Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Can J Physiol Pharmacol ; 102(3): 161-179, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38226807

RESUMO

Cardiac disorders remain the leading cause of mortality worldwide. Current clinical strategies, including drug therapy, surgical interventions, and organ transplantation offer limited benefits to patients without regenerating the damaged myocardium. Over the past decade, stem cell therapy has generated a keen interest owing to its unique self-renewal and immune privileged characteristics. Furthermore, the ability of stem cells to differentiate into specialized cell types, has made them a popular therapeutic tool against various diseases. This comprehensive review provides an overview of therapeutic potential of different types of stem cells in reference to cardiovascular diseases. Furthermore, it sheds light on the advantages and limitations associated with each cell type. An in-depth analysis of the challenges associated with stem cell research and the hurdles for its clinical translation and their possible solutions have also been elaborated upon. It examines the controversies surrounding embryonic stem cells and the emergence of alternative approaches, such as the use of induced pluripotent stem cells for cardiac therapeutic applications. Overall, this review serves as a valuable resource for researchers, clinicians, and policymakers involved in the field of regenerative medicine, guiding the development of safe and effective stem cell-based therapies to revolutionize patient care.


Assuntos
Cardiopatias , Coração , Humanos , Cardiopatias/terapia , Cardiopatias/metabolismo , Transplante de Células-Tronco , Regeneração , Células-Tronco Embrionárias
2.
Biochim Biophys Acta Mol Basis Dis ; 1869(7): 166806, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37437748

RESUMO

Autophagy maintains cellular homeostasis and plays a crucial role in managing pathological conditions including ischemic myocardial injury leading to heart failure (HF). Despite treatments, no intervention can replace lost cardiomyocytes. Stem cell therapy offers potential for post-myocardial infarction repair but struggles with poor cell retention due to immune rejection. In the search for effective therapies, stem cell-derived extracellular vesicles (EVs), especially exosomes, have emerged as promising tools. These tiny bioactive molecule carriers play vital roles in intercellular communication and tissue engineering. They offer numerous therapeutic benefits including modulating immune responses, promoting tissue repair, and boosting angiogenesis. Additionally, biomaterials provide a conducive 3D microenvironment for cell, exosome, and biomolecule delivery, and enhance heart muscle strength, making it a comprehensive cardiac repair strategy. In this regard, the current review delves into the intricate application of extracellular vesicles (EVs) and biomaterials for managing autophagy in the heart muscle during cardiac injury. Central to our investigation is the exploration of how these elements interact within the context of cardiac repair and regeneration. Additionally, this review also casts light on the formidable challenges that plague this field, such as the issues of safety, efficacy, controlled delivery, and acceptance of these therapeutic strategies for effective clinical translation. Addressing these challenges is crucial for unlocking the full therapeutic potential of EV and biomaterial-based therapies and ensuring their successful translation from bench to bedside.


Assuntos
Exossomos , Traumatismos Cardíacos , Humanos , Materiais Biocompatíveis/uso terapêutico , Miócitos Cardíacos , Células-Tronco , Autofagia
4.
Sci Adv ; 8(14): eabl4370, 2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35394834

RESUMO

A "Leap-of-Faith" approach is used to treat patients with previously unknown ultrarare pathogenic mutations, often based on evidence from patients having dissimilar but more prevalent mutations. This uncertainty reflects the need to develop personalized prescreening platforms for these patients to assess drug efficacy before considering clinical trial enrollment. In this study, we report an 18-year-old patient with ultrarare Leigh-like syndrome. This patient had previously participated in two clinical trials with unfavorable responses. We established an induced pluripotent stem cell (iPSC)-based platform for this patient, and assessed the efficacy of a panel of drugs. The iPSC platform validated the safety and efficacy of the screened drugs. The efficacy of three of the screened drugs was also investigated in the patient. After 3 years of treatment, the drugs were effective in shifting the metabolic profile of this patient toward healthy control. Therefore, this personalized iPSC-based platform can act as a prescreening tool to help in decision-making with respect to patient's participation in future clinical trials.


Assuntos
Células-Tronco Pluripotentes Induzidas , Adolescente , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo
5.
Mol Cell Biochem ; 477(6): 1681-1695, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35235124

RESUMO

A significantly high percentage of hospitalized COVID-19 patients with diabetes mellitus (DM) had severe conditions and were admitted to ICU. In this review, we have delineated the plausible molecular mechanisms that could explain why there are increased clinical complications in patients with DM that become critically ill when infected with SARS-CoV2. RNA viruses have been classically implicated in manifestation of new onset diabetes. SARS-CoV2 infection through cytokine storm leads to elevated levels of pro-inflammatory cytokines creating an imbalance in the functioning of T helper cells affecting multiple organs. Inflammation and Th1/Th2 cell imbalance along with Th17 have been associated with DM, which can exacerbate SARS-CoV2 infection severity. ACE-2-Ang-(1-7)-Mas axis positively modulates ß-cell and cardiac tissue function and survival. However, ACE-2 receptors dock SARS-CoV2, which internalize and deplete ACE-2 and activate Renin-angiotensin system (RAS) pathway. This induces inflammation promoting insulin resistance that has positive effect on RAS pathway, causes ß-cell dysfunction, promotes inflammation and increases the risk of cardiovascular complications. Further, hyperglycemic state could upregulate ACE-2 receptors for viral infection thereby increasing the severity of the diabetic condition. SARS-CoV2 infection in diabetic patients with heart conditions are linked to worse outcomes. SARS-CoV2 can directly affect cardiac tissue or inflammatory response during diabetic condition and worsen the underlying heart conditions.


Assuntos
COVID-19 , Doenças Cardiovasculares , Diabetes Mellitus , Enzima de Conversão de Angiotensina 2 , COVID-19/complicações , Sobrevivência Celular , Síndrome da Liberação de Citocina , Humanos , RNA Viral , SARS-CoV-2
6.
Can J Physiol Pharmacol ; 99(2): 140-150, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33559528

RESUMO

Ischemic heart disease is among the primary causes of cardiovascular-related deaths worldwide. Conventional treatments including surgical interventions and medical therapies aid in preventing further damage to heart muscle but are unable to provide a permanent solution. In recent years, stem cell therapy has emerged as an attractive alternative to restore damaged myocardium after myocardial injury. Allogeneic (donor-derived) mesenchymal stem cells (MSCs) have shown great promise in preclinical and clinical studies, making them the most widely accepted candidates for cardiac cell therapy. MSCs promote cardiac repair by modulating host immune system and secreting various soluble factors, of which prostaglandin E2 (PGE2) is an important one. PGE2 plays a significant role in regulating cardiac remodeling following myocardial injury. In this review, we provide an overview of allogeneic MSCs as candidates for myocardial regeneration with a focus on the role of the PGE2/cyclooxygenase-2 (COX2) pathway in mediating these effects.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Dinoprostona/metabolismo , Transplante de Células-Tronco Mesenquimais , Animais , Humanos , Transplante Homólogo
7.
Adv Funct Mater ; 31(46): 2106786, 2021 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-35153642

RESUMO

MXene nanomaterials have sparked significant interest among interdisciplinary researchers to tackle today's medical challenges. In particular, colloidal MXene quantum dots (MQDs) offer the high specific surface area and compositional flexibility of MXene while providing improvements to aqueous stability and material-cell interactions. The current study for the first time reports the development and application of immunoengineered tantalum-carbide (Ta4C3T x ) MQDs for in vivo treatment of transplant vasculopathy. This report comes at a critical juncture in the field as poor long-term safety of other MXene compositions challenge the eventual clinical translatability of these materials. Using rational design and synthesis strategies, the Ta4C3T x MQDs leverage the intrinsic anti-inflammatory and antiapoptotic properties of tantalum to provide a novel nanoplatform for biomedical engineering. In particular, these MQDs are synthesized with high efficiency and purity using a facile hydrofluoric acid-free protocol and are enriched with different bioactive functional groups and stable surface TaO2 and Ta2O5. Furthermore, MQDs are spontaneously uptaken into antigen-presenting endothelial cells and alter surface receptor expression to reduce their activation of allogeneic T-lymphocytes. Finally, when applied in vivo, Ta4C3T x MQDs ameliorate the cellular and structural changes of early allograft vasculopathy. These findings highlight the robust potential of tailored Ta4C3T x MQDs for future applications in medicine.

8.
FASEB J ; 34(11): 15236-15251, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32959405

RESUMO

Allogeneic mesenchymal stem cells (MSCs) from young and healthy donors are reported to hold the potential to treat several immunological and degenerative disorders. However, recent data from animal studies and clinical trials demonstrate that immunogenicity and poor survival of transplanted MSCs impaired the efficacy of cells for regenerative applications. It is reported that initially immunoprivileged under in vitro conditions, MSCs are targeted by the host immune system after transplantation in the ischemic tissues in vivo. We performed in vitro (in MSCs) and in vivo (in the rat model of myocardial infarction [MI]) studies to elucidate the mechanisms responsible for the change in the immunophenotype of MSCs from immunoprivileged to immunogenic under ischemic conditions. We have recently reported that a soluble factor prostaglandin E2 (PGE2) preserves the immunoprivilege of allogeneic MSCs. In the current study, we found that PGE2 levels, which were elevated during normoxia, decreased in MSCs following exposure to hypoxia. Further, we found that proteasome-mediated degradation of cyclooxygenase-2 (COX2, rate-limiting enzyme in PGE2 biosynthesis) in hypoxic MSCs is responsible for PGE2 decrease and loss of immunoprivilege of MSCs. While investigating the mechanisms of COX2 degradation in hypoxic MSCs, we found that in normoxic MSCs, COP9 signalosome subunit 5 (CSN5) binds to COX2 and prevents its degradation by the proteasome. However, exposure to hypoxia leads to a decrease in CSN5 levels and its binding to COX2, rendering COX2 protein susceptible to proteasome-mediated degradation. This subsequently causes PGE2 downregulation and loss of immunoprivilege of MSCs. Maintaining COX2 levels in MSCs preserves immunoprivilege in vitro and improves the survival of transplanted MSCs in a rat model of MI. These data provide novel mechanistic evidence that PGE2 is downregulated in hypoxic MSCs which is responsible for the post-transplantation rejection of allogeneic MSCs. Therefore, our data suggest that the new strategies that target CSN5-COX2 signaling may improve survival and utility of transplanted allogeneic MSCs in the ischemic heart.


Assuntos
Ciclo-Oxigenase 2/química , Hipóxia/fisiopatologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/imunologia , Animais , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Ratos , Ratos Sprague-Dawley , Transplante Homólogo
9.
FASEB J ; 34(9): 12860-12876, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32770803

RESUMO

Allogeneic mesenchymal stem cells (MSCs) from young and healthy donors are immunoprivileged and have the potential to treat numerous degenerative diseases. However, recent reviews of clinical trials report poor long-term survival of transplanted cells in the recipient that turned down the enthusiasm regarding MSC therapies. Increasing evidence now confirm that though initially immunoprivileged, MSCs eventually become immunogenic after transplantation in the ischemic or hypoxic environment of diseased tissues and are rejected by the host immune system. We performed in vitro (in rat and human cells) and in vivo (in a rat model) investigations to understand the mechanisms of the immune switch in the phenotype of MSCs. The immunoprivilege of MSCs is preserved by the absence of cell surface immune antigen, major histocompatibility complex II (MHC-II) molecule. We found that the ATPase subunit of 19S proteasome "Sug1" regulates MHC-II biosynthesis in MSCs. Exposure to hypoxia upregulates Sug1 in MSCs and its binding to class II transactivator (CIITA), a coactivator of MHC-II transcription. Sug1 binding to CIITA in hypoxic MSCs promotes the acetylation and K63 ubiquitination of CIITA leading to its activation and translocation to the nucleus, and ultimately MHC-II upregulation. In both rat and human MSCs, knocking down Sug1 inactivated MHC-II and preserved immunoprivilege even following hypoxia. In a rat model of myocardial infarction, transplantation of Sug1-knockdown MSCs in ischemic heart preserved immunoprivilege and improved the survival of transplanted cells. Therefore, the current study provides novel mechanisms of post-transplantation loss of immunoprivilege of MSCs. This study may help in facilitating better planning for future clinical trials.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , Hipóxia , Transplante de Células-Tronco Mesenquimais , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transativadores/metabolismo , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Privilégio Imunológico , Leucócitos/citologia , Leucócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Wistar
10.
Cell Death Dis ; 11(6): 419, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32499535

RESUMO

Allogeneic mesenchymal stem cells (MSCs) are immunoprivileged and are being investigated in phase I and phase II clinical trials to treat different degenerative and autoimmune diseases. In spite of encouraging outcome of initial trials, the long-term poor survival of transplanted cells in the host tissue has declined the overall enthusiasm. Recent analyses of allogeneic MSCs based studies confirm that after transplantation in the hypoxic or ischemic microenvironment of diseased tissues, MSCs become immunogenic and are rejected by recipient immune system. The immunoprivilege of MSCs is preserved by absence or negligible expression of cell surface antigen, human leukocyte antigen (HLA)-DRα. We found that in normoxic MSCs, 26S proteasome degrades HLA-DRα and maintains immunoprivilege of MSCs. The exposure to hypoxia leads to inactivation of 26S proteasome and formation of immunoproteasome in MSCs, which is associated with upregulation and activation of HLA-DRα, and as a result, MSCs become immunogenic. Furthermore, inhibition of immunoproteasome formation in hypoxic MSCs preserves the immunoprivilege. Therefore, hypoxia-induced shift in the phenotype of proteasome from 26S toward immunoproteasome triggers loss of immunoprivilege of allogeneic MSCs. The outcome of the current study may provide molecular targets to plan interventions to preserve immunoprivilege of allogeneic MSCs in the hypoxic or ischemic environment.


Assuntos
Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Complexo de Endopeptidases do Proteassoma/imunologia , Hipóxia Celular/imunologia , Regulação para Baixo , Cadeias alfa de HLA-DR/metabolismo , Humanos , Fenótipo , Subunidades Proteicas/metabolismo , Proteólise , Regulação para Cima
11.
Adv Healthc Mater ; 8(16): e1900569, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31265217

RESUMO

Inflammation is tightly linked to tissue injury. In regenerative medicine, immune activation plays a key role in rejection of transplanted stem cells and reduces the efficacy of stem cell therapies. Next-generation smart biomaterials are reported to possess multiple biologic properties for tissue repair. Here, the first use of 0D titanium carbide (Ti3 C2 ) MXene quantum dots (MQDs) for immunomodulation is presented with the goal of enhancing material-based tissue repair after injury. MQDs possess intrinsic immunomodulatory properties and selectively reduce activation of human CD4+ IFN-γ+ T-lymphocytes (control 87.1 ± 2.0%, MQDs 68.3 ± 5.4%) while promoting expansion of immunosuppressive CD4+ CD25+ FoxP3+ regulatory T-cells (control 5.5 ± 0.7%, MQDs 8.5 ± 0.8%) in a stimulated lymphocyte population. Furthermore, MQDs are biocompatible with bone marrow-derived mesenchymal stem cells and induced pluripotent stem cell-derived fibroblasts. Finally, Ti3 C2 MQDs are incorporated into a chitosan-based hydrogel to create a 3D platform with enhanced physicochemical properties for stem cell delivery and tissue repair. This composite hydrogel demonstrates increased conductivity while maintaining injectability and thermosensitivity. These findings suggest that this new class of biomaterials may help bridge the translational gap in material and stem cell-based therapies for tissue repair and treatment of inflammatory and degenerative diseases.


Assuntos
Imunomodulação/efeitos dos fármacos , Pontos Quânticos , Medicina Regenerativa/métodos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
12.
Cell Death Dis ; 10(2): 90, 2019 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692516

RESUMO

Bone marrow-derived allogeneic (donor derived) mesenchymal stem cells (MSCs) are immunoprivileged and are considered to be prominent candidates for regenerative therapy for numerous degenerative diseases. Even though the outcome of initial allogeneic MSCs based clinical trials was encouraging, the overall enthusiasm, of late, has dimmed down. This is due to failure of long-term survival of transplanted cells in the recipient. In fact, recent analyses of allogeneic MSC-based studies demonstrated that cells after transplantation turned immunogenic and were subsequently rejected by host immune system. The current study reveals a novel mechanism of immune switch in MSCs. We demonstrate that hypoxia, a common denominator of ischemic tissues, induces an immune shift in MSCs from immunoprivileged to immunogenic state. The immunoprivilege of MSCs is preserved by downregulation or the absence of major histocompatibility complex class II (MHC-II) molecules. We found that 26S proteasome-mediated intracellular degradation of MHC-II helps maintain the absence of MHC-II expression on cell surface in normoxic MSCs and preserves their immunoprivilege. The exposure to hypoxia leads to dissociation of 19S and 20S subunits, and inactivation of 26S proteasome. This prevented the degradation of MHC-II and, as a result, the MSCs became immunogenic. Furthermore, we found that hypoxia-induced decrease in the levels of a chaperon protein HSP90α is responsible for inactivation of 26S proteasome. Maintaining HSP90α levels in hypoxic MSCs preserved the immunoprivilege of MSCs. Therefore, hypoxia-induced inactivation of 26S proteasome assembly instigates loss of immunoprivilege of allogeneic mesenchymal stem cells. Maintaining 26S proteasome activity in mesenchymal stem cells preserves their immunoprivilege.


Assuntos
Hipóxia Celular/imunologia , Células-Tronco Mesenquimais/imunologia , Complexo de Endopeptidases do Proteassoma/genética , Humanos
13.
Can J Physiol Pharmacol ; 97(6): 536-543, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30422687

RESUMO

Increasing reports of successful and safe application of bone marrow derived mesenchymal stem cells (BM-MSCs) for cell therapy are pouring in from numerous studies. However poor survival of transplanted cells in the recipient has impaired the benefits of BM-MSCs based therapies. Therefore cell product preparation procedures pertaining to MSC therapy need to be optimized to improve the survival of transplanted cells. One of the important ex vivo procedures in the preparation of cells for therapy is passaging of BM-MSCs to ensure a suitable number of cells for transplantation, which may affect the turnover of proteins involved in regulation of cell survival and (or) death pathways. In the current study, we investigated the effect of an increase in passage number of BM-MSCs in cell culture on the intracellular protein turnover (protein synthesis, processing, and degradation machinery). We performed proteomic analysis of BM-MSCs at different passages. There was no significant difference observed in the ribosomal, protein processing, and proteasomal pathways related proteins in BM-MSCs with an increase in passage number from P3 to P7. Therefore, expansion of MSCs in the cell culture in clinically relevant passages (Passage 3-7) does not affect the quality of MSCs in terms of intracellular protein synthesis and turnover.


Assuntos
Células-Tronco Mesenquimais/citologia , Biossíntese de Proteínas , Proteômica , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Retículo Endoplasmático/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais , Ratos , Ratos Sprague-Dawley , Ribossomos/metabolismo
14.
Compr Physiol ; 9(1): 75-125, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30549015

RESUMO

Cardiovascular disease leading to heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Improved pharmacological and interventional coronary procedures have led to improved outcomes following acute myocardial infarction. This success has translated into an unforeseen increased incidence in HF. This review summarizes the signaling pathways implicated in the transition to HF following cardiac injury. In addition, we provide an update on cell death signaling and discuss recent advances in cardiac fibrosis as an independent event leading to HF. Finally, we discuss cell-based therapies and their possible use to avert the deteriorating nature of HF. © 2019 American Physiological Society. Compr Physiol 9:75-125, 2019.


Assuntos
Insuficiência Cardíaca/metabolismo , Medicina Regenerativa/métodos , Transdução de Sinais , Engenharia Tecidual/métodos , Animais , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/terapia , Humanos , Miocárdio/metabolismo , Miocárdio/patologia
15.
Sci Rep ; 8(1): 15069, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305684

RESUMO

Abnormal conduction and improper electrical impulse propagation are common in heart after myocardial infarction (MI). The scar tissue is non-conductive therefore the electrical communication between adjacent cardiomyocytes is disrupted. In the current study, we synthesized and characterized a conductive biodegradable scaffold by incorporating graphene oxide gold nanosheets (GO-Au) into a clinically approved natural polymer chitosan (CS). Inclusion of GO-Au nanosheets in CS scaffold displayed two fold increase in electrical conductivity. The scaffold exhibited excellent porous architecture with desired swelling and controlled degradation properties. It also supported cell attachment and growth with no signs of discrete cytotoxicity. In a rat model of MI, in vivo as well as in isolated heart, the scaffold after 5 weeks of implantation showed a significant improvement in QRS interval which was associated with enhanced conduction velocity and contractility in the infarct zone by increasing connexin 43 levels. These results corroborate that implantation of novel conductive polymeric scaffold in the infarcted heart improved the cardiac contractility and restored ventricular function. Therefore, our approach may be useful in planning future strategies to construct clinically relevant conductive polymer patches for cardiac patients with conduction defects.


Assuntos
Medicamentos de Ervas Chinesas/química , Ouro/química , Grafite/química , Contração Miocárdica , Infarto do Miocárdio , Nanoestruturas/química , Alicerces Teciduais/química , Animais , Masculino , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Ratos , Ratos Wistar
16.
Stem Cell Res Ther ; 9(1): 121, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29720263

RESUMO

BACKGROUND: Bone marrow-derived allogeneic mesenchymal stem cells (MSCs) from young healthy donors are immunoprivileged and their clinical application for regenerative medicine is under evaluation. However, data from preclinical and initial clinical trials indicate that allogeneic MSCs after transplantation provoke a host immune response and are rejected. In the current study, we evaluated the effect of an increase in passage number in cell culture on immunoprivilege of the MSCs. Since only limited numbers of MSCs can be sourced at a time from a donor, it is imperative to expand them in culture to meet the necessary numbers required for cell therapy. Presently, the most commonly used passages for transplantation include passages (P)3-7. Therefore, in this study we included clinically relevant passages, i.e., P3, P5, and P7, for evaluation. METHODS: The immunoprivilege of MSCs was assessed with the mixed leukocyte reaction assay, where rat MSCs were cocultured with peripheral blood leukocytes for 72 h. Leukocyte-mediated cytotoxicity, apoptosis (Bax/Bcl-xl ratio), leukocyte proliferation, and alterations in cellular bioenergetics in MSCs were assessed after the coculture. Furthermore, the expression of various oxidized phospholipids (oxidized phosphatidylcholine (ox-PC)) was analyzed in MSCs using a lipidomic platform. To determine if the ox-PCs were acting in tandem with downstream intracellular protein alterations, we performed proteome analysis using a liquid chromatography/mass spectrometry (LC/MS) proteomic platform. RESULTS: Our data demonstrate that MSCs were immunoprivileged at all three passages since coculture with leukocytes did not affect the survival of MSCs at P3, P5, and P7. We also found that, with an increase in the passage number of MSCs, leukocytes did not cause any significant effect on cellular bioenergetics (basal respiration rate, spare respiratory capacity, maximal respiration, and coupling efficiency). Interestingly, in our omics data, we detected alterations in some of the ox-PCs and proteins in MSCs at different passages; however, these changes were not significant enough to affect their immunoprivilege. CONCLUSIONS: The outcome of this study demonstrates that an increase in passage number (from P3 to P7) in the cell culture does not have any significant effect on the immunoprivilege of MSCs.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Proteômica/métodos , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Humanos , Masculino , Ratos , Ratos Sprague-Dawley
17.
Methods Mol Biol ; 1553: 15-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28229404

RESUMO

Mesenchymal stem cells (MSCs) provide an opportunity to bring the field of regenerative medicine to realization. A lot of clinical trials are presently trying to establish their applicability in real-world scenarios. Some of the biggest challenges encountered in bringing MSCs from bench to bedside are the number of MSCs required, their procurement from various sources, and the batch-to-batch variability. This often leads to inconclusive results within and between different studies. Therefore, we have hereby proposed a simple protocol to source mesenchymal stem cells through differentiation of embryonic stem cells.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Adultas , Animais , Biomarcadores , Técnicas de Cultura de Células , Linhagem Celular , Separação Celular/métodos , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Fibroblastos , Imunofenotipagem/métodos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Medicina Regenerativa
18.
Methods Mol Biol ; 1553: 241-248, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28229421

RESUMO

This chapter is based on a simplified method to validate the current preservation procedure of mesenchymal stem cells (MSCs). Currently, there are various media available for freezing and thus preserving the MSCs, making it hard to decide which agent will be apt for cellular requirements. The study describes the effect of two different compositions of freezing media used in regular cell culture experiments, on the morphology, proliferation, and doubling rate of MSCs. Commonly used agents for the cryopreservation of MSCs include DMSO (Dimethyl Sulfoxide) and FBS (Fetal Bovine Serum) and DMEM (Dulbecco's Modified Eagle Medium). To ascertain that the currently used agents do not lead to major changes in the MSC morphology and proliferation, the cells are frozen using the above-mentioned agents in different groups and then their effects analyzed. Thus, the chapter helps to decide what reagents can suit the MSCs, hence minimizing the laboratory to laboratory variability of their characteristics.


Assuntos
Criopreservação/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Separação Celular/métodos , Sobrevivência Celular , Células Cultivadas , Crioprotetores , Camundongos , Ratos
19.
Can J Physiol Pharmacol ; 93(10): 835-41, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26443930

RESUMO

Cardiac injury and loss of cardiomyocytes is a causative as well as a resultant condition of cardiovascular disorders, which are the leading cause of death throughout the world. This loss of cardiomyocytes cannot be completely addressed through the currently available drugs being administered, which mainly function only in relieving the symptoms. There is a huge potential being investigated for regenerative and cell replacement therapies through recruiting stem cells of various origins namely embryonic, reprogramming/induction, and adult tissue. These sources are being actively studied for translation to clinical scenarios. In this review, we attempt to discuss some of these promising scenarios, including the clinical trials and the obstacles that need to be overcome, and hope to address the direction in which stem cell therapy is heading.


Assuntos
Transplante de Medula Óssea/métodos , Reprogramação Celular , Cardiopatias/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Miócitos Cardíacos/transplante , Transplante de Medula Óssea/efeitos adversos , Cardiopatias/patologia , Humanos , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências
20.
Stem Cell Res Ther ; 6: 148, 2015 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-26296856

RESUMO

INTRODUCTION: Doxorubicin (DOX) is a well-known anticancer drug. However its clinical use has been limited due to cardiotoxic effects. One of the major concerns with DOX therapy is its toxicity in patients who are frail, particularly diabetics. Several studies suggest that mesenchymal stem cells (MSCs) have the potential to restore cardiac function after DOX-induced injury. However, limited data are available on the effects of MSC therapy on DOX-induced cardiac dysfunction in diabetics. Our objective was to test the efficacy of bone marrow-derived (BM-MSCs) and adipose-derived MSCs (AT-MSCs) from age-matched humans in a non-immune compromised rat model. METHODS: Diabetes mellitus was induced in rats by streptozotocin injection (STZ, 65 mg/kg b.w, i.p.). Diabetic rats were treated with DOX (doxorubicin hydrochloride, 2.5 mg/kg b.w, i.p) 3 times/wk for 2 weeks (DOX group); or with DOX+ GFP labelled BM-MSCs (2x106cells, i.v.) or with DOX + GFP labelled AT-MSCs (2x106cells, i.v.). Echocardiography and Langendorff perfusion analyses were carried out to determine the heart function. Immunostaining and western blot analysis of the heart tissue was carried out for CD31 and to assess inflammation and fibrosis. Statistical analysis was carried out using SPSS and data are expressed as mean ± SD. RESULTS: Glucose levels in the STZ treated groups were significantly greater than control group. After 4 weeks of intravenous injection, the presence of injected MSCs in the heart was confirmed through fluorescent microscopy and real time PCR for ALU transcripts. Both BM-MSCs and AT-MSCs injection prevented DOX-induced deterioration of %FS, LVDP, dp/dt max and rate pressure product. Staining for CD31 showed a significant increase in the number of capillaries in BM-MSCs and AT-MSCs treated animals in comparison to DOX treated group. Assessment of the inflammation and fibrosis revealed a marked reduction in the DOX-induced increase in immune cell infiltration, collagen deposition and αSMA in the BM-MSCs and AT-MSCs groups. CONCLUSIONS: In conclusion BM-MSCs and AT-MSCs were equally effective in mitigating DOX-induced cardiac damage by promoting angiogenesis, decreasing the infiltration of immune cells and collagen deposition.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Doxorrubicina/toxicidade , Traumatismos Cardíacos/terapia , Coração/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Adulto , Animais , Células Cultivadas , Diabetes Mellitus Experimental , Feminino , Traumatismos Cardíacos/induzido quimicamente , Humanos , Masculino , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA