Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 326(5): F862-F875, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38511222

RESUMO

IgA nephropathy (IgAN) is characterized by glomerular deposition of immune complexes (ICs) consisting of IgA1 with O-glycans deficient in galactose (Gd-IgA1) and Gd-IgA1-specific IgG autoantibodies. These ICs induce kidney injury, and in the absence of disease-specific therapy, up to 40% of patients with IgAN progress to kidney failure. IgA1 with its clustered O-glycans is unique to humans, which hampered development of small-animal models of IgAN. Here, we used a model wherein engineered ICs (EICs) formed from human Gd-IgA1 and recombinant human IgG autoantibody are injected into nude mice to induce glomerular injury mimicking human IgAN. In this model, we assessed the protective effects of sparsentan, a single-molecule dual endothelin angiotensin receptor antagonist (DEARA) versus vehicle on EIC-induced glomerular proliferation and dysregulation of gene expression in the kidney. Oral administration of sparsentan (60 or 120 mg/kg daily) to mice intravenously injected with EIC attenuated the EIC-induced glomerular hypercellularity. Furthermore, analysis of changes in the whole kidney transcriptome revealed that key inflammatory and proliferative biological genes and pathways that are upregulated in this EIC model of IgAN were markedly reduced by sparsentan, including complement genes, integrin components, members of the mitogen-activated protein kinase family, and Fc receptor elements. Partial overlap between mouse and human differentially expressed genes in IgAN further supported the translational aspect of the immune and inflammatory components from our transcriptional findings. In conclusion, our data indicate that in the mouse model of IgAN, sparsentan targets immune and inflammatory processes leading to protection from mesangial hypercellularity.NEW & NOTEWORTHY The mechanisms by which deposited IgA1 immune complexes cause kidney injury during early phases of IgA nephropathy are poorly understood. We used an animal model we recently developed that involves IgA1-IgG immune complex injections and determined pathways related to the induced mesangioproliferative changes. Treatment with sparsentan, a dual inhibitor of endothelin type A and angiotensin II type 1 receptors, ameliorated the induced mesangioproliferative changes and the associated alterations in the expression of inflammatory genes and networks.


Assuntos
Complexo Antígeno-Anticorpo , Modelos Animais de Doenças , Glomerulonefrite por IGA , Imunoglobulina A , Imunoglobulina G , Glomérulos Renais , Animais , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/tratamento farmacológico , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/patologia , Glomerulonefrite por IGA/metabolismo , Imunoglobulina A/metabolismo , Imunoglobulina A/imunologia , Glomérulos Renais/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Redes Reguladoras de Genes , Camundongos Nus , Humanos , Camundongos , Proliferação de Células/efeitos dos fármacos
2.
J Pathol ; 260(3): 353-364, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37256677

RESUMO

Alport syndrome (AS), a type IV collagen disorder, leads to glomerular disease and, in some patients, hearing loss. AS is treated with inhibitors of the renin-angiotensin system; however, a need exists for novel therapies, especially those addressing both major pathologies. Sparsentan is a single-molecule dual endothelin type-A and angiotensin II type 1 receptor antagonist (DEARA) under clinical development for focal segmental glomerulosclerosis and IgA nephropathy. We report the ability of sparsentan to ameliorate both renal and inner ear pathologies in an autosomal-recessive Alport mouse model. Sparsentan significantly delayed onset of glomerulosclerosis, interstitial fibrosis, proteinuria, and glomerular filtration rate decline. Sparsentan attenuated glomerular basement membrane defects, blunted mesangial filopodial invasion into the glomerular capillaries, increased lifespan more than losartan, and lessened changes in profibrotic/pro-inflammatory gene pathways in both the glomerular and the renal cortical compartments. Notably, treatment with sparsentan, but not losartan, prevented accumulation of extracellular matrix in the strial capillary basement membranes in the inner ear and reduced susceptibility to hearing loss. Improvements in lifespan and in renal and strial pathology were observed even when sparsentan was initiated after development of renal pathologies. These findings suggest that sparsentan may address both renal and hearing pathologies in Alport syndrome patients. © 2023 Travere Therapeutics, Inc and The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Orelha Interna , Nefrite Hereditária , Animais , Camundongos , Nefrite Hereditária/metabolismo , Receptores de Angiotensina/metabolismo , Receptores de Angiotensina/uso terapêutico , Membrana Basal Glomerular/metabolismo , Colágeno Tipo IV/genética , Orelha Interna/metabolismo , Orelha Interna/patologia , Endotelinas/metabolismo , Endotelinas/uso terapêutico
3.
Cell ; 132(3): 474-86, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18267077

RESUMO

Mitotic spindle orientation and plane of cleavage in mammals is a determinant of whether division yields progenitor expansion and/or birth of new neurons during radial glial progenitor cell (RGPC) neurogenesis, but its role earlier in neuroepithelial stem cells is poorly understood. Here we report that Lis1 is essential for precise control of mitotic spindle orientation in both neuroepithelial stem cells and radial glial progenitor cells. Controlled gene deletion of Lis1 in vivo in neuroepithelial stem cells, where cleavage is uniformly vertical and symmetrical, provokes rapid apoptosis of those cells, while radial glial progenitors are less affected. Impaired cortical microtubule capture via loss of cortical dynein causes astral and cortical microtubules to be greatly reduced in Lis1-deficient cells. Increased expression of the LIS/dynein binding partner NDEL1 restores cortical microtubule and dynein localization in Lis1-deficient cells. Thus, control of symmetric division, essential for neuroepithelial stem cell proliferation, is mediated through spindle orientation determined via LIS1/NDEL1/dynein-mediated cortical microtubule capture.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Embrião de Mamíferos/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Células Neuroepiteliais/citologia , Fuso Acromático/metabolismo , Células-Tronco/citologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Encéfalo/citologia , Encéfalo/embriologia , Ciclo Celular , Movimento Celular , Proliferação de Células , Dineínas/metabolismo , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Modelos Biológicos , Neurônios/citologia
4.
Mol Psychiatry ; 26(12): 7641-7651, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34341515

RESUMO

Early detection and intervention are believed to be key to facilitating better outcomes in children with autism, yet the impact of age at treatment start on the outcome is poorly understood. While clinical traits such as language ability have been shown to predict treatment outcome, whether or not and how information at the genomic level can predict treatment outcome is unknown. Leveraging a cohort of toddlers with autism who all received the same standardized intervention at a very young age and provided a blood sample, here we find that very early treatment engagement (i.e., <24 months) leads to greater gains while controlling for time in treatment. Pre-treatment clinical behavioral measures predict 21% of the variance in the rate of skill growth during early intervention. Pre-treatment blood leukocyte gene expression patterns also predict the rate of skill growth, accounting for 13% of the variance in treatment slopes. Results indicated that 295 genes can be prioritized as driving this effect. These treatment-relevant genes highly interact at the protein level, are enriched for differentially histone acetylated genes in autism postmortem cortical tissue, and are normatively highly expressed in a variety of subcortical and cortical areas important for social communication and language development. This work suggests that pre-treatment biological and clinical behavioral characteristics are important for predicting developmental change in the context of early intervention and that individualized pre-treatment biology related to histone acetylation may be key.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Transtorno Autístico/genética , Comunicação , Intervenção Educacional Precoce/métodos , Expressão Gênica , Humanos , Resultado do Tratamento
5.
BMC Genomics ; 22(1): 69, 2021 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-33478392

RESUMO

BACKGROUND: Both RNA-Seq and sample freeze-thaw are ubiquitous. However, knowledge about the impact of freeze-thaw on downstream analyses is limited. The lack of common quality metrics that are sufficiently sensitive to freeze-thaw and RNA degradation, e.g. the RNA Integrity Score, makes such assessments challenging. RESULTS: Here we quantify the impact of repeated freeze-thaw cycles on the reliability of RNA-Seq by examining poly(A)-enriched and ribosomal RNA depleted RNA-seq from frozen leukocytes drawn from a toddler Autism cohort. To do so, we estimate the relative noise, or percentage of random counts, separating technical replicates. Using this approach we measured noise associated with RIN and freeze-thaw cycles. As expected, RIN does not fully capture sample degradation due to freeze-thaw. We further examined differential expression results and found that three freeze-thaws should extinguish the differential expression reproducibility of similar experiments. Freeze-thaw also resulted in a 3' shift in the read coverage distribution along the gene body of poly(A)-enriched samples compared to ribosomal RNA depleted samples, suggesting that library preparation may exacerbate freeze-thaw-induced sample degradation. CONCLUSION: The use of poly(A)-enrichment for RNA sequencing is pervasive in library preparation of frozen tissue, and thus, it is important during experimental design and data analysis to consider the impact of repeated freeze-thaw cycles on reproducibility.


Assuntos
Criopreservação , RNA , Congelamento , Humanos , Reprodutibilidade dos Testes , Análise de Sequência de RNA
6.
Mol Psychiatry ; 25(9): 2175-2188, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-30104728

RESUMO

Early-onset neurodevelopmental conditions (e.g., autism) affect males more frequently than females. Androgens may play a role in this male-bias by sex-differentially impacting early prenatal brain development, particularly neural circuits that later develop specialized roles in social cognition. Here, we find that increasing prenatal testosterone in humans is associated with later reduction of functional connectivity between social brain default mode (DMN) subsystems in adolescent males, but has no effect in females. Since testosterone can work directly via the androgen receptor (AR) or indirectly via the estrogen receptor through aromatase conversion to estradiol, we further examined how a potent non-aromatizable androgen, dihydrotestosterone (DHT), acts via the AR to influence gene expression in human neural stem cells (hNSC)-particularly for genes of high-relevance for DMN circuitry. DHT dysregulates a number of genes enriched for syndromic causes of autism and intellectual disability and for genes that in later development are expressed in anatomical patterns that highly correspond to the cortical midline DMN subsystem. DMN-related and DHT-affected genes (e.g., MEF2C) are involved in a number of synaptic processes, many of which impact excitation-inhibition balance. Androgens have male-specific prenatal influence over social brain circuitry in humans and may be relevant towards explaining some component of male-bias in early-onset neurodevelopmental conditions.


Assuntos
Androgênios , Di-Hidrotestosterona , Adolescente , Encéfalo , Estradiol , Feminino , Humanos , Masculino , Testosterona
7.
Mol Psychiatry ; 24(1): 88-107, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29934544

RESUMO

Autism spectrum disorder (ASD) has captured the attention of scientists, clinicians and the lay public because of its uncertain origins and striking and unexplained clinical heterogeneity. Here we review genetic, genomic, cellular, postmortem, animal model, and cell model evidence that shows ASD begins in the womb. This evidence leads to a new theory that ASD is a multistage, progressive disorder of brain development, spanning nearly all of prenatal life. ASD can begin as early as the 1st and 2nd trimester with disruption of cell proliferation and differentiation. It continues with disruption of neural migration, laminar disorganization, altered neuron maturation and neurite outgrowth, disruption of synaptogenesis and reduced neural network functioning. Among the most commonly reported high-confidence ASD (hcASD) genes, 94% express during prenatal life and affect these fetal processes in neocortex, amygdala, hippocampus, striatum and cerebellum. A majority of hcASD genes are pleiotropic, and affect proliferation/differentiation and/or synapse development. Proliferation and subsequent fetal stages can also be disrupted by maternal immune activation in the 1st trimester. Commonly implicated pathways, PI3K/AKT and RAS/ERK, are also pleiotropic and affect multiple fetal processes from proliferation through synapse and neural functional development. In different ASD individuals, variation in how and when these pleiotropic pathways are dysregulated, will lead to different, even opposing effects, producing prenatal as well as later neural and clinical heterogeneity. Thus, the pathogenesis of ASD is not set at one point in time and does not reside in one process, but rather is a cascade of prenatal pathogenic processes in the vast majority of ASD toddlers. Despite this new knowledge and theory that ASD biology begins in the womb, current research methods have not provided individualized information: What are the fetal processes and early-age molecular and cellular differences that underlie ASD in each individual child? Without such individualized knowledge, rapid advances in biological-based diagnostic, prognostic, and precision medicine treatments cannot occur. Missing, therefore, is what we call ASD Living Biology. This is a conceptual and paradigm shift towards a focus on the abnormal prenatal processes underlying ASD within each living individual. The concept emphasizes the specific need for foundational knowledge of a living child's development from abnormal prenatal beginnings to early clinical stages. The ASD Living Biology paradigm seeks this knowledge by linking genetic and in vitro prenatal molecular, cellular and neural measurements with in vivo post-natal molecular, neural and clinical presentation and progression in each ASD child. We review the first such study, which confirms the multistage fetal nature of ASD and provides the first in vitro fetal-stage explanation for in vivo early brain overgrowth. Within-child ASD Living Biology is a novel research concept we coin here that advocates the integration of in vitro prenatal and in vivo early post-natal information to generate individualized and group-level explanations, clinically useful prognoses, and precision medicine approaches that are truly beneficial for the individual infant and toddler with ASD.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/metabolismo , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Masculino , Fenótipo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
8.
Mol Psychiatry ; 22(6): 820-835, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27378147

RESUMO

Autism spectrum disorders (ASD) are common, complex and heterogeneous neurodevelopmental disorders. Cellular and molecular mechanisms responsible for ASD pathogenesis have been proposed based on genetic studies, brain pathology and imaging, but a major impediment to testing ASD hypotheses is the lack of human cell models. Here, we reprogrammed fibroblasts to generate induced pluripotent stem cells, neural progenitor cells (NPCs) and neurons from ASD individuals with early brain overgrowth and non-ASD controls with normal brain size. ASD-derived NPCs display increased cell proliferation because of dysregulation of a ß-catenin/BRN2 transcriptional cascade. ASD-derived neurons display abnormal neurogenesis and reduced synaptogenesis leading to functional defects in neuronal networks. Interestingly, defects in neuronal networks could be rescued by insulin growth factor 1 (IGF-1), a drug that is currently in clinical trials for ASD. This work demonstrates that selection of ASD subjects based on endophenotypes unraveled biologically relevant pathway disruption and revealed a potential cellular mechanism for the therapeutic effect of IGF-1.


Assuntos
Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Técnicas de Cultura de Tecidos/métodos , Adolescente , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Encéfalo/metabolismo , Proliferação de Células/genética , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/uso terapêutico , Masculino , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Neurônios/fisiologia , beta Catenina/metabolismo
9.
Mol Syst Biol ; 11(12): 841, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26668231

RESUMO

Genetic mechanisms underlying abnormal early neural development in toddlers with Autism Spectrum Disorder (ASD) remain uncertain due to the impossibility of direct brain gene expression measurement during critical periods of early development. Recent findings from a multi-tissue study demonstrated high expression of many of the same gene networks between blood and brain tissues, in particular with cell cycle functions. We explored relationships between blood gene expression and total brain volume (TBV) in 142 ASD and control male toddlers. In control toddlers, TBV variation significantly correlated with cell cycle and protein folding gene networks, potentially impacting neuron number and synapse development. In ASD toddlers, their correlations with brain size were lost as a result of considerable changes in network organization, while cell adhesion gene networks significantly correlated with TBV variation. Cell cycle networks detected in blood are highly preserved in the human brain and are upregulated during prenatal states of development. Overall, alterations were more pronounced in bigger brains. We identified 23 candidate genes for brain maldevelopment linked to 32 genes frequently mutated in ASD. The integrated network includes genes that are dysregulated in leukocyte and/or postmortem brain tissue of ASD subjects and belong to signaling pathways regulating cell cycle G1/S and G2/M phase transition. Finally, analyses of the CHD8 subnetwork and altered transcript levels from an independent study of CHD8 suppression further confirmed the central role of genes regulating neurogenesis and cell adhesion processes in ASD brain maldevelopment.


Assuntos
Transtorno do Espectro Autista/genética , Encéfalo/patologia , Proteínas de Ciclo Celular/genética , Redes Reguladoras de Genes , Mutação , Transtorno do Espectro Autista/patologia , Encéfalo/crescimento & desenvolvimento , Adesão Celular , Proteínas de Ciclo Celular/sangue , Pré-Escolar , Biologia Computacional , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Lactente , Masculino
10.
PLoS Genet ; 8(3): e1002592, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22457638

RESUMO

Autism is a highly heritable neurodevelopmental disorder, yet the genetic underpinnings of the disorder are largely unknown. Aberrant brain overgrowth is a well-replicated observation in the autism literature; but association, linkage, and expression studies have not identified genetic factors that explain this trajectory. Few studies have had sufficient statistical power to investigate whole-genome gene expression and genotypic variation in the autistic brain, especially in regions that display the greatest growth abnormality. Previous functional genomic studies have identified possible alterations in transcript levels of genes related to neurodevelopment and immune function. Thus, there is a need for genetic studies involving key brain regions to replicate these findings and solidify the role of particular functional pathways in autism pathogenesis. We therefore sought to identify abnormal brain gene expression patterns via whole-genome analysis of mRNA levels and copy number variations (CNVs) in autistic and control postmortem brain samples. We focused on prefrontal cortex tissue where excess neuron numbers and cortical overgrowth are pronounced in the majority of autism cases. We found evidence for dysregulation in pathways governing cell number, cortical patterning, and differentiation in young autistic prefrontal cortex. In contrast, adult autistic prefrontal cortex showed dysregulation of signaling and repair pathways. Genes regulating cell cycle also exhibited autism-specific CNVs in DNA derived from prefrontal cortex, and these genes were significantly associated with autism in genome-wide association study datasets. Our results suggest that CNVs and age-dependent gene expression changes in autism may reflect distinct pathological processes in the developing versus the mature autistic prefrontal cortex. Our results raise the hypothesis that genetic dysregulation in the developing brain leads to abnormal regional patterning, excess prefrontal neurons, cortical overgrowth, and neural dysfunction in autism.


Assuntos
Fatores Etários , Transtorno Autístico , Variações do Número de Cópias de DNA , Regulação da Expressão Gênica , Córtex Pré-Frontal , Adolescente , Adulto , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Autopsia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Criança , Pré-Escolar , Variações do Número de Cópias de DNA/genética , Feminino , Deleção de Genes , Redes Reguladoras de Genes , Genoma Humano , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/patologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Transdução de Sinais/genética
11.
PLoS Genet ; 7(3): e1001331, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21423666

RESUMO

Heterozygous LIS1 mutations are the most common cause of human lissencephaly, a human neuronal migration defect, and DCX mutations are the most common cause of X-linked lissencephaly. LIS1 is part of a protein complex including NDEL1 and 14-3-3ε that regulates dynein motor function and microtubule dynamics, while DCX stabilizes microtubules and cooperates with LIS1 during neuronal migration and neurogenesis. Targeted gene mutations of Lis1, Dcx, Ywhae (coding for 14-3-3ε), and Ndel1 lead to neuronal migration defects in mouse and provide models of human lissencephaly, as well as aid the study of related neuro-developmental diseases. Here we investigated the developing brain of these four mutants and wild-type mice using expression microarrays, bioinformatic analyses, and in vivo/in vitro experiments to address whether mutations in different members of the LIS1 neuronal migration complex lead to similar and/or distinct global gene expression alterations. Consistent with the overall successful development of the mutant brains, unsupervised clustering and co-expression analysis suggested that cell cycle and synaptogenesis genes are similarly expressed and co-regulated in WT and mutant brains in a time-dependent fashion. By contrast, focused co-expression analysis in the Lis1 and Ndel1 mutants uncovered substantial differences in the correlation among pathways. Differential expression analysis revealed that cell cycle, cell adhesion, and cytoskeleton organization pathways are commonly altered in all mutants, while synaptogenesis, cell morphology, and inflammation/immune response are specifically altered in one or more mutants. We found several commonly dysregulated genes located within pathogenic deletion/duplication regions, which represent novel candidates of human mental retardation and neurocognitive disabilities. Our analysis suggests that gene expression and pathway analysis in mouse models of a similar disorder or within a common pathway can be used to define novel candidates for related human diseases.


Assuntos
Encéfalo/metabolismo , Movimento Celular , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Adesão Celular , Ciclo Celular , Polaridade Celular/genética , Citoesqueleto/metabolismo , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fenótipo , Proteína Quinase C/metabolismo , Transdução de Sinais , Sinapses/fisiologia , Regulação para Cima
12.
J Cell Sci ; 124(Pt 5): 745-54, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21285247

RESUMO

Little is known about the role of cell-cell adhesion in the development of mineralized tissues. Here we report that PERP, a tetraspan membrane protein essential for epithelial integrity, regulates enamel formation. PERP is necessary for proper cell attachment and gene expression during tooth development, and its expression is controlled by P63, a master regulator of stratified epithelial development. During enamel formation, PERP is localized to the interface between the enamel-producing ameloblasts and the stratum intermedium (SI), a layer of cells subjacent to the ameloblasts. Perp-null mice display dramatic enamel defects, which are caused, in part, by the detachment of ameloblasts from the SI. Microarray analysis comparing gene expression in teeth of wild-type and Perp-null mice identified several differentially expressed genes during enamel formation. Analysis of these genes in ameloblast-derived LS8 cells upon knockdown of PERP confirmed the role for PERP in the regulation of gene expression. Together, our data show that PERP is necessary for the integrity of the ameloblast-SI interface and that a lack of Perp causes downregulation of genes that are required for proper enamel formation.


Assuntos
Adesão Celular/fisiologia , Esmalte Dentário/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Expressão Gênica , Proteínas de Membrana/metabolismo , Odontogênese/fisiologia , Ameloblastos/citologia , Ameloblastos/fisiologia , Animais , Células Cultivadas , Desmossomos/metabolismo , Desmossomos/ultraestrutura , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Análise em Microsséries , Dente/anatomia & histologia , Dente/crescimento & desenvolvimento , Dente/metabolismo
13.
Orphanet J Rare Dis ; 18(1): 13, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36650582

RESUMO

BACKGROUND: Cerebrotendinous xanthomatosis (CTX) is a rare recessive genetic disease characterized by disruption of bile acid synthesis due to inactivation of the CYP27A1 gene. Treatment is available in the form of bile acid replacement. CTX is likely underdiagnosed, and prevalence estimates based on case diagnosis are probably inaccurate. Large population-based genomic databases are a valuable resource to estimate prevalence of rare recessive diseases as an orthogonal unbiased approach building upon traditional epidemiological studies. METHODS: We leveraged the Hardy-Weinberg principle and allele frequencies from gnomAD to calculate CTX prevalence. ClinVar and HGMD were used to identify high-confidence pathogenic missense variants and to calculate a disease-specific cutoff. Variant pathogenicity was also assessed by the VarSome implementation of the ACMG/AMP algorithm and the REVEL in silico predictor. RESULTS: CTX prevalence estimates were highest in Asians (1:44,407-93,084) and lowest in the Finnish population (1:3,388,767). Intermediate estimates were found in Europeans, Americans, and Africans/African Americans (1:70,795-233,597). The REVEL-predicted pathogenic variants accounted for a greater increase in prevalence estimates for Europeans, Americans, and Africans/African Americans compared with Asians. We identified the most frequent alleles designated pathogenic in ClinVar (p.Gly472Ala, p.Arg395Cys), labeled pathogenic based on sequence consequence (p.Met1?), and predicted to be pathogenic by REVEL (p.Met383Lys, p.Arg448His) across populations. Also, we provide a prospective geographic map of estimated disease distribution based on CYP27A1 variation queries performed by healthcare providers from selected specialties. CONCLUSIONS: Prevalence estimates calculated herein support and expand upon existing evidence indicating underdiagnosis of CTX, suggesting that improved detection strategies are needed. Increased awareness of CTX is important for early diagnosis, which is essential for patients as early treatment significantly slows or prevents disease progression.


Assuntos
Xantomatose Cerebrotendinosa , Humanos , Xantomatose Cerebrotendinosa/epidemiologia , Xantomatose Cerebrotendinosa/genética , Alelos , Prevalência , Estudos Prospectivos , Ácidos e Sais Biliares
14.
Semin Cell Dev Biol ; 21(8): 823-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20688183

RESUMO

Lissencephaly is a severe human neuronal migration defect characterized by a smooth cerebral surface, mental retardation and seizures. The two most common genes mutated in patients with lissencephaly are LIS1 and DCX. LIS1 was the first gene cloned that was important for neuronal migration in any organism, and heterozygous mutations or deletions of LIS1 are found in the majority of patients with lissencephaly, while DCX mutations were found in males with X-linked lissencephaly. In this review, we will discuss how an understanding of the molecular and cellular pathways disrupted in model organisms with Lis1 and Dcx mutations or knock-down not only provide insights into the normal processes of neuronal migration, including neurogenesis, but they also may lead to potential novel therapeutic strategies for these severe cortical malformations.


Assuntos
Modelos Animais de Doenças , Lisencefalia/genética , Lisencefalia/terapia , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Animais , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/genética , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/metabolismo , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/terapia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Humanos , Lisencefalia/metabolismo , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurogênese , Neuropeptídeos/metabolismo , Ratos
15.
J Neurosci ; 30(8): 3002-12, 2010 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-20181597

RESUMO

Heterozygous LIS1 mutations and males with loss of the X-linked DCX result in lissencephaly, a neuronal migration defect. LIS1 regulates nuclear translocation and mitotic division of neural progenitor cells, while the role of DCX in cortical development remains poorly understood. Here, we uncovered novel neuronal migration and proliferation defects in the Dcx mutant embryonic brains. Although cortical organization was fairly well preserved, Dcx(ko/Y) neurons displayed defective migration velocities similar to Lis1(+/ko) neurons when characterized by time-lapse video-microscopy of embryonic cortical slices. Dcx(ko/Y) migrating neurons displayed novel multidirectional movements with abnormal morphology and increased branching. Surprisingly, Dcx(ko/Y) radial glial cells displayed spindle orientation abnormalities similar to Lis1(+/ko) cells that in turn lead to moderate proliferation defects both in vivo and in vitro. We found functional genetic interaction of the two genes, with the combined effects of Lis1 haploinsufficiency and Dcx knock-out leading to more severe neuronal migration and proliferation phenotypes in the Lis1(+/ko);Dcx(ko/Y) male double mutant compared with the single mutants, resulting in cortical disorganization and depletion of the progenitor pool. Thus, we provide definitive evidence for a critical role for Dcx in neuronal migration and neurogenesis, as well as for the in vivo genetic interaction of the two genes most commonly involved in human neuronal migration defects.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Encéfalo/anormalidades , Diferenciação Celular/genética , Movimento Celular/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurogênese/genética , Neuropeptídeos/genética , Animais , Encéfalo/citologia , Forma Celular/genética , Células Cultivadas , Córtex Cerebral/anormalidades , Córtex Cerebral/citologia , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Malformações do Sistema Nervoso/genética , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Esferoides Celulares , Células-Tronco/citologia , Células-Tronco/metabolismo
16.
Sci Adv ; 7(36): eabh1663, 2021 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-34516910

RESUMO

Cortical regionalization develops via genomic patterning along anterior-posterior (A-P) and dorsal-ventral (D-V) gradients. Here, we find that normative A-P and D-V genomic patterning of cortical surface area (SA) and thickness (CT), present in typically developing and autistic toddlers with good early language outcome, is absent in autistic toddlers with poor early language outcome. Autistic toddlers with poor early language outcome are instead specifically characterized by a secondary and independent genomic patterning effect on CT. Genes involved in these effects can be traced back to midgestational A-P and D-V gene expression gradients and different prenatal cell types (e.g., progenitor cells and excitatory neurons), are functionally important for vocal learning and human-specific evolution, and are prominent in prenatal coexpression networks enriched for high-penetrance autism risk genes. Autism with poor early language outcome may be explained by atypical genomic cortical patterning starting in prenatal development, which may detrimentally affect later regional functional specialization and circuit formation.

17.
Clin Cancer Res ; 27(5): 1391-1398, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33262136

RESUMO

PURPOSE: Circulating tumor cells (CTC) are under investigation as a minimally invasive liquid biopsy that may improve risk stratification and treatment selection. CTCs uniquely allow for digital pathology of individual malignant cell morphology and marker expression. We compared CTC features and T-cell counts with survival endpoints in a cohort of patients with metastatic genitourinary cancer treated with combination immunotherapy. EXPERIMENTAL DESIGN: Markers evaluated included pan-CK/CD45/PD-L1/DAPI for CTCs and CD4/CD8/Ki-67/DAPI for T cells. ANOVA was used to compare CTC burden and T-cell populations across timepoints. Differences in survival and disease progression were evaluated using the maximum log-rank test. RESULTS: From December 2016 to January 2019, 183 samples from 81 patients were tested. CTCs were found in 75% of patients at baseline. CTC burden was associated with shorter overall survival (OS) at baseline (P = 0.022), but not on-therapy. Five morphologic subtypes were detected, and the presence of two specific subtypes with unique cellular features at baseline and on-therapy was associated with worse OS (0.9-2.3 vs. 28.2 months; P < 0.0001-0.013). Increasing CTC heterogeneity on-therapy had a trend toward worse OS (P = 0.045). PD-L1+ CTCs on-therapy were associated with worse OS (P < 0.01, cycle 2). Low baseline and on-therapy CD4/CD8 counts were also associated with poor OS and response category. CONCLUSIONS: Shorter survival may be associated with high CTC counts at baseline, presence of specific CTC morphologic subtypes, PD-L1+ CTCs, and low %CD4/8 T cells in patients with metastatic genitourinary cancer. A future study is warranted to validate the prognostic utility of CTC heterogeneity and detection of specific CTC morphologies.


Assuntos
Biomarcadores Tumorais/análise , Imunoterapia/métodos , Células Neoplásicas Circulantes/patologia , Linfócitos T/imunologia , Neoplasias Urogenitais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Linfócitos T/classificação , Neoplasias Urogenitais/imunologia , Neoplasias Urogenitais/terapia , Adulto Jovem
18.
J Neurosci ; 29(49): 15520-30, 2009 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-20007476

RESUMO

Haploinsufficiency of LIS1 results in lissencephaly, a human neuronal migration disorder. LIS1 is a microtubule- (MT) and centrosome- [microtubule organizing center (MTOC)] associated protein that regulates nucleokinesis via the regulation of dynein motor function and localization. NDEL1 (NudE isoform, NudE like) interacts with LIS1/dynein complex, and is phosphorylated by CDK5/P35. Previous reports using siRNA-mediated knock-down demonstrated similar critical roles for LIS1 and NDEL1 during neuronal migration, but neuronal migration has not been studied in genetic mutants for Lis1 and Ndel1 where protein levels are uniform in all cells. Brains from mice with complete loss of Lis1 and Ndel1 displayed severe cortical layering and hippocampal defects, but Lis1 mutants had more severe defects. Neuronal migration speed was reduced and neurite lengths were elongated in proportion to the reduction of LIS1 and NDEL1 protein levels in embryonic day 14.5 mutant cortical slices compared to wild type, using two-photon confocal time lapse videomicroscopy. Additionally, mice with 35% of wild-type NDEL1 levels displayed diverse branched migration modes with multiple leading processes, suggesting defects in adhesion and/or polarity. Complete loss of Lis1 or Ndel1 resulted in the total inhibition of nuclear movement in cortical slice assays, and in neurosphere assays, the percentage of migrating neurons with correctly polarized MTOC location was significantly reduced while nuclear-centrosomal distance was extended. Neurite lengths were increased after complete loss Ndel1 but reduced after complete loss of Lis1. Thus, Lis1 and Ndel1 are essential for normal cortical neuronal migration, neurite outgrowth, and function of the MTOC in a dose-dependent manner.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Córtex Cerebral/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Neuritos/fisiologia , Neurônios/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/fisiologia , Proteínas de Transporte/genética , Contagem de Células , Núcleo Celular/fisiologia , Centrossomo/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Hipocampo/citologia , Hipocampo/embriologia , Hipocampo/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/fisiologia , Mutação , Neurônios/citologia , Células-Tronco/citologia , Células-Tronco/fisiologia
19.
Nat Neurosci ; 22(10): 1624-1634, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31551593

RESUMO

Hundreds of genes are implicated in autism spectrum disorder (ASD), but the mechanisms through which they contribute to ASD pathophysiology remain elusive. Here we analyzed leukocyte transcriptomics from 1- to 4-year-old male toddlers with ASD or typical development from the general population. We discovered a perturbed gene network that includes highly expressed genes during fetal brain development. This network is dysregulated in human induced pluripotent stem cell-derived neuron models of ASD. High-confidence ASD risk genes emerge as upstream regulators of the network, and many risk genes may impact the network by modulating RAS-ERK, PI3K-AKT and WNT-ß-catenin signaling pathways. We found that the degree of dysregulation in this network correlated with the severity of ASD symptoms in the toddlers. These results demonstrate how the heterogeneous genetics of ASD may dysregulate a core network to influence brain development at prenatal and very early postnatal ages and, thereby, the severity of later ASD symptoms.


Assuntos
Transtorno do Espectro Autista/genética , Redes Reguladoras de Genes/genética , Transtorno do Espectro Autista/patologia , Encéfalo/embriologia , Encéfalo/patologia , Pré-Escolar , Desenvolvimento Fetal/genética , Humanos , Lactente , Leucócitos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Mutação/genética , Células-Tronco Neurais , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Via de Sinalização Wnt/genética , beta Catenina/genética
20.
Eur J Hum Genet ; 16(8): 880-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18337728

RESUMO

We report a patient with mild pachygyria, ascertained during a screening of subjects with abnormal neuronal migration and/or epilepsy, having a 7q11.23 duplication reciprocal to the Williams-Beuren critical region (WBCR) deletion. He exhibited speech delay and mental retardation together to type II trigonocephaly and other abnormalities. The proband's mother carried the same imbalance, though her phenotype was milder and no abnormal conformation of the cranium was reported. She had suffered a few seizures in infancy, as already described in other duplicated subjects. This genomic imbalance, now described in 17 subjects, including one parent for each of the four probands, is associated with a variable phenotype. Speech impairment is present in most cases; no distinctive facial gestalt is recognizable; seizures have been reported in four subjects and brain magnetic resonance, performed in eight cases, resulted abnormal in six, while detected abnormal neuronal migration in two. Although the clinical description of additional cases is needed to delineate a definite phenotypic core for WBCR duplications, trigonocephaly, also reported in another dup(7)(q11.23) patient, is possibly a trait that, together with speech impairment, may call for clinically oriented specific screening. Abnormal development of the cerebral cortex, reported also in the Williams-Beuren deletion, suggests that at least one gene is present in the critical region whose deletion/duplication impairs neuronal migration.


Assuntos
Cromossomos Humanos Par 7/genética , Ossos Faciais/anormalidades , Duplicação Gênica , Deficiência Intelectual/genética , Transtornos do Desenvolvimento da Linguagem/genética , Síndrome de Williams/genética , Adolescente , Adulto , Criança , Pré-Escolar , Deleção Cromossômica , Feminino , Humanos , Hibridização in Situ Fluorescente , Deficiência Intelectual/patologia , Cariotipagem , Transtornos do Desenvolvimento da Linguagem/patologia , Masculino , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Síndrome de Williams/diagnóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA