Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Circulation ; 148(5): 405-425, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37409482

RESUMO

BACKGROUND: Adeno-associated virus (AAV) has emerged as one of the best tools for cardiac gene delivery due to its cardiotropism, long-term expression, and safety. However, a significant challenge to its successful clinical use is preexisting neutralizing antibodies (NAbs), which bind to free AAVs, prevent efficient gene transduction, and reduce or negate therapeutic effects. Here we describe extracellular vesicle-encapsulated AAVs (EV-AAVs), secreted naturally by AAV-producing cells, as a superior cardiac gene delivery vector that delivers more genes and offers higher NAb resistance. METHODS: We developed a 2-step density-gradient ultracentrifugation method to isolate highly purified EV-AAVs. We compared the gene delivery and therapeutic efficacy of EV-AAVs with an equal titer of free AAVs in the presence of NAbs, both in vitro and in vivo. In addition, we investigated the mechanism of EV-AAV uptake in human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and mouse models in vivo using a combination of biochemical techniques, flow cytometry, and immunofluorescence imaging. RESULTS: Using cardiotropic AAV serotypes 6 and 9 and several reporter constructs, we demonstrated that EV-AAVs deliver significantly higher quantities of genes than AAVs in the presence of NAbs, both to human left ventricular and human induced pluripotent stem cell-derived cardiomyocytes in vitro and to mouse hearts in vivo. Intramyocardial delivery of EV-AAV9-sarcoplasmic reticulum calcium ATPase 2a to infarcted hearts in preimmunized mice significantly improved ejection fraction and fractional shortening compared with AAV9-sarcoplasmic reticulum calcium ATPase 2a delivery. These data validated NAb evasion by and therapeutic efficacy of EV-AAV9 vectors. Trafficking studies using human induced pluripotent stem cell-derived cells in vitro and mouse hearts in vivo showed significantly higher expression of EV-AAV6/9-delivered genes in cardiomyocytes compared with noncardiomyocytes, even with comparable cellular uptake. Using cellular subfraction analyses and pH-sensitive dyes, we discovered that EV-AAVs were internalized into acidic endosomal compartments of cardiomyocytes for releasing and acidifying AAVs for their nuclear uptake. CONCLUSIONS: Together, using 5 different in vitro and in vivo model systems, we demonstrate significantly higher potency and therapeutic efficacy of EV-AAV vectors compared with free AAVs in the presence of NAbs. These results establish the potential of EV-AAV vectors as a gene delivery tool to treat heart failure.


Assuntos
Vesículas Extracelulares , Células-Tronco Pluripotentes Induzidas , Humanos , Camundongos , Animais , Dependovirus/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Vetores Genéticos , Células-Tronco Pluripotentes Induzidas/metabolismo , Anticorpos Neutralizantes , Vesículas Extracelulares/metabolismo
2.
bioRxiv ; 2023 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-37034724

RESUMO

Transition between activation and quiescence programs in hematopoietic stem and progenitor cells (HSC/HSPCs) is perceived to be governed intrinsically and by microenvironmental co-adaptation. However, HSC programs dictating both transition and adaptability, remain poorly defined. Single cell multiome analysis divulging differential transcriptional activity between distinct HSPC states, indicated for the exclusive absence of Fli-1 motif from quiescent HSCs. We reveal that Fli-1 activity is essential for HSCs during regenerative hematopoiesis. Fli-1 directs activation programs while manipulating cellular sensory and output machineries, enabling HSPCs co-adoptability with a stimulated vascular niche. During regenerative conditions, Fli-1 presets and enables propagation of niche-derived Notch1 signaling. Constitutively induced Notch1 signaling is sufficient to recuperate functional HSC impairments in the absence of Fli-1. Applying FLI-1 modified-mRNA transduction into lethargic adult human mobilized HSPCs, enables their vigorous niche-mediated expansion along with superior engraftment capacities. Thus, decryption of stem cell activation programs offers valuable insights for immune regenerative medicine.

3.
Methods Mol Biol ; 2573: 77-87, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36040588

RESUMO

Modified mRNA (modRNA) is a safe and effective vector for gene-based therapies. Notably, the safety of modRNA has been validated through COVID-19 vaccines which incorporate modRNA technology to translate spike proteins. Alternative gene delivery methods using plasmids, lentiviruses, adenoviruses, and adeno-associated viruses have suffered from key challenges such as genome integration, delayed and uncontrolled expression, and immunogenic responses. However, modRNA poses no risk of genome integration, has transient and rapid expression, and lacks an immunogenic response. Our lab utilizes modRNA-based therapies to promote cardiac regeneration following myocardial infarction and heart failure. We have also developed and refined an optimized and economical method for synthesis of modRNA. Here, we provide an updated methodology with improved translational efficiency for in vitro and in vivo application.


Assuntos
Vacinas contra COVID-19 , COVID-19 , COVID-19/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Pharmaceutics ; 13(10)2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34683969

RESUMO

Advances in the using in vitro transcribed (IVT) modRNA in the past two decades, especially the tremendous recent success of mRNA vaccines against SARS-CoV-2, have brought increased attention to IVT mRNA technology. Despite its well-known use in infectious disease vaccines, IVT modRNA technology is being investigated mainly in cancer immunotherapy and protein replacement therapy, with ongoing clinical trials in both areas. One of the main barriers to progressing mRNA therapeutics to the clinic is determining how to deliver mRNA to target cells and protect it from degradation. Over the years, many different vehicles have been developed to tackle this issue. Desirable vehicles must be safe, stable and preferably organ specific for successful mRNA delivery to clinically relevant cells and tissues. In this review we discuss various mRNA delivery platforms, with particular focus on attempts to create organ-specific vehicles for therapeutic mRNA delivery.

5.
Cardiovasc Drugs Ther ; 34(6): 871-880, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32822006

RESUMO

Despite various clinical modalities available for patients, heart disease remains among the leading causes of mortality and morbidity worldwide. Genetic medicine, particularly mRNA, has broad potential as a therapeutic. More specifically, mRNA-based protein delivery has been used in the fields of cancer and vaccination, but recent changes to the structural composition of mRNA have led the scientific community to swiftly embrace it as a new drug to deliver missing genes to injured myocardium and many other organs. Modified mRNA (modRNA)-based gene delivery features transient but potent protein translation and low immunogenicity, with minimal risk of insertional mutagenesis. In this review, we compared and listed the advantages of modRNA over traditional vectors for cardiac therapy, with particular focus on using modRNA therapy in cardiac repair. We present a comprehensive overview of modRNA's role in cardiomyocyte (CM) proliferation, cardiac vascularization, and prevention of cardiac apoptosis. We also emphasize recent advances in modRNA delivery strategies and discuss the challenges for its clinical translation.


Assuntos
Terapia Genética , Cardiopatias/terapia , RNA Mensageiro/uso terapêutico , Animais , Proliferação de Células , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Mensageiro/efeitos adversos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica , Regeneração , Fatores de Risco , Resultado do Tratamento
6.
Nat Commun ; 11(1): 4311, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32855420

RESUMO

Pulmonary disease increases the risk of developing abdominal aortic aneurysms (AAA). However, the mechanism underlying the pathological dialogue between the lungs and aorta is undefined. Here, we find that inflicting acute lung injury (ALI) to mice doubles their incidence of AAA and accelerates macrophage-driven proteolytic damage of the aortic wall. ALI-induced HMGB1 leaks and is captured by arterial macrophages thereby altering their mitochondrial metabolism through RIPK3. RIPK3 promotes mitochondrial fission leading to elevated oxidative stress via DRP1. This triggers MMP12 to lyse arterial matrix, thereby stimulating AAA. Administration of recombinant HMGB1 to WT, but not Ripk3-/- mice, recapitulates ALI-induced proteolytic collapse of arterial architecture. Deletion of RIPK3 in myeloid cells, DRP1 or MMP12 suppression in ALI-inflicted mice repress arterial stress and brake MMP12 release by transmural macrophages thereby maintaining a strengthened arterial framework refractory to AAA. Our results establish an inter-organ circuitry that alerts arterial macrophages to regulate vascular remodeling.


Assuntos
Lesão Pulmonar Aguda/complicações , Aneurisma da Aorta Abdominal/patologia , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Remodelação Vascular , Lesão Pulmonar Aguda/patologia , Animais , Aorta Abdominal/citologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Dinaminas/antagonistas & inibidores , Dinaminas/metabolismo , Humanos , Macrófagos/citologia , Metaloproteinase 12 da Matriz/genética , Metaloproteinase 12 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Dinâmica Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Cultura Primária de Células , Proteólise/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Estudos Retrospectivos , Regulação para Cima
7.
Circulation ; 138(25): 2919-2930, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566018

RESUMO

BACKGROUND: For more than a decade, Sca-1+ cells within the mouse heart have been widely recognized as a stem cell population with multipotency that can give rise to cardiomyocytes, endothelial cells, and smooth muscle cells in vitro and after cardiac grafting. However, the developmental origin and authentic nature of these cells remain elusive. METHODS: Here, we used a series of high-fidelity genetic mouse models to characterize the identity and regenerative potential of cardiac resident Sca-1+ cells. RESULTS: With these novel genetic tools, we found that Sca-1 does not label cardiac precursor cells during early embryonic heart formation. Postnatal cardiac resident Sca-1+ cells are in fact a pure endothelial cell population. They retain endothelial properties and exhibit minimal cardiomyogenic potential during development, normal aging and upon ischemic injury. CONCLUSIONS: Our study provides definitive insights into the nature of cardiac resident Sca-1+ cells. The observations challenge the current dogma that cardiac resident Sca-1+ cells are intrinsic stem cells for myocardial development, renewal, and repair, and suggest that the mechanisms of transplanted Sca-1+ cells in heart repair need to be reassessed.


Assuntos
Células-Tronco Adultas/fisiologia , Antígenos Ly/metabolismo , Células Endoteliais/fisiologia , Coração/embriologia , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/fisiologia , Animais , Antígenos Ly/genética , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Células Cultivadas , Desenvolvimento Embrionário , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Modelos Animais , Regeneração , Transplante de Células-Tronco , Cicatrização
8.
Circulation ; 135(1): 59-72, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27803039

RESUMO

BACKGROUND: Epicardial adipose tissue volume and coronary artery disease are strongly associated, even after accounting for overall body mass. Despite its pathophysiological significance, the origin and paracrine signaling pathways that regulate epicardial adipose tissue's formation and expansion are unclear. METHODS: We used a novel modified mRNA-based screening approach to probe the effect of individual paracrine factors on epicardial progenitors in the adult heart. RESULTS: Using 2 independent lineage-tracing strategies in murine models, we show that cells originating from the Wt1+ mesothelial lineage, which includes epicardial cells, differentiate into epicardial adipose tissue after myocardial infarction. This differentiation process required Wt1 expression in this lineage and was stimulated by insulin-like growth factor 1 receptor (IGF1R) activation. IGF1R inhibition within this lineage significantly reduced its adipogenic differentiation in the context of exogenous, IGF1-modified mRNA stimulation. Moreover, IGF1R inhibition significantly reduced Wt1 lineage cell differentiation into adipocytes after myocardial infarction. CONCLUSIONS: Our results establish IGF1R signaling as a key pathway that governs epicardial adipose tissue formation in the context of myocardial injury by redirecting the fate of Wt1+ lineage cells. Our study also demonstrates the power of modified mRNA -based paracrine factor library screening to dissect signaling pathways that govern progenitor cell activity in homeostasis and disease.


Assuntos
Adipócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Infarto do Miocárdio/patologia , Pericárdio/citologia , Receptor IGF Tipo 1/metabolismo , Adipócitos/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Comunicação Parácrina , Reação em Cadeia da Polimerase em Tempo Real , Receptor IGF Tipo 1/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas WT1
9.
Cold Spring Harb Perspect Med ; 5(1): a014035, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25301935

RESUMO

Over the past two decades, a host of new molecular pathways have been uncovered that guide mammalian heart development and disease. The ability to genetically manipulate these pathways in vivo have largely been dependent on the generation of genetically engineered mouse model systems or the transfer of exogenous genes in a variety of DNA vectors (plasmid, adenoviral, adeno-associated viruses, antisense oligonucleotides, etc.). Recently, a new approach to manipulate the gene program of the adult mammalian heart has been reported that will quickly allow the high-efficiency expression of virtually any protein in the intact heart of mouse, rat, porcine, nonhuman primate, and human heart cells via the generation of chemically modified mRNA (modRNA). The technology platform has important implications for delineating the specific paracrine cues that drive human cardiogenesis, and the pathways that might trigger heart regeneration via the rapid generation of modRNA libraries of paracrine factors for direct in vivo administration. In addition, the strategy can be extended to a variety of other cardiovascular tissues and solid organs across multiple species, and recent improvements in the core technology have supported moving toward the first human studies of modRNA in the next 2 years. These recent advances are reviewed along with projections of the potential impact of the technology for a host of other biomedical problems in the cardiovascular system.


Assuntos
Cardiopatias/terapia , Coração/fisiologia , RNA Mensageiro/síntese química , Animais , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Camundongos , Músculo Esquelético/fisiologia , Miocárdio/citologia , Miócitos Cardíacos/fisiologia , Comunicação Parácrina/fisiologia , Proteínas/metabolismo , Ratos , Regeneração/fisiologia , Células-Tronco/fisiologia , Suínos , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Stem Cell Res ; 13(3 Pt B): 693-704, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25043723

RESUMO

The heart has a limited capacity for regeneration following injury. Recent strategies to promote heart regeneration have largely focused on autologous and allogeneic cell-based therapy, where the transplanted cells have been suggested to secrete unknown paracrine factors that are envisioned to promote endogenous repair and/or mobilization of endogenous heart progenitors. Here, we discuss the importance of paracrine mechanisms in facilitating replication of endogenous epicardial progenitor cells in the adult heart and signaling their subsequent reactivation and de novo differentiation into functional cell types such as endothelial cells and cardiomyocytes. Moreover, we discuss the use of a novel modified RNA technology in delivering such therapeutic paracrine factors into myocardium following injury. These studies suggest that modified mRNA may be a valuable experimental tool for the precise in vivo identification of paracrine factors and their downstream signaling that may promote heart repair, cardiac muscle replication, and/or heart progenitor mobilization. In addition, these studies lay the foundation for a new clinically tractable technology for a cell-free approach to promote heart regeneration.


Assuntos
Coração/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , RNA Mensageiro/metabolismo , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Técnicas de Transferência de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , RNA Mensageiro/genética , Regeneração , Transplante de Células-Tronco , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/genética
11.
Cell Res ; 23(10): 1172-86, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24018375

RESUMO

Distinct families of multipotent heart progenitors play a central role in the generation of diverse cardiac, smooth muscle and endothelial cell lineages during mammalian cardiogenesis. The identification of precise paracrine signals that drive the cell-fate decision of these multipotent progenitors, and the development of novel approaches to deliver these signals in vivo, are critical steps towards unlocking their regenerative therapeutic potential. Herein, we have identified a family of human cardiac endothelial intermediates located in outflow tract of the early human fetal hearts (OFT-ECs), characterized by coexpression of Isl1 and CD144/vWF. By comparing angiocrine factors expressed by the human OFT-ECs and non-cardiac ECs, vascular endothelial growth factor (VEGF)-A was identified as the most abundantly expressed factor, and clonal assays documented its ability to drive endothelial specification of human embryonic stem cell (ESC)-derived Isl1+ progenitors in a VEGF receptor-dependent manner. Human Isl1-ECs (endothelial cells differentiated from hESC-derived ISL1+ progenitors) resemble OFT-ECs in terms of expression of the cardiac endothelial progenitor- and endocardial cell-specific genes, confirming their organ specificity. To determine whether VEGF-A might serve as an in vivo cell-fate switch for human ESC-derived Isl1-ECs, we established a novel approach using chemically modified mRNA as a platform for transient, yet highly efficient expression of paracrine factors in cardiovascular progenitors. Overexpression of VEGF-A promotes not only the endothelial specification but also engraftment, proliferation and survival (reduced apoptosis) of the human Isl1+ progenitors in vivo. The large-scale derivation of cardiac-specific human Isl1-ECs from human pluripotent stem cells, coupled with the ability to drive endothelial specification, engraftment, and survival following transplantation, suggest a novel strategy for vascular regeneration in the heart.


Assuntos
Células Endoteliais/citologia , Coração/embriologia , Proteínas com Homeodomínio LIM/genética , Miocárdio/citologia , Células-Tronco/citologia , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Diferenciação Celular , Células Cultivadas , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Proteínas com Homeodomínio LIM/análise , Proteínas com Homeodomínio LIM/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/análise , Fatores de Transcrição/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/genética
12.
Nat Biotechnol ; 31(10): 898-907, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24013197

RESUMO

In a cell-free approach to regenerative therapeutics, transient application of paracrine factors in vivo could be used to alter the behavior and fate of progenitor cells to achieve sustained clinical benefits. Here we show that intramyocardial injection of synthetic modified RNA (modRNA) encoding human vascular endothelial growth factor-A (VEGF-A) results in the expansion and directed differentiation of endogenous heart progenitors in a mouse myocardial infarction model. VEGF-A modRNA markedly improved heart function and enhanced long-term survival of recipients. This improvement was in part due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types. Direct in vivo comparison with DNA vectors and temporal control with VEGF inhibitors revealed the greatly increased efficacy of pulse-like delivery of VEGF-A. Our results suggest that modRNA is a versatile approach for expressing paracrine factors as cell fate switches to control progenitor cell fate and thereby enhance long-term organ repair.


Assuntos
Linhagem da Célula , Infarto do Miocárdio/terapia , Miocárdio/patologia , RNA Mensageiro/metabolismo , Regeneração , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Apoptose , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/patologia , Técnicas de Transferência de Genes , Humanos , Cinética , Luciferases/metabolismo , Camundongos , Modelos Biológicos , Músculo Esquelético/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , RNA Mensageiro/genética , Transplante de Células-Tronco , Análise de Sobrevida , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Blood ; 120(8): 1647-57, 2012 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-22776817

RESUMO

Immature dendritic cells (imDCs) can have a tolerizing effect under normal conditions or after transplantation. However, because of the significant heterogeneity of this cell population, it is extremely difficult to study the mechanisms that mediate the tolerance induced or to harness the application of imDCs for clinical use. In the present study, we describe the generation of a highly defined population of imDCs from hematopoietic progenitors and the direct visualization of the fate of TCR-transgenic alloreactive CD4(+) and CD8(+) T cells after encountering cognate or noncognate imDCs. Whereas CD4(+) T cells were deleted via an MHC-independent mechanism through the NO system, CD8(+) T-cell deletion was found to occur through a unique MHC-dependent, perforin-based killing mechanism involving activation of TLR7 and signaling through Triggering Receptor-1 Expressed on Myeloid cells (TREM-1). This novel subpopulation of perforin-expressing imDCs was also detected in various lymphoid tissues in normal animals and its frequency was markedly enhanced after GM-CSF administration.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Granzimas/imunologia , Células-Tronco Hematopoéticas/imunologia , Glicoproteínas de Membrana/imunologia , Perforina/imunologia , Receptores Imunológicos/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Técnicas de Cultura de Células/métodos , Células Cultivadas , Células Dendríticas/citologia , Feminino , Células-Tronco Hematopoéticas/citologia , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos C57BL , Receptor Gatilho 1 Expresso em Células Mieloides , Quinases da Família src/imunologia
14.
Stem Cells ; 27(11): 2865-74, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19750539

RESUMO

Although mesenchymal stromal cells (MSCs) exhibit marked immunoregulatory activity through multiple mechanisms, their potential to completely evade rejection upon transplantation into allogeneic recipients is controversial. To directly address this controversy, the survival of luciferase-labeled MSCs (Luc(+) MSCs) was evaluated by imaging in allogeneic recipients. This analysis showed that although MSCs exhibited longer survival compared to fibroblasts (Fib), their survival was significantly shorter compared to that exhibited in syngeneic or in immune-deficient Balb-Nude or non-obese diabetic severe combined immunodeficiency (NOD-SCID) recipients. Graft rejection in re-challenge experiments infusing Luc(+) Fib into mice, which had previously rejected Luc(+) MSCs, indicated potential induction of immune memory by the MSCs. This was further analyzed in T-cell antigen receptor (TCR) transgeneic mice in which either CD4 TEA mice or CD8 T cells (2C mice) bear a TCR transgene against a specific MHC I or MHC II, respectively. Thus, following a re-challenge with MSCs expressing the cognate MHC haplotype, an enhanced percentage of 2C CD8(+) or TEA CD4(+) T cells exhibited a memory phenotype (CD122(+), CD44(+), and CD62L(low)). Collectively, these results demonstrate that MSCs are not intrinsically immune-privileged, and under allogeneic settings, these cells induce rejection, which is followed by an immune memory. Considering that the use of allogeneic or even a third party ("off the shelf") MSCs is commonly advocated for a variety of clinical applications, our results strongly suggest that long-term survival of allogeneic MSCs likely represents a major challenge.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/fisiologia , Células Estromais/imunologia , Células Estromais/fisiologia , Transplante Homólogo/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/fisiologia , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Células Estromais/citologia , Linfócitos T/imunologia
15.
PLoS One ; 3(4): e1939, 2008 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-18398465

RESUMO

The cell lineage tree of a multicellular organism represents its history of cell divisions from the very first cell, the zygote. A new method for high-resolution reconstruction of parts of such cell lineage trees was recently developed based on phylogenetic analysis of somatic mutations accumulated during normal development of an organism. In this study we apply this method in mice to reconstruct the lineage trees of distinct cell types. We address for the first time basic questions in developmental biology of higher organisms, namely what is the correlation between the lineage relation among cells and their (1) function, (2) physical proximity and (3) anatomical proximity. We analyzed B-cells, kidney-, mesenchymal- and hematopoietic-stem cells, as well as satellite cells, which are adult skeletal muscle stem cells isolated from their niche on the muscle fibers (myofibers) from various skeletal muscles. Our results demonstrate that all analyzed cell types are intermingled in the lineage tree, indicating that none of these cell types are single exclusive clones. We also show a significant correlation between the physical proximity of satellite cells within muscles and their lineage. Furthermore, we show that satellite cells obtained from a single myofiber are significantly clustered in the lineage tree, reflecting their common developmental origin. Lineage analysis based on somatic mutations enables performing high resolution reconstruction of lineage trees in mice and humans, which can provide fundamental insights to many aspects of their development and tissue maintenance.


Assuntos
Linhagem da Célula , Células-Tronco/citologia , Animais , Linfócitos B/citologia , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Rim/citologia , Células Matadoras Naturais/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/citologia , Mutação , Oócitos/metabolismo , Células Satélites de Músculo Esquelético/citologia
16.
Nephrol Dial Transplant ; 23(6): 2071-80, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18344240

RESUMO

BACKGROUND: In epithelial and endothelial cells, detachment from the matrix results in anoikis, a form of apoptosis, whereas stromal and cancer cells are often anchorage independent. The classical anoikis model is based on static 3D epithelial cell culture conditions (STCK). METHODS: We characterized a new model of renal, stromal and mesenchymal stem cell (MSC) matrix deprivation, based on slow rotation cell culture conditions (ROCK). This model induces anoikis using a low shear stress, laminar flow. The mechanism of cell death was determined via FACS (fluorescence-activated cell sorting) analysis for annexin V and propidium iodide uptake and via DNA laddering. RESULTS: While only renal epithelial cells progressively died in STCK, the ROCK model could induce apoptosis in stromal and transformed cells; cell survival decreased in ROCK versus STCK to 40%, 52%, 62% and 7% in human fibroblast, rat MSC, renal cell carcinoma (RCC) and human melanoma cell lines, respectively. Furthermore, while ROCK induced primarily apoptosis in renal epithelial cells, necrosis was more prevalent in transformed and cancer cells [necrosis/apoptosis ratio of 72.7% in CaKi-1 RCC cells versus 4.3% in MDCK (Madin-Darby canine kidney) cells]. The ROCK-mediated shift to necrosis in RCC cells was further accentuated 3.4-fold by H(2)O(2)-mediated oxidative stress while in adherent HK-2 renal epithelial cells, oxidative stress enhanced apoptosis. ROCK conditions could also unveil a similar pattern in the LZ100 rat MSC line where in ROCK 44% less apoptosis was observed versus STCK and 45% less apoptosis versus monolayer conditions. Apoptosis in response to oxidative stress was also attenuated in the rat MSC line in ROCK, thereby highlighting rat MSC transformation. CONCLUSIONS: The ROCK matrix-deficiency cell culture model may provide a valuable insight into the mechanism of renal and MSC cell death in response to matrix deprivation.


Assuntos
Apoptose/fisiologia , Células Mesangiais/citologia , Células-Tronco Mesenquimais/citologia , Estresse Oxidativo/fisiologia , Animais , Anoikis/fisiologia , Carcinoma de Células Renais/patologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , Células Cultivadas , Cães , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Matriz Extracelular , Imunofluorescência , Humanos , Melanoma/patologia , Células Mesangiais/fisiologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Modelos Teóricos , Probabilidade , Ratos , Espécies Reativas de Oxigênio/análise , Sensibilidade e Especificidade , Estresse Mecânico
17.
Cloning Stem Cells ; 9(2): 157-75, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17579550

RESUMO

Transplantation of adult mesenchymal stem cells (MSCs) could provide a basis for tissue regeneration. MSCs are typically isolated from bone marrow (BM) based on their preferential adherence to plastic, although with low efficiency in terms of yield and purity. Extensive expansion is needed to reach a significant number of MSCs for any application. Fibrin microbeads (FMB) were designed to attach mesenchymal cells and to provide a matrix for their expansion. The current study was aimed at isolating a high yield of purified BM-derived mouse MSCs based on their preferential adherence and proliferation on FMB in suspension cultures. MSCs could be downloaded to plastics or further expanded on FMB. The yield of MSCs obtained by the FMB isolation technique was about one order of magnitude higher than that achieved by plastic adherence, suggesting that these cells are more abundant than previously reported. FMB-isolated cells were classified as MSCs by their fibroblastic morphology, self-renewal ability, and expression profile of their surface antigens, as examined by flow cytometry and immunostaining. In cell culture, the isolated MSCs could be induced to differentiate into three different mesodermal lineages, as demonstrated by histochemical stains and by RT-PCR analyses of tissue-specific genes. MSCs were also able to differentiate into osteocytes while still cultured on FMB. Our results suggest that FMB might serve as an efficient platform for the isolation, expansion, and differentiation of mouse BM-derived MSCs to be subsequently implanted for tissue regeneration.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fibrina/metabolismo , Células-Tronco Mesenquimais/citologia , Microesferas , Animais , Antígenos de Diferenciação/metabolismo , Células da Medula Óssea/fisiologia , Adesão Celular/fisiologia , Separação Celular , Células Cultivadas , Células-Tronco Mesenquimais/fisiologia , Camundongos
18.
J Am Soc Nephrol ; 17(12): 3300-14, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17093069

RESUMO

Tissue engineering and cell therapy approaches aim to take advantage of the repopulating ability and plasticity of multipotent stem cells to regenerate lost or diseased tissue. Recently, stage-specific embryonic kidney progenitor tissue was used to regenerate nephrons. Through fluorescence-activated cell sorting, microarray analysis, in vitro differentiation assays, mixed lymphocyte reaction, and a model of ischemic kidney injury, this study sought to identify and characterize multipotent organ stem/progenitor cells in the adult kidney. Herein is reported the existence of nontubular cells that express stem cell antigen-1 (Sca-1). This population of small cells includes a CD45-negative fraction that lacks hematopoietic stem cell and lineage markers and resides in the renal interstitial space. In addition, these cells are enriched for beta1-integrin, are cytokeratin negative, and show minimal expression of surface markers that typically are found on bone marrow-derived mesenchymal stem cells. Global gene profiling reveals enrichment for many genes downstream of developmental signaling molecules and self-renewal pathways, such as TGF-beta/bone morphogenic protein, Wnt, or fibroblast growth factor, as well as for those that are involved in specification of mesodermal lineages (myocyte enhancer factor 2A, YY1-associated factor 2, and filamin-beta). In vitro, they are plastic adherent and slowly proliferating and result in inhibition of alloreactive CD8(+) T cells, indicative of an immune-privileged behavior. Furthermore, clonal-derived lines can be differentiated into myogenic, osteogenic, adipogenic, and neural lineages. Finally, when injected directly into the renal parenchyma, shortly after ischemic/reperfusion injury, renal Sca-1(+)Lin(-) cells, derived from ROSA26 reporter mice, adopt a tubular phenotype and potentially could contribute to kidney repair. These data define a unique phenotype for adult kidney-derived cells, which have potential as stem cells and may contribute to the regeneration of injured kidneys.


Assuntos
Antígenos Ly/isolamento & purificação , Rim/citologia , Antígenos Comuns de Leucócito/isolamento & purificação , Proteínas de Membrana/isolamento & purificação , Células-Tronco Multipotentes/citologia , Animais , Diferenciação Celular , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Análise em Microsséries , Células-Tronco Multipotentes/imunologia , Fenótipo
19.
Tissue Eng ; 12(8): 2343-54, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16968174

RESUMO

Fibrin microbeads (FMB), made of extensively cross-linked dense and partially denatured fibrin, were used as a matrix for efficient isolation of mesenchymal stem cells (MSC) from rat bone marrow (BM). After 2 days of incubation of FMB with whole BM in suspension, a high number of cells of mesenchymal origin attached to the FMB. On the 14th day after their transfer to plastic, the yield of the cells isolated via FMB was approximately 3-4 times higher than that obtained by currently used protocols based solely on plastic adhesion. This implies that the number of MSC in BM may be higher than previously reported. FACS analyses and immunostaining showed the mesenchymal characteristics of these cells by positive staining for fibronectin, vimentin, CD49E, and CD29. Immediately after isolation, less than 20% of the cells still expressed the hematopoietic markers CD11b and CD45. Most of these cells were eventually eliminated after further expansion of the isolated cells on plastic. Cells isolated via FMB were expanded in culture for more than 4 months and could be defined as MSC along this time period based on their ability to differentiate into precursors of mesenchymal tissues, such as osteogenic, adipogenic, and chondrogenic cells. Similar differentiation plasticity was observed in clones derived from single cells from whole MSC populations isolated via FMB. Based on our results we propose that FMB can serve as a 3-dimensional biodegradable matrix for isolation, differentiation, and possibly implantation of MSC for tissue regeneration.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Separação Celular , Fibrina , Células-Tronco Mesenquimais/fisiologia , Microesferas , Animais , Células da Medula Óssea/citologia , Masculino , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Wistar , Engenharia Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA