Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.198
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
São Paulo; s.n; 2023. 27 p.
Tese em Português | ColecionaSUS, SMS-SP, HSPM-Producao, SMS-SP | ID: biblio-1532438

RESUMO

Introdução: A displasia fibrosa (DF) do osso é uma desordem congênita, rara, que corresponde de 5 a 10% dos tumores ósseos benignos, não hereditária, que cursa com amplo espectro de apresentação, variando do assintomático à dor óssea, fraturas de repetição, deformidades ósseas (fêmur em cajado de pastor e fácies leonina) e compressão de nervos cranianos. Histologicamente é composta de estroma fibroso celular de baixo a moderado grau circundando trabéculas ósseas de formato irregular sem borda osteoblástica. Todos os casos contêm a mutação GNAS1. A DF apresenta duas formas: a monostótica, mais comum (70-80%), e a poliostótica, mais rara (20-30%), que quando acompanhada de manchas café-com-leite e puberdade precoce constitui a síndrome de McCune-Albright ou Síndrome de Mazabraud em casos mais raros. O tratamento pode ser feito com medicamentos como bifosfonato ou de forma cirúrgica, objetivando-se a correção das lesões com curetagem e enxertia óssea ou como iremos mostrar a seguir, pela Técnica de Masquelet. Este trabalho relata o caso de um menino de 20 anos de idade cujos sinais e sintomas conduziam ao diagnóstico de DF sendo realizado tratamento com Técnica de Masquelet e follow up de 18 meses. Além disso, faz revisão de literatura sobre uma doença pouco comum, com variada gama de diagnósticos diferenciais. Objetivo: relatar um caso de displasia fibrosa com tratamento cirúrgico de enxerto autólogo de fíbula pela Técnica de Masquelet. Método: relato de caso de paciente do Ambulatório de Especialidade do Hospital do Servidor Público Municipal, de 20 anos de idade, que foi acompanhado por 1 ano e meio apresentando um tumor ósseo na tíbia compatível com diagnóstico de displasia fibrosa, que ao longo desse período foi submetido à Técnica de Masquelet. Conclusão: É pouco descrito na literatura o tratamento de displasia fibrosa pela Técnica de Masquelet, que mostrou ter ótimo resultado funcional para o paciente estudado. Palavras-chave: Displasia Fibrosa Óssea. Displasia Fibrosa. Técnica de Masquelet. Técnica de Membrana Induzida.


Assuntos
Humanos , Masculino , Adulto , Osteomielite/terapia , Pseudoartrose/terapia , Tíbia/cirurgia , Transplante Autólogo/métodos , Osso e Ossos/fisiopatologia , Fraturas Ósseas/congênito , Fêmur/cirurgia , Displasia Fibrosa Poliostótica , Fíbula/cirurgia , Mutação/fisiologia , Neoplasias/cirurgia
2.
Genes (Basel) ; 12(11)2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34828274

RESUMO

Mitochondria are specialized organelles involved in energy production that have retained their own genome throughout evolutionary history. The mitochondrial genome (mtDNA) is maternally inherited and requires coordinated regulation with nuclear genes to produce functional enzyme complexes that drive energy production. Each mitochondrion contains 5-10 copies of mtDNA and consequently, each cell has several hundreds to thousands of mtDNAs. Due to the presence of multiple copies of mtDNA in a mitochondrion, mtDNAs with different variants may co-exist, a condition called heteroplasmy. Heteroplasmic variants can be clonally expanded, even in post-mitotic cells, as replication of mtDNA is not tied to the cell-division cycle. Heteroplasmic variants can also segregate during germ cell formation, underlying the inheritance of some mitochondrial mutations. Moreover, the uneven segregation of heteroplasmic variants is thought to underlie the heterogeneity of mitochondrial variation across adult tissues and resultant differences in the clinical presentation of mitochondrial disease. Until recently, however, the mechanisms mediating the relation between mitochondrial genetic variation and disease remained a mystery, largely due to difficulties in modeling human mitochondrial genetic variation and diseases. The advent of induced pluripotent stem cells (iPSCs) and targeted gene editing of the nuclear, and more recently mitochondrial, genomes now provides the ability to dissect how genetic variation in mitochondrial genes alter cellular function across a variety of human tissue types. This review will examine the origins of mitochondrial heteroplasmic variation and propagation, and the tools used to model mitochondrial genetic diseases. Additionally, we discuss how iPSC technologies represent an opportunity to advance our understanding of human mitochondrial genetics in disease.


Assuntos
DNA Mitocondrial/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/terapia , Animais , Edição de Genes/métodos , Heterogeneidade Genética , Genoma Mitocondrial , Heteroplasmia/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Doenças Mitocondriais/patologia , Mutação/fisiologia , Transplante de Células-Tronco/métodos
3.
Artigo em Inglês | MEDLINE | ID: mdl-34820595

RESUMO

Pathogenic germline variants (PGVs) in cancer susceptibility genes are usually identified through germline testing of DNA from blood or saliva: their detection can affect treatment options and potential risk-reduction strategies for patient relatives. PGV can also be identified in tumor sequencing assays, which, when performed without patient-matched normal specimens, render determination of variants' germline or somatic origin critical. METHODS: Tumor-only sequencing data from 1,608 patients were retrospectively analyzed to infer germline versus somatic status of variants using an information-theoretic, gene-independent approach. Loss of heterozygosity was also determined. Predicted mutational models were compared with clinical germline testing results. Statistical measures were computed to evaluate performance. RESULTS: Tumor-only sequencing detected 3,988 variants across 70 cancer susceptibility genes for which germline testing data were available. We imputed germline versus somatic status for > 75% of all detected variants, with a sensitivity of 65%, specificity of 88%, and overall accuracy of 86% for pathogenic variants. False omission rate was 3%, signifying minimal error in misclassifying true PGV. A higher portion of PGV in known hereditary tumor suppressors were found to be retained with loss of heterozygosity in the tumor specimens (72%) compared with variants of uncertain significance (58%). CONCLUSION: Analyzing tumor-only data in the context of specimens' tumor cell content allows precise, systematic exclusion of somatic variants and suggests a balance between type 1 and 2 errors for identification of patients with candidate PGV for standard germline testing. Although technical or systematic errors in measuring variant allele frequency could result in incorrect inference, misestimation of specimen purity could result in inferring somatic variants as germline in somatically mutated tumor suppressor genes. A user-friendly bioinformatics application facilitates objective analysis of tumor-only data in clinical settings.


Assuntos
Mutação/genética , Neoplasias/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Células Germinativas , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Mutação/fisiologia , Sequenciamento Completo do Genoma/métodos
4.
Biochem Pharmacol ; 192: 114715, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34339714

RESUMO

Glucose-dependent insulinotropic polypeptide (GIP) and its receptor (GIPR) are part of the incretin system that regulates glucose homeostasis. A series of GIPR residues putatively important for ligand binding and receptor activation were mutated and pharmacologically evaluated using GIPR selective agonists in cAMP accumulation, ERK1/2 phosphorylation (pERK1/2) and ß-arrestin 2 recruitment assays. The impact of mutation on ligand efficacy was determined by operational modelling of experimental data for each mutant, with results mapped onto the full-length, active-state GIPR structure. Two interaction networks, comprising transmembrane helix (TM) 7, TM1 and TM2, and extracellular loop (ECL) 2, TM5 and ECL3 were revealed, respectively. Both networks were critical for Gαs-mediated cAMP accumulation and the recruitment of ß-arrestin 2, however, cAMP response was more sensitive to alanine substitution, with most mutated residues displaying reduced signaling. Unlike the other two assays, activation of ERK1/2 was largely independent of the network involving ECL2, TM5 and ECL3, indicating that pERK1/2 is at least partially distinct from Gαs or ß-arrestin pathways and this network is also crucial for potential biased agonism at GIPR. Collectively, our work advances understanding of the structure-function relationship of GIPR and provides a framework for the design and/or interpretation of GIP analogues with unique signaling profiles.


Assuntos
Polipeptídeo Inibidor Gástrico/genética , Polipeptídeo Inibidor Gástrico/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Células COS , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Polipeptídeo Inibidor Gástrico/química , Polipeptídeo Inibidor Gástrico/farmacologia , Células HEK293 , Humanos , Mutação/fisiologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Estrutura Secundária de Proteína , Receptores dos Hormônios Gastrointestinais/química
5.
Front Neural Circuits ; 15: 700968, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34366796

RESUMO

Autism spectrum disorder (ASD) is a neurodevelopmental disorder defined by altered social interaction and communication, and repetitive, restricted, inflexible behaviors. Approximately 1.5-2% of the general population meet the diagnostic criteria for ASD and several brain regions including the cortex, amygdala, cerebellum and basal ganglia have been implicated in ASD pathophysiology. The midbrain dopamine system is an important modulator of cellular and synaptic function in multiple ASD-implicated brain regions via anatomically and functionally distinct dopaminergic projections. The dopamine hypothesis of ASD postulates that dysregulation of dopaminergic projection pathways could contribute to the behavioral manifestations of ASD, including altered reward value of social stimuli, changes in sensorimotor processing, and motor stereotypies. In this review, we examine the support for the idea that cell-autonomous changes in dopaminergic function are a core component of ASD pathophysiology. We discuss the human literature supporting the involvement of altered dopamine signaling in ASD including genetic, brain imaging and pharmacologic studies. We then focus on genetic mouse models of syndromic neurodevelopmental disorders in which single gene mutations lead to increased risk for ASD. We highlight studies that have directly examined dopamine neuron number, morphology, physiology, or output in these models. Overall, we find considerable support for the idea that the dopamine system may be dysregulated in syndromic ASDs; however, there does not appear to be a consistent signature and some models show increased dopaminergic function, while others have deficient dopamine signaling. We conclude that dopamine dysregulation is common in syndromic forms of ASD but that the specific changes may be unique to each genetic disorder and may not account for the full spectrum of ASD-related manifestations.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Dopamina/genética , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutação/fisiologia
7.
Cancer Med ; 10(14): 4977-4993, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34076361

RESUMO

BACKGROUND: Lung adenocarcinoma (LUAD), as the most common subtype of lung cancer, is the leading cause of cancer deaths in the world. The accumulation of driver gene mutations enables cancer cells to gradually acquire growth advantage. Therefore, it is important to understand the functions and interactions of driver gene mutations in cancer progression. METHODS: We obtained gene mutation data and gene expression profile of 506 LUAD tumors from The Cancer Genome Atlas (TCGA). The subtypes of tumors with driver gene mutations were identified by consensus cluster analysis. RESULTS: We found 21 significantly mutually exclusive pairs consisting of 20 genes among 506 LUAD patients. Because of the increased transcriptomic heterogeneity of mutations, we identified subtypes among tumors with non-silent mutations in driver genes. There were 494 mutually exclusive pairs found among driver gene mutations within different subtypes. Furthermore, we identified functions of mutually exclusive pairs based on the hypothesis of functional redundancy of mutual exclusivity. These mutually exclusive pairs were significantly enriched in nuclear division and humoral immune response, which played crucial roles in cancer initiation and progression. We also found 79 mutually exclusive triples among subtypes of tumors with driver gene mutations, which were key roles in cell motility and cellular chemical homeostasis. In addition, two mutually exclusive triples and one mutually exclusive triple were associated with the overall survival and disease-specific survival of LUAD patients, respectively. CONCLUSIONS: We revealed novel mutual exclusivity and generated a comprehensive functional landscape of driver gene mutations, which could offer a new perspective to understand the mechanisms of cancer development and identify potential biomarkers for LUAD therapy.


Assuntos
Adenocarcinoma de Pulmão/genética , Progressão da Doença , Neoplasias Pulmonares/genética , Mutação/genética , Transcriptoma/genética , Adenocarcinoma de Pulmão/mortalidade , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Heterogeneidade Genética , Fenômenos Genéticos , Humanos , Neoplasias Pulmonares/mortalidade , Mutação/fisiologia
8.
Biochem Pharmacol ; 188: 114583, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33915156

RESUMO

Somatic gain-of-function mutations within estrogen receptor alpha (ERα) are highly associated with hormone therapy resistance in breast cancer. However, current understanding of abnormal activity of ERα mutants and their relevant targeted intervention is still very limited. Herein, we developed a new, real-time, and reliably Gaussia luciferase-based protein-fragment complementation assay (GLPCA) for evaluating ERα mutants activities. We found that, compared with ER WT, ERα mutants (Y537S/N and D538G) exhibit high ligand-independent activity, suggesting the gain-of-function phenotype of these ERα mutants. Notably, Y537S, the most common ERα mutant type, has the highest intrinsic activation. We then collected and screened a natural product library for potential ERα antagonists via GLPCA and identified celastrol and gambogic acid as new antagonists of the ERα Y537S mutant. Moreover, interactions between these two compounds and the ERα Y537S mutant were confirmed by molecular docking and cellular thermal shift assay. Importantly, we further demonstrated that celastrol and gambogic acid exhibit synergistic antiproliferative and pro-apoptotic effects when combined with an approved CDK4/6 inhibitor abemaciclib in breast cancer cells expressing ERα Y537S. In summary, GLPCA provides a powerful platform for exploring innovative functional biology and drug discovery of antagonists targeting ERα mutants.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Mutação/efeitos dos fármacos , Triterpenos Pentacíclicos/uso terapêutico , Xantonas/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Relação Dose-Resposta a Droga , Receptor alfa de Estrogênio/genética , Feminino , Células HEK293 , Humanos , Células MCF-7 , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação/fisiologia , Triterpenos Pentacíclicos/farmacologia , Estrutura Secundária de Proteína , Xantonas/farmacologia
9.
Cell Biol Int ; 45(8): 1720-1732, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33847415

RESUMO

Keratin 8/18, the predominant keratin pair of simple epithelia, is known to be aberrantly expressed in several squamous cell carcinomas (SCCs), where its expression is often correlated with increased invasion, neoplastic progression, and poor prognosis. The majority of keratin 8/18 structural and regulatory functions are governed by posttranslational modifications, particularly phosphorylation. Apart from filament reorganization, cellular processes including cell cycle, cell growth, cellular stress, and apoptosis are known to be orchestrated by K8 phosphorylation at specific residues in the head and tail domains. Even though deregulation of K8 phosphorylation at two significant sites (Serine73 /Serine431 ) has been implicated in neoplastic progression of SCCs by various in vitro studies, including ours, it is reported to be highly context-dependent. Therefore, to delineate the precise role of Kereatin 8 phosphorylation in cancer initiation and progression, we have developed the tissue-specific transgenic mouse model expressing Keratin 8 wild type and phosphodead mutants under Keratin 14 promoter. Subjecting these mice to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-mediated skin carcinogenesis revealed that Keratin 8 phosphorylation may lead to an early onset of tumors compared to Keratin 8 wild-type expressing mice. Conclusively, the transgenic mouse model developed in the present study ascertained a positive impact of Keratin 8 phosphorylation on the neoplastic transformation of skin-squamous cells.


Assuntos
Carcinogênese/metabolismo , Queratina-8/metabolismo , Mutação/fisiologia , Neoplasias Cutâneas/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Eletroporação/métodos , Células HEK293 , Humanos , Queratina-8/genética , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação/fisiologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
10.
Cell Calcium ; 96: 102384, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33676318

RESUMO

BACKGROUND: Colorectal cancer (CRC) metastases are the main cause of CRC mortality. Intracellular Ca2+ regulates cell migration and invasion, key factors for metastases. Ca2+ also activates Ca2+-dependent potassium channels which in turn affect Ca2+ driving force. We have previously reported that the expression of the Ca2+ activated potassium channel KCNN4 (SK4) is higher in CRC primary tumors compared to normal tissues. Here, we aimed to investigate the role of SK4 in the physiology of CRC. RESULTS: SK4 protein expression is enhanced in CRC tissues compared to normal colon tissues, with a higher level of KCNN4 in CRC patients with KRAS mutations. At the cellular level, we found that SK4 regulates the membrane potential of HCT116 cells. We also found that its inhibition reduced store operated Ca2+ entry (SOCE) and constitutive Ca2+ entry (CCE), while reducing cell migration. We also found that the activity of SK4 is linked to resistance pathways such as KRAS mutation and the expression of NRF2 and HIF-1α. In addition, the pharmacological inhibition of SK4 reduced intracellular reactive oxygen species (ROS) production, NRF2 expression and HIF1α stabilization. CONCLUSION: Our results suggest that SK4 contributes to colorectal cancer cell migration and invasion by modulating both Ca2+ entry and ROS regulation. Therefore, SK4 could be a potential target to reduce metastasis in KRAS-mutated CRC.


Assuntos
Cálcio/metabolismo , Movimento Celular/fisiologia , Neoplasias Colorretais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/biossíntese , Mutação/fisiologia , Proteínas Proto-Oncogênicas p21(ras) , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Bases de Dados Genéticas , Células HCT116 , Células HT29 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirazóis/farmacologia
11.
Cell Cycle ; 20(7): 631-646, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33722167

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a deadly neuromuscular disorder caused by progressive motor neuron loss in the brain and spinal cord. Over the past decades, a number of genetic mutations have been identified that cause or are associated with ALS disease progression. Numerous genes harbor ALS mutations, and they encode proteins displaying a wide range of physiological functions, with limited overlap. Despite the divergent functions, mutations in these genes typically trigger protein aggregation, which can confer gain- and/or loss-of-function to a number of essential cellular processes. Nuclear processes such as mRNA splicing and the response to DNA damage are significantly affected in ALS patients. Cytoplasmic organelles such as mitochondria are damaged by ALS mutant proteins. Processes that maintain cellular homeostasis such as autophagy, nonsense-mediated mRNA decay and nucleocytoplasmic transport, are also impaired by ALS mutations. Here, we review the multiple mechanisms by which mutations in major ALS-associated genes, such as TARDBP, C9ORF72 and FUS, lead to impairment of essential cellular processes.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteína C9orf72/genética , Morte Celular/fisiologia , Proteínas de Ligação a DNA/genética , Mutação/fisiologia , Proteína FUS de Ligação a RNA/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Autofagia/fisiologia , Proteína C9orf72/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Humanos , Proteína FUS de Ligação a RNA/metabolismo
12.
Biochem Pharmacol ; 185: 114407, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33421376

RESUMO

The tumor suppressor protein p53 participates in the control of key biological functions such as cell death, metabolic homeostasis and immune function, which are closely related to various diseases such as tumors, metabolic disorders, infection and neurodegeneration. The p53 gene is also mutated in approximately 50% of human cancer cells. Mutant p53 proteins escape from the ubiquitination-dependent degradation, gain oncogenic function and promote the carcinogenesis, malignant progression, metastasis and chemoresistance. Therefore, the stability of both wild type and mutant p53 needs to be precisely regulated to maintain normal functions and targeting the p53 stability is one of the therapeutic strategies against cancer. Here, we focus on compound-induced degradation of p53 by both the ubiquitination-dependent proteasome and autophagy-lysosome degradation pathways. We also review other posttranslational modifications which control the stability of p53 and the biological functions involved in these processes. This review provides the current theoretical basis for the regulation of p53 abundance and its possible applications in different diseases.


Assuntos
Antineoplásicos/metabolismo , Neoplasias/metabolismo , Proteólise , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Humanos , Mutação/efeitos dos fármacos , Mutação/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Estabilidade Proteica/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/fisiologia
13.
Exp Cell Res ; 399(2): 112456, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33417921

RESUMO

Identifying patient mutations driving skeletal development disorders has driven our understanding of bone development. Integrin adhesion deficiency disease is caused by a Kindlin-3 (fermitin family member 3) mutation, and its inactivation results in bleeding disorders and osteopenia. In this study, we uncover a role for Kindlin-3 in the differentiation of bone marrow mesenchymal stem cells (BMSCs) down the chondrogenic lineage. Kindlin-3 expression increased with chondrogenic differentiation, similar to RUNX2. BMSCs isolated from a Kindlin-3 deficient patient expressed chondrocyte markers, including SOX9, under basal conditions, which were further enhanced with chondrogenic differentiation. Rescue of integrin activation by a constitutively activated ß3 integrin construct increased adhesion to multiple extracellular matrices and reduced SOX9 expression to basal levels. Growth plates from mice expressing a mutated Kindlin-3 with the integrin binding site ablated demonstrated alterations in chondrocyte maturation similar to that seen with the human Kindlin-3 deficient BMSCs. These findings suggest that Kindlin-3 expression mirrors RUNX2 during chondrogenesis.


Assuntos
Condrogênese/genética , Proteínas do Citoesqueleto/genética , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/fisiologia , Proteínas de Neoplasias/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/fisiologia
14.
Neurochirurgie ; 67(1): 76-82, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30554773

RESUMO

Adult medulloblastomas are orphan diseases that differ from their pediatric counterpart. Most are classified as classic or desmoplastic and fall in the SHH subgroup, mainly with loss-of-function mutations in PTCH1 and some by TP53-mutation due to underlying germline mutation. Activation of the WNT pathway is sporadic, although underlying Turcot syndrome may be present. One-third of tumors are issued from group 4. Most adult studies are small non-randomized retrospective heterogeneous studies performed at a single center with short follow-up. Standard craniospinal irradiation followed by maintenance chemotherapy (CCNU, cisplatin-vincristine) results in a 4-year event-free survival (EFS) and overall survival (OS) of 68% and 89% respectively in standard-risk adults, and in a 4-year EFS and OS of 50% and 90%, respectively in high-risk adults. Several pooled analyses point out the potential role of chemotherapy in adults. The feasibility of pediatric protocols in adults is sometimes hampered because of blood and peripheral nerve toxicity. In the near future, subgroups of medulloblastomas may be treated by personalized therapies. With prolonged follow-up, adults fare worse. Long-term sequelae and second line treatment are not well defined in adults. Prospective studies are ongoing to define optimal first-line and relapse treatments.


Assuntos
Neoplasias Cerebelares/diagnóstico , Neoplasias Cerebelares/genética , Meduloblastoma/diagnóstico , Meduloblastoma/genética , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Cerebelares/terapia , Criança , Pré-Escolar , Cisplatino/uso terapêutico , Feminino , Humanos , Masculino , Meduloblastoma/terapia , Mutação/fisiologia , Procedimentos Neurocirúrgicos/métodos , Procedimentos Neurocirúrgicos/tendências , Intervalo Livre de Progressão , Estudos Prospectivos , Radioterapia/métodos , Radioterapia/tendências , Estudos Retrospectivos , Vincristina/uso terapêutico , Adulto Jovem
15.
Neurochem Int ; 144: 104940, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33333210

RESUMO

Pediatric brain tumors are the leading cancer-related cause of death in children and adolescents in the United States, affecting on average 1 in 2000 children per year. Recent advances in cancer genomics have led to profound discoveries about the underlying molecular biology and ontogeny of these tumors. In particular, these studies have revealed epigenetic dysregulation to be one of the main hallmarks of pediatric brain tumorigenesis. In this review, we will highlight a number of important recent findings about the nature of this dysregulation in different types of pediatric brain tumors as well as examine their implications for preclinical research and clinical practice. Specifically, we discuss the emergence of methylation signatures as tools for tumor stratification/classification while also highlighting the importance of mutations that directly affect the epigenome and clarifying their impact on risk stratification and pediatric brain tumor biology. We then incorporate recent advances in our understanding of pediatric brain tumor cellular architecture and emphasize the link between epigenetic dysregulation and the "stalled" development seen in many of these malignant neoplasms. Lastly, we explore recentwork investigating the use of these mutated epigenomic regulators as therapeutic targets and extrapolate their utility in overcoming this "stalling" to halt tumor growth.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Mutação/fisiologia , Fatores Etários , Neoplasias Encefálicas/metabolismo , Criança , Humanos
17.
J Neuroinflammation ; 17(1): 290, 2020 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-33023623

RESUMO

BACKGROUND: Evidence shows significant heterogeneity in astrocyte gene expression and function. We previously demonstrated that brain-derived neurotrophic factor (BDNF) exerts protective effects on whole brain primary cultured rat astrocytes treated with 3-nitropropionic acid (3NP), a mitochondrial toxin widely used as an in vitro model of Huntington's disease (HD). Therefore, we now investigated 3NP and BDNF effects on astrocytes from two areas involved in HD: the striatum and the entire cortex, and their involvement in neuron survival. METHODS: We prepared primary cultured rat cortical or striatal astrocytes and treated them with BDNF and/or 3NP for 24 h. In these cells, we assessed expression of astrocyte markers, BDNF receptor, and glutamate transporters, and cytokine release. We prepared astrocyte-conditioned medium (ACM) from cortical and striatal astrocytes and tested its effect on a cellular model of HD. RESULTS: BDNF protected astrocytes from 3NP-induced death, increased expression of its own receptor, and activation of ERK in both cortical and striatal astrocytes. However, BDNF modulated glutamate transporter expression differently by increasing GLT1 and GLAST expression in cortical astrocytes but only GLT1 expression in striatal astrocytes. Striatal astrocytes released higher amounts of tumor necrosis factor-α than cortical astrocytes in response to 3NP but BDNF decreased this effect in both populations. 3NP decreased transforming growth factor-ß release only in cortical astrocytes, whereas BDNF treatment increased its release only in striatal astrocytes. Finally, we evaluated ACM effect on a cellular model of HD: the rat striatal neuron cell line ST14A expressing mutant human huntingtin (Q120) or in ST14A cells expressing normal human huntingtin (Q15). Neither striatal nor cortical ACM modified the viability of Q15 cells. Only ACM from striatal astrocytes treated with BDNF and ACM from 3NP + BDNF-treated striatal astrocytes protected Q120 cells, whereas ACM from cortical astrocytes did not. CONCLUSIONS: Data suggest that cortical and striatal astrocytes respond differently to mitochondrial toxin 3NP and BDNF. Moreover, striatal astrocytes secrete soluble neuroprotective factors in response to BDNF that selectively protect neurons expressing mutant huntingtin implicating that BDNF modulation of striatal astrocyte function has therapeutic potential against neurodegeneration.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/toxicidade , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Proteína Huntingtina/biossíntese , Nitrocompostos/toxicidade , Propionatos/toxicidade , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Feminino , Expressão Gênica , Humanos , Proteína Huntingtina/genética , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mutação/efeitos dos fármacos , Mutação/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Ratos , Ratos Wistar
18.
Nat Rev Mol Cell Biol ; 21(12): 715-728, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32968234

RESUMO

In the 20 years since human embryonic stem cells, and subsequently induced pluripotent stem cells, were first described, it has become apparent that during long-term culture these cells (collectively referred to as 'pluripotent stem cells' (PSCs)) can acquire genetic changes, which commonly include gains or losses of particular chromosomal regions, or mutations in certain cancer-associated genes, especially TP53. Such changes raise concerns for the safety of PSC-derived cellular therapies for regenerative medicine. Although acquired genetic changes may not be present in a cell line at the start of a research programme, the low sensitivity of current detection methods means that mutations may be difficult to detect if they arise but are present in only a small proportion of the cells. In this Review, we discuss the types of mutations acquired by human PSCs and the mechanisms that lead to their accumulation. Recent work suggests that the underlying mutation rate in PSCs is low, although they also seem to be particularly susceptible to genomic damage. This apparent contradiction can be reconciled by the observations that, in contrast to somatic cells, PSCs are programmed to die in response to genomic damage, which may reflect the requirements of early embryogenesis. Thus, the common genetic variants that are observed are probably rare events that give the cells with a selective growth advantage.


Assuntos
Evolução Clonal/genética , Acúmulo de Mutações , Células-Tronco Pluripotentes/metabolismo , Técnicas de Cultura de Células/métodos , Técnicas de Cultura de Células/normas , Diferenciação Celular/genética , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Células Cultivadas , Evolução Clonal/fisiologia , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Mutação/fisiologia , Células-Tronco Pluripotentes/fisiologia
19.
PLoS Comput Biol ; 16(9): e1008185, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32925942

RESUMO

Cells adjust their metabolism in response to mutations, but how this reprogramming depends on the genetic context is not well known. Specifically, the absence of individual enzymes can affect reprogramming, and thus the impact of mutations in cell growth. Here, we examine this issue with an in silico model of Saccharomyces cerevisiae's metabolism. By quantifying the variability in the growth rate of 10000 different mutant metabolisms that accumulated changes in their reaction fluxes, in the presence, or absence, of a specific enzyme, we distinguish a subset of modifier genes serving as buffers or potentiators of variability. We notice that the most potent modifiers refer to the glycolysis pathway and that, more broadly, they show strong pleiotropy and epistasis. Moreover, the evidence that this subset depends on the specific growing condition strengthens its systemic underpinning, a feature only observed before in a toy model of a gene-regulatory network. Some of these enzymes also modulate the effect that biochemical noise and environmental fluctuations produce in growth. Thus, the reorganization of metabolism induced by mutations has not only direct physiological implications but also transforms the influence that other mutations have on growth. This is a general result with implications in the development of cancer therapies based on metabolic inhibitors.


Assuntos
Redes Reguladoras de Genes/genética , Redes e Vias Metabólicas , Mutação , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/fisiologia , Mutação/genética , Mutação/fisiologia , Fenótipo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Biologia de Sistemas
20.
Mol Hum Reprod ; 26(10): 727-737, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32777047

RESUMO

The FMR1 gene contains a polymorphic CGG trinucleotide sequence within its 5' untranslated region. More than 200 CGG repeats (termed a full mutation) underlie the severe neurodevelopmental condition fragile X syndrome, while repeat lengths that range between 55 and 200 (termed a premutation) result in the conditions fragile X-associated tremor/ataxia syndrome and fragile X-associated premature ovarian insufficiency (FXPOI). Premutations in FMR1 are the most common monogenic cause of premature ovarian insufficiency and are routinely tested for clinically; however, the mechanisms that contribute to the pathology are still largely unclear. As studies in this field move towards unravelling the molecular mechanisms involved in FXPOI aetiology, we review the evidence surrounding the two main theories which describe an RNA toxic gain-of-function mechanism, resulting in the loss of function of RNA-binding proteins, or a protein-based mechanism, where repeat-associated non-AUG translation leads to the formation of an abnormal polyglycine containing protein, called FMRpolyG.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/fisiologia , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/metabolismo , Animais , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Humanos , Mutação/fisiologia , Peptídeos/genética , Peptídeos/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA