengdu, China
| | - Mei-Ling Ge
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| | - Bi-Rong Dong
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| |
Collapse
|
184
|
Liao S, Zhang Y, Ting S, Zhen Z, Luo F, Zhu Z, Jiang Y, Sun S, Lai WH, Lian Q, Tse HF. Potent immunomodulation and angiogenic effects of mesenchymal stem cells versus cardiomyocytes derived from pluripotent stem cells for treatment of heart failure. Stem Cell Res Ther 2019; 10:78. [PMID: 30845990 PMCID: PMC6407247 DOI: 10.1186/s13287-019-1183-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Optimal cell type as cell-based therapies for heart failure (HF) remains unclear. We sought to compare the safety and efficacy of direct intramyocardial transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) and human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) in a porcine model of HF. METHODS Eight weeks after induction of HF with myocardial infarction (MI) and rapid pacing, animals with impaired left ventricular ejection fraction (LVEF) were randomly assigned to receive direct intramyocardial injection of saline (MI group), 2 × 108 hESC-CMs (hESC-CM group), or 2 × 108 hiPSC-MSCs (hiPSC-MSC group). The hearts were harvested for immunohistochemical evaluation after serial echocardiography and hemodynamic evaluation and ventricular tachyarrhythmia (VT) induction by in vivo programmed electrical stimulation. RESULTS At 8 weeks post-transplantation, LVEF, left ventricular maximal positive pressure derivative, and end systolic pressure-volume relationship were significantly higher in the hiPSC-MSC group but not in the hESC-CM group compared with the MI group. The incidence of early spontaneous ventricular tachyarrhythmia (VT) episodes was higher in the hESC-CM group but the incidence of inducible VT was similar among the different groups. Histological examination showed no tumor formation but hiPSC-MSCs exhibited a stronger survival capacity by activating regulatory T cells and reducing the inflammatory cells. In vitro study showed that hiPSC-MSCs were insensitive to pro-inflammatory interferon-gamma-induced human leukocyte antigen class II expression compared with hESC-CMs. Moreover, hiPSC-MSCs also significantly enhanced angiogenesis compared with other groups via increasing expression of distinct angiogenic factors. CONCLUSIONS Our results demonstrate that transplantation of hiPSC-MSCs is safe and does not increase proarrhythmia or tumor formation and superior to hESC-CMs for the improvement of cardiac function in HF. This is due to their immunomodulation that improves in vivo survival and enhanced angiogenesis via paracrine effects.
Collapse
Affiliation(s)
- Songyan Liao
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Yuelin Zhang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China.,Department of Emergency, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Sherwin Ting
- Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), Singapore, 138668, Singapore
| | - Zhe Zhen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Fan Luo
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China
| | - Ziyi Zhu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China
| | - Yu Jiang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Sijia Sun
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Qizhou Lian
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China. .,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China.
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Rm 1928, Block K, Hong Kong SAR, China. .,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen, China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China. .,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China.
| |
Collapse
|
185
|
Buja LM. Cardiac repair and the putative role of stem cells. J Mol Cell Cardiol 2019; 128:96-104. [DOI: 10.1016/j.yjmcc.2019.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/30/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2023]
|
186
|
Premer C, Kanelidis AJ, Hare JM, Schulman IH. Rethinking Endothelial Dysfunction as a Crucial Target in Fighting Heart Failure. Mayo Clin Proc Innov Qual Outcomes 2019; 3:1-13. [PMID: 30899903 PMCID: PMC6408687 DOI: 10.1016/j.mayocpiqo.2018.12.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is characterized by nitric oxide dysregulation and an altered redox state. Oxidative stress and inflammatory markers prevail, thus promoting atherogenesis and hypertension, important risk factors for the development and progression of heart failure. There has been a reemerging interest in the role that endothelial dysfunction plays in the failing circulation. Accordingly, patients with heart failure are being clinically assessed for endothelial dysfunction via various methods, including flow-mediated vasodilation, peripheral arterial tonometry, quantification of circulating endothelial progenitor cells, and early and late endothelial progenitor cell outgrowth measurements. Although the mechanisms underlying endothelial dysfunction are intimately related to cardiovascular disease and heart failure, it remains unclear whether targeting endothelial dysfunction is a feasible strategy for ameliorating heart failure progression. This review focuses on the pathophysiology of endothelial dysfunction, the mechanisms linking endothelial dysfunction and heart failure, and the various diagnostic methods currently used to measure endothelial function, ultimately highlighting the therapeutic implications of targeting endothelial dysfunction for the treatment of heart failure.
Collapse
Key Words
- Ach, acetylcholine
- CAD, coronary artery disease
- CVD, cardiovascular disease
- ECFC, endothelial colony-forming cell
- EDHF, endothelium-derived hyperpolarizing factor
- EPC, endothelial progenitor cell
- EPC-CFU, EPC–colony-forming unit
- FMD, flow-mediated vasodilation
- H2O2, hydrogen peroxide
- HF, heart failure
- HFpEF, HF with preserved ejection fraction
- HFrEF, HF with reduced ejection fraction
- IVUS, intravascular ultrasound
- LVEF, left ventricular ejection fraction
- NO, nitric oxide
- NOS, NO synthase
- PAT, peripheral arterial tonometry
- QCA, quantitative coronary angiography
- ROS, reactive oxygen species
- cGMP, cyclic guanosine monophosphate
- eNOS, endothelial nitric oxide synthase
Collapse
Affiliation(s)
- Courtney Premer
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL.,Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
187
|
Fisher SA, Cutler A, Doree C, Brunskill SJ, Stanworth SJ, Navarrete C, Girdlestone J. Mesenchymal stromal cells as treatment or prophylaxis for acute or chronic graft-versus-host disease in haematopoietic stem cell transplant (HSCT) recipients with a haematological condition. Cochrane Database Syst Rev 2019; 1:CD009768. [PMID: 30697701 PMCID: PMC6353308 DOI: 10.1002/14651858.cd009768.pub2] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recipients of allogeneic haematopoietic stem cell transplants (HSCT) can develop acute or chronic, or both forms of graft-versus-host disease (a/cGvHD), whereby immune cells of the donor attack host tissues. Steroids are the primary treatment, but patients with severe, refractory disease have limited options and a poor prognosis. Mesenchymal stromal cells (MSCs) exhibit immunosuppressive properties and are being tested in clinical trials for their safety and efficacy in treating many immune-mediated disorders. GvHD is one of the first areas in which MSCs were clinically applied, and it is important that the accumulating evidence is systematically reviewed to assess whether their use is favoured. OBJECTIVES To determine the evidence for the safety and efficacy of MSCs for treating immune-mediated inflammation post-transplantation of haematopoietic stem cells. SEARCH METHODS We searched for randomised controlled trials (RCTs) in the Cochrane Central Register of Controlled Trials (CENTRAL, the Cochrane Library 2018, Issue 12), MEDLINE (from 1946), Embase (from 1974), CINAHL (from 1937), Web of Science: Conference Proceedings Citation Index-Science (CPCI-S) (from 1990) and ongoing trial databases to 6 December 2018. No constraints were placed on language or publication status. SELECTION CRITERIA We included RCTs of participants with a haematological condition who have undergone an HSCT as treatment for their condition and were randomised to MSCs (intervention arm) or no MSCs (comparator arm), to prevent or treat GvHD. We also included RCTs which compared different doses of MSCs or MSCs of different sources (e.g. bone marrow versus cord). We included MSCs co-transplanted with haematopoietic stem cells as well as MSCs administered post-transplantation of haematopoietic stem cells. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane.We employed a random-effects model for all analyses due to expected clinical heterogeneity arising from differences in participant characteristics and interventions. MAIN RESULTS We identified 12 completed RCTs (879 participants), and 13 ongoing trials (1532 enrolled participants planned). Of 12 completed trials, 10 compared MSCs versus no MSCs and two compared different doses of MSCs. One trial was in people with thalassaemia major, the remaining trials were for haematological malignancies. Seven trials administered MSCs to prevent GvHD, whereas five trials gave MSCs to treat GvHD.In the comparison of MSCs with no MSCs, cells were administered at a dose of between 105 and 107 cells/kg in either a single dose (six trials) or in multiple doses (four trials) over a period of three days to four months. The dose-comparison trials compared 2 x 106 cells/kg with 8 x 106 cells/kg in two infusions, or 1 x 106 cells/kg with 3 x 106 cells/kg in a single infusion.The median duration of follow-up in seven trials which administered MSCs prophylactically ranged from 10 to 60 months. In three trials of MSCs as treatment for aGvHD, participants were followed up for 90 or 100 days. In two trials of MSCs as treatment for cGvHD, the mean duration of follow-up was 13.4 months (MSC group) and 23.6 months (control group) in one trial, and 56 weeks in the second trial. Five trials included adults only, six trials included adults and children, and one trial included children only. In eight trials which reported the gender distribution, the percentage of females ranged from 20% to 59% (median 35.8%).The overall quality of the included studies was low: randomisation methods were poorly reported and several of the included studies were subject to a high risk of performance bias and reporting bias. One trial which started in 2008 has not been published and the progress of this trial in unknown, leading to potential publication bias. The quality of evidence was therefore low or very low for all outcomes due to a high risk of bias as well as imprecision due to the low number of overall participants, and in some cases evidence based on a single study. We found that MSCs may make little or no difference in the risk of all-cause mortality in either prophylactic trials (HR 0.85, 95% CI 0.50 to 1.42; participants = 301; studies = 5; I2 = 34% ; low-quality evidence) or therapeutic trials (HR 1.12, 95% CI 0.80 to 1.56; participants = 244; studies = 1; very low-quality evidence), and no difference in the risk of relapse of malignant disease (prophylactic trials: RR 1.08, 95% CI 0.73 to 1.59; participants = 323; studies = 6; I2 = 0%; low-quality evidence) compared with no MSCs. MSCs were well-tolerated, no infusion-related toxicity or ectopic tissue formation was reported. No study reported health-related quality of life. In prophylactic trials, MSCs may reduce the risk of chronic GvHD (RR 0.66, 95% CI 0.49 to 0.89; participants = 283; studies = 6; I2 = 0%; low-quality evidence). This means that only 310 (95% CI 230 to 418) in every 1000 patients in the MSC arm are expected to develop chronic GvHD compared to 469 in the control arm. However, MSCs may make little or no difference to the risk of aGvHD (RR 0.86, 95% CI 0.63 to 1.17; participants = 247; studies = 6; I2 = 0%; low-quality evidence). In GvHD therapeutic trials, we are very uncertain whether MSCs improve complete response of either aGvHD (RR 1.16, 95% CI 0.79 to 1.70, participants = 260, studies = 1; very low-quality evidence) or cGvHD (RR 5.00, 95%CI 0.75 to 33.21, participants = 40, studies = 1; very low-quality evidence).In two trials which compared different doses of MSCs, we found no evidence of any differences in outcomes. AUTHORS' CONCLUSIONS MSCs are an area of intense research activity, and an increasing number of trials have been undertaken or are planned. Despite a number of reports of positive outcomes from the use of MSCs for treating acute GvHD, the evidence to date from RCTs has not supported the conclusion that they are an effective therapy. There is low-quality evidence that MSCs may reduce the risk of cGvHD. New trial evidence will be incorporated into future updates of this review, which may better establish a role for MSCs in the prevention or treatment of GvHD.
Collapse
Affiliation(s)
- Sheila A Fisher
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Antony Cutler
- NHS Blood and TransplantHistocompatibility & Immunogenetics Research GroupLondonUK
| | - Carolyn Doree
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Susan J Brunskill
- NHS Blood and TransplantSystematic Review InitiativeLevel 2, John Radcliffe HospitalHeadingtonOxfordOxonUKOX3 9BQ
| | - Simon J Stanworth
- Oxford University Hospitals NHS Foundation Trust and University of OxfordNational Institute for Health Research (NIHR) Oxford Biomedical Research CentreJohn Radcliffe Hospital, Headley WayHeadingtonOxfordUKOX3 9BQ
| | | | - John Girdlestone
- University College LondonDivision of Infection and ImmunityLondonUK
- NHS Blood and TransplantStem Cells and ImmunotherapiesHeadley WayOxfordUKOX3 9BQ
| | | |
Collapse
|
188
|
Liu YW, Fang YH, Su CT, Hwang SM, Liu PY, Wu SN. The biochemical and electrophysiological profiles of amniotic fluid-derived stem cells following Wnt signaling modulation cardiac differentiation. Cell Death Discov 2019; 5:59. [PMID: 30701091 PMCID: PMC6349909 DOI: 10.1038/s41420-019-0143-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/31/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Owing to the beneficial properties of amniotic fluid-derived stem cells (AFSCs), including pluripotency and the lack of ethical issues associated with embryonic stem cells (ESCs), they should be a promising cell source for regenerative medicine. However, how to differentiate AFSCs into contracting cardiomyocytes has not been established. In this study, a well-established, direct cardiac differentiation protocol involving the modulation of Wnt signaling was used to differentiate Oct 3/4+ AFSCs into cardiomyocytes. By day 14 of cardiomyocyte differentiation, these AFSCs expressed cardiac-specific genes (i.e., cardiac troponin T and myosin light chain 2v) and proteins but could not spontaneously contract. Using the patch-clamp technique, we further characterized the electrophysiological properties of human ESC-derived cardiomyocytes (hESC-CMs) and differentiated AFSCs. We used different configurations to investigate membrane potentials and ion currents in differentiated AFSCs and hESC-CMs. Under cell-attached voltage- or whole-cell current-clamp modes, we recorded spontaneous action currents (ACs) or action potentials (APs) in hESC-CMs but not in differentiated AFSCs. Compared to hESC-CMs, differentiated AFSCs showed significantly diminished activity of both BKCa and IKCa channels, which might lead to a lack of spontaneous ACs and APs in differentiated AFSCs. These results indicated that this well-established Wnt signaling modulating cardiac differentiation protocol was insufficient to induce the differentiation of functional cardiomyocytes from Oct 3/4+ AFSCs. Therefore, AFSC may not be an ideal candidate for cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Rd. North District, Tainan, 70403 Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsein Fang
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Ting Su
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yun-Lin, Taiwan
| | - Shiaw-Min Hwang
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Ping-Yen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, 138 Sheng-Li Rd. North District, Tainan, 70403 Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, 1 University Rd, East District, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
189
|
Hagmeijer MH, Vonk LA, Kouwenhoven JW, Custers RJ, Bleys RL, Krych AJ, Saris DB. Surgical Feasibility of a One-Stage Cell-Based Arthroscopic Procedure for Meniscus Regeneration: A Cadaveric Study. Tissue Eng Part C Methods 2018; 24:688-696. [PMID: 30398399 PMCID: PMC7615694 DOI: 10.1089/ten.tec.2018.0240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
IMPACT STATEMENT Meniscus injury remains the most common indication for orthopedic surgery, but loss of functioning meniscus tissue is strongly correlated with development of early osteoarthritis. However, current clinical options for tissue engineering of the meniscus are limited. This study demonstrates the feasibility of combining human meniscus cells with mesenchymal stromal cells to enhance a meniscus scaffold for meniscus regeneration in a one-stage solution for partial meniscal deficiency.
Collapse
Affiliation(s)
- Michella H. Hagmeijer
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lucienne A. Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan-Willem Kouwenhoven
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roel J.H. Custers
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ronald L. Bleys
- Department of Anatomy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aaron J. Krych
- Department of Orthopedic Surgery and Sports Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel B.F. Saris
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Orthopedic Surgery and Sports Medicine, Mayo Clinic, Rochester, Minnesota
- MIRA Institute for Technical Medicine, University Twente, Enschede, The Netherlands. Investigation performed at the University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
190
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J Cell Mol Med 2018; 23:720-730. [PMID: 30484934 PMCID: PMC6349184 DOI: 10.1111/jcmm.14035] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 10/20/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is a common, severe emergency case in clinics, with high incidence, significant mortality and increased costs. Despite development in the understanding of its pathophysiology, the therapeutic choices are still confined to dialysis and renal transplantation. Considering their antiapoptotic, immunomodulatory, antioxidative and pro‐angiogenic effects, mesenchymal stem cells (MSCs) may be a promising candidate for AKI management. Based on these findings, some clinical trials have been performed, but the results are contradictory (NCT00733876, NCT01602328). The low engraftment, poor survival rate, impaired paracrine ability and delayed administration of MSCs are the four main reasons for the limited clinical efficacy. Investigators have developed a series of preconditioning strategies to improve MSC survival rates and paracrine ability. In this review, by summarizing these encouraging studies, we intend to provide a comprehensive understanding of various preconditioning strategies on AKI therapy and improve the prognosis of AKI patients by regenerative medicine.
Collapse
Affiliation(s)
- Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ping Zhang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Hua Jiang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
191
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
192
|
Poltavtseva RA, Poltavtsev AV, Lutsenko GV, Svirshchevskaya EV. Myths, reality and future of mesenchymal stem cell therapy. Cell Tissue Res 2018; 375:563-574. [PMID: 30456646 DOI: 10.1007/s00441-018-2961-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell (MSC) therapy represents an alternative approach for tissue regeneration and inflammation control. In spite of a huge amount of preclinical data that has been accumulated on the therapeutic properties of MSCs, there are many conflicting results, possibly due to differences in the properties of MSCs obtained from different sources or underestimated mechanisms of MSC in vivo behavior. This review consolidates the in vivo effects of MSC therapy, discusses the fate of MSCs after intravascular and local delivery and proposes possible trends in MSC therapy.
Collapse
Affiliation(s)
- R A Poltavtseva
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology" Ministry of Healthcare of the Russian Federation, Oparin St, 4, Moscow, Russian Federation, 117997
| | - A V Poltavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997
| | - G V Lutsenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997
| | - E V Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997.
| |
Collapse
|
193
|
Affiliation(s)
- Jay H Traverse
- From the Minneapolis Cardiology Associates, Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, MN; and Cardiovascular Division, University of Minnesota School of Medicine, Minneapolis.
| |
Collapse
|
194
|
Schulman IH, Balkan W, Hare JM. Mesenchymal Stem Cell Therapy for Aging Frailty. Front Nutr 2018; 5:108. [PMID: 30498696 PMCID: PMC6249304 DOI: 10.3389/fnut.2018.00108] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic diseases and degenerative conditions are strongly linked with the geriatric syndrome of frailty and account for a disproportionate percentage of the health care budget. Frailty increases the risk of falls, hospitalization, institutionalization, disability, and death. By definition, frailty syndrome is characterized by declines in lean body mass, strength, endurance, balance, gait speed, activity and energy levels, and organ physiologic reserve. Collectively, these changes lead to the loss of homeostasis and capability to withstand stressors and resulting vulnerabilities. There is a strong link between frailty, inflammation, and the impaired ability to repair tissue injury due to decreases in endogenous stem cell production. Although exercise and nutritional supplementation provide benefit to frail patients, there are currently no specific therapies for frailty. Bone marrow-derived allogeneic mesenchymal stem cells (MSCs) provide therapeutic benefits in heart failure patients irrespective of age. MSCs contribute to cellular repair and tissue regeneration through their multilineage differentiation capacity, immunomodulatory, and anti-inflammatory effects, homing and migratory capacity to injury sites, and stimulatory effect on endogenous tissue progenitors. The advantages of using MSCs as a therapeutic strategy include standardization of isolation and culture expansion techniques and safety in allogeneic transplantation. Based on this evidence, we performed a randomized, double-blinded, dose-finding study in elderly, frail individuals and showed that intravenously delivered allogeneic MSCs are safe and produce significant improvements in physical performance measures and inflammatory biomarkers. We thus propose that frailty can be treated and the link between frailty and chronic inflammation offers a potential therapeutic target, addressable by cell therapy.
Collapse
Affiliation(s)
- Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States.,Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
195
|
Haider KH. Bone marrow cell therapy and cardiac reparability: better cell characterization will enhance clinical success. Regen Med 2018; 13:457-475. [PMID: 29985118 DOI: 10.2217/rme-2017-0134] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nearly two decades of experimental and clinical research with bone marrow cells have paved the way for Phase III pivotal trials in larger groups of heart patients. Despite immense advancements, a multitude of factors are hampering the acceptance of bone marrow cell-based therapy for routine clinical use. These include uncertainties regarding purification and characterization of the cell preparation, delivery protocols, mechanistic understanding and study end points and their methods of assessment. Clinical data show mediocre outcomes in terms of sustained cardiac pump function. This review reasons that the modest outcomes observed in trials thus far are based on quality of the cell preparation with a focus on the chronological aging of cells when autologous cells are used for transplantation in elderly patients.
Collapse
Affiliation(s)
- Khawaja H Haider
- Department of Basic Sciences, Sulaiman AlRajhi Medical School, Al Qassim, Al Bukayria, 51941, Kingdom of Saudi Arabia
| |
Collapse
|
196
|
Ru nanoparticles coated with γ-Fe2O3 promoting and monitoring the differentiation of human mesenchymal stem cells via MRI tracking. Colloids Surf B Biointerfaces 2018; 170:701-711. [DOI: 10.1016/j.colsurfb.2018.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/13/2023]
|
197
|
Dolati S, Yousefi M, Mahdipour M, Afrasiabi Rad A, Pishgahi A, Nouri M, Jodati AR. Mesenchymal stem cell and bone marrow mononuclear cell therapy for cardiomyopathy: From bench to bedside. J Cell Biochem 2018; 120:45-55. [DOI: 10.1002/jcb.27531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Student’s Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Abbas Afrasiabi Rad
- Cardiovascular Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Cardiac Surgery Tabriz University of Medical Tabriz Iran
| | - Alireza Pishgahi
- Department of Physical Medicine and Rehabilitation Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Science Tabriz Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Ahmad Reza Jodati
- Cardiovascular Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Cardiac Surgery Tabriz University of Medical Tabriz Iran
| |
Collapse
|
198
|
Combining Stem Cell Therapy for Advanced Heart Failure and Ventricular Assist Devices: A Review. ASAIO J 2018. [DOI: 10.1097/mat.0000000000000782] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
199
|
Kobayashi K, Suzuki K. Mesenchymal Stem/Stromal Cell-Based Therapy for Heart Failure ― What Is the Best Source? ―. Circ J 2018; 82:2222-2232. [DOI: 10.1253/circj.cj-18-0786] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kazuya Kobayashi
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London
| |
Collapse
|
200
|
Teraa M, Gremmels H, Wijnand JGJ, Verhaar MC. Cell Therapy for Chronic Limb-Threatening Ischemia: Current Evidence and Future Directions. Stem Cells Transl Med 2018; 7:842-846. [PMID: 30070050 PMCID: PMC6265636 DOI: 10.1002/sctm.18-0025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/16/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022] Open
Abstract
Cell‐based therapies have gained interest as a potential treatment method in cardiovascular disease in the past two decades, peripheral artery disease amongst others. Initial pre‐clinical and small pilot clinical studies showed promising effects of cell therapy in peripheral artery disease and chronic limb‐threatening ischemia in particular. However, these promising results were not corroborated in larger high quality blinded randomized trials. This has led to a shift of the field towards more sophisticated cell products, especially mesenchymal stromal cells. Mesenchymal stromal cells have some important benefits, making these cells ideal for regenerative medicine, e.g., potential for allogeneic application, loss of disease‐mediated cell dysfunction, reduced production costs, off‐the‐shelf availability. Future high quality and large clinical studies have to prove the efficacy of mesenchymal stromal cells in the treatment of peripheral artery disease. Stem Cells Translational Medicine2018;7:842–846
Collapse
Affiliation(s)
- Martin Teraa
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joep G J Wijnand
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|