Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Am J Physiol Renal Physiol ; 326(6): F981-F987, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38545650

RESUMO

Cystinosis is an autosomal recessive lysosomal storage disorder, caused by mutations in the CTNS gene, resulting in an absent or altered cystinosin (CTNS) protein. Cystinosin exports cystine out of the lysosome, with a malfunction resulting in cystine accumulation and a defect in other cystinosin-mediated pathways. Cystinosis is a systemic disease, but the kidneys are the first and most severely affected organs. In the kidney, the disease initially manifests as a generalized dysfunction in the proximal tubules (also called renal Fanconi syndrome). MFSD12 is a lysosomal cysteine importer that directly affects the cystine levels in melanoma cells, HEK293T cells, and cystinosis patient-derived fibroblasts. In this study, we aimed to evaluate MFSD12 mRNA levels in cystinosis patient-derived proximal tubular epithelial cells (ciPTECs) and to study the effect of MFSD12 knockout on cystine levels. We showed similar MFSD12 mRNA expression in patient-derived ciPTECs in comparison with the control cells. CRISPR MFSD12 knockout in a patient-derived ciPTEC (CTNSΔ57kb) resulted in significantly reduced cystine levels. Furthermore, we evaluated proximal tubular reabsorption after injection of mfsd12a translation-blocking morpholino (TB MO) in a ctns-/- zebrafish model. This resulted in decreased cystine levels but caused a concentration-dependent increase in embryo dysmorphism. Furthermore, the mfsd12a TB MO injection did not improve proximal tubular reabsorption or megalin expression. In conclusion, MFSD12 mRNA depletion reduced cystine levels in both tested models without improvement of the proximal tubular function in the ctns-/- zebrafish embryo. In addition, the apparent toxicity of higher mfsd12a TB MO concentrations on the zebrafish development warrants further evaluation.NEW & NOTEWORTHY In this study, we show that MFSD12 depletion with either CRISPR/Cas9-mediated gene editing or a translation-blocking morpholino significantly reduced cystine levels in cystinosis ciPTECs and ctns-/- zebrafish embryos, respectively. However, we observed no improvement in the proximal tubular reabsorption of dextran in the ctns-/- zebrafish embryos injected with mfsd12a translation-blocking morpholino. Furthermore, a negative effect of the mfsd12a morpholino on the zebrafish development warrants further investigation.


Assuntos
Cistina , Cistinose , Modelos Animais de Doenças , Túbulos Renais Proximais , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Cistinose/metabolismo , Cistinose/genética , Cistinose/patologia , Humanos , Cistina/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Células Epiteliais/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas CRISPR-Cas
2.
J Clin Immunol ; 42(8): 1638-1652, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35829840

RESUMO

PURPOSE: Mendelian susceptibility to mycobacterial disease (MSMD) is caused by inborn errors of IFN-γ immunity. The most frequent genetic defects are found in IL12 or a subunit of its receptor. IL23R deficiency in MSMD has only been reported once, in two pediatric patients from the same kindred with isolated disseminated Bacille Calmette-Guérin disease. We evaluated the impact of a homozygous stop mutation in IL23R (R381X), identified by whole exome sequencing, in an adult patient with disseminated non-tuberculous mycobacterial disease. METHODS: We performed functional validation of the R381X mutation by evaluating IL23R expression and IL-23 signaling (STAT3 phosphorylation, IFN-γ production) in primary cells (PBMCs, EBV-B cells) and cell lines (HeLa) with or without back-complementation of wild-type IL23R. RESULTS: We report on a 48-year-old male with disseminated non-tuberculous mycobacterial disease. We identified and characterized a homozygous loss-of-function stop mutation underlying IL23R deficiency, resulting in near absent expression of membrane bound IL23R. IL23R deficiency was characterized by impaired IL-23-mediated IFN-γ secretion in CD4+, CD8+ T, and mucosal-associated invariant T (MAIT) cells, and low frequencies of circulating Th17 (CD3+CD45RA-CCR4+CXCR3-RORγT+), Th1* (CD45RA-CCR4-CXCR3+RORγT+), and MAIT (CD3+CD8+Vα7.2+CD161+) cells. Although the patient did not have a history of recurrent fungal infections, impaired Th17 differentiation and blunted IL-23-mediated IL-17 secretion in PBMCs were observed. CONCLUSION: We demonstrate that impaired IL-23 immunity caused by a homozygous R381X mutation in IL23R underlies MSMD, corroborating earlier findings with a homozygous p.C115Y IL23R mutation. Our report further supports a model of redundant contribution of IL-23- to IL-17-mediated anti-fungal immunity.1.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Infecções por Mycobacterium , Masculino , Adulto , Humanos , Criança , Pessoa de Meia-Idade , Interleucina-17/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Infecções por Mycobacterium/etiologia , Infecções por Mycobacterium não Tuberculosas/genética , Infecções por Mycobacterium não Tuberculosas/complicações , Mutação/genética , Interleucina-23 , Predisposição Genética para Doença , Receptores de Interleucina/genética
4.
Mol Ther ; 29(12): 3383-3397, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34174440

RESUMO

Hematopoietic stem cell gene therapy is emerging as a promising therapeutic strategy for many diseases of the blood and immune system. However, several individuals who underwent gene therapy in different trials developed hematological malignancies caused by insertional mutagenesis. Preclinical assessment of vector safety remains challenging because there are few reliable assays to screen for potential insertional mutagenesis effects in vitro. Here we demonstrate that genotoxic vectors induce a unique gene expression signature linked to stemness and oncogenesis in transduced murine hematopoietic stem and progenitor cells. Based on this finding, we developed the surrogate assay for genotoxicity assessment (SAGA). SAGA classifies integrating retroviral vectors using machine learning to detect this gene expression signature during the course of in vitro immortalization. On a set of benchmark vectors with known genotoxic potential, SAGA achieved an accuracy of 90.9%. SAGA is more robust and sensitive and faster than previous assays and reliably predicts a mutagenic risk for vectors that led to leukemic severe adverse events in clinical trials. Our work provides a fast and robust tool for preclinical risk assessment of gene therapy vectors, potentially paving the way for safer gene therapy trials.


Assuntos
Terapia Genética , Vetores Genéticos , Animais , Dano ao DNA , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Células-Tronco Hematopoéticas , Humanos , Aprendizado de Máquina , Camundongos , Mutagênese Insercional
5.
Retrovirology ; 17(1): 23, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727480

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

7.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255702

RESUMO

(1) Background: Neurogenesis is considered to be a potential brain repair mechanism and is enhanced in stroke. It is difficult to reconstruct the neurogenesis process only from the histological sections taken from different animals at different stages of brain damage and restoration. Study of neurogenesis would greatly benefit from development of tissue-specific visualization probes. (2) Purpose: The study aimed to explore if overexpression of ferritin, a nontoxic iron-binding protein, under a doublecortin promoter can be used for non-invasive visualization of neurogenesis using magnetic resonance imaging (MRI). (3) Methods: Ferritin heavy chain (FerrH) was expressed in the adeno-associated viral backbone (AAV) under the doublecortin promoter (pDCX), specific for young neurons, in the viral construct AAV-pDCX-FerrH. Expression of the enhanced green fluorescent protein (eGFP) was used as an expression control (AAV-pDCX-eGFP). The viral vectors or phosphate-buffered saline (PBS) were injected intracerebrally into 18 adult male Sprague-Dawley rats. Three days before injection, rats underwent transient middle-cerebral-artery occlusion or sham operation. Animals were subjected to In vivo MRI study before surgery and on days 7, 14, 21, and 28 days after injection using a Bruker BioSpec 11.7 T scanner. Brain sections obtained on day 28 after injection were immunostained for ferritin, young (DCX) and mature (NeuN) neurons, and activated microglia/macrophages (CD68). Additionally, RT-PCR was performed to confirm ferritin expression. (4) Results: T2* images in post-ischemic brains of animals injected with AAV-pDCX-FerrH showed two distinct zones of MRI signal hypointensity in the ipsilesioned hemisphere starting from 14 days after viral injection-in the ischemic lesion and near the lateral ventricle and subventricular zone (SVZ). In sham-operated animals, only one zone of hypointensity near the lateral ventricle and SVZ was revealed. Immunochemistry showed that ferritin-expressing cells in ischemic lesions were macrophages (88.1%), while ferritin-expressing cells near the lateral ventricle in animals both after ischemia and sham operation were mostly mature (55.7% and 61.8%, respectively) and young (30.6% and 7.1%, respectively) neurons. RT-PCR confirmed upregulated expression of ferritin in the caudoputamen and corpus callosum. Surprisingly, in animals injected with AAV-pDCX-eGFP we similarly observed two zones of hypointensity on T2* images. Cellular studies also showed the presence of mature (81.5%) and young neurons (6.1%) near the lateral ventricle in both postischemic and sham-operated animals, while macrophages in ischemic lesions were ferritin-positive (98.2%). (5) Conclusion: Ferritin overexpression induced by injection of AAV-pDCX-FerrH was detected by MRI using T2*-weighted images, which was confirmed by immunochemistry showing ferritin in young and mature neurons. Expression of eGFP also caused a comparable reduced MR signal intensity in T2*-weighted images. Additional studies are needed to investigate the potential and tissue-specific features of the use of eGFP and ferritin expression in MRI studies.


Assuntos
Ferritinas/genética , Neurogênese/genética , Neurônios/metabolismo , Acidente Vascular Cerebral/genética , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Corpo Caloso/diagnóstico por imagem , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Modelos Animais de Doenças , Proteína Duplacortina , Vetores Genéticos/farmacologia , Humanos , Infarto da Artéria Cerebral Média , Ventrículos Laterais/diagnóstico por imagem , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Imageamento por Ressonância Magnética , Masculino , Microglia/metabolismo , Microglia/patologia , Proteínas Associadas aos Microtúbulos/genética , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia
8.
Trends Biochem Sci ; 40(2): 108-16, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555456

RESUMO

To achieve productive infection, retroviruses such as HIV stably integrate their reverse transcribed RNA genome into a host chromosome. Each retroviral family preferentially integrates near a unique subset of genomic features. HIV integrase (IN) is targeted to the body of active transcription units through interaction with lens epithelium-derived growth factor (LEDGF/p75). We describe the successful effort to develop inhibitors of the interaction between IN and LEDGF/p75, referred to as LEDGINs. Gammaretroviruses display a distinct integration pattern. Recently, BET (bromo- and extraterminal domain) proteins were identified as the LEDGF/p75 counterparts that target the integration of gammaretroviruses. The identification of the chromatin-readers LEDGF/p75 and BET as cellular cofactors that orchestrate lentiviral or gammaretroviral integration opens new avenues to developing safer viral vectors for gene therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por HIV/tratamento farmacológico , Inibidores de Integrase de HIV/administração & dosagem , Fatores de Transcrição/metabolismo , Integração Viral/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Cromatina/efeitos dos fármacos , Gammaretrovirus/efeitos dos fármacos , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Infecções por HIV/virologia , Integrase de HIV/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Lentivirus/efeitos dos fármacos , Lentivirus/genética , Lentivirus/patogenicidade , Fatores de Transcrição/genética , Integração Viral/efeitos dos fármacos
9.
Retrovirology ; 16(1): 8, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30940165

RESUMO

BACKGROUND: Persistence of latent, replication-competent provirus is the main impediment towards the cure of HIV infection. One of the critical questions concerning HIV latency is the role of integration site selection in HIV expression. Inhibition of the interaction between HIV integrase and its chromatin tethering cofactor LEDGF/p75 is known to reduce integration and to retarget residual provirus to regions resistant to reactivation. LEDGINs, small molecule inhibitors of the interaction between HIV integrase and LEDGF/p75, provide an interesting tool to study the underlying mechanisms. During early infection, LEDGINs block the interaction with LEDGF/p75 and allosterically inhibit the catalytic activity of IN (i.e. the early effect). When present during virus production, LEDGINs interfere with proper maturation due to enhanced IN oligomerization in the progeny virions (i.e. the late effect). RESULTS: We studied the effect of LEDGINs present during virus production on the transcriptional state of the residual virus. Infection of cells with viruses produced in the presence of LEDGINs resulted in a residual reservoir that was refractory to activation. Integration of residual provirus was less favored near epigenetic markers associated with active transcription. However, integration near H3K36me3 and active genes, both targeted by LEDGF/p75, was not affected. Also in primary cells, LEDGIN treatment induced a reservoir resistant to activation due to a combined early and late effect. CONCLUSION: LEDGINs present a research tool to study the link between integration and transcription, an essential question in retrovirology. LEDGIN treatment during virus production altered integration of residual provirus in a LEDGF/p75-independent manner, resulting in a reservoir that is refractory to activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , HIV-1/fisiologia , Fatores de Transcrição/genética , Integração Viral , Latência Viral , Replicação Viral , Linhagem Celular , Células Cultivadas , Integrase de HIV/genética , HIV-1/genética , Humanos , Leucócitos Mononucleares/virologia , Ligação Proteica , Provírus/fisiologia , Ativação Viral
10.
Int J Mol Sci ; 20(5)2019 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-30841538

RESUMO

Progressive muscle degeneration followed by dilated cardiomyopathy is a hallmark of muscular dystrophy. Stem cell therapy is suggested to replace diseased myofibers by healthy myofibers, although so far, we are faced by low efficiencies of migration and engraftment of stem cells. Chemokines are signalling proteins guiding cell migration and have been shown to tightly regulate muscle tissue repair. We sought to determine which chemokines are expressed in dystrophic muscles undergoing tissue remodelling. Therefore, we analysed the expression of chemokines and chemokine receptors in skeletal and cardiac muscles from Sarcoglycan-α null, Sarcoglycan-ß null and immunodeficient Sgcß-null mice. We found that several chemokines are dysregulated in dystrophic muscles. We further show that one of these, platelet-derived growth factor-B, promotes interstitial stem cell migration. This finding provides perspective to an approachable mechanism for improving stem cell homing towards dystrophic muscles.


Assuntos
Movimento Celular , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mioblastos/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Animais , Células Cultivadas , Quimiocinas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Sarcoglicanas/genética , Sarcoglicanas/metabolismo
11.
Circulation ; 136(8): 747-761, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28611091

RESUMO

BACKGROUND: Cardiovascular diseases remain the predominant cause of death worldwide, with the prevalence of heart failure continuing to increase. Despite increased knowledge of the metabolic alterations that occur in heart failure, novel therapies to treat the observed metabolic disturbances are still lacking. METHODS: Mice were subjected to pressure overload by means of angiotensin-II infusion or transversal aortic constriction. MicroRNA-146a was either genetically or pharmacologically knocked out or genetically overexpressed in cardiomyocytes. Furthermore, overexpression of dihydrolipoyl succinyltransferase (DLST) in the murine heart was performed by means of an adeno-associated virus. RESULTS: MicroRNA-146a was upregulated in whole heart tissue in multiple murine pressure overload models. Also, microRNA-146a levels were moderately increased in left ventricular biopsies of patients with aortic stenosis. Overexpression of microRNA-146a in cardiomyocytes provoked cardiac hypertrophy and left ventricular dysfunction in vivo, whereas genetic knockdown or pharmacological blockade of microRNA-146a blunted the hypertrophic response and attenuated cardiac dysfunction in vivo. Mechanistically, microRNA-146a reduced its target DLST-the E2 subcomponent of the α-ketoglutarate dehydrogenase complex, a rate-controlling tricarboxylic acid cycle enzyme. DLST protein levels significantly decreased on pressure overload in wild-type mice, paralleling a decreased oxidative metabolism, whereas DLST protein levels and hence oxidative metabolism were partially maintained in microRNA-146a knockout mice. Moreover, overexpression of DLST in wild-type mice protected against cardiac hypertrophy and dysfunction in vivo. CONCLUSIONS: Altogether we show that the microRNA-146a and its target DLST are important metabolic players in left ventricular dysfunction.


Assuntos
Aciltransferases/biossíntese , Cardiomegalia/metabolismo , Regulação Enzimológica da Expressão Gênica , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , Disfunção Ventricular Esquerda/metabolismo , Aciltransferases/genética , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Cardiomegalia/prevenção & controle , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Ratos , Ratos Endogâmicos Lew , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/prevenção & controle
12.
J Clin Immunol ; 37(8): 801-810, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28993958

RESUMO

Hypomorphic IKBKG mutations in males are typically associated with anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). Some mutations cause immunodeficiency without EDA (NEMO-ID). The immunological profile associated with these NEMO-ID variants is not fully documented. We present a 2-year-old patient with suspected immunodeficiency in which a hemizygous p.Glu57Lys IKBKG variant was identified. At the age of 1 year, he had an episode of otitis media that evolved into a bilateral mastoiditis (Pseudomonas spp). Hypogammaglobulinemia, specific (polysaccharide) antibody deficiency, and low switched memory B cell subsets were noticed. The mother was heterozygous for the variant but had no signs of incontinentia pigmenti. Patient peripheral blood mononuclear cells produced low amounts of IL-6 after stimulation with IL-1ß, Pam3CSK4, and FSL-1. In patient fibroblasts, IκB-α was degraded normally upon stimulation with IL-1ß or TNF-α. Transduction of wild-type and variant NEMO in NEMO-/- deficient SV40 fibroblasts revealed a slight but significant reduction of IL-6 production upon stimulation with IL-1ß and TNF-α. In conclusion, we demonstrated that p.Glu57Lys leads to specific immunological defects in vitro. No other pathogenic PID variants were identified through whole exome sequencing. As rare polymorphisms have been described in IKBKG and polygenic inheritance remains an option in the presented case, this study emphasizes the need for thorough functional and genetic evaluation when encountering and interpreting suspected disease-causing NEMO-ID variants.


Assuntos
Imunodeficiência de Variável Comum/genética , Displasia Ectodérmica/genética , Fibroblastos/fisiologia , Quinase I-kappa B/genética , Mutação/genética , Infecções por Pseudomonas/diagnóstico , Pseudomonas/fisiologia , Agamaglobulinemia , Células Cultivadas , Pré-Escolar , Imunodeficiência de Variável Comum/diagnóstico , Displasia Ectodérmica/diagnóstico , Hemizigoto , Humanos , Quinase I-kappa B/metabolismo , Interleucina-6/metabolismo , Masculino , Mastoidite , Otite , Polimorfismo Genético
13.
J Virol ; 90(10): 5205-5209, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962222

RESUMO

Recent advances in fluorescence microscopy allow three-dimensional analysis of HIV-1 preintegration complexes in the nuclei of infected cells. To extend this investigation to gammaretroviruses, we engineered a fluorescent Moloney murine leukemia virus (MLV) system consisting of MLV-integrase fused to enhanced green fluorescent protein (MLV-IN-EGFP). A comparative analysis of lentiviral (HIV-1) and gammaretroviral (MLV) fluorescent complexes in the nuclei of infected cells revealed their different spatial distributions. This research tool has the potential to achieve new insight into the nuclear biology of these retroviruses.


Assuntos
Núcleo Celular/ultraestrutura , Núcleo Celular/virologia , HIV-1/fisiologia , Vírus da Leucemia Murina de Moloney/fisiologia , Animais , Proteínas de Fluorescência Verde/genética , HIV-1/genética , HIV-1/ultraestrutura , Células HeLa , Humanos , Integrases/genética , Camundongos , Microscopia de Fluorescência , Vírus da Leucemia Murina de Moloney/ultraestrutura , Integração Viral
15.
Bioessays ; 37(11): 1202-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26293289

RESUMO

Here, we review genomic target site selection during retroviral integration as a multistep process in which specific biases are introduced at each level. The first asymmetries are introduced when the virus takes a specific route into the nucleus. Next, by co-opting distinct host cofactors, the integration machinery is guided to particular chromatin contexts. As the viral integrase captures a local target nucleosome, specific contacts introduce fine-grained biases in the integration site distribution. In vivo, the established population of proviruses is subject to both positive and negative selection, thereby continuously reshaping the integration site distribution. By affecting stochastic proviral expression as well as the mutagenic potential of the virus, integration site choice may be an inherent part of the evolutionary strategies used by different retroviruses to maximise reproductive success.


Assuntos
HIV-1/genética , Vírus da Leucemia Murina/genética , Integração Viral/genética , Latência Viral/genética , Animais , Capsídeo/metabolismo , DNA Viral/genética , Infecções por HIV/patologia , Humanos , Integrases/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia
16.
Am J Respir Crit Care Med ; 193(3): 288-98, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26509335

RESUMO

RATIONALE: Gene therapy holds promise for a curative mutation-independent treatment applicable to all patients with cystic fibrosis (CF). The various viral vector-based clinical trials conducted in the past have demonstrated safety and tolerance of different vectors, but none have led to a clear and persistent clinical benefit. Recent clinical breakthroughs in recombinant adeno-associated viral vector (rAAV)-based gene therapy encouraged us to reexplore an rAAV approach for CF. OBJECTIVES: We evaluated the preclinical potential of rAAV gene therapy for CF to restore chloride and fluid secretion in two complementary models: intestinal organoids derived from subjects with CF and a CF mouse model, an important milestone toward the development of a clinical rAAV candidate for CF gene therapy. METHODS: We engineered an rAAV vector containing a truncated CF transmembrane conductance regulator (CFTRΔR) combined with a short promoter (CMV173) to ensure optimal gene expression. A rescue in chloride and fluid secretion after rAAV-CFTRΔR treatment was assessed by forskolin-induced swelling in CF transmembrane conductance regulator (CFTR)-deficient organoids and by nasal potential differences in ΔF508 mice. MEASUREMENTS AND MAIN RESULTS: rAAV-CFTRΔR transduction of human CFTR-deficient organoids resulted in forskolin-induced swelling, indicating a restoration of CFTR function. Nasal potential differences demonstrated a clear response to low chloride and forskolin perfusion in most rAAV-CFTRΔR-treated CF mice. CONCLUSIONS: Our study provides robust evidence that rAAV-mediated gene transfer of a truncated CFTR functionally rescues the CF phenotype across the nasal mucosa of CF mice and in patient-derived organoids. These results underscore the clinical potential of rAAV-CFTRΔR in offering a cure for all patients with CF in the future.


Assuntos
Fibrose Cística/terapia , Dependovirus , Terapia Genética/métodos , Vetores Genéticos , Intestinos , Organoides , Animais , Líquidos Corporais/metabolismo , Canais de Cloreto/genética , Cloretos/metabolismo , Colforsina/farmacologia , Fibrose Cística/genética , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Genótipo , Células HeLa , Humanos , Camundongos , Organoides/metabolismo , Transdução Genética
17.
Proc Natl Acad Sci U S A ; 110(14): 5636-41, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23513220

RESUMO

Recent advances in fluorescence microscopy provided tools for the investigation and the analysis of the viral replication steps in the cellular context. In the HIV field, the current visualization systems successfully achieve the fluorescent labeling of the viral envelope and proteins, but not the genome. Here, we developed a system able to visualize the proviral DNA of HIV-1 through immunofluorescence detection of repair foci for DNA double-strand breaks specifically induced in the viral genome by the heterologous expression of the I-SceI endonuclease. The system for Single-Cell Imaging of HIV-1 Provirus, named SCIP, provides the possibility to individually track integrated-viral DNA within the nuclei of infected cells. In particular, SCIP allowed us to perform a topological analysis of integrated viral DNA revealing that HIV-1 preferentially integrates in the chromatin localized at the periphery of the nuclei.


Assuntos
HIV-1/ultraestrutura , Microscopia de Fluorescência/métodos , Provírus/ultraestrutura , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Quebras de DNA de Cadeia Dupla , Primers do DNA/genética , Reparo do DNA/genética , Desoxirribonucleases de Sítio Específico do Tipo II , Humanos , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Proteínas de Saccharomyces cerevisiae , Análise de Célula Única/métodos
18.
J Neurosci ; 34(49): 16518-32, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25471588

RESUMO

Alpha-synuclein (αSYN) aggregation plays a pivotal role in the pathogenesis of Parkinson's disease and other synucleinopathies. In this multistep process, oligomerization of αSYN monomers is the first step in the formation of fibrils and intracytoplasmic inclusions. Although αSYN oligomers are generally considered to be the culprit of these diseases, the methodology currently available to follow-up oligomerization in cells and in brain is inadequate. We developed a split firefly luciferase complementation system to visualize oligomerization of viral vector-encoded αSYN fusion proteins. αSYN oligomerization resulted in successful luciferase complementation in cell culture and in mouse brain. Oligomerization of αSYN was monitored noninvasively with bioluminescence imaging in the mouse striatum and substantia nigra up to 8 months after injection. Moreover, the visualized αSYN oligomers retained their toxic and aggregation properties in both model systems. Next, the effect of two small molecules, FK506 and (-)-epigallocatechin-3-gallate (EGCG), known to inhibit αSYN fibril formation, was investigated. FK506 inhibited the observed αSYN oligomerization both in cell culture and in mouse brain. In conclusion, the split firefly luciferase-αSYN complementation assay will increase our insight in the role of αSYN oligomers in synucleinopathies and opens new opportunities to evaluate potential αSYN-based neuroprotective therapies.


Assuntos
Corpo Estriado/metabolismo , Luciferases de Vaga-Lume/genética , Medições Luminescentes/métodos , Neuroimagem/métodos , Agregados Proteicos/efeitos dos fármacos , Substância Negra/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animais , Catequina/análogos & derivados , Catequina/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Corpo Estriado/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Humanos , Luciferases de Vaga-Lume/metabolismo , Camundongos , Fármacos Neuroprotetores/farmacologia , Substância Negra/efeitos dos fármacos , Tacrolimo/farmacologia , alfa-Sinucleína/genética
19.
J Biol Chem ; 289(36): 25351-61, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25063804

RESUMO

Transportin-SR2 (Tnpo3, TRN-SR2), a human karyopherin encoded by the TNPO3 gene, has been identified as a cellular cofactor of HIV-1 replication, specifically interacting with HIV-1 integrase (IN). Whether this interaction mediates the nuclear import of HIV remains controversial. We previously characterized the TRN-SR2 binding interface in IN and introduced mutations at these positions to corroborate the biological relevance of the interaction. The pleiotropic nature of IN mutations complicated the interpretation. Indeed, all previously tested IN interaction mutants also affected RT. Here we report on a virus with a pair of IN mutations, IN(R263A/K264A), that significantly reduce interaction with TRN-SR2. The virus retains wild-type reverse transcription activity but displays a block in nuclear import and integration, as measured by quantitative PCR. The defect in integration of this mutant resulted in a smaller increase in the number of two-long terminal repeat circles than for virus specifically blocked at integration by raltegravir or catalytic site mutations (IN(D64N/D116N/E152Q)). Finally, using an eGFP-IN-labeled HIV fluorescence-based import assay, the defect in nuclear import was corroborated. These data altogether underscore the importance of the HIV-IN TRN-SR2 protein-protein interaction for HIV nuclear import and validate the IN/TRN-SR2 interaction interface as a promising target for future antiviral therapy.


Assuntos
Integrase de HIV/genética , HIV-1/genética , Mutação , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Sítios de Ligação/genética , Ligação Competitiva , Western Blotting , Núcleo Celular/virologia , Integrase de HIV/química , Integrase de HIV/metabolismo , HIV-1/enzimologia , HIV-1/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Replicação Viral/genética , beta Carioferinas/química
20.
Retrovirology ; 12: 16, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25809198

RESUMO

BACKGROUND: The dynamic interaction between HIV and its host governs the replication of the virus and the study of the virus-host interplay is key to understand the viral lifecycle. The host factor lens epithelium-derived growth factor (LEDGF/p75) tethers the HIV preintegration complex to the chromatin through a direct interaction with integrase (IN). Small molecules that bind the LEDGF/p75 binding pocket of the HIV IN dimer (LEDGINs) block HIV replication through a multimodal mechanism impacting early and late stage replication including HIV maturation. Furthermore, LEDGF/p75 has been identified as a Pol interaction partner. This raised the question whether LEDGF/p75 besides acting as a molecular tether in the target cell, also affects late steps of HIV replication. RESULTS: LEDGF/p75 is recruited into HIV-1 particles through direct interaction with the viral IN (or Pol polyprotein) and is a substrate for HIV-1 protease. Incubation in the presence of HIV-1 protease inhibitors resulted in detection of full-length LEDGF/p75 in purified viral particles. We also demonstrate that inhibition of LEDGF/p75-IN interaction by specific mutants or LEDGINs precludes incorporation of LEDGF/p75 in virions, underscoring the specificity of the uptake. LEDGF/p75 depletion did however not result in altered LEDGIN potency. CONCLUSION: Together, these results provide evidence for an IN/Pol mediated uptake of LEDGF/p75 in viral particles and a specific cleavage by HIV protease. Understanding of the possible role of LEDGF/p75 or its cleavage fragments in the viral particle awaits further experimentation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Fatores de Transcrição/metabolismo , Integração Viral , Replicação Viral , Produtos do Gene pol do Vírus da Imunodeficiência Humana/metabolismo , Protease de HIV/metabolismo , Humanos , Proteólise
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa