Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Am J Hum Genet ; 109(10): 1789-1813, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36152629

RESUMO

Chromosome 16p11.2 reciprocal genomic disorder, resulting from recurrent copy-number variants (CNVs), involves intellectual disability, autism spectrum disorder (ASD), and schizophrenia, but the responsible mechanisms are not known. To systemically dissect molecular effects, we performed transcriptome profiling of 350 libraries from six tissues (cortex, cerebellum, striatum, liver, brown fat, and white fat) in mouse models harboring CNVs of the syntenic 7qF3 region, as well as cellular, transcriptional, and single-cell analyses in 54 isogenic neural stem cell, induced neuron, and cerebral organoid models of CRISPR-engineered 16p11.2 CNVs. Transcriptome-wide differentially expressed genes were largely tissue-, cell-type-, and dosage-specific, although more effects were shared between deletion and duplication and across tissue than expected by chance. The broadest effects were observed in the cerebellum (2,163 differentially expressed genes), and the greatest enrichments were associated with synaptic pathways in mouse cerebellum and human induced neurons. Pathway and co-expression analyses identified energy and RNA metabolism as shared processes and enrichment for ASD-associated, loss-of-function constraint, and fragile X messenger ribonucleoprotein target gene sets. Intriguingly, reciprocal 16p11.2 dosage changes resulted in consistent decrements in neurite and electrophysiological features, and single-cell profiling of organoids showed reciprocal alterations to the proportions of excitatory and inhibitory GABAergic neurons. Changes both in neuronal ratios and in gene expression in our organoid analyses point most directly to calretinin GABAergic inhibitory neurons and the excitatory/inhibitory balance as targets of disruption that might contribute to changes in neurodevelopmental and cognitive function in 16p11.2 carriers. Collectively, our data indicate the genomic disorder involves disruption of multiple contributing biological processes and that this disruption has relative impacts that are context specific.


Assuntos
Transtorno do Espectro Autista , Transtornos Cromossômicos , Deficiência Intelectual , Animais , Transtorno do Espectro Autista/genética , Calbindina 2/genética , Córtex Cerebral , Deleção Cromossômica , Transtornos Cromossômicos/genética , Cromossomos Humanos Par 16/genética , Variações do Número de Cópias de DNA , Genômica , Humanos , Deficiência Intelectual/genética , Camundongos , Neurônios , RNA
2.
Cell ; 135(3): 401-6, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18984149

RESUMO

Autism is a complex genetic disorder, but single-gene disorders with a high prevalence of autism offer insight into its pathogenesis. Recent evidence suggests that some molecular defects in autism may interfere with the mechanisms of synaptic protein synthesis. We propose that aberrant synaptic protein synthesis may represent one possible pathway leading to autistic phenotypes, including cognitive impairment and savant abilities.


Assuntos
Transtorno Autístico/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Biossíntese de Proteínas , Animais , Cognição , Humanos , Camundongos , Plasticidade Neuronal , Sinapses/metabolismo
4.
Proc Natl Acad Sci U S A ; 115(32): 8185-8190, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038001

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by a mutation in the X-linked methyl-CpG-binding protein 2 (MECP2). There is currently no disease-specific treatment, but MECP2 restoration through reactivation of the inactive X (Xi) has been of considerable interest. Progress toward an Xi-reactivation therapy has been hampered by a lack of suitable female mouse models. Because of cellular mosaicism due to random X-chromosome inactivation (XCI), Mecp2+/- heterozygous females develop only mild RTT. Here, we create an improved female mouse model by introducing a mutation in Tsix, the antisense regulator of XCI allelic choice. Tsix-Mecp2 mice show reduced MECP2 mosaicism and closely phenocopy the severely affected Mecp2-null males. Tsix-Mecp2 females demonstrate shortened lifespan, motor weakness, tremors, and gait disturbance. Intriguingly, they also exhibit repetitive behaviors, as is often seen in human RTT, including excessive grooming and biting that result in self-injury. With a Tsix allelic series, we vary MECP2 levels in brain and demonstrate a direct, but nonlinear correlation between MECP2 levels and phenotypic improvement. As little as 5-10% MECP2 restoration improves neuromotor function and extends lifespan five- to eightfold. Our study thus guides future pharmacological strategies and suggests that partial MECP2 restoration could have disproportionate therapeutic benefit.


Assuntos
Modelos Animais de Doenças , Genes Ligados ao Cromossomo X/genética , Longevidade/genética , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , RNA Longo não Codificante/genética , Síndrome de Rett/genética , Alelos , Animais , Comportamento Animal , Encéfalo/patologia , Feminino , Heterozigoto , Humanos , Masculino , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mosaicismo , Atividade Motora/genética , Mutação , Fenótipo , RNA Longo não Codificante/metabolismo , Síndrome de Rett/mortalidade , Síndrome de Rett/patologia , Inativação do Cromossomo X
5.
Proc Natl Acad Sci U S A ; 115(4): E668-E675, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29282321

RESUMO

The X-chromosome harbors hundreds of disease genes whose associated diseases predominantly affect males. However, a subset, including neurodevelopmental disorders, Rett syndrome (RTT), fragile X syndrome, and CDKL5 syndrome, also affects females. These disorders lack disease-specific treatment. Because female cells carry two X chromosomes, an emerging treatment strategy has been to reawaken the healthy allele on the inactive X (Xi). Here, we focus on methyl-CpG binding protein 2 (MECP2) restoration for RTT and combinatorially target factors in the interactome of Xist, the noncoding RNA responsible for X inactivation. We identify a mixed modality approach combining an Xist antisense oligonucleotide and a small-molecule inhibitor of DNA methylation, which, together, achieve 30,000-fold MECP2 up-regulation from the Xi in cultured cells. Combining a brain-specific genetic Xist ablation with short-term 5-aza-2'-deoxycytidine (Aza) treatment models the synergy in vivo without evident toxicity. The Xi is selectively reactivated. These experiments provide proof of concept for a mixed modality approach for treating X-linked disorders in females.


Assuntos
Azacitidina/análogos & derivados , Terapia Genética/métodos , Proteína 2 de Ligação a Metil-CpG/genética , Oligonucleotídeos Antissenso/uso terapêutico , Síndrome de Rett/terapia , Animais , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Encéfalo/metabolismo , Linhagem Celular , Metilação de DNA/efeitos dos fármacos , Decitabina , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Síndrome de Rett/genética , Inativação do Cromossomo X
6.
Immunol Invest ; 49(7): 726-743, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32299258

RESUMO

Background: T cells present in chronic inflammatory tissues such as nasal polyps (from chronic rhinosinusitis patients) have been demonstrated to be hypo-responsive to activation via the TCR, similar to tumor-specific T cells in multiple different human tumor microenvironments. While immunosuppressive exosomes have been known to contribute to the failure of the tumor-associated T cells to respond optimally to activation stimuli, it is not known whether they play a similar role in chronic inflammatory microenvironments. In the current study, we investigate whether exosomes derived from chronic inflammatory microenvironments contribute to the immune suppression of T cells. Methods: Exosomes were isolated by ultracentrifugation and characterized by size and composition using nanoparticle tracking analysis, scanning electron microscopy, antibody arrays and flow exometry. Immunosuppressive ability of the exosomes was measured by quantifying its effect on activation of T cells, using nuclear translocation of NFκB as an activation endpoint. Results: Exosomes were isolated and characterized from two different types of chronic inflammatory tissues - nasal polyps from chronic rhinosinusitis patients and synovial fluid from rheumatoid arthritis patients. These exosomes arrest the activation of T cells stimulated via the TCR. This immune suppression, like that which is seen in tumor microenvironments, is dependent in part upon a lipid, ganglioside GD3, which is expressed on the exosomal surface. Conclusion: Immunosuppressive exosomes present in non-malignant chronic inflammatory tissues represent a new T cell checkpoint, and potentially represent a novel therapeutic target to enhance the response to current therapies and prevent disease recurrences.


Assuntos
Microambiente Celular/imunologia , Exossomos/metabolismo , Imunomodulação , Inflamação/etiologia , Inflamação/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Artrite/etiologia , Artrite/metabolismo , Biomarcadores , Doença Crônica , Suscetibilidade a Doenças , Exossomos/ultraestrutura , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestrutura , Humanos , Imuno-Histoquímica , Imunofenotipagem , Inflamação/patologia , Metabolismo dos Lipídeos , Ativação Linfocitária/imunologia , NF-kappa B/metabolismo , Pólipos Nasais/etiologia , Pólipos Nasais/metabolismo , Pólipos Nasais/patologia , Transporte Proteico , Transdução de Sinais , Líquido Sinovial/metabolismo
7.
J Immunol ; 201(12): 3750-3758, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30446565

RESUMO

The tumor microenvironment is rendered immunosuppressive by a variety of cellular and acellular factors that represent potential cancer therapeutic targets. Although exosomes isolated from ovarian tumor ascites fluids have been previously reported to induce a rapid and reversible T cell arrest, the factors present on or within exosomes that contribute to immunosuppression have not been fully defined. In this study, we establish that GD3, a ganglioside expressed on the surface of exosomes isolated from human ovarian tumor ascites fluids, is causally linked to the functional arrest of T cells activated through their TCR. This arrest is inhibited by Ab blockade of exosomal GD3 or by the removal of GD3+ exosomes. Empty liposomes expressing GD3 on the surface also inhibit the activation of T cells, establishing that GD3 contributes to the functional arrest of T cells independent of factors present in exosomes. Finally, we demonstrate that the GD3-mediated arrest of the TCR activation is dependent upon sialic acid groups, because their enzymatic removal from exosomes or liposomes results in a loss of inhibitory capacity. Collectively, these data define GD3 as a potential immunotherapeutic target.


Assuntos
Líquido Ascítico/metabolismo , Exossomos/metabolismo , Gangliosídeos/metabolismo , Imunoterapia/métodos , Ácido N-Acetilneuramínico/metabolismo , Neoplasias Ovarianas/metabolismo , Linfócitos T/imunologia , Ascite , Células Cultivadas , Feminino , Humanos , Tolerância Imunológica , NF-kappa B/metabolismo , Estadiamento de Neoplasias , Neoplasias Ovarianas/imunologia , Microambiente Tumoral
9.
J Neurosci ; 33(28): 11606-17, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843529

RESUMO

Mutations in the PSEN1 gene encoding Presenilin-1 (PS1) are the predominant cause of familial Alzheimer's disease (FAD), but the underlying mechanisms remain unresolved. To reconcile the dominant action of pathogenic PSEN1 mutations with evidence that they confer a loss of mutant protein function, we tested the hypothesis that PSEN1 mutations interfere with γ-secretase activity in a dominant-negative manner. Here, we show that pathogenic PSEN1 mutations act in cis to impair mutant PS1 function and act in trans to inhibit wild-type PS1 function. Coexpression of mutant and wild-type PS1 at equal gene dosage in presenilin-deficient mouse embryo fibroblasts resulted in trans-dominant-negative inhibition of wild-type PS1 activity, suppressing γ-secretase-dependent cleavage of APP and Notch. Surprisingly, mutant PS1 could stimulate production of Aß42 by wild-type PS1 while decreasing its production of Aß40. Mutant and wild-type PS1 efficiently coimmunoprecipitated, suggesting that mutant PS1 interferes with wild-type PS1 activity via physical interaction. These results support the conclusion that mutant PS1 causes wild-type PS1 to adopt an altered conformation with impaired catalytic activity and substrate specificity. Our findings reveal a novel mechanism of action for pathogenic PSEN1 mutations and suggest that dominant-negative inhibition of presenilin activity plays an important role in FAD pathogenesis.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Mutação/genética , Fragmentos de Peptídeos/antagonistas & inibidores , Presenilina-1/antagonistas & inibidores , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Animais , Células Cultivadas , Genes Dominantes/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/biossíntese , Presenilina-1/biossíntese , Presenilina-1/genética , Presenilina-1/fisiologia
10.
J Biol Chem ; 288(24): 17051-6, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23649621

RESUMO

Administration of recombinant factor VIII (FVIII), an important co-factor in blood clotting cascade, elicits unwanted anti-FVIII antibodies in hemophilia A (HA) patients. Previously, FVIII associated with phosphatidylserine (PS) showed significant reduction in the anti-FVIII antibody response in HA mice. The reduction in the immune response to FVIII-PS could be due either to a failure of the immune system to recognize the antigen (i.e. immunological ignorance) or to an active induction of an antigen-specific nonresponsiveness (i.e. immunological tolerance). If it were a result of tolerance, one would predict that pre-exposure to FVIII-PS would render the mice hypo-responsive to a subsequent FVIII challenge. Here, we have demonstrated that naive HA mice that were pretreated with FVIII-PS showed a significantly reduced FVIII immune response to further challenge with native FVIII and that this decreased responsiveness could be adoptively transferred to other mice. An increase in number of FoxP3-expressing CD4(+) regulatory T-cells (Treg) was observed for the FVIII-PS-immunized group as compared with animals that received FVIII alone, suggesting the involvement of Treg in PS-mediated hypo-responsiveness. The PS-mediated reduction in antibody response was reversed by the co-administration of function-blocking anti-TGF-ß antibody with FVIII-PS. The decreased response to FVIII induced by FVIII-PS was determined to be antigen-specific because the immune response to another non-cross-reactive antigen (ovalbumin) was not altered. These results are consistent with the notion that FVIII-PS is tolerogenic and suggest that immunization with this tolerogenic form of the protein could be a useful treatment option to minimize immunogenicity of FVIII and other protein-based therapeutics.


Assuntos
Fator VIII/imunologia , Hemofilia A/terapia , Fosfatidilserinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Transferência Adotiva , Animais , Anticorpos Neutralizantes/sangue , Fator VIII/administração & dosagem , Fatores de Transcrição Forkhead/metabolismo , Hemofilia A/imunologia , Humanos , Tolerância Imunológica , Camundongos , Camundongos Knockout , Fosfatidilserinas/administração & dosagem , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante
11.
PLoS Genet ; 7(9): e1002248, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21912526

RESUMO

The Tsx gene resides at the X-inactivation center and is thought to encode a protein expressed in testis, but its function has remained mysterious. Given its proximity to noncoding genes that regulate X-inactivation, here we characterize Tsx and determine its function in mice. We find that Tsx is actually noncoding and the long transcript is expressed robustly in meiotic germ cells, embryonic stem cells, and brain. Targeted deletion of Tsx generates viable offspring and X-inactivation is only mildly affected in embryonic stem cells. However, mutant embryonic stem cells are severely growth-retarded, differentiate poorly, and show elevated cell death. Furthermore, male mice have smaller testes resulting from pachytene-specific apoptosis and a maternal-specific effect results in slightly smaller litters. Intriguingly, male mice lacking Tsx are less fearful and have measurably enhanced hippocampal short-term memory. Combined, our study indicates that Tsx performs general functions in multiple cell types and links the noncoding locus to stem and germ cell development, learning, and behavior in mammals.


Assuntos
Encéfalo/crescimento & desenvolvimento , Proteínas/genética , RNA não Traduzido/genética , Animais , Apoptose/genética , Proteínas de Ligação a DNA , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/crescimento & desenvolvimento , Hipocampo/fisiologia , Masculino , Memória de Curto Prazo/fisiologia , Camundongos , Proteínas/metabolismo , Proteínas de Ligação a RNA , Deleção de Sequência , Testículo/anatomia & histologia , Inativação do Cromossomo X
12.
J Neurosci ; 32(15): 5085-96, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496554

RESUMO

Mutations in the presenilin-1 (PSEN1) gene are associated with familial Alzheimer's disease and frontotemporal dementia (FTD). Interestingly, neuropathological analysis of a Belgian FTD family carrying a PSEN1 c.548G>T mutation confirmed neurodegeneration in the absence of amyloid plaques. To investigate the impact of the c.548G>T mutation on presenilin-1 (PS1) function in vivo, we introduced this mutation into the genomic Psen1 locus. The resulting c.548G>T knock-in (KI) mice are viable but express markedly lower levels of Psen1 mRNA and protein in the brain. This reduction is due to production of aberrantly spliced transcripts lacking either exon 6 or exons 6 and 7 and their subsequent degradation via non-sense-mediated decay (NMD); inhibition of NMD by cycloheximide treatment stabilized these transcripts and restored the level of Psen1 mRNA in KI/KI brains. Interestingly, the reduction of Psen1 mRNA expression and the degradation of aberrant Psen1 splice products occur exclusively in the brain but not in other tissues. Consistent with decreased Psen1 expression, γ-secretase activity was strongly reduced in the cerebral cortex of KI mice, as measured by de novo γ-secretase-mediated cleavage of APP and Notch. Moreover, PS1 expressed from Psen1 cDNA carrying the c.548G>T mutation displayed normal γ-secretase activity in cultured cells, indicating that the corresponding p.183G>V amino acid substitution does not affect γ-secretase activity. Finally, Psen1 c.548G>T(KI/KI);Psen2(-/-) mice exhibited mild spatial memory deficits in the Morris water maze task. Together, our findings demonstrate that the c.548G>T mutation results in a brain-specific loss of presenilin function due to decreased Psen1 mRNA expression.


Assuntos
Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Mutação/fisiologia , Presenilina-1/genética , RNA Mensageiro/genética , Alelos , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Northern Blotting , Western Blotting , Encéfalo/patologia , Cicloeximida/farmacologia , Ensaio de Imunoadsorção Enzimática , Demência Frontotemporal/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Camundongos , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Proteínas tau/genética , Proteínas tau/metabolismo
13.
Cancer Immun ; 13: 14, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23882159

RESUMO

Human memory T cells present in ovarian tumor ascites fluids fail to respond normally to stimulation via the T cell receptor (TCR). This immunosuppression is manifested by decreases in NF-κB and NFAT activation, IFN-γ production, and cell proliferation in response to TCR stimulation with immobilized antibodies to CD3 and CD28. The anergy of the tumor-associated T cells (TATs) is mediated by soluble factors present in ovarian tumor ascites fluids. The non-responsiveness of the T cells is quickly reversed when the cells are assayed in the absence of the ascites fluid, and is rapidly reestablished when a cell-free ascites fluid is added back to the T cells. Based upon the observed normal phosphorylation patterns of the TCR proximal signaling molecules, the inhibition of NF-κB, and NFAT activation in response to TCR stimulation, as well as the ability of the diacylglycerol analog PMA and the ionophore ionomycin to bypass the ascites fluid-induced TCR signaling arrest, the site of the arrest in the activation cascade appears to be at or just upstream of PLC-γ. An identical TCR signaling arrest pattern was observed when T cells derived from normal donor peripheral blood were incubated with either malignant or nonmalignant (cirrhotic) ascites fluids. The immunosuppressive activity of ascites fluids reported here suggests that soluble factors acting directly or indirectly upon T cells present within tumors contribute to the anergy that has previously been observed in T cells derived from malignant and nonmalignant inflammatory microenvironments. The soluble immunosuppressive factors represent potential therapeutic targets for ovarian cancer.


Assuntos
NF-kappa B/imunologia , Fatores de Transcrição NFATC/imunologia , Neoplasias Ovarianas/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Ascite/imunologia , Ascite/patologia , Feminino , Humanos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais
14.
Cancer Immun ; 13: 11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23885217

RESUMO

Despite an initial response to chemotherapy, most patients with ovarian cancer eventually progress and succumb to their disease. Understanding why effector T cells that are known to infiltrate the tumor do not eradicate the disease after cytoreduction is critically important to the development of novel therapeutic strategies to augment tumor immunity and improve patient outcomes. Such studies have been hampered by the lack of a suitable in vivo model. We report here a simple and reliable model system in which ovarian tumor cell aggregates implanted intraperitoneally into severely immunodeficient NSG mice establish tumor microenvironments within the omentum. The rapid establishment of tumor xenografts within this small anatomically well-defined site enables the recovery, characterization, and quantification of tumor and tumor-associated T cells. We validate here the ability of the omental tumor xenograft (OTX) model to quantify changes in tumor cell number in response to therapy, to quantify changes in the tumor vasculature, and to demonstrate and study the immunosuppressive effects of the tumor microenvironment. Using the OTX model, we show that the tumor-associated T cells originally present within the tumor tissues are anergic and that fully functional autologous T cells injected into tumor-bearing mice localize within the tumor xenograft. The transferred T cells remain functional for up to 3 days within the tumor microenvironment but become unresponsive to activation after 7 days. The OTX model provides for the first time the opportunity to study in vivo the cellular and molecular events contributing to the arrest in T cell function in human ovarian tumors.


Assuntos
Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/imunologia , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Imunoquímica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Linfócitos T/patologia , Microambiente Tumoral
15.
Proc Natl Acad Sci U S A ; 107(21): 9879-84, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20457918

RESUMO

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. LRRK2 is a large protein containing a small GTPase domain and a kinase domain, but its physiological role is unknown. To identify the normal function of LRRK2 in vivo, we generated two independent lines of germ-line deletion mice. The dopaminergic system of LRRK2(-/-) mice appears normal, and numbers of dopaminergic neurons and levels of striatal dopamine are unchanged. However, LRRK2(-/-) kidneys, which suffer the greatest loss of LRRK compared with other organs, develop striking accumulation and aggregation of alpha-synuclein and ubiquitinated proteins at 20 months of age. The autophagy-lysosomal pathway is also impaired in the absence of LRRK2, as indicated by accumulation of lipofuscin granules as well as altered levels of LC3-II and p62. Furthermore, loss of LRRK2 dramatically increases apoptotic cell death, inflammatory responses, and oxidative damage. Collectively, our findings show that LRRK2 plays an essential and unexpected role in the regulation of protein homeostasis during aging, and suggest that LRRK2 mutations may cause Parkinson's disease and cell death via impairment of protein degradation pathways, leading to alpha-synuclein accumulation and aggregation over time.


Assuntos
Envelhecimento , Apoptose , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitinação , alfa-Sinucleína/metabolismo , Animais , Autofagia , Dopamina/metabolismo , Homeostase , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Oxirredução , Proteínas Serina-Treonina Quinases/deficiência
16.
Hum Gene Ther ; 34(19-20): 1049-1063, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37578141

RESUMO

Autosomal dominant Alzheimer's disease (ADAD) is a rare early-onset form of Alzheimer's disease, caused by dominant mutations in one of three genes: presenilin 1, presenilin 2, and amyloid ß precursor protein (APP). Mutations in the presenilin 1 gene (PSEN1) account for the majority of cases, and individuals who inherit a single-mutant PSEN1 allele go on to develop early-onset dementia, ultimately leading to death. The presenilin 1 protein (PS1) is the catalytic subunit of the γ-secretase protease, a tetrameric protease responsible for cleavage of numerous transmembrane proteins, including Notch and the APP. Inclusion of a mutant PS1 subunit in the γ-secretase complex leads to a loss of enzyme function and a preferential reduction of shorter forms of Aß peptides over longer forms, an established biomarker of ADAD progression in human patients. In this study, we describe the development of a gene therapy vector expressing a wild-type (WT) copy of human PSEN1 to ameliorate the loss of function associated with PSEN1 mutations. We have carried out studies in mouse models using a recombinant AAV9 vector to deliver the PSEN1 gene directly into the central nervous system (CNS) and shown that we can normalize γ-secretase function and slow neurodegeneration in both PSEN1 conditional knockout and PSEN1 mutant knockin models. We have also carried out biodistribution studies in nonhuman primates (NHPs) and demonstrated the ability to achieve broad PS1 protein expression throughout the cortex and the hippocampus, two regions known to be critically involved in ADAD progression. These studies demonstrate preclinical proof of concept that expression of a WT human PSEN1 gene in cells harboring a dominant PSEN1 mutation can correct the γ-secretase dysfunction. In addition, direct administration of the recombinant AAV9 into the NHP brain can achieve broad expression at levels predicted to provide efficacy in the clinic.


Assuntos
Doença de Alzheimer , Animais , Camundongos , Humanos , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Distribuição Tecidual , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Mutação , Terapia Genética
17.
J Immunol ; 185(5): 2681-92, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20686130

RESUMO

Fibroblasts are a dominant cell type in most human solid tumors. The possibility that fibroblasts have the capacity to interact with and modulate the function of tumor-associated T lymphocytes makes them a potential therapeutic target. To address this question, primary cultures of fibroblasts derived from human lung tumors were established and cultured with T cells derived from the same tumor. The tumor fibroblasts significantly enhance the production of IFN-gamma and IL-17A by the tumor-associated T cells following a CD3/CD28-induced activation of the T cells. This enhancement was fibroblast cell dose-dependent and did not require direct contact between the two cell types. Tumor-associated fibroblast-conditioned media similarly enhanced both IFN-gamma and IL-17A in activated T cells, and this enhancement was significantly reduced by Abs to IL-6. Conditioned media derived from activated lymphocyte cultures significantly enhanced IL-6 production by tumor fibroblasts. A similar enhancement of IFN-gamma and IL-17A was observed when activated T cells from a normal donor were cultivated with skin fibroblasts derived from the same donor. These results establish that fibroblasts and autologous lymphocytes, whether derived from the tumor microenvironment or from nonmalignant tissues, have the capacity to reciprocally interact and modulate function. In contrast to other reports, fibroblasts are shown to have an immunostimulatory effect upon activated T lymphocytes. The ability of fibroblasts to enhance two T cell cytokines known to have an impact upon tumor progression suggests that fibroblasts play an important role in tumor pathogenesis that could be exploited therapeutically.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Comunicação Celular/imunologia , Fibroblastos/imunologia , Fibroblastos/patologia , Neoplasias Pulmonares/imunologia , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Separação Celular , Técnicas de Cocultura , Relação Dose-Resposta Imunológica , Fibroblastos/metabolismo , Humanos , Interferon gama/biossíntese , Interleucina-17/biossíntese , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Tumorais Cultivadas
18.
J Biol Chem ; 285(29): 22350-9, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20460383

RESUMO

Mutations in presenilin-1 and presenilin-2 (PS1 and PS2) are the most common cause of familial Alzheimer disease. PS1 and PS2 are the presumptive catalytic components of the multisubunit gamma-secretase complex, which proteolyzes a number of type I transmembrane proteins, including the amyloid precursor protein (APP) and Notch. APP processing by gamma-secretase produces beta-amyloid peptides (Abeta40 and Abeta42) that accumulate in the Alzheimer disease brain. Here we identify a pathogenic L435F mutation in PS1 in two affected siblings with early-onset familial Alzheimer disease characterized by deposition of cerebral cotton wool plaques. The L435F mutation resides in a conserved C-terminal PAL sequence implicated in active site conformation and catalytic activity. The impact of PS1 mutations in and around the PAL motif on gamma-secretase activity was assessed by expression of mutant PS1 in mouse embryo fibroblasts lacking endogenous PS1 and PS2. Surprisingly, the L435F mutation caused a nearly complete loss of gamma-secretase activity, including >90% reductions in the generation of Abeta40, Abeta42, and the APP and Notch intracellular domains. Two nonpathogenic PS1 mutations, P433L and L435R, caused essentially complete loss of gamma-secretase activity, whereas two previously identified pathogenic PS1 mutations, P436Q and P436S, caused partial loss of function with substantial reductions in production of Abeta40, Abeta42, and the APP and Notch intracellular domains. These results argue against overproduction of Abeta42 as an essential property of presenilin proteins bearing pathogenic mutations. Rather, our findings provide support for the hypothesis that pathogenic mutations cause a general loss of presenilin function.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Mutação/genética , Placa Amiloide/enzimologia , Placa Amiloide/genética , Presenilina-1/genética , Idade de Início , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/patologia , Substituição de Aminoácidos/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Caderinas/metabolismo , Membrana Celular/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Pessoa de Meia-Idade , Proteínas Mutantes/metabolismo , Placa Amiloide/patologia , Presenilina-1/deficiência , Processamento de Proteína Pós-Traducional , Receptores Notch/metabolismo
19.
Blood ; 113(16): 3809-12, 2009 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-19196657

RESUMO

The incorporation of rituximab, a chimeric anti-CD20 monoclonal antibody, into the therapeutic armamentarium for patients with follicular lymphoma (FL) has significantly improved treatment outcome for such patients. Despite the almost universal application of this therapy, however, its exact mechanism of action has not been completely defined. One proposed mechanism is that of a "vaccinal" effect, whereby FL cell kill by rituximab results in the elicitation of an FL-specific T-cell response. The demonstration that rituximab can even elicit such a response in patients has, to our knowledge, never been shown. We analyzed the response against the immunoglobulin expressed by the FL before and after rituximab monotherapy in 5 FL patients and found an increase in FL idiotype-specific T cells after rituximab in 4 of 5 patients. Our data thus provide "proof of principle" for the ability of passive immunotherapy with rituximab to elicit an active FL-specific cellular response.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Antineoplásicos/imunologia , Formação de Anticorpos/efeitos dos fármacos , Antineoplásicos/administração & dosagem , Linfoma Folicular/imunologia , Linfócitos T/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Murinos , Antineoplásicos/imunologia , Feminino , Humanos , Imunidade Celular/efeitos dos fármacos , Imunoterapia , Linfoma Folicular/terapia , Masculino , Rituximab
20.
Clin Transl Immunology ; 10(2): e1246, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33552509

RESUMO

OBJECTIVES: With a rapidly growing list of candidate immune-based cancer therapeutics, there is a critical need to generate highly reliable animal models to preclinically evaluate the efficacy of emerging immune-based therapies, facilitating successful clinical translation. Our aim was to design and validate a novel in vivo model (called Xenomimetic or 'X' mouse) that allows monitoring of the ability of human tumor-specific T cells to suppress tumor growth following their entry into the tumor. METHODS: Tumor xenografts are established rapidly in the greater omentum of globally immunodeficient NOD-scid IL2Rγnull (NSG) mice following an intraperitoneal injection of melanoma target cells expressing tumor neoantigen peptides, as well as green fluorescent protein and/or luciferase. Changes in tumor burden, as well as in the number and phenotype of adoptively transferred patient-derived tumor neoantigen-specific T cells in response to immunotherapy, are measured by imaging to detect fluorescence/luminescence and flow cytometry, respectively. RESULTS: The tumors progress rapidly and disseminate in the mice unless patient-derived tumor-specific T cells are introduced. An initial T cell-mediated tumor arrest is later followed by a tumor escape, which correlates with the upregulation of the checkpoint molecules programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (LAG3) on T cells. Treatment with immune-based therapies that target these checkpoints, such as anti-PD-1 antibody (nivolumab) or interleukin-12 (IL-12), prevented or delayed the tumor escape. Furthermore, IL-12 treatment suppressed PD-1 and LAG3 upregulation on T cells. CONCLUSION: Together, these results validate the X-mouse model and establish its potential to preclinically evaluate the therapeutic efficacy of immune-based therapies.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa