Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Nucleic Acids ; 28: 859-874, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35694211

RESUMO

Arginase deficiency is associated with prominent neuromotor features, including spastic diplegia, clonus, and hyperreflexia; intellectual disability and progressive neurological decline are other signs. In a constitutive murine model, we recently described leukodystrophy as a significant component of the central nervous system features of arginase deficiency. In the present studies, we sought to examine if the administration of a lipid nanoparticle carrying human ARG1 mRNA to constitutive knockout mice could prevent abnormalities in myelination associated with arginase deficiency. Imaging of the cingulum, striatum, and cervical segments of the corticospinal tract revealed a drastic reduction of myelinated axons; signs of degenerating axons were also present with thin myelin layers. Lipid nanoparticle/ARG1 mRNA administration resulted in both light and electron microscopic evidence of a dramatic recovery of myelin density compared with age-matched controls; oligodendrocytes were seen to be extending processes to wrap many axons. Abnormally thin myelin layers, when myelination was present, were resolved with intermittent mRNA administration, indicative of not only a greater density of myelinated axons but also an increase in the thickness of the myelin sheath. In conclusion, lipid nanoparticle/ARG1 mRNA administration in arginase deficiency prevents the associated leukodystrophy and restores normal oligodendrocyte function.

2.
Mol Ther Methods Clin Dev ; 25: 278-296, 2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35505663

RESUMO

Creatine deficiency disorders are inborn errors of creatine metabolism, an energy homeostasis molecule. One of these, guanidinoacetate N-methyltransferase (GAMT) deficiency, has clinical characteristics that include features of autism, self-mutilation, intellectual disability, and seizures, with approximately 40% having a disorder of movement; failure to thrive can also be a component. Along with low creatine levels, guanidinoacetic acid (GAA) toxicity has been implicated in the pathophysiology of the disorder. Present-day therapy with oral creatine to control GAA lacks efficacy; seizures can persist. Dietary management and pharmacological ornithine treatment are challenging. Using an AAV-based gene therapy approach to express human codon-optimized GAMT in hepatocytes, in situ hybridization, and immunostaining, we demonstrated pan-hepatic GAMT expression. Serial collection of blood demonstrated a marked early and sustained reduction of GAA with normalization of plasma creatine; urinary GAA levels also markedly declined. The terminal time point demonstrated marked improvement in cerebral and myocardial creatine levels. In conjunction with the biochemical findings, treated mice gained weight to nearly match their wild-type littermates, while behavioral studies demonstrated resolution of abnormalities; PET-CT imaging demonstrated improvement in brain metabolism. In conclusion, a gene therapy approach can result in long-term normalization of GAA with increased creatine in guanidinoacetate N-methyltransferase deficiency and at the same time resolves the behavioral phenotype in a murine model of the disorder. These findings have important implications for the development of a new therapy for this abnormality of creatine metabolism.

3.
Yale J Biol Med ; 94(4): 545-557, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34970092

RESUMO

CPS1 deficiency is an inborn error of metabolism caused by loss-of-function mutations in the CPS1 gene, catalyzing the initial reaction of the urea cycle. Deficiency typically leads to toxic levels of plasma ammonia, cerebral edema, coma, and death, with the only curative treatment being liver transplantation; due to limited donor availability and the invasiveness and complications of the procedure, however, alternative therapies are needed. Induced pluripotent stem cells offer an alternative cell source to partial or whole liver grafts that theoretically would not require immune suppression regimens and additionally are amenable to genetic modifications. Here, we genetically modified CPS1 deficient patient-derived stem cells to constitutively express human codon optimized CPS1 from the AAVS1 safe harbor site. While edited stem cells efficiently differentiated to hepatocyte-like cells, they failed to metabolize ammonia more efficiently than their unedited counterparts. This unexpected result appears to have arisen in part due to transgene promoter methylation, and thus transcriptional silencing, in undifferentiated cells, impacting their capacity to restore the complete urea cycle function upon differentiation. As pluripotent stem cell strategies are being expanded widely for potential cell therapies, these results highlight the need for strict quality control and functional analysis to ensure the integrity of cell products.


Assuntos
Células-Tronco Pluripotentes Induzidas , Carbamoil-Fosfato Sintase (Amônia)/genética , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genômica , Homeostase , Humanos , Nitrogênio
4.
Mol Ther ; 28(7): 1717-1730, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32359471

RESUMO

The urea cycle enzyme carbamoyl phosphate synthetase 1 (CPS1) catalyzes the initial step of the urea cycle; bi-allelic mutations typically present with hyperammonemia, vomiting, ataxia, lethargy progressing into coma, and death due to brain edema if ineffectively treated. The enzyme deficiency is particularly difficult to treat; early recognition is essential to minimize injury to the brain. Even under optimal conditions, therapeutic interventions are of limited scope and efficacy, with most patients developing long-term neurologic sequelae. One significant encumberment to gene therapeutic development is the size of the CPS1 cDNA, which, at 4.5 kb, nears the packaging capacity of adeno-associated virus (AAV). Herein we developed a split AAV (sAAV)-based approach, packaging the large transgene and its regulatory cassette into two separate vectors, thereby delivering therapeutic CPS1 by a dual vector system with testing in a murine model of the disorder. Cps1-deficient mice treated with sAAVs survive long-term with markedly improved ammonia levels, diminished dysregulation of circulating amino acids, and increased hepatic CPS1 expression and activity. In response to acute ammonia challenging, sAAV-treated female mice rapidly incorporated nitrogen into urea. This study demonstrates the first proof-of-principle that sAAV-mediated therapy is a viable, potentially clinically translatable approach to CPS1 deficiency, a devastating urea cycle disorder.


Assuntos
Carbamoil-Fosfato Sintase (Amônia)/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/terapia , Dependovirus/genética , Ureia/metabolismo , Amônia/metabolismo , Animais , Doença da Deficiência da Carbamoil-Fosfato Sintase I/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/metabolismo , Empacotamento do DNA , Modelos Animais de Doenças , Feminino , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Estudo de Prova de Conceito
5.
J Inherit Metab Dis ; 42(6): 1044-1053, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30835861

RESUMO

The enzyme carbamoyl phosphate synthetase 1 (CPS1; EC 6.3.4.16) forms carbamoyl phosphate from bicarbonate, ammonia, and adenosine triphosphate (ATP) and is activated allosterically by N-acetylglutamate. The neonatal presentation of bi-allelic mutations of CPS1 results in hyperammonemia with reduced citrulline and is reported as the most challenging nitrogen metabolism disorder to treat. As therapeutic interventions are limited, patients often develop neurological injury or die from hyperammonemia. Survivors remain vulnerable to nitrogen overload, being at risk for repetitive neurological injury. With transgenic technology, our lab developed a constitutive Cps1 mutant mouse and reports its characterization herein. Within 24 hours of birth, all Cps1 -/- mice developed hyperammonemia and expired. No CPS1 protein by Western blot or immunostaining was detected in livers nor was Cps1 mRNA present. CPS1 enzymatic activity was markedly decreased in knockout livers and reduced in Cps1+/- mice. Plasma analysis found markedly reduced citrulline and arginine and markedly increased glutamine and alanine, both intermolecular carriers of nitrogen, along with elevated ammonia, taurine, and lysine. Derangements in multiple other amino acids were also detected. While hepatic amino acids also demonstrated markedly reduced citrulline, arginine, while decreased, was not statistically significant; alanine and lysine were markedly increased while glutamine was trending towards significance. In conclusion we have determined that this constitutive neonatal mouse model of CPS1 deficiency replicates the neonatal human phenotype and demonstrates the key biochemical features of the disorder. These mice will be integral for addressing the challenges of developing new therapeutic approaches for this, at present, poorly treated disorder.


Assuntos
Carbamoil-Fosfato Sintase (Amônia)/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/complicações , Doença da Deficiência da Carbamoil-Fosfato Sintase I/mortalidade , Glutamina/sangue , Hiperamonemia , Animais , Animais Recém-Nascidos , Carbamoil-Fosfato Sintase (Amônia)/deficiência , Doença da Deficiência da Carbamoil-Fosfato Sintase I/sangue , Doença da Deficiência da Carbamoil-Fosfato Sintase I/genética , Hiperamonemia/sangue , Hiperamonemia/complicações , Hiperamonemia/genética , Hiperamonemia/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação
6.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30724386

RESUMO

The urea cycle and glutamine synthetase (GS) are the two main pathways for waste nitrogen removal and their deficiency results in hyperammonemia. Here, we investigated the efficacy of liver-specific GS overexpression for therapy of hyperammonemia. To achieve hepatic GS overexpression, we generated a helper-dependent adenoviral (HDAd) vector expressing the murine GS under the control of a liver-specific expression cassette (HDAd-GS). Compared to mice injected with a control vector expressing an unrelated reporter gene (HDAd-alpha-fetoprotein), wild-type mice with increased hepatic GS showed reduced blood ammonia levels and a concomitant increase of blood glutamine after intraperitoneal injections of ammonium chloride, whereas blood urea was unaffected. Moreover, injection of HDAd-GS reduced blood ammonia levels at baseline and protected against acute hyperammonemia following ammonia challenge in a mouse model with conditional hepatic deficiency of carbamoyl phosphate synthetase 1 (Cps1), the initial and rate-limiting step of ureagenesis. In summary, we found that upregulation of hepatic GS reduced hyperammonemia in wild-type and Cps1-deficient mice, thus confirming a key role of GS in ammonia detoxification. These results suggest that hepatic GS augmentation therapy has potential for treatment of both primary and secondary forms of hyperammonemia.


Assuntos
Amônia/metabolismo , Terapia Genética/métodos , Glutamato-Amônia Ligase/genética , Hiperamonemia/genética , Hiperamonemia/terapia , Fígado/metabolismo , Amônia/toxicidade , Animais , Carbamoil-Fosfato Sintase (Amônia)/genética , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Doença da Deficiência da Carbamoil-Fosfato Sintase I/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/metabolismo , Doença da Deficiência da Carbamoil-Fosfato Sintase I/terapia , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Glutamato-Amônia Ligase/metabolismo , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos/genética
7.
Mol Genet Metab ; 124(4): 243-253, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29801986

RESUMO

Carbamoyl phosphate synthetase 1 (CPS1) is a urea cycle enzyme that forms carbamoyl phosphate from bicarbonate, ammonia and ATP. Bi-allelic mutations of the CPS1 gene result in a urea cycle disorder presenting with hyperammonemia, often with reduced citrulline, and without orotic aciduria. CPS1 deficiency is particularly challenging to treat and lack of early recognition typically results in early neonatal death. Therapeutic interventions have limited efficacy and most patients develop long-term neurologic sequelae. Using transgenic techniques, we generated a conditional Cps1 knockout mouse. By loxP/Cre recombinase technology, deletion of the Cps1 locus was achieved in adult transgenic animals using a Cre recombinase-expressing adeno-associated viral vector. Within four weeks from vector injection, all animals developed hyperammonemia without orotic aciduria and died. Minimal CPS1 protein was detectable in livers. To investigate the efficacy of gene therapy for CPS deficiency following knock-down of hepatic endogenous CPS1 expression, we injected these mice with a helper-dependent adenoviral vector (HDAd) expressing the large murine CPS1 cDNA under control of the phosphoenolpyruvate carboxykinase promoter. Liver-directed HDAd-mediated gene therapy resulted in survival, normalization of plasma ammonia and glutamine, and 13% of normal Cps1 expression. A gender difference in survival suggests that female mice may require higher hepatic CPS1 expression. We conclude that this conditional murine model recapitulates the clinical and biochemical phenotype detected in human patients with CPS1 deficiency and will be useful to investigate ammonia-mediated neurotoxicity and for the development of cell- and gene-based therapeutic approaches.


Assuntos
Carbamoil-Fosfato Sintase (Amônia)/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/terapia , Terapia Genética , Hiperamonemia/terapia , Amônia/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/uso terapêutico , Doença da Deficiência da Carbamoil-Fosfato Sintase I/genética , Doença da Deficiência da Carbamoil-Fosfato Sintase I/metabolismo , Doença da Deficiência da Carbamoil-Fosfato Sintase I/patologia , Carbamoil-Fosfato/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Glutamina/metabolismo , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patologia , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Mutação , Orotato Fosforribosiltransferase/deficiência , Orotato Fosforribosiltransferase/genética , Orotidina-5'-Fosfato Descarboxilase/deficiência , Orotidina-5'-Fosfato Descarboxilase/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/genética , Erros Inatos do Metabolismo da Purina-Pirimidina/patologia
8.
Mol Genet Metab ; 124(2): 114-123, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29724658

RESUMO

The transplantation, engraftment, and expansion of primary hepatocytes have the potential to be an effective therapy for metabolic disorders of the liver including those of nitrogen metabolism. To date, such methods for the treatment of urea cycle disorders in murine models has only been minimally explored. Arginase deficiency, an inherited disorder of nitrogen metabolism that presents in the first two years of life, has the potential to be treated by such methods. To explore the potential of this approach, we mated the conditional arginase deficient mouse with a mouse model deficient in fumarylacetoacetate hydrolase (FAH) and with Rag2 and IL2-Rγ mutations to give a selective advantage to transplanted (normal) human hepatocytes. On day -1, a uroplasminogen-expressing adenoviral vector was administered intravenously followed the next day with the transplantation of 1 × 106 human hepatocytes (or vehicle alone) by intrasplenic injection. As the initial number of administered hepatocytes would be too low to prevent hepatotoxicity-induced mortality, NTBC cycling was performed to allow for hepatocyte expansion and repopulation. While all control mice died, all except one human hepatocyte transplanted mice survived. Four months after hepatocyte transplantation, 2 × 1011 genome copies of AAV-TBG-Cre recombinase was administered IV to disrupt endogenous hepatic arginase expression. While all control mice died within the first month, human hepatocyte transplanted mice did well. Ammonia and amino acids, analyzed in both groups before and after disruption of endogenous arginase expression, while well-controlled in the transplanted group, were markedly abnormal in the controls. Ammonium challenging further demonstrated the durability and functionality of the human repopulated liver. In conclusion, these studies demonstrate that human hepatocyte repopulation in the murine liver can result in effective treatment of arginase deficiency.


Assuntos
Arginase/fisiologia , Predisposição Genética para Doença , Hepatócitos/transplante , Hepatopatias/terapia , Doenças Metabólicas/terapia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Hepatócitos/citologia , Humanos , Hepatopatias/enzimologia , Hepatopatias/patologia , Masculino , Doenças Metabólicas/enzimologia , Doenças Metabólicas/patologia , Camundongos , Camundongos Knockout
9.
Dis Markers ; 2017: 7506976, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28781416

RESUMO

We analyzed the effect of transcribed noncoding RNA centromeric satellites on chromosome segregation in normal human and murine stem and fibrosarcoma cells. The overexpression of different centromeric alphoid DNAs in all cell lines induced a marked increase in chromosome mis-segregation in anaphase. Overexpression of centromeric mouse minor satellite also increased chromosome instability in the murine stem but not in human cells. Analysis of chromosome segregation in vivo showed disturbances in the mitotic progression, which was frequently unresolved. Live cell imaging revealed that overexpression of centromeric satellites resulted in several different chromosomal morphological errors in the cell nuclei. Our findings correlated with other reports that several centromeric noncoding RNAs are detected in different carcinoma cells and their expression resulted in segregation errors. Our study furnishes further insights into a novel source of genomic instability in human and murine cells. It has recently been shown that noncoding centromeric RNAs are present in some form of cancer, and thus, overexpression of several types of centromeric noncoding RNAs may be useful as a specific maker for neoplastic cells.


Assuntos
Instabilidade Cromossômica , RNA não Traduzido/genética , Células-Tronco/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Centrômero/metabolismo , Segregação de Cromossomos , Humanos , Camundongos , Células-Tronco/citologia
10.
Stem Cell Reports ; 6(1): 95-108, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26771355

RESUMO

The generation of tissue-specific cell types from human embryonic stem cells (hESCs) is critical for the development of future stem cell-based regenerative therapies. Here, we identify CD13 and ROR2 as cell-surface markers capable of selecting early cardiac mesoderm emerging during hESC differentiation. We demonstrate that the CD13+/ROR2+ population encompasses pre-cardiac mesoderm, which efficiently differentiates to all major cardiovascular lineages. We determined the engraftment potential of CD13+/ROR2+ in small (murine) and large (porcine) animal models, and demonstrated that CD13+/ROR2+ progenitors have the capacity to differentiate toward cardiomyocytes, fibroblasts, smooth muscle, and endothelial cells in vivo. Collectively, our data show that CD13 and ROR2 identify a cardiac lineage precursor pool that is capable of successful engraftment into the porcine heart. These markers represent valuable tools for further dissection of early human cardiac differentiation, and will enable a detailed assessment of human pluripotent stem cell-derived cardiac lineage cells for potential clinical applications.


Assuntos
Antígenos CD13/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Mesoderma/metabolismo , Miocárdio/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Animais , Antígenos CD13/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica/métodos , Células-Tronco Embrionárias Humanas/citologia , Humanos , Mesoderma/citologia , Camundongos , Músculo Liso/citologia , Músculo Liso/metabolismo , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Suínos , Fatores de Tempo , Transplante Heterólogo
11.
Stem Cells Transl Med ; 5(1): 67-74, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26582908

RESUMO

UNLABELLED: Given the limited regenerative capacity of the heart, cellular therapy with stem cell-derived cardiac cells could be a potential treatment for patients with heart disease. However, reliable imaging techniques to longitudinally assess engraftment of the transplanted cells are scant. To address this issue, we used ferumoxytol as a labeling agent of human embryonic stem cell-derived cardiac progenitor cells (hESC-CPCs) to facilitate tracking by magnetic resonance imaging (MRI) in a large animal model. Differentiating hESCs were exposed to ferumoxytol at different time points and varying concentrations. We determined that treatment with ferumoxytol at 300 µg/ml on day 0 of cardiac differentiation offered adequate cell viability and signal intensity for MRI detection without compromising further differentiation into definitive cardiac lineages. Labeled hESC-CPCs were transplanted by open surgical methods into the left ventricular free wall of uninjured pig hearts and imaged both ex vivo and in vivo. Comprehensive T2*-weighted images were obtained immediately after transplantation and 40 days later before termination. The localization and dispersion of labeled cells could be effectively imaged and tracked at days 0 and 40 by MRI. Thus, under the described conditions, ferumoxytol can be used as a long-term, differentiation-neutral cell-labeling agent to track transplanted hESC-CPCs in vivo using MRI. SIGNIFICANCE: The development of a safe and reproducible in vivo imaging technique to track the fate of transplanted human embryonic stem cell-derived cardiac progenitor cells (hESC-CPCs) is a necessary step to clinical translation. An iron oxide nanoparticle (ferumoxytol)-based approach was used for cell labeling and subsequent in vivo magnetic resonance imaging monitoring of hESC-CPCs transplanted into uninjured pig hearts. The present results demonstrate the use of ferumoxytol labeling and imaging techniques in tracking the location and dispersion of cell grafts, highlighting its utility in future cardiac stem cell therapy trials.


Assuntos
Rastreamento de Células/métodos , Células-Tronco Embrionárias , Óxido Ferroso-Férrico/farmacologia , Imageamento por Ressonância Magnética , Mioblastos Cardíacos , Transplante de Células-Tronco , Células-Tronco Embrionárias/diagnóstico por imagem , Células-Tronco Embrionárias/transplante , Compostos Férricos/farmacologia , Óxido Ferroso-Férrico/farmacocinética , Xenoenxertos , Humanos , Mioblastos Cardíacos/diagnóstico por imagem , Mioblastos Cardíacos/transplante , Radiografia
12.
Hum Mol Genet ; 20(15): 2905-13, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21593218

RESUMO

We present a novel and efficient non-integrating gene expression system in human embryonic stem cells (hESc) utilizing human artificial chromosomes (HAC), which behave as autonomous endogenous host chromosomes and segregate correctly during cell division. HAC are important vectors for investigating the organization and structure of the kinetochore, and gene complementation. HAC have so far been obtained in immortalized or tumour-derived cell lines, but never in stem cells, thus limiting their potential therapeutic application. In this work, we modified the herpes simplex virus type 1 amplicon system for efficient transfer of HAC DNA into two hESc. The deriving stable clones generated green fluorescent protein gene-expressing HAC at high frequency, which were stably maintained without selection for 3 months. Importantly, no integration of the HAC DNA was observed in the hESc lines, compared with the fibrosarcoma-derived control cells, where the exogenous DNA frequently integrated in the host genome. The hESc retained pluripotency, differentiation and teratoma formation capabilities. This is the first report of successfully generating gene expressing de novo HAC in hESc, and is a significant step towards the genetic manipulation of stem cells and potential therapeutic applications.


Assuntos
Cromossomos Artificiais Humanos/metabolismo , Células-Tronco Embrionárias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Cromossomos Artificiais Humanos/genética , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Imunofluorescência , Herpesvirus Humano 1/genética , Humanos
13.
Stem Cell Rev Rep ; 7(2): 471-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21188651

RESUMO

Prolonged in vitro culture of human embryonic stem (hES) cells can result in chromosomal abnormalities believed to confer a selective advantage. This potential occurrence has crucial implications for the appropriate use of hES cells for research and therapeutic purposes. In view of this, time-point karyotypic evaluation to assess genetic stability is recommended as a necessary control test to be carried out during extensive 'passaging'. Standard techniques currently used for the cytogenetic assessment of ES cells include G-banding and/or Fluorescence in situ Hybridization (FISH)-based protocols for karyotype analysis, including M-FISH and SKY. Critical for both banding and FISH techniques are the number and quality of metaphase spreads available for analysis at the microscope. Protocols for chromosome preparation from hES and human induced pluripotent stem (hiPS) cells published so far appear to differ considerably from one laboratory to another. Here we present an optimized technique, in which both the number and the quality of chromosome metaphase spreads were substantially improved when compared to current standard techniques for chromosome preparations. We believe our protocol represents a significant advancement in this line of work, and has the required attributes of simplicity and consistency to be widely accepted as a reference method for high quality, fast chromosomal analysis of human ES and iPS cells.


Assuntos
Células-Tronco Embrionárias/classificação , Células-Tronco Pluripotentes Induzidas/classificação , Cariotipagem/métodos , Técnicas de Cultura de Células , Cromossomos Humanos , Demecolcina/química , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Hibridização in Situ Fluorescente , Indicadores e Reagentes/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Nocodazol/química , Coloração e Rotulagem/métodos
14.
BMC Infect Dis ; 10: 7, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20064274

RESUMO

BACKGROUND: We have previously reported a HIV-1 subtype A infection in a community of injection drug users (IDUs) in Karachi, Pakistan. We now show that this infection among the IDUs may have originated from a single source. METHODS: Phylogenetic analysis was performed of partial gag sequences, generated using PCR, from 26 HIV-positive IDU samples. RESULTS: Our results showed formation of a tight monophyletic group with an intra-sequence identity of < 98% indicating a "founder effect". Our data indicate that the HIV-1 epidemic in this community of IDUs may have been transmitted by an HIV positive overseas contract worker who admitted to having contact with commercial sex workers during stay abroad. CONCLUSION: Specific measures need to implemented to control transmission of HIV infection in Pakistan through infected migrant workers.


Assuntos
Usuários de Drogas , Efeito Fundador , Infecções por HIV/epidemiologia , HIV-1/genética , Adulto , HIV-1/classificação , Humanos , Masculino , Paquistão/epidemiologia , Filogenia , Vigilância da População , RNA Viral/genética , Alinhamento de Sequência , Análise de Sequência de RNA , Trabalho Sexual , Migrantes , Adulto Jovem
15.
PLoS One ; 3(9): e3191, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18784834

RESUMO

BACKGROUND: Genetic analysis of a viral infection helps in following its spread in a given population, in tracking the routes of infection and, where applicable, in vaccine design. Additionally, sequence analysis of the viral genome provides information about patterns of genetic divergence that may have occurred during viral evolution. OBJECTIVE: In this study we have analyzed the subtypes of Human Immunodeficiency Virus -1 (HIV-1) circulating in a diverse sample population of Nairobi, Kenya. METHODOLOGY: 69 blood samples were collected from a diverse subject population attending the Aga Khan University Hospital in Nairobi, Kenya. Total DNA was extracted from peripheral blood mononuclear cells (PBMCs), and used in a Polymerase Chain Reaction (PCR) to amplify the HIV gag gene. The PCR amplimers were partially sequenced, and alignment and phylogenetic analysis of these sequences was performed using the Los Alamos HIV Database. RESULTS: Blood samples from 69 HIV-1 infected subjects from varying ethnic backgrounds were analyzed. Sequence alignment and phylogenetic analysis showed 39 isolates to be subtype A, 13 subtype D, 7 subtype C, 3 subtype AD and CRF01_AE, 2 subtype G and 1 subtype AC and 1 AG. Deeper phylogenetic analysis revealed HIV subtype A sequences to be highly divergent as compared to subtypes D and C. CONCLUSION: Our analysis indicates that HIV-1 subtypes in the Nairobi province of Kenya are dominated by a genetically diverse clade A. Additionally, the prevalence of highly divergent, complex subtypes, intersubtypes, and the recombinant forms indicates viral mixing in Kenyan population, possibly as a result of dual infections.


Assuntos
Infecções por HIV/virologia , HIV-1/genética , Adolescente , Adulto , DNA Viral/genética , Feminino , Genoma Viral , Infecções por HIV/sangue , Humanos , Quênia , Masculino , Pessoa de Meia-Idade , Filogenia , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Globinas beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...