Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 77: 103407, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38552357

RESUMO

We employed a Sendai virus-based reprogramming method to transform human lymphoblastoid cell lines (LCL) derived from two individuals diagnosed with phenylketonuria (PKU) into induced pluripotent stem cells (iPSC). This reprogramming process involved the expression of the four Yamanaka factors: KLF4, OCT4, SOX2, and C-MYC. The resulting patient-specific iPSCs exhibited a normal karyotype and expressed endogenous pluripotent markers NANOG and OCT-4. Notably, these iPSCs demonstrated strong differentiation capabilities, giving rise to cell populations representing the ectoderm, endoderm, and mesoderm germ layers.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Fator 4 Semelhante a Kruppel , Fenilcetonúrias , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fenilcetonúrias/metabolismo , Fenilcetonúrias/patologia , Linhagem Celular , Masculino , Linfócitos/metabolismo , Reprogramação Celular
2.
Stem Cell Res ; 77: 103405, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38555716

RESUMO

Phenylketonuria is a rare autosomal recessive metabolic disorder mainly due to a significant reduction in the enzyme phenylalanine hydroxylase, resulting in elevation of phenylalanine in the blood. Here, we have established two fibroblast-derived induced pluripotent stem cell lines using Sendai virus-based reprogramming. The established induced pluripotent stem cell lines exhibited a normal karyotype and expressed markers of pluripotency assessed through quantitative PCR, flow cytometry and immunocytochemistry. These cell lines also demonstrated the ability to differentiate into the three primary germ layers of the human body, including ectoderm, endoderm, and mesoderm.


Assuntos
Diferenciação Celular , Fibroblastos , Células-Tronco Pluripotentes Induzidas , Fenilcetonúrias , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fenilcetonúrias/metabolismo , Fenilcetonúrias/patologia , Fibroblastos/metabolismo , Linhagem Celular , Masculino , Criança
3.
Sci Rep ; 10(1): 6919, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332814

RESUMO

To accelerate the cardiac drug discovery pipeline, we set out to develop a platform that would be capable of quantifying tissue-level functions such as contractile force and be amenable to standard multiwell-plate manipulations. We report a 96-well-based array of 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues - termed Cardiac MicroRings (CaMiRi) - in custom 3D-print-molded multiwell plates capable of contractile force measurement. Within each well, two elastomeric microcantilevers are situated above a circumferential ramp. The wells are seeded with cell-laden collagen, which, in response to the gradual slope of the circumferential ramp, self-organizes around tip-gated microcantilevers to form contracting CaMiRi. The contractile force exerted by the CaMiRi is measured and calculated using the deflection of the cantilevers. Platform responses were robust and comparable across wells, and we used it to determine an optimal tissue formulation. We validated the contractile force response of CaMiRi using selected cardiotropic compounds with known effects. Additionally, we developed automated protocols for CaMiRi seeding, image acquisition, and analysis to enable the measurement of contractile force with increased throughput. The unique tissue fabrication properties of the platform, and the consequent effects on tissue function, were demonstrated upon adding hPSC-derived epicardial cells to the system. This platform represents an open-source contractile force screening system useful for drug screening and tissue engineering applications.


Assuntos
Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Animais , Automação , Cardiotônicos/farmacologia , Células Cultivadas , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Camundongos , Contração Miocárdica/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Impressão Tridimensional
4.
Tissue Eng Part C Methods ; 24(3): 146-157, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29239281

RESUMO

Human pluripotent stem cells (hPSCs) are viewed as promising candidates for applications in regenerative medicine and therapy due to their proliferative and pluripotent properties. However, obtaining clinically significant numbers of hPSCs remains a limiting factor and impedes their use in therapeutic applications. Conventionally, hPSCs are cultured on two-dimensional surfaces coated with a suitable substrate, such as Matrigel™. This method, however, requires a large surface area to generate sufficient cell numbers to meet clinical needs and is therefore impractical as a manufacturing platform for cell expansion. In addition, the use of enzymes for cell detachment and small molecule inhibitors to increase plating efficiency may impact future cell behavior when used for routine subculturing. In this study, we describe a protocol to generate and maintain hPSC aggregates in a three-dimensional suspension culture by utilizing thermoresponsive nanobridges. The property of the polymer used in the nanobridges enables passaging and expansion through a temperature change in combination with mechanically applied shear to dissociate aggregates; thus, we eliminate the need of enzymes or small molecules for cell dissociation and viability, respectively. Utilizing this platform, maintenance of human embryonic stem cells for three continuous passages demonstrated high expression levels in key pluripotent markers.


Assuntos
Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Embrionárias Humanas/citologia , Nanotecnologia/métodos , Polímeros/química , Temperatura , Proliferação de Células , Células Cultivadas , Células-Tronco Embrionárias Humanas/fisiologia , Humanos
5.
Can J Cardiol ; 30(11): 1335-49, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25442434

RESUMO

Despite advances in coronary artery disease treatment and prevention, myocardial damage due to acute myocardial infarction (MI) remains a major cause of morbidity and mortality in the population. Cell-based clinical trials to treat MI have focused on cells derived from the bone marrow or those potentially possessing functional similarities such as skeletal myoblasts or cardiac progenitors isolated from heart biopsies. Any benefits provided by these cells in improving heart function, left ventricular ejection fraction, or extending life expectancy after MI have been credited mostly to paracrine effects. Functional restoration of damaged myocardium will require a functional cell type with similar phenotype and characteristics of the damaged tissue that can also integrate, survive, and electrically couple to the host. Human pluripotent stem cells (hPSCs) have the ability to differentiate into multiple cell types of the adult body. hPSC-derived cardiomyocytes represent a promising target population for cell-based therapies for MI because they are scalable and the product can be defined with a specific set of release criteria. The purpose of this article is to review the rationale for cell therapy in heart disease, discuss the properties of hPSC cardiomyocytes that define their usefulness for regenerative therapy, consider manufacturing issues and preclinical investigation, and finally examine the steps required to establish effective clinical implementation. Pluripotent stem cell-derived cardiomyocyte-based therapies have enormous potential to revolutionize the management of heart disease; expedient but careful development is needed to ensure that this potential is fully realized.


Assuntos
Ensaios Clínicos como Assunto/métodos , Cardiopatias/terapia , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco/métodos , Cardiopatias/patologia , Humanos
6.
Biomacromolecules ; 15(3): 844-55, 2014 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-24571238

RESUMO

The development of robust suspension cultures of human embryonic stem cells (hESCs) without the use of cell membrane disrupting enzymes or inhibitors is critical for future clinical applications in regenerative medicine. We have achieved this by using long, flexible, and thermoresponsive polymer worms decorated with a recombinant vitronectin subdomain that bridge hESCs, aiding in hESC's natural ability to form embryoid bodies (EBs) and satisfying their inherent requirement for cell-cell and cell-extracellular matrix contact. When the EBs reached an optimal upper size where cytokine and nutrient penetration becomes limiting, these long and flexible polymer worms facilitated EB breakdown via a temperature shift from 37 to 25 °C. The thermoresponsive nature of the worms enabled a cyclical dissociation and propagation of the cells. Repeating the process for three cycles (over eighteen days) provided a >30-fold expansion in cell number while maintaining pluripotency, thereby providing a simple, nondestructive process for the 3D expansion of hESC.


Assuntos
Técnicas de Cultura de Células , Corpos Embrioides/química , Células-Tronco Embrionárias/citologia , Matriz Extracelular/química , Diferenciação Celular/genética , Proliferação de Células , Corpos Embrioides/citologia , Humanos , Polímeros/química , Medicina Regenerativa , Temperatura
7.
Clin Kidney J ; 6(1): 21-28, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27818747

RESUMO

BACKGROUND: The use of generic formulations of immunosuppressive drugs in place of brand name drugs offers considerable cost savings. Brand name tacrolimus (Prograf®) came off patent in April 2008. However, published evidence supporting therapeutic equivalence of generic formulations of tacrolimus in solid organ transplantation is lacking. The South West Transplant Centre switched from administering Prograf® to a generic formulation (Adoport®) for de novo transplant recipients in November 2010. This study sought to compare the clinical outcomes of renal transplant recipients administered Prograf® with those receiving Adoport®. METHODS: Data regarding patient characteristics and clinical outcomes were collected retrospectively for all patients undergoing renal transplantation at the South West Transplant Centre between 8 November 2009 and 8 November 2011 to whom tacrolimus was prescribed. RESULTS: A total of 48 patients received Prograf® and 51 received Adoport®. At 6 months, no statistically significant differences were identified in the rates of patient survival, graft survival, acute allograft rejection, delayed graft function, calcineurin inhibitor toxicity or cytomegalovirus infection occurring within the two groups. CONCLUSIONS: This is the first study to compare the clinical outcomes of patients receiving Adoport® with those receiving brand name tacrolimus. We report comparable clinical outcomes at 6 months in patients receiving either Prograf® or Adoport® from the time of renal transplantation. These early outcome data therefore support the use of Adoport® in place of Prograf® as a potential cost-saving measure.

8.
PLoS One ; 7(12): e52214, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284940

RESUMO

Human embryonic stem cell (hESC) derivatives show promise as viable cell therapy options for multiple disorders in different tissues. Recent advances in stem cell biology have lead to the reliable production and detailed molecular characterisation of a range of cell-types. However, the role of mitochondria during differentiation has yet to be fully elucidated. Mitochondria mediate a cells response to altered energy requirements (e.g. cardiomyocyte contraction) and, as such, the mitochondrial phenotype is likely to change during the dynamic process of hESC differentiation. We demonstrate that manipulating mitochondrial biogenesis alters mesendoderm commitment. To investigate mitochondrial localisation during early lineage specification of hESCs we developed a mitochondrial reporter line, KMEL2, in which sequences encoding the green fluorescent protein (GFP) are targeted to the mitochondria. Differentiation of KMEL2 lines into the three germ layers showed that the mitochondria in these differentiated progeny are GFP positive. Therefore, KMEL2 hESCs facilitate the study of mitochondria in a range of cell types and, importantly, permit real-time analysis of mitochondria via the GFP tag.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Mitocôndrias/metabolismo , Linhagem Celular , Citometria de Fluxo , Imunofluorescência , Humanos , Cariótipo , Fosforilação Oxidativa
10.
Stem Cell Res ; 6(1): 1-12, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21075697

RESUMO

Use of stem cells, whether adult or embryonic for clinical applications to treat diseases such as Parkinson's, macular degeneration or Type I diabetes will require a homogenous population of mature, terminally differentiated cells. A current area of intense interest is the development of defined surfaces for stem cell derivation, maintenance, proliferation and subsequent differentiation, which are capable of replicating the complex cellular environment existing in vivo. During development many cellular cues result from integrin signalling induced by the local extracellular matrix. There are 24 known integrin heterodimers comprised of one of 18 α subunits and one of 8 ß subunits and these have a diverse range of functions mediating cell-cell adhesion, growth factor receptor responses and intracellular signalling cascades for cell migration, differentiation, survival and proliferation. We discuss here a brief summary of defined conditions for human embryonic stem cell culture together with a description of integrin function and signalling pathways. The importance of integrin expression during development is highlighted as critical for lineage specific cell function and how consideration of the integrin expression profile should be made while differentiating stem cells for use in therapy. In addition this review summarises the known integrin expression profiles for human embryonic stem cells and 3 common adult stem cell types: mesenchymal, haematopoietic and neural. We then outline some of the possible technologies available for investigating cell-extracellular matrix interactions and subsequent integrin mediated cell responses.


Assuntos
Técnicas de Cultura de Células , Terapia Baseada em Transplante de Células e Tecidos , Integrinas/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Humanos , Integrinas/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Células-Tronco/citologia
11.
Biomaterials ; 31(32): 8281-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20674971

RESUMO

Human embryonic stem cells (hESC) are expected to provide revolutionary therapeutic applications and drug discovery technologies. In order for this to be achieved a reproducible, defined animal component free culture system is required for the scale-up production of undifferentiated hESC. In this work we have investigated the applicability of a recombinantly produced domain of human vitronectin as an extracellular matrix alternative to the common standards Geltrex or Matrigel. In addition we have validated an ascorbate free media capable of supporting CD30(low) populations of hESC through a multi-factorial analysis of bFGF and Activin A. The recombinant vitronectin domain combined with the ascorbate free media were capable of supporting 3 cell lines, MEL1, MEL2 and hES3 for 10 or more passages while maintaining hESC pluripotency markers and differentiation capacity. The culture method outlined here provides a platform for future investigation into growth factor and extracellular matrix effects on hESC maintenance prior to bioreactor scale-up.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Proteínas Recombinantes/metabolismo , Vitronectina/metabolismo , Sequência de Aminoácidos , Reatores Biológicos , Adesão Celular , Diferenciação Celular , Linhagem Celular , Meios de Cultura/química , Meios de Cultura/metabolismo , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Somatomedinas/genética , Somatomedinas/isolamento & purificação , Somatomedinas/metabolismo , Fatores de Tempo , Vitronectina/genética , Vitronectina/isolamento & purificação
12.
Biomaterials ; 31(19): 5137-42, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20378164

RESUMO

Unlocking the clinical potential of stem cell based therapies requires firstly elucidation of the biological mechanisms which direct stem cell fate decisions and thereafter, technical advances which allow these processes to be driven in a fully defined culture environment. Strategies for the generation of defined surfaces for human embryonic stem cell (hESC) and mesenchymal stem cell (MSC) culture remain in their infancy. In this paper we outline a simple, effective and efficient method for presenting proteins or peptides on an otherwise non-fouling Layer-by-Layer (LbL) self-assembled surface of hyaluronic acid (HA) and chitosan (CHI). We are able to generate a surface that has both good temporal stability and the ability to direct biological outcomes based on its defined surface composition. Surface functionalization is achieved through suspending the selected extracellular matrix (ECM) protein domain or extracted full-length protein in buffer containing a cross-linking agent (N-hydroxysulfosuccinimide/N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride) over the LbL HA-CHI surface and then allowing the solvent to evaporate overnight. This simple, but important step results in remarkable protein deposition efficiencies often exceeding 50%, whereas traditional cross-linking methods result in such poor deposition of non-collagenous proteins that a.) quantification of bound amounts of protein is outside the resolution of commonly utilized protein assays, and b.) these surfaces are both unable to support cell attachment and growth. The utility of the protein-modified HA-CHI surfaces is demonstrated through the identification of specific hESC attachment efficiencies and through directing MSC osteogenic outcomes on these fully defined surfaces. This simple and scalable method is shown to enable the development of defined stem cell culture conditions, as well as the elucidation of the fundamental biological processes necessary for the realization of stem cell based therapies.


Assuntos
Materiais Biocompatíveis/química , Moléculas de Adesão Celular/química , Proteínas da Matriz Extracelular/química , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Engenharia Tecidual/métodos , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Quitina/química , Quitosana/química , Humanos , Ácido Hialurônico/química , Teste de Materiais , Células-Tronco Mesenquimais/fisiologia , Camundongos , Células NIH 3T3 , Osteoblastos/fisiologia , Propriedades de Superfície
13.
Biotechniques ; 47(1): 599-606, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19594444

RESUMO

Enumeration of human embryonic stem cell (hESC) numbers through single cell digestion can be time consuming especially in high-throughput or multi-factorial analysis containing 50+ samples. We have developed a reproducible, cost-effective method of counting hESCs in clumps circumventing the need to manually dissociate each sample to single cells. The method is based on the DNA binding capacity of propidium iodide (PI) and subsequent fluorescent signal detection. Standard curves generated for cell numbers versus PI fluorescence as single cells or clumps showed an almost identical relationship in the lines of best fit. The reproducibility of the assay was first demonstrated by seeding hESC clumps at specific cell densities ranging 0.05[#x02013]2x105 cells/well and then secondly by using the assay to count cell numbers after different growth conditions. Validation tests showed that consistent seeding densities are important in maintaining undifferentiated hESC culture and that the assay can be used to estimate relative cell numbers and growth curves with high accuracy.


Assuntos
Técnicas de Cultura de Células/métodos , Corantes/metabolismo , DNA/metabolismo , Células-Tronco Embrionárias/citologia , Propídio/metabolismo , Biomarcadores/análise , Contagem de Células/economia , Linhagem Celular , Separação Celular , Detergentes/farmacologia , Endopeptidases/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Octoxinol/farmacologia , Reprodutibilidade dos Testes , Fatores de Tempo , Tripsina/farmacologia
14.
Stem Cells Dev ; 18(8): 1135-40, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19397457

RESUMO

Use of flow cytometry to detect pluripotency markers on or in human embryonic stem cells (hESCs) is a powerful analytical tool. However, current staining methodologies for high-content analysis of large numbers of samples utilize large quantities of primary and secondary antibodies, are time consuming, and may suffer from sample-to-sample variability. To circumvent these issues, we have developed a reproducible, quick, and cost-effective method of staining 12 populations of hESCs grown under different conditions by labeling each with a unique optical signature (UOS). The UOS for each population is achieved by combining different combinations and concentrations of 3 esterase activated, live cell, fluorescent indicators. The individually stained populations are then combined and an aliquot of the hESC samples stained for pluripotency or other markers of interest in the far-red region of the spectrum. Based on the unique fluorescent intensity and emission wavelengths of each population, the characteristics of each population are decoded in software after flow cytometric analysis. We have validated both our staining procedure and decoding methods by mixing populations of differentiated and undifferentiated hESCs and successfully quantifying differences in the pluripotency markers SSEA-4, Tra-1-60, GCTM2, and CD9 between the 12 different populations. Our multiplexing approach allows for the addition of internal controls and reduces sample-to-sample variation, while offering a significant reduction in time and reagent consumption. We anticipate that this method will be of great benefit to laboratories conducting high-content flow cytometric analysis of hESCs.


Assuntos
Células-Tronco Embrionárias/química , Citometria de Fluxo/métodos , Coloração e Rotulagem/métodos , Biomarcadores/análise , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Células-Tronco Embrionárias/citologia , Humanos , Células-Tronco Pluripotentes/química , Células-Tronco Pluripotentes/citologia
15.
J Proteome Res ; 6(9): 3796-807, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17655345

RESUMO

The unique pluripotential characteristic of human embryonic stem cells heralds their use in fields such as medicine, biotechnology, biopharmaceuticals, and developmental biology. However, the current availability of sufficient quantities of embryonic stem cells for such applications is limited, and generating sufficient numbers for downstream therapeutic applications is a key concern. In the absence of feeder layers or their conditioned media, human embryonic stem cells readily differentiate to form embryoid bodies, indicating that trophic factors secreted by the feeder layers are required for long-term proliferation and maintenance of pluripotency. Adding further complexity to the elucidation of the factors required for the maintenance of pluripotency is the variability of different fibroblast feeder layers (of mouse or human origin) to effectively support human embryonic stem cells. Currently, the deficiency of knowledge concerning the exact identity of factors within the pathways for self-renewal illustrates that a number of factors may be required to support pluripotent, undifferentiated growth of human embryonic stem cells. This study utilized a proteomic analysis (multidimensional chromatography coupled to tandem mass spectrometry) to isolate and identify proteins in the conditioned media of three mitotically inactivated fibroblast lines (human fetal, human neonatal, and mouse embryonic fibroblasts) used to support the undifferentiated growth of human embryonic stem cells. One-hundred seventy-five unique proteins were identified between the three cell lines using a

Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Proteômica/métodos , Animais , Bovinos , Diferenciação Celular , Cromatografia Líquida/métodos , Meios de Cultivo Condicionados/farmacologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Espectrometria de Massas , Camundongos , Fator de Crescimento Transformador beta/metabolismo
16.
Proteomics ; 5(4): 978-89, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15712233

RESUMO

The pathways involved in the maintenance of human embryonic stem (hES) cells remain largely unknown, although some signaling pathways have been identified in mouse embryonic stem (mES) cells. Fibroblast feeder layers are used to maintain the undifferentiated growth of hES cells and an examination of the conditioned media (CM) of human neonatal fibroblasts (HNFs) could provide insights into the maintenance of hES cells. The neonatal foreskin fibroblast line (HNF02) used in this study was shown to have a normal 2n = 46, XY chromosomal complement and to support the undifferentiated growth of the Embryonic Stem Cell International Pte. Ltd.-hES3 cell line. The CM of HNF02 was examined using two-dimensional liquid chromatography-tandem mass spectrometry (2-D LCMS) and two-dimensional electrophoresis (2-DE) followed by matrix-assisted laser desorption/ionization-time of flight tandem mass spectrometry (2-DE/MALDI). A total of 102 proteins were identified, 19 by 2-DE/MALDI, 53 by 2-D LCMS and 30 by both techniques. These proteins were classified into 15 functional groups. Proteins identified in the extracellular matrix and differentiation and growth factor functional categories were considered most likely to be involved in the maintenance of hES cell growth, differentiation and pluripotency as these groups contained proteins involved in a variety of events including cell adhesion, cell proliferation and inhibition of cell proliferation, Wnt signaling and inhibition of bone morphogenetic proteins.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Proteômica/métodos , Células-Tronco/citologia , Proteínas Morfogenéticas Ósseas/química , Adesão Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Cromatografia Líquida , Técnicas de Cocultura , Citogenética , Eletroforese em Gel Bidimensional , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Humanos , Concentração de Íons de Hidrogênio , Recém-Nascido , Cariotipagem , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...