Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Stem Cells Int ; 2017: 6406025, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29090011

RESUMO

The interactions between stem cells and their surrounding microenvironment are pivotal to determine tissue homeostasis and stem cell renewal or differentiation and regeneration in vivo. Ever since they were postulated in 1978, stem cell niches have been identified and characterized in many germline and adult tissues. Comprehensive studies over the last decades helped to clarify the critical components of stem cell niches that include cellular, extracellular, biochemical, molecular, and physical regulators. This knowledge has direct impact on their inherent regenerative potential. Clinical applications demand readily available cell sources that, under controlled conditions, provide a specific therapeutic function. Thus, translational medicine aims at optimizing in vitro or in vivo the various components and complex architecture of the niche to exploit its therapeutic potential. Accordingly, the objective is to recreate the natural niche microenvironment during cell therapy process development and closely comply with the requests of regulatory authorities. In this paper, we review the most recent advances of translational medicine approaches that target the adult stem cell natural niche microenvironment for regenerative medicine applications.

2.
Stem Cells Int ; 2017: 2653142, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29075297

RESUMO

Already during embryonic development, the heart and the lung are thoroughly connected organs. Their interdependence allows our survival in the terrestrial environment by coupling cardiac output and gas exchange. The knowledge on developmental processes involving stem and progenitor cells is crucial to understand the onset of human cardiopulmonary diseases. The precise identification of various adult endogenous progenitors is still incomplete. Thus, caution should be exercised on newly available stem cell-based treatments until specific mechanisms of action are disclosed. The objective is to provide in the nearest future feasible and safer cell therapeutics for the complex pathological condition of human cardiopulmonary diseases. In this paper, we highlight the significant knowledge advancement concerning stem and progenitor cells in the cardiopulmonary field: from embryonic development to adult progenitors until early preclinical models for cardiopulmonary regeneration.

3.
Adv Exp Med Biol ; 951: 163-178, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27837563

RESUMO

Cardiovascular diseases, including heart failure, are the most frequent cause of death annually, even higher than any other pathologies. Specifically, patients who suffer from myocardial infarction may encounter adverse remodeling processes of the heart that can ultimately lead to heart failure. Prognosis of patients affected by heart failure is very poor with 5-year mortality close to 50 %. Despite the impressive progress in the clinical treatment of heart failure in recent years, heart transplantation is still required to avoid death as the result of the inexorable decline in cardiac function. Unfortunately, the availability of donor human hearts for transplantation largely fails to cover the number of potential recipient requests. From this urgent unmet clinical need the interest in stem cell applications for heart regeneration made its start, and has rapidly grown in the last decades. Indeed, the discovery and application of stem and progenitor cells as therapeutic agents has raised substantial interest with the objective of reversing these processes, and ultimately inducing cardiac regeneration. In this scenario, the role of biobanking may play a remarkable role to provide cells at the right time according to the patient's clinical needs, mostly for autologous use in the acute setting of myocardial infarction, largely reducing the time needed for cell preparation and expansion before administration.


Assuntos
Criopreservação/métodos , Células-Tronco Embrionárias/citologia , Insuficiência Cardíaca/terapia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco , Animais , Bancos de Espécimes Biológicos/provisão & distribuição , Crioprotetores/farmacologia , Modelos Animais de Doenças , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Insuficiência Cardíaca/patologia , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/transplante , Humanos , Macaca nemestrina , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/transplante , Regeneração/fisiologia , Medicina Regenerativa/métodos , Transplante Autólogo
4.
Circ Res ; 111(7): 894-906, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22851539

RESUMO

RATIONALE: According to the immortal DNA strand hypothesis, dividing stem cells selectively segregate chromosomes carrying the old template DNA, opposing accumulation of mutations resulting from nonrepaired replication errors and attenuating telomere shortening. OBJECTIVE: Based on the premise of the immortal DNA strand hypothesis, we propose that stem cells retaining the old DNA would represent the most powerful cells for myocardial regeneration. METHODS AND RESULTS: Division of human cardiac stem cells (hCSCs) by nonrandom and random segregation of chromatids was documented by clonal assay of bromodeoxyuridine-tagged hCSCs. Additionally, their growth properties were determined by a series of in vitro and in vivo studies. We report that a small class of hCSCs retain during replication the mother DNA and generate 2 daughter cells, which carry the old and new DNA, respectively. hCSCs with immortal DNA form a pool of nonsenescent cells with longer telomeres and higher proliferative capacity. The self-renewal and long-term repopulating ability of these cells was shown in serial-transplantation assays in the infarcted heart; these cells created a chimeric organ, composed of spared rat and regenerated human cardiomyocytes and coronary vessels, leading to a remarkable restoration of cardiac structure and function. The documentation that hCSCs divide by asymmetrical and symmetrical chromatid segregation supports the view that the human heart is a self-renewing organ regulated by a compartment of resident hCSCs. CONCLUSIONS: The impressive recovery in ventricular hemodynamics and anatomy mediated by clonal hCSCs carrying the "mother" DNA underscores the clinical relevance of this stem cell class for the management of heart failure in humans.


Assuntos
Cromátides/fisiologia , Segregação de Cromossomos/fisiologia , Coração/fisiologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Regeneração/fisiologia , Transplante de Células-Tronco , Células-Tronco/citologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Bromodesoxiuridina , Proliferação de Células , Células Cultivadas , Criança , Pré-Escolar , Cromátides/ultraestrutura , DNA/fisiologia , Feminino , Humanos , Técnicas In Vitro , Lactente , Masculino , Pessoa de Meia-Idade , Modelos Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Células-Tronco/fisiologia , Telômero/ultraestrutura , Adulto Jovem
5.
N Engl J Med ; 364(19): 1795-806, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21561345

RESUMO

BACKGROUND: Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS: Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS: Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS: Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).


Assuntos
Pulmão/citologia , Células-Tronco/fisiologia , Adulto , Animais , Células Clonais , Feminino , Humanos , Pulmão/embriologia , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes , Proteínas Proto-Oncogênicas c-kit/análise , Regeneração , Transplante de Células-Tronco , Células-Tronco/química
6.
J Cardiovasc Transl Res ; 4(2): 200-10, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21327755

RESUMO

Multipotent mesenchymal stromal cells (MSCs) are a type of adult stem cells that can be easily isolated from various tissues and expanded in vitro. Many reports on their pluripotency and possible clinical applications have raised hopes and interest in MSCs. In an attempt to unify the terminology and the criteria to label a cell as MSC, in 2006 the International Society for Cellular Therapy (ISCT) proposed a standard set of rules to define the identity of these cells. However, MSCs are still extracted from different tissues, by diverse isolation protocols, are cultured and expanded in different media and conditions. All these variables may have profound effects on the selection of cell types and the composition of heterogeneous subpopulations, on the selective expansion of specific cell populations with totally different potentials and ergo, on the long-term fate of the cells upon in vitro culture. Therefore, specific molecular and cellular markers that identify MSCs subsets as well as standardization of expansion protocols for these cells are urgently needed. Here, we briefly discuss new useful markers and recent data supporting the rapidly emerging concept that many different types of progenitor cells are found in close association with blood vessels. This knowledge may promote the necessary technical improvements required to reduce variability and promote higher efficacy and safety when isolating and expanding these cells for therapeutic use. In the light of the discussed data, particularly the identification of new markers, and advances in the understanding of fundamental MSC biology, we also suggest a revision of the 2006 ISCT criteria.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/fisiologia , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Multipotentes/fisiologia , Tecido Adiposo/irrigação sanguínea , Adulto , Células-Tronco Adultas/metabolismo , Biomarcadores/metabolismo , Separação Celular/métodos , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/metabolismo , Terminologia como Assunto
7.
J Cardiovasc Transl Res ; 4(2): 192-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21052883

RESUMO

The 2010 edition of the Lugano Stem Cell Meeting, under the auspices of the Swiss center of excellence in cardiovascular diseases "Cardiocentro Ticino" and the Swiss Stem Cell Foundation, offered an update on clinical, translational, and biotechnological advances in regenerative science and medicine pertinent to cardiovascular applications. Highlights from the international forum ranged from innate mechanisms of heart repair, safety, and efficacy of ongoing and completed clinical trials, novel generations of stem cell biologics, bioengineered platforms, and regulatory processes. In the emerging era of regenerative medicine, accelerating the critical path from discovery to product development will require integrated multidisciplinary teams to ensure timely translation of new knowledge into validated algorithms for practice adoption.


Assuntos
Cardiopatias/cirurgia , Miocárdio/patologia , Medicina Regenerativa/métodos , Transplante de Células-Tronco , Distinções e Prêmios , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Regeneração , Medicina Regenerativa/legislação & jurisprudência , Transplante de Células-Tronco/legislação & jurisprudência , Resultado do Tratamento
8.
Circ Res ; 107(11): 1374-86, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-21088285

RESUMO

RATIONALE: The turnover of cardiomyocytes in the aging female and male heart is currently unknown, emphasizing the need to define human myocardial biology. OBJECTIVE: The effects of age and gender on the magnitude of myocyte regeneration and the origin of newly formed cardiomyocytes were determined. METHODS AND RESULTS: The interaction of myocyte replacement, cellular senescence, growth inhibition, and apoptosis was measured in normal female (n=32) and male (n=42) human hearts collected from patients 19 to 104 years of age who died from causes other than cardiovascular diseases. A progressive loss of telomeric DNA in human cardiac stem cells (hCSCs) occurs with aging and the newly formed cardiomyocytes inherit short telomeres and rapidly reach the senescent phenotype. Our data provide novel information on the superior ability of the female heart to sustain the multiple variables associated with the development of the senescent myopathy. At all ages, the female heart is equipped with a larger pool of functionally competent hCSCs and younger myocytes than the male myocardium. The replicative potential is higher and telomeres are longer in female hCSCs than in male hCSCs. In the female heart, myocyte turnover occurs at a rate of 10%, 14%, and 40% per year at 20, 60, and 100 years of age, respectively. Corresponding values in the male heart are 7%, 12%, and 32% per year, documenting that cardiomyogenesis involves a large and progressively increasing number of parenchymal cells with aging. From 20 to 100 years of age, the myocyte compartment is replaced 15 times in women and 11 times in men. CONCLUSIONS: The human heart is a highly dynamic organ regulated by a pool of resident hCSCs that modulate cardiac homeostasis and condition organ aging.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Coração/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/fisiologia , Morte Celular/fisiologia , Células Cultivadas , Feminino , Coração/anatomia & histologia , Humanos , Masculino , Pessoa de Meia-Idade , Caracteres Sexuais , Adulto Jovem
9.
J Cardiovasc Transl Res ; 3(2): 128-34, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20560026
10.
Circ Res ; 105(11): 1128-40, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19815820

RESUMO

RATIONALE: Chronic rejection, accelerated coronary atherosclerosis, myocardial infarction, and ischemic heart failure determine the unfavorable evolution of the transplanted heart in humans. OBJECTIVE: Here we tested whether the pathological manifestations of the transplanted heart can be corrected partly by a strategy that implements the use of cardiac progenitor cells from the recipient to repopulate the donor heart with immunocompatible cardiomyocytes and coronary vessels. METHODS AND RESULTS: A large number of cardiomyocytes and coronary vessels were created in a rather short period of time from the delivery, engraftment, and differentiation of cardiac progenitor cells from the recipient. A proportion of newly formed cardiomyocytes acquired adult characteristics and was integrated structurally and functionally within the transplant. Similarly, the regenerated arteries, arterioles, and capillaries were operative and contributed to the oxygenation of the chimeric myocardium. Attenuation in the extent of acute damage by repopulating cardiomyocytes and vessels decreased significantly the magnitude of myocardial scarring preserving partly the integrity of the donor heart. CONCLUSIONS: Our data suggest that tissue regeneration by differentiation of recipient cardiac progenitor cells restored a significant portion of the rejected donor myocardium. Ultimately, immunosuppressive therapy may be only partially required improving quality of life and lifespan of patients with cardiac transplantation.


Assuntos
Rejeição de Enxerto/patologia , Transplante de Coração , Histocompatibilidade , Miócitos Cardíacos/citologia , Regeneração/imunologia , Células-Tronco/citologia , Animais , Sequência de Bases , Diferenciação Celular/fisiologia , Fusão Celular , Vasos Coronários/citologia , Cães , Feminino , Genótipo , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/imunologia , Proteínas de Fluorescência Verde/genética , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/patologia , Imunossupressores/uso terapêutico , Masculino , Dados de Sequência Molecular , Miocárdio/patologia , Células-Tronco/fisiologia
11.
Proc Natl Acad Sci U S A ; 106(37): 15885-90, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19717420

RESUMO

Primitive cells capable of generating small resistance arterioles and capillary structures in the injured myocardium have been identified repeatedly. However, these cells do not form large conductive coronary arteries that would have important implications in the management of the ischemic heart. In the current study, we determined whether the human heart possesses a class of progenitor cells that regulates the growth of endothelial cells (ECs) and smooth muscle cells (SMCs) and vasculogenesis. The expression of vascular endothelial growth-factor receptor 2 (KDR) was used, together with the stem cell antigen c-kit, to isolate and expand a resident coronary vascular progenitor cell (VPC) from human myocardial samples. Structurally, vascular niches composed of c-kit-KDR-positive VPCs were identified within the walls of coronary vessels. The VPCs were connected by gap junctions to ECs, SMCs, and fibroblasts that operate as supporting cells. In vitro, VPCs were self-renewing and clonogenic and differentiated predominantly into ECs and SMCs and partly into cardiomyocytes. To establish the functional import of VPCs, a critical stenosis was created in immunosuppressed dogs, and tagged human VPCs were injected in proximity to the constricted artery. One month later, there was an increase in coronary blood flow (CBF) distal to the stenotic artery, resulting in functional improvement of the ischemic myocardium. Regenerated large, intermediate, and small human coronary arteries and capillaries were found. In conclusion, the human heart contains a pool of VPCs that can be implemented clinically to form functionally competent coronary vessels and improve CBF in patients with ischemic cardiomyopathy.


Assuntos
Células-Tronco Adultas/citologia , Vasos Coronários/citologia , Actinas/metabolismo , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Conexina 43/metabolismo , Circulação Coronária , Estenose Coronária/patologia , Estenose Coronária/fisiopatologia , Estenose Coronária/terapia , Vasos Coronários/fisiopatologia , Cães , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Perfilação da Expressão Gênica , Humanos , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Regeneração/genética , Regeneração/fisiologia , Transplante Heterólogo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Antioxid Redox Signal ; 11(3): 589-600, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18717627

RESUMO

Redox effector protein-1 (Ref-1) plays an essential role in DNA repair and redox regulation of several transcription factors. In the present study, we examined the role of Ref-1 in maintaining the redox status and survivability of adult cardiac stem cells challenged with a subtoxic level of H2O2 under inhibition of Ref-1 by RNA interference. Treatment of cardiac stem cells with a low concentration of H2O2 induced Ref-1-mediated survival signaling through phosphorylation of Akt. However, Ref-1 inhibition followed by H2O2 treatment extensively induced the level of intracellular reactive oxygen species (ROS) through activation of the components of NADPH oxidase, like p22( phox ), p47( phox ), and Nox4. Cardiac differentiation markers (Nkx2.5, MEF2C, and GATA4), and cell death by apoptosis were significantly elevated in Ref-1 siRNA followed by H2O2-treated stem cells. Further, inhibition of Ref-1 increased the level of p53 but decreased the phosphorylation of Akt, a molecule involved in survival signaling. Treatment with ROS scavenger N-acetyl-L-cysteine attenuated Ref-1 siRNA-mediated activation of NADPH oxidase and cardiac differentiation. Taken together, these results indicate that Ref-1 plays an important role in maintaining the redox status of cardiac stem cells and protects them from oxidative injury-mediated cell death and differentiation.


Assuntos
Diferenciação Celular , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/antagonistas & inibidores , Miocárdio/citologia , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/citologia , Adulto , Sequência de Bases , Primers do DNA , Humanos , Marcação In Situ das Extremidades Cortadas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Transl Med ; 5: 55, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17974012

RESUMO

BACKGROUND: The stromal vascular fraction (SVF) is a heterogeneous cell population derived from the adipose tissue. There is still a lack of information concerning the characterization of the cell subpopulations constituting the SVF as well as its mesenchymal and haematopoietic potential. Furthermore there are great variations in its phenotypical characterization. METHODS: Composition of SVF was investigated by FACS analysis, cytological and "in vitro" assays. We studied CD34+ population by combining FACS with human CFC (colony-forming-cell haematopoietic assay). The endothelial fraction was investigated by quantifying the co-expression of specific markers (CD146, CD105, CD31 and UEA-1). Mesenchymal potential was assessed by CFU-F assay and cultured AT-MSC were characterized by a 5-color FACS analysis. The multipotent differentiation potential (osteogenic, adipogenic and chondrogenic) was investigated both at cellular and molecular level. RESULTS: We identified in the SVF two CD34+ populations with a marked difference in the intensity of antigen expression, the majority of the cells expressing CD34 at low intensity. Moreover, two CD146+ cell populations were clearly distinguishable in the SVF:a CD146 dim accounting for 9.9% of the total SVF cells and a CD146+ bright cell population accounting for about 39.3%. The frequency of CFC clones was comparable with the one reported for peripheral blood. Endothelial cells account for about 7.7% of the SVF cells. AT-MSC differenced in the osteogenic adipogenic and chondrogenic lineage. CONCLUSION: The SVF is not a homogeneous cell population, and its final composition could be influenced both by the flow cytometric technique analysis and the SVF extraction steps. The CFU-F frequency in the SVF was 1/4880, a value about seven times greater than the data reported for bone marrow. The antigenic profile of AT-MSC was comparable with bone-marrow derived MSC. AT-MSC were able to differentiate along the osteogenic adipogenic and chondrogenic lineages. The data here reported, further contribute to the characterization of SVF, a tissue providing an alternative as a source of MSC for clinical applications.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/irrigação sanguínea , Antígenos CD34/imunologia , Sequência de Bases , Citometria de Fluxo , Humanos , Células-Tronco Mesenquimais/imunologia , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Int J Oncol ; 26(5): 1159-68, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15809705

RESUMO

The region 6q27 from human chromosome 6 has been reported to contain one or more tumor suppressor genes on the basis of cytogenetic, molecular and functional studies. We have recently carried out a detailed analysis of a candidate gene from 6q27 to evaluate its putative role as a tumor suppressor gene involved in ovarian cancer pathogenesis. The RNASET2 gene was shown to behave as a class II tumor suppressor and abolish the tumorigenic potential of an ovarian cancer-derived cell line. In this study, we have started the cellular and biochemical characterization of RNASET2 and showed that it is a secreted glycoprotein. Moreover, we have extended our previous studies by evaluating the effect of RNASET2 on the metastatic behavior of the highly-invasive ovarian cancer cell line HEY3MET2. From such analysis, RNASET2 was found to significantly decrease the metastatic potential of this cell line in vivo. Moreover, RNASET2-mediated suppression of tumorigenesis and metastasis was not affected by a double point mutation targeted to the putative ribonuclease catalytic sites, suggesting that tumor suppression by RNASET2 is not mediated by its ribonuclease activity. The potential biological implications of this unexpected finding are discussed in relation to the current evolutionary models.


Assuntos
Genes Supressores de Tumor/fisiologia , Metástase Neoplásica/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ribonucleases/genética , Ribonucleases/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Cromossomos Humanos Par 6/genética , Feminino , Humanos , Invasividade Neoplásica , Mutação Puntual , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA