Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Biochim Biophys Acta ; 1863(7 Pt B): 1728-48, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26524115

RESUMO

Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Assuntos
Pesquisa Biomédica/métodos , Fármacos Cardiovasculares/farmacologia , Linhagem da Célula , Descoberta de Drogas/métodos , Cardiopatias/tratamento farmacológico , Ensaios de Triagem em Larga Escala , Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Testes de Toxicidade/métodos , Fármacos Cardiovasculares/toxicidade , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Genótipo , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fenótipo , Medição de Risco
2.
Bioessays ; 35(3): 281-98, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22886688

RESUMO

The emphasis in human pluripotent stem cell (hPSC) technologies has shifted from cell therapy to in vitro disease modelling and drug screening. This review examines why this shift has occurred, and how current technological limitations might be overcome to fully realise the potential of hPSCs. Details are provided for all disease-specific human induced pluripotent stem cell lines spanning a dozen dysfunctional organ systems. Phenotype and pharmacology have been examined in only 17 of 63 lines, primarily those that model neurological and cardiac conditions. Drug screening is most advanced in hPSC-cardiomyocytes. Responses for almost 60 agents include examples of how careful tests in hPSC-cardiomyocytes have improved on existing in vitro assays, and how these cells have been integrated into high throughput imaging and electrophysiology industrial platforms. Such successes will provide an incentive to overcome bottlenecks in hPSC technology such as improving cell maturity and industrial scalability whilst reducing cost.


Assuntos
Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Células-Tronco Pluripotentes/metabolismo , Animais , Ensaios de Triagem em Larga Escala , Humanos , Fenótipo , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco
3.
Biochim Biophys Acta ; 1830(6): 3517-24, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23403134

RESUMO

BACKGROUND: Online label-free monitoring of in-vitro differentiation of stem cells remains a major challenge in stem cell research. In this paper we report the use of Raman micro-spectroscopy (RMS) to measure time- and spatially-resolved molecular changes in intact embryoid bodies (EBs) during in-vitro cardiogenic differentiation. METHODS: EBs formed by aggregation of human embryonic stem cells (hESCs) were cultured in defined medium to induce differentiation towards cardiac phenotype and maintained in purpose-built micro-bioreactors on the Raman microscope for 5days (between days 5 and 9 of differentiation) and spatially-resolved spectra were recorded at 24h intervals. RESULTS: The Raman spectra showed that the onset of spontaneous beating of EBs at day 7 coincided with an increase in the intensity of the Raman bands at 1340cm(-1), 1083cm(-1), 937cm(-1), 858cm(-1), 577cm(-1) and 482cm(-1). The spectral maps corresponding to these bands had a high positive correlation with the expression of the cardiac-specific α-actinin obtained by immuno-fluorescence imaging of the same EBs. The spectral markers obtained here are also in agreement with previous studies performed on individual live hESC-derived CMs. CONCLUSIONS: The intensity profile of these Raman bands can be used for label-free in-situ monitoring of EBs to estimate the efficacy of cardiogenic differentiation. GENERAL SIGNIFICANCE: As the acquisition of the time-course Raman spectra did not affect the viability or the differentiation potential of the hESCs, this study demonstrates the feasibility of using RMS for on-line non-invasive continuous monitoring of such processes inside bioreactor culture systems.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias , Miócitos Cardíacos , Análise Espectral Raman/métodos , Actinina/biossíntese , Antígenos de Diferenciação/biossíntese , Técnicas de Cultura de Células , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo
4.
Mol Ther Methods Clin Dev ; 32(2): 101271, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38946936

RESUMO

Hematopoietic stem cell gene therapy (HSCGT) is a promising therapeutic strategy for the treatment of neurodegenerative, metabolic disorders. The approach involves the ex vivo introduction of a missing gene into patients' own stem cells via lentiviral-mediated transduction (TD). Once transplanted back into a fully conditioned patient, these genetically modified HSCs can repopulate the blood system and produce the functional protein, previously absent or non-functional in the patient, which can then cross-correct other affected cells in somatic organs and the central nervous system. We previously developed an HSCGT approach for the treatment of Mucopolysaccharidosis type II (MPSII) (Hunter syndrome), a debilitating pediatric lysosomal disorder caused by mutations in the iduronate-2-sulphatase (IDS) gene, leading to the accumulation of heparan and dermatan sulfate, which causes severe neurodegeneration, skeletal abnormalities, and cardiorespiratory disease. In HSCGT proof-of-concept studies using lentiviral IDS fused to a brain-targeting peptide ApoEII (IDS.ApoEII), we were able to normalize brain pathology and behavior of MPSII mice. Here we present an optimized and validated good manufacturing practice hematopoietic stem cell TD protocol for MPSII in preparation for first-in-man studies. Inclusion of TEs LentiBOOST and protamine sulfate significantly improved TD efficiency by at least 3-fold without causing adverse toxicity, thereby reducing vector quantity required.

5.
Cells Tissues Organs ; 195(6): 484-94, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22123583

RESUMO

Bone marrow-derived mesenchymal stem cells (MSCs) are widely used as a cellular model of bone formation, and can mineralize in vitro in response to osteogenic medium (OM). It is unclear, however, whether this property is specific to cells of mesenchymal origin. We analysed the OM response in 3 non-osteogenic lines, HEK293, HeLa and NTera, compared to MSCs. Whereas HEK293 cells failed to respond to OM conditions, the 2 carcinoma-derived lines NTera and HeLa deposited a calcium phosphate mineral comparable to that present in MSC cultures. However, unlike MSCs, HeLa and NTera cultures did so in the absence of dexamethasone. This discrepancy was confirmed, as bone morphogenetic protein inhibition obliterated the OM response in MSCs but not in HeLa or NTera, indicating that these 2 models can deposit mineral through a mechanism independent of established dexamethasone or bone morphogenetic protein signalling.


Assuntos
Osso e Ossos/citologia , Calcificação Fisiológica , Minerais/metabolismo , Osteogênese , Fosfatase Alcalina/metabolismo , Biomarcadores/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Calcificação Fisiológica/efeitos dos fármacos , Linhagem Celular , Meios de Cultura/farmacologia , Dexametasona/farmacologia , Citometria de Fluxo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/metabolismo , Osteogênese/efeitos dos fármacos , Fatores de Tempo
6.
J Neuromuscul Dis ; 3(3): 309-332, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27854224

RESUMO

Muscular dystrophies (MDs) are clinically and molecularly a highly heterogeneous group of single-gene disorders that primarily affect striated muscles. Cardiac disease is present in several MDs where it is an important contributor to morbidity and mortality. Careful monitoring of cardiac issues is necessary but current management of cardiac involvement does not effectively protect from disease progression and cardiac failure. There is a critical need to gain new knowledge on the diverse molecular underpinnings of cardiac disease in MDs in order to guide cardiac treatment development and assist in reaching a clearer consensus on cardiac disease management in the clinic. Animal models are available for the majority of MDs and have been invaluable tools in probing disease mechanisms and in pre-clinical screens. However, there are recognized genetic, physiological, and structural differences between human and animal hearts that impact disease progression, manifestation, and response to pharmacological interventions. Therefore, there is a need to develop parallel human systems to model cardiac disease in MDs. This review discusses the current status of cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSC) to model cardiac disease, with a focus on Duchenne muscular dystrophy (DMD) and myotonic dystrophy (DM1). We seek to provide a balanced view of opportunities and limitations offered by this system in elucidating disease mechanisms pertinent to human cardiac physiology and as a platform for treatment development or refinement.


Assuntos
Cardiopatias/fisiopatologia , Células-Tronco Pluripotentes Induzidas , Distrofia Muscular de Duchenne/fisiopatologia , Miócitos Cardíacos/citologia , Distrofia Miotônica/fisiopatologia , Cardiopatias/etiologia , Humanos , Modelos Cardiovasculares , Distrofias Musculares/etiologia , Distrofias Musculares/genética , Distrofias Musculares/fisiopatologia , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/genética , Distrofia Miotônica/complicações , Distrofia Miotônica/genética , Células-Tronco Pluripotentes
7.
Stem Cells Dev ; 25(6): 439-52, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26906236

RESUMO

Automated planar patch clamp systems are widely used in drug evaluation studies because of their ability to provide accurate, reliable, and reproducible data in a high-throughput manner. Typically, CHO and HEK tumorigenic cell lines overexpressing single ion channels are used since they can be harvested as high-density, homogenous, single-cell suspensions. While human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are physiologically more relevant, these cells are fragile, have complex culture requirements, are inherently heterogeneous, and are expensive to produce, which has restricted their use on automated patch clamp (APC) devices. Here, we used high efficiency differentiation protocols to produce cardiomyocytes from six different hPSC lines for analysis on the Patchliner (Nanion Technologies GmbH) APC platform. We developed a two-step cell preparation protocol that yielded cell catch rates and whole-cell breakthroughs of ∼80%, with ∼40% of these cells allowing electrical activity to be recorded. The protocol permitted formation of long-lasting (>15 min), high quality seals (>2 GΩ) in both voltage- and current-clamp modes. This enabled density of sodium, calcium, and potassium currents to be evaluated, along with dose-response curves to their respective channel inhibitors, tetrodotoxin, nifedipine, and E-4031. Thus, we show the feasibility of using the Patchliner platform for automated evaluation of the electrophysiology and pharmacology of hPSC-CMs, which will enable considerable increase in throughput for reliable and efficient drug evaluation.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp/métodos , Células-Tronco Pluripotentes/citologia , Cultura Primária de Células/métodos , Potenciais de Ação , Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/fisiologia , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Sódio/metabolismo , Bloqueadores dos Canais de Sódio/farmacologia
8.
Dis Model Mech ; 8(5): 457-66, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25791035

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD), and is characterized by progressive weakness in skeletal and cardiac muscles. Currently, dilated cardiomyopathy due to cardiac muscle loss is one of the major causes of lethality in late-stage DMD patients. To study the molecular mechanisms underlying dilated cardiomyopathy in DMD heart, we generated cardiomyocytes (CMs) from DMD and healthy control induced pluripotent stem cells (iPSCs). DMD iPSC-derived CMs (iPSC-CMs) displayed dystrophin deficiency, as well as the elevated levels of resting Ca(2+), mitochondrial damage and cell apoptosis. Additionally, we found an activated mitochondria-mediated signaling network underlying the enhanced apoptosis in DMD iPSC-CMs. Furthermore, when we treated DMD iPSC-CMs with the membrane sealant Poloxamer 188, it significantly decreased the resting cytosolic Ca(2+) level, repressed caspase-3 (CASP3) activation and consequently suppressed apoptosis in DMD iPSC-CMs. Taken together, using DMD patient-derived iPSC-CMs, we established an in vitro model that manifests the major phenotypes of dilated cardiomyopathy in DMD patients, and uncovered a potential new disease mechanism. Our model could be used for the mechanistic study of human muscular dystrophy, as well as future preclinical testing of novel therapeutic compounds for dilated cardiomyopathy in DMD patients.


Assuntos
Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Adolescente , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Distrofia Muscular de Duchenne/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Poloxâmero/farmacologia , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/genética
9.
Stem Cells Dev ; 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23786351

RESUMO

With an incidence of ~1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harbouring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ~30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.

10.
Stem Cells Dev ; 22(20): 2714-24, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23829870

RESUMO

With an incidence of ∼1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harboring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ∼30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.


Assuntos
Distrofina/genética , Expressão Gênica/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Distrofia Muscular de Duchenne/genética , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Sequência de Bases , Diferenciação Celular , Criança , Distrofina/metabolismo , Éxons , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Dados de Sequência Molecular , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Cultura Primária de Células
11.
J Bone Joint Surg Am ; 95(8): 736-43, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23595073

RESUMO

BACKGROUND: Strategies to improve mechanical strength, neovascularization, and the regenerative capacity of allograft include both the addition of skeletal stem cells and the investigation of novel biomaterials to reduce and ultimately obviate the need for allograft altogether. Use of bone cement is a common method of stabilizing implants in conjunction with impacted allograft. Curing cement, however, can reach temperatures in excess of 70°C, which is potentially harmful to skeletal stem cells. The aim of this study was to investigate the effects of setting bone cement on the survival of human adult skeletal stem cells within tissue-engineered allograft and a novel allograft substitute. METHODS: Milled allograft and a polymer graft substitute were seeded with skeletal stem cells, impacted into a graduated chamber, and exposed to curing bone cement. Sections were removed at 5-mm increments from the allograft-cement interface. A quantitative WST-1 assay was performed on each section as a measure of remaining cell viability. A second stage of the experiment involved assessment of methods to potentially enhance cell survival, including pretreating the allograft or polymer by either cooling to 5°C or coating with 1% Laponite, or both. RESULTS: There was a significant drop in cellular activity in the sections taken from within 0.5 cm of the cement interface in both the allograft and the polymer (p < 0.05), although there was still measurable cellular activity. Pretreatment methods did not significantly improve cell survival in any group. CONCLUSIONS: While the addition of bone cement reduced cellular viability of tissue-engineered constructs, this reduction occurred only in close proximity to the cement and measurable numbers of skeletal stem cells were observed, confirming the potential for cell population recovery.


Assuntos
Cimentos Ósseos , Substitutos Ósseos , Transplante Ósseo , Células-Tronco/fisiologia , Engenharia Tecidual , Idoso , Idoso de 80 Anos ou mais , Técnicas de Cultura de Células , Sobrevivência Celular , Durapatita , Humanos , Masculino , Poliésteres , Alicerces Teciduais
12.
J Biomed Mater Res A ; 100(12): 3211-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22707404

RESUMO

Impaction bone grafting (IBG) with human allograft remains the preferred approach for replacement of lost bone stock during revision hip surgery. Associated problems include cost, disease transmission, and stem subsidence. Synthetic grafts are therefore appealing, and ideally display similar mechanical characteristics as allograft, but with enhanced ability to form de novo bone. High and low molecular weight forms of three different polymers [poly(DL-lactide) (P(DL) LA), poly(DL-lactide-co-glycolide) (P(DL) LGA), and poly(ε-caprolactone) (PCL)] were milled, impacted into discs, and then examined in a shear testing rig, in comparison to allograft. In addition, skeletal stem cells (SSCs) were combined with each of the milled polymers, followed by impaction and examination for cell viability and number, via fluorostaining and biochemical assays. The shear strengths of high/low mwt P(DL) LA, and high/low mwt P(DL) LGA were significantly higher than allograft (p < 0.01). High/low mwt PCL had significantly lower shear strengths (p < 0.01). WST-1 assay and fluorstaining indicated significantly increased cell viability on high mwt P(DL) LA and high mwt P(DL) LGA over allograft (p < 0.05). Mechanical and biochemical analysis indicated improved properties of high mwt P(DL) LA and high mwt P(DL) LGA over allograft. This study indicates the potential of these polymers for use as substitute human allograft, creating a living composition with SSC for application in IBG.


Assuntos
Materiais Biocompatíveis/farmacologia , Transplante Ósseo , Teste de Materiais/métodos , Fenômenos Mecânicos/efeitos dos fármacos , Polímeros/química , Idoso , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Varredura , Resistência ao Cisalhamento/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Transplante Homólogo , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA