Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Inherit Metab Dis ; 46(5): 874-905, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37078180

RESUMO

Lysosomal Storage Disorders (LSDs) are a diverse group of inherited, monogenic diseases caused by functional defects in specific lysosomal proteins. The lysosome is a cellular organelle that plays a critical role in catabolism of waste products and recycling of macromolecules in the body. Disruption to the normal function of the lysosome can result in the toxic accumulation of storage products, often leading to irreparable cellular damage and organ dysfunction followed by premature death. The majority of LSDs have no curative treatment, with many clinical subtypes presenting in early infancy and childhood. Over two-thirds of LSDs present with progressive neurodegeneration, often in combination with other debilitating peripheral symptoms. Consequently, there is a pressing unmet clinical need to develop new therapeutic interventions to treat these conditions. The blood-brain barrier is a crucial hurdle that needs to be overcome in order to effectively treat the central nervous system (CNS), adding considerable complexity to therapeutic design and delivery. Enzyme replacement therapy (ERT) treatments aimed at either direct injection into the brain, or using blood-brain barrier constructs are discussed, alongside more conventional substrate reduction and other drug-related therapies. Other promising strategies developed in recent years, include gene therapy technologies specifically tailored for more effectively targeting treatment to the CNS. Here, we discuss the most recent advances in CNS-targeted treatments for neurological LSDs with a particular emphasis on gene therapy-based modalities, such as Adeno-Associated Virus and haematopoietic stem cell gene therapy approaches that encouragingly, at the time of writing are being evaluated in LSD clinical trials in increasing numbers. If safety, efficacy and improved quality of life can be demonstrated, these therapies have the potential to be the new standard of care treatments for LSD patients.


Assuntos
Doenças por Armazenamento dos Lisossomos , Qualidade de Vida , Humanos , Criança , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/terapia , Doenças por Armazenamento dos Lisossomos/metabolismo , Terapia Genética , Encéfalo/metabolismo , Lisossomos , Terapia de Reposição de Enzimas
2.
Neuropathol Appl Neurobiol ; 45(2): 119-140, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29679380

RESUMO

AIMS: Resident and peripherally derived glioma associated microglia/macrophages (GAMM) play a key role in driving tumour progression, angiogenesis, invasion and attenuating host immune responses. Differentiating these cells' origins is challenging and current preclinical models such as irradiation-based adoptive transfer, parabiosis and transgenic mice have limitations. We aimed to develop a novel nonmyeloablative transplantation (NMT) mouse model that permits high levels of peripheral chimerism without blood-brain barrier (BBB) damage or brain infiltration prior to tumour implantation. METHODS: NMT dosing was determined in C57BL/6J or Pep3/CD45.1 mice conditioned with concentrations of busulfan ranging from 25 mg/kg to 125 mg/kg. Donor haematopoietic cells labelled with eGFP or CD45.2 were injected via tail vein. Donor chimerism was measured in peripheral blood, bone marrow and spleen using flow cytometry. BBB integrity was assessed with anti-IgG and anti-fibrinogen antibodies. Immunocompetent chimerised animals were orthotopically implanted with murine glioma GL-261 cells. Central and peripheral cell contributions were assessed using immunohistochemistry and flow cytometry. GAMM subpopulation analysis of peripheral cells was performed using Ly6C/MHCII/MerTK/CD64. RESULTS: NMT achieves >80% haematopoietic chimerism by 12 weeks without BBB damage and normal life span. Bone marrow derived cells (BMDC) and peripheral macrophages accounted for approximately 45% of the GAMM population in GL-261 implanted tumours. Existing markers such as CD45 high/low proved inaccurate to determine central and peripheral populations while Ly6C/MHCII/MerTK/CD64 reliably differentiated GAMM subpopulations in chimerised and unchimerised mice. CONCLUSION: NMT is a powerful method for dissecting tumour microglia and macrophage subpopulations and can guide further investigation of BMDC subsets in glioma and neuro-inflammatory diseases.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Macrófagos/patologia , Microglia/patologia , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Pediatr Transplant ; 19(2): 211-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25546609

RESUMO

In vivo T-cell depletion, using alemtuzumab therapy prior to SCT, can reduce the incidence of GVHD. This treatment has a potential to delay immune reconstitution resulting in increased morbidity due to viral illnesses. We retrospectively analyzed data on all pediatric patients with non-malignant disorders who received alemtuzumab-based conditioning regimens in our center over the last 10 yr (n = 91). Our data show an OS of 91.2%. The incidence of acute (grade 2-4) GVHD was 18.7% and that of chronic GVHD 5.5%. Viremia due to adenovirus, EBV and CMV was seen in 19.8%, 64.8% and 39.6% patients, respectively, with only two deaths attributed to viral infection (adenovirus). Chimerism level at three month was predictive of graft outcome. Nine patients, who had graft failure after first SCT, were salvaged with a second SCT using RIC and same donor (if available). Based on these results, we conclude that the use of in vivo T-cell depletion is safe, achieves good chimerism and does not lead to increased morbidity and mortality due to viral infections. It is associated with a reduced incidence of chronic GVHD.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/métodos , Linfócitos T/imunologia , Adenoviridae/metabolismo , Adolescente , Alemtuzumab , Anemia Aplástica/terapia , Criança , Pré-Escolar , Feminino , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Imunossupressores/uso terapêutico , Incidência , Lactente , Masculino , Doenças Metabólicas/terapia , Estudos Retrospectivos , Quimeras de Transplante , Condicionamento Pré-Transplante , Transplante Homólogo , Resultado do Tratamento , Doadores não Relacionados , Viremia/fisiopatologia , Adulto Jovem
4.
Pediatr Transplant ; 15(5): 505-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21504523

RESUMO

Norovirus infection is a major cause of nonbacterial gastroenteritis. In immunocompetent individuals the illness caused by norovirus is mostly self limiting. Excretion of norovirus has been reported to be prolonged in the immunocompromised including adult HSCT recipients. We report a case series of 13 children who received HSCT and required prolonged parenteral and enteral nutrition due to severe gut dysfunction accompanying protracted norovirus excretion that was monitored by RT-PCR. The median duration of viral excretion was 150 days (range 60-380) and the eventual clearance of norovirus from feces was closely associated with donor T cell recovery in the peripheral blood. There was no disease manifestation beyond the gut but the severity and length of norovirus associated illness suggests that HSCT should be delayed where possible in patients excreting the virus prior to conditioning therapy.


Assuntos
Infecções por Caliciviridae/complicações , Infecções por Caliciviridae/diagnóstico , Transplante de Células-Tronco Hematopoéticas/métodos , Norovirus/genética , Criança , Pré-Escolar , Diarreia/patologia , Fezes , Feminino , Citometria de Fluxo/métodos , Gastroenterite/virologia , Humanos , Imunossupressores/uso terapêutico , Lactente , Masculino , Ciências da Nutrição , Apoio Nutricional , RNA Viral/metabolismo , Linfócitos T/citologia , Condicionamento Pré-Transplante/métodos
5.
Gene Ther ; 13(2): 117-26, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16163377

RESUMO

Immune responses against an introduced transgenic protein are a potential risk in many gene replacement strategies to treat genetic disease. We have developed a gene delivery approach for hemophilia B based on lentiviral expression of human factor IX in purified hematopoietic stem cells. In both normal C57Bl/6J and hemophilic 129/Sv recipient mice, we observed the production of therapeutic levels of human factor IX, persisting for at least a year with tolerance to human factor IX antigen. Secondary and tertiary recipients also demonstrate long-term production of therapeutic levels of human factor IX and tolerance, even at very low levels of donor chimerism. Furthermore, in hemophilic mice, partial functional correction of treated mice and phenotypic rescue is achieved. These data show the potential of a stem cell approach to gene delivery to tolerize recipients to a secreted foreign transgenic protein and, with appropriate modification, may be of use in developing treatments for other genetic disorders.


Assuntos
Fator IX/genética , Terapia Genética/métodos , HIV-1/genética , Hemofilia B/terapia , Transplante de Células-Tronco/métodos , Animais , Antígenos/imunologia , Células Cultivadas , Fator IX/metabolismo , Expressão Gênica , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/terapia , Genótipo , Proteínas de Fluorescência Verde/genética , Hemofilia B/sangue , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Células-Tronco/metabolismo , Células-Tronco/virologia , Fatores de Tempo , Transdução Genética/métodos , Transgenes
6.
Cell Prolif ; 37(1): 1-21, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14871234

RESUMO

The liver is normally proliferatively quiescent, but hepatocyte loss through partial hepatectomy, uncomplicated by virus infection or inflammation, invokes a rapid regenerative response from all cell types in the liver to perfectly restore liver mass. Moreover, hepatocyte transplants in animals have shown that a certain proportion of hepatocytes in foetal and adult liver can clonally expand, suggesting that hepatoblasts/hepatocytes are themselves the functional stem cells of the liver. More severe liver injury can activate a potential stem cell compartment located within the intrahepatic biliary tree, giving rise to cords of bipotential transit amplifying cells (oval cells), that can ultimately differentiate into hepatocytes and biliary epithelial cells. A third population of stem cells with hepatic potential resides in the bone marrow; these haematopoietic stem cells may contribute to the albeit low renewal rate of hepatocytes, but can make a more significant contribution to regeneration under a very strong positive selection pressure. In such instances, cell fusion rather than transdifferentiation appears to be the underlying mechanism by which the haematopoietic genome becomes reprogrammed.


Assuntos
Fígado/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Humanos
8.
BioDrugs ; 15(9): 615-34, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11580305

RESUMO

Since the discovery of the cystic fibrosis transmembrane conductance regulator (CFTR) gene nearly 12 years ago, cystic fibrosis (CF) has become one of the most intensively investigated monogenetic disorders considered approachable by gene therapy. This has resulted in over 20 clinical trials currently under way, concluded or awaiting approval. Despite the initial promise of gene therapy for CF, and the demonstration of successful gene transfer to the nose and airways of individuals, it has not so far been as effective as initially projected. Here we discuss the rationale behind CF gene therapy and dissect the vast array of literature representing the work that ultimately brought about the current phase I/II clinical trials. In the context of human trials, we review the limitations of current vector systems for CF gene therapy. We come to the conclusion that at present none of the application methods and vector systems are able to achieve the level and persistence of CFTR gene expression in the affected epithelia of CF patients that is required for therapeutic success. We also outline the challenges that must be overcome and describe some of the novel approaches to be taken in order to attain the curative therapy that was originally envisaged for this disease.


Assuntos
Adenoviridae , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/terapia , Terapia Genética , Vetores Genéticos/uso terapêutico , Retroviridae , Ensaios Clínicos como Assunto , Fibrose Cística/genética , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA