Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Lab Invest ; 104(4): 100327, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38237738

RESUMO

Impaired endometrial decidualization is the primary cause of recurrent implantation failure (RIF). RNA methylation modification, especially NSUN family mediated m5C, is crucial for various physiological events, such as maternal-to-zygotic transition, gametogenesis, embryonic development, organismal lifespan, and cell cycle. However, the regulatory mechanisms between NSUN family mediated m5C modification and RIF remain unknown. We acquired NSUN2 expression data of 15 human endometrium samples at proliferative and secretory stages from reproductive cell atlas. The overall pattern of m5C sites and genes was elucidated through m5C-BS-seq, whereas the overall m5C levels in different groups were revealed by dot blot assay. BrdU and western blotting assays were carried out to evaluate the role of NSUN2 in proliferation and autophagy. The effects of NSUN2-mediated m5C modification on embryo attachment were evaluated by an in vitro model of a confluent monolayer of Ishikawa cells cocultured with BeWo spheroids, and its downstream targets were evaluated by real-time reverse-transcription PCR and western blotting in Ishikawa cells. The molecular mechanism for NSUN2 regulating its downstream targets' expression was determined by Cut&Tag and coimmunoprecipitation assays. NSUN2 was increased in SOX9+ cells and widespread in epithelial cell type at the proliferative stage by previous single-cell RNA sequencing data. NSUN2 overexpression (NSUN2OE) in the Ishikawa cell line elevated m5C levels and promoted cell proliferation and autophagy. NSUN2OE reduced attachment efficiency of BeWo cell spheres. Overexpressed NSUN2 was found to increase STAT1 and MMP14 mRNA expressions by inducing exon skipping. NSUN2 interacted with CLDN4 through m5C modification, and NSUN2OE or NSUN2 knockdown resulted in a similar variation tendency of CLDN4. Overexpression of NSUN2 increased CLDN4 H3K9ac modification by downregulating SIRT4 expression at the protein level, leading to the upregulation of CLDN4 mRNA expression. Our results uncovered a novel intricate regulatory mechanism between NSUN2-mediated m5C and RIF and suggested a potential new therapeutic strategy for RIF.


Assuntos
Implantação do Embrião , Endométrio , Gravidez , Feminino , Humanos , Implantação do Embrião/genética , Metilação , Linhagem Celular , RNA Mensageiro/metabolismo , Metiltransferases/metabolismo
2.
Cell Biosci ; 12(1): 212, 2022 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36587229

RESUMO

BACKGROUND: Pathogenic mutations in WRN are a cause of premature aging disease Werner syndrome (WS). Besides accelerated aging phenotypes and cancer predisposition, patients with WS also display underdevelopment in the skeletal system, characterized by short stature, light body weight and unusually thin extremities. The reasons for these developmental defects are not completely understood and the underlying molecular mechanism remains to be elucidated. RESULTS: In this study, WRN was found to modulate transcription of short stature homeobox gene SHOX. Loss of WRN resulted in insufficient expression of SHOX, the gene dose of which is critical for driving chondrocyte differentiation. WRN could bind the G-quadruplex (G4) structures in the SHOX promoter and stimulate transcription. Aberrant formation of G4 structures in WRN-deficient cells impeded normal transcription of SHOX, thus resulting in impaired chondrogenesis. Chondrogenesis could be rescued by overexpression of WRN helicase or SHOX, suggesting that SHOX is a downstream target of WRN. Gene editing of the G4 structures in the SHOX promoter could increase SHOX expression, therefore rescuing the impaired chondrogenesis in WRN-deficient cells. CONCLUSIONS: Our data suggest that dysgenesis of the developing bone in WS might be caused by SHOX insufficiency. Aberrant formation of G4 structures in SHOX promoter suppresses SHOX expression and impairs chondrogenesis. Targeted mutagenesis in the G4 structures enhances SHOX expression and thus providing an opportunity to rescue the chondrogenic defect.

4.
Nat Commun ; 13(1): 5456, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114168

RESUMO

Werner Syndrome (WS) is an autosomal recessive disorder characterized by premature aging due to mutations of the WRN gene. A classical sign in WS patients is short stature, but the underlying mechanisms are not well understood. Here we report that WRN is indispensable for chondrogenesis, which is the engine driving the elongation of bones and determines height. Zebrafish lacking wrn exhibit impairment of bone growth and have shorter body stature. We pinpoint the function of WRN to its helicase domain. We identify short-stature homeobox (SHOX) as a crucial and direct target of WRN and find that the WRN helicase core regulates the transcriptional expression of SHOX via unwinding G-quadruplexes. Consistent with this, shox-/- zebrafish exhibit impaired bone growth, while genetic overexpression of SHOX or shox expression rescues the bone developmental deficiency induced in WRN/wrn-null mutants both in vitro and in vivo. Collectively, we have identified a previously unknown function of WRN in regulating bone development and growth through the transcriptional regulation of SHOX via the WRN helicase domain, thus illuminating a possible approach for new therapeutic strategies.


Assuntos
Quadruplex G , Síndrome de Werner , Animais , Desenvolvimento Ósseo , Proteínas de Ligação a DNA/metabolismo , Genes Homeobox , RecQ Helicases/genética , RecQ Helicases/metabolismo , Síndrome de Werner/genética , Helicase da Síndrome de Werner/genética , Helicase da Síndrome de Werner/metabolismo , Peixe-Zebra/genética
5.
JHEP Rep ; 2(6): 100179, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33134908

RESUMO

BACKGROUND & AIMS: The paradox of hepatic insulin resistance describes the inability for liver to respond to bioenergetics hormones in suppressing gluconeogenesis whilst maintaining lipid synthesis. Here, we report the deficiency of miR-192-3p in the livers of mice with diabetes and its role in alleviating hepatic steatosis. METHODS: As conventional pre-microRNA (miRNA) stem-loop overexpression only boosts guiding strand (i.e. miR-192-5p) expression, we adopted an artificial AAV(DJ)-directed, RNA Pol III promoter-driven miRNA hairpin construct for star-strand-specific overexpression in the liver. Liver steatosis and insulin resistance markers were evaluated in primary hepatocytes, mice with diabetes, and mice with excessive carbohydrate consumption. RESULTS: Functional loss of miR-192-3p in liver exacerbated hepatic micro-vesicular steatosis and insulin resistance in either mice with diabetes or wild-type mice with excessive fructose consumption. Liver-specific overexpression of miR-192-3p effectively halted hepatic steatosis and ameliorated insulin resistance in these mice models. Likewise, hepatocytes overexpressing miR-192-3p exhibited improved lipid accumulation, accompanied with decreases in lipogenesis and lipid-accumulation-related transcripts. Mechanistically, glucocorticoid receptor (GCR, also known as nuclear receptor subfamily 3, group C, member 1 [NR3C1]) was demonstrated to be negatively regulated by miR-192-3p. The effect of miR-192-3p on mitigating micro-vesicular steatosis was ablated by the reactivation of NR3C1. CONCLUSIONS: The star strand miR-192-3p was an undermined glycerolipid regulator involved in controlling fat accumulation and insulin sensitivity in liver through blockade of hepatic GCR signalling; this miRNA may serve as a potential therapeutic option for the common co-mobility of diabetic mellitus and fatty liver disease. LAY SUMMARY: The potential regulatory activity of star strand microRNA (miRNA) species has been substantially underestimated. In this study, we investigate the role and mechanism of an overlooked star strand miRNA (miR-192-3p) in regulating hepatic steatosis and insulin signalling in the livers of mice with diabetes and mice under excessive carbohydrate consumption.

6.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33137927

RESUMO

Parkinson disease (PD) is a neurological movement disorder resulting primarily from damage to and degeneration of the nigrostriatal dopaminergic pathway. The pathway consists of neural populations in the substantia nigra that project to the striatum of the brain where they release dopamine. Diagnosis of PD is based on the presence of impaired motor features such as asymmetric or unilateral resting tremor, bradykinesia, and rigidity. Nonmotor features including cognitive impairment, sleep disorders, and autonomic dysfunction are also present. No cure for PD has been discovered, and treatment strategies focus on symptomatic management through restoration of dopaminergic activity. However, proposed cell replacement therapies are promising because midbrain dopaminergic neurons have been shown to restore dopaminergic neurotransmission and functionally rescue the dopamine-depleted striatum. In this review, we summarize our current understanding of the molecular pathogenesis of neurodegeneration in PD and discuss the development of new therapeutic strategies that have led to the initiation of exploratory clinical trials. We focus on the applications of stem cells for the treatment of PD and discuss how stem cell research has contributed to an understanding of PD, predicted the efficacy of novel neuroprotective therapeutics, and highlighted what we believe to be the critical areas for future research.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Doença de Parkinson/terapia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Animais , Humanos
7.
Aging Cell ; 19(5): e13116, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32320127

RESUMO

WRN mutation causes a premature aging disease called Werner syndrome (WS). However, the mechanism by which WRN loss leads to progeroid features evident with impaired tissue repair and regeneration remains unclear. To determine this mechanism, we performed gene editing in reprogrammed induced pluripotent stem cells (iPSCs) derived from WS fibroblasts. Gene correction restored the expression of WRN. WRN+/+ mesenchymal stem cells (MSCs) exhibited improved pro-angiogenesis. An analysis of paracrine factors revealed that hepatocyte growth factor (HGF) was downregulated in WRN-/- MSCs. HGF insufficiency resulted in poor angiogenesis and cutaneous wound healing. Furthermore, HGF was partially regulated by PI3K/AKT signaling, which was desensitized in WRN-/- MSCs. Consistently, the inhibition of the PI3K/AKT pathway in WRN+/+ MSC resulted in reduced angiogenesis and poor wound healing. Our findings indicate that the impairment in the pro-angiogenic function of WS-MSCs is due to HGF insufficiency and PI3K/AKT dysregulation, suggesting trophic disruption between stromal and epithelial cells as a mechanism for WS pathogenesis.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Patológica/metabolismo , Helicase da Síndrome de Werner/genética , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Senescência Celular , Edição de Genes , Humanos , Células-Tronco Mesenquimais/patologia , Neovascularização Patológica/patologia
8.
Cell Death Dis ; 11(2): 107, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034125

RESUMO

Mouse embryonic stem cells (ESCs) are isolated from the inner cell mass of blastocysts, and they exist in different states of pluripotency-naïve and primed states. Pten is a well-known tumor suppressor. Here, we generated Pten-/- mouse ESCs with the CRISPR-Cas9 system and verified that Pten-/- ESCs maintained naïve pluripotency by blocking Gsk3ß activity. Serum/LIF and 2i (MAPK and GSK3 inhibitors) conditions are commonly used for ESC maintenance. We show that the Pten-inhibitor SF1670 contributed to sustaining mouse ESCs and that Pten activation by the S380A, T382A, and T383A mutations (Pten-A3) suppressed the pluripotency of ESCs. The in vivo teratoma formation ability of SF1670-treated ESCs increased, while the Pten-A3 mutations suppressed teratoma formation. Furthermore, the embryoid bodies derived from Pten-deficient ESCs or SF1670-treated wild-type ESCs showed greater expression of ectoderm and pluripotency markers. These results suggest that Pten-mediated Gsk3ß modulates the naïve pluripotency of ESCs and that Pten ablation regulates the lineage-specific differentiation.


Assuntos
Diferenciação Celular , Linhagem da Célula , Glicogênio Sintase Quinase 3 beta/metabolismo , Células-Tronco Embrionárias Murinas/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Animais , Linhagem Celular , Corpos Embrioides/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Glicogênio Sintase Quinase 3 beta/genética , Camundongos , Camundongos Nus , Mutação , PTEN Fosfo-Hidrolase/genética , Fenótipo , Transdução de Sinais , Teratoma/enzimologia , Teratoma/genética , Teratoma/patologia
9.
Front Oncol ; 9: 486, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31245291

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at a post-transcriptional level. We examined the role of miR-126 in granulosa cell tumor (GCT) of the ovaries. In tissues from malignant GCT patients miR-126 expression was repressed. We showed that miR-126 could inhibit proliferation, migration, hormone production and promote apoptosis of cancerous granulosa cells (GCs) in vitro. The role of miR-126 as "tumor suppressor" was confirmed by using a tumor formation model in vivo. By RNA-seq, immunohistochemical staining (IHC), Western blot and luciferase reporter assay, we identified and confirmed EGFL7 as a direct functional target of miR-126 in cancer GCs. Furthermore, we found that the AKT signaling pathway was associated with miR-126 and EGFL7 in cancer GCs. Taken together, our results demonstrate a function of miR-126 in the suppression of GCT development via the regulation of EGFL7.

10.
Stem Cells Dev ; 28(13): 833-845, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31020917

RESUMO

Doxycycline (DOX), an antibacterial drug, has been widely used in the inducible gene expression system. However, its effect was largely ignored when studying functions of the inducible transgene. By using a DOX-inducible Tet-ON system, we identified that DOX alone dramatically promoted dopaminergic (DA) neuron differentiation from human pluripotent stem cells (hPSCs), whereas the studied gene had no significant effects after considering the confounding factor DOX. These findings suggest that the effect of DOX should be taken into consideration when it is used in the inducible system especially during DA neuron differentiation from hPSCs. Meanwhile, it also suggests that DOX can be used as an efficient and inexpensive molecule to increase DA neuron differentiation efficacy from hPSCs for cell therapy.


Assuntos
Antibacterianos/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Doxiciclina/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese , Linhagem Celular , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Transgenes
11.
Biogerontology ; 20(3): 255-269, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30666569

RESUMO

Aging is a natural and unavoidable part of life. However, aging is also the primary driver of the dominant human diseases, such as cardiovascular disease, cancer, and neurodegenerative diseases, including Alzheimer's disease. Unraveling the sophisticated molecular mechanisms of the human aging process may provide novel strategies to extend 'healthy aging' and the cure of human aging-related diseases. Werner syndrome (WS), is a heritable human premature aging disease caused by mutations in the gene encoding the Werner (WRN) DNA helicase. As a classical premature aging disease, etiological exploration of WS can shed light on the mechanisms of normal human aging and facilitate the development of interventional strategies to improve healthspan. Here, we summarize the latest progress of the molecular understandings of WRN protein, highlight the advantages of using different WS model systems, including Caenorhabditis elegans, Drosophila melanogaster and induced pluripotent stem cell (iPSC) systems. Further studies on WS will propel drug development for WS patients, and possibly also for normal age-related diseases.


Assuntos
Envelhecimento/patologia , Síndrome de Werner/patologia , Animais , Caenorhabditis elegans/fisiologia , Drosophila melanogaster/fisiologia , Humanos , Modelos Biológicos , Mutação , Síndrome de Werner/genética , Síndrome de Werner/terapia
12.
Curr Drug Targets ; 20(1): 16-28, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30156157

RESUMO

In previous studies, oxidative stress damage has been solely considered to be the mechanism of ovarian aging, and several antioxidants have been used to delay ovarian aging. But recently, more reports have found that endoplasmic reticulum stress, autophagy, sirtuins, mitochondrial dysfunction, telomeres, gene mutation, premature ovarian failure, and polycystic ovary syndrome are all closely related to ovarian aging, and these factors all interact with oxidative stress. These novel insights on ovarian aging are summarized in this review. Furthermore, as a pleiotropic molecule, melatonin is an important antioxidant and used as drugs for several diseases treatment. Melatonin regulates not only oxidative stress, but also the various molecules, and normal and pathological processes interact with ovarian functions and aging. Hence, the mechanism of ovarian aging and the extensive role of melatonin in the ovarian aging process are described herein. This systematic review supply new insights into ovarian aging and the use of melatonin to delay its onset, further supply a novel drug of melatonin for ovarian aging treatment.


Assuntos
Envelhecimento/efeitos dos fármacos , Melatonina/antagonistas & inibidores , Ovário/efeitos dos fármacos , Envelhecimento/metabolismo , Autofagia/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Melatonina/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ovário/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Sirtuínas/metabolismo
13.
Cell Death Dis ; 9(11): 1076, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348959

RESUMO

We have previously reported that microRNA-10 family could disturb normal development of granulosa cells (GC) during follicle formation. In the current study, the effect of miR-10a on granulosa cell tumor (GCT), a subtype of ovarian cancer, was examined. Strong miR-10a signal was detected in tissues from malignant GCT patients. Forced expression of miR-10a significantly promoted cell proliferation, migration, invasion, ovarian hormone production, and repressed anticancer drug-induced apoptosis in vitro. The oncogenic role of miR-10a was further validated in an orthotopic GCT model in vivo. In addition, by using CRISPR-Cas9 system, the aggressive phenotype was repressed in miR-10a knockout cancer GC. By using a heterotopic mice model, the oncogenic role of miR-10a was confirmed in vivo. RNA-seq, FISH, western blot, luciferase reporter assay were used to identified PTEN, a well-known anti-GCT gene, as direct functional target of miR-10a in cancer GC; Akt and Wnt were also found as two associated oncogenic pathways of miR-10a in cancer GC. Taken together, our results demonstrate that the miR-10a could promote GCT development via synergistically regulating PTEN, Akt, and Wnt pathways.


Assuntos
Tumor de Células da Granulosa/genética , Células da Granulosa/patologia , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Proto-Oncogênicas c-akt/genética , Via de Sinalização Wnt/genética , Animais , Apoptose/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/genética , Transdução de Sinais/genética
14.
Sci Rep ; 8(1): 9630, 2018 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-29941943

RESUMO

MicroRNAs (miRNAs) are known to be crucial players in governing the differentiation of human induced pluripotent stem cells (hiPSCs). Despite their utter importance, identifying key lineage specifiers among the myriads of expressed miRNAs remains challenging. We believe that the current practice in mining miRNA specifiers via delineating dynamic fold-changes only is inadequate. Our study, therefore, provides evidence to pronounce "lineage specificity" as another important attribute to qualify for these lineage specifiers. Adopted hiPSCs were differentiated into representative lineages (hepatic, nephric and neuronal) over all three germ layers whilst the depicted miRNA expression changes compiled into an integrated atlas. We demonstrated inter-lineage analysis shall aid in the identification of key miRNAs with lineage-specificity, while these shortlisted candidates were collectively known as "lineage-specific miRNAs". Subsequently, we followed through the fold-changes along differentiation via computational analysis to identify miR-192 and miR-372-3p, respectively, as representative candidate key miRNAs for the hepatic and nephric lineages. Indeed, functional characterization validated that miR-192 and miR-372-3p regulate lineage differentiation via modulation of the expressions of lineage-specific genes. In summary, our presented miRNA atlas is a resourceful ore for the mining of key miRNAs responsible for lineage specification.


Assuntos
Diferenciação Celular , Linhagem da Célula , Células-Tronco Pluripotentes Induzidas/citologia , MicroRNAs/genética , Mineração de Dados , Regulação da Expressão Gênica , Hepatócitos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/citologia , Células-Tronco Neurais/citologia
15.
FEBS Open Bio ; 8(4): 502-512, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29632804

RESUMO

Induced pluripotent stem cells (iPSCs) are useful for modeling neuron development and related diseases. Cortical interneurons are essential players in neuropsychiatric diseases such as autism. miRNAs are a class of pivotal regulators in neural differentiation. Using a previously established model of cortical interneuron differentiation from human embryonic stem cells, we profiled miRNAs involved in differentiation from human iPSCs. A number of miRNAs were modulated in the differentiation process. This study captured the temporal in vitro neurogenesis from iPSCs to mature cortical interneurons. The specific miRNAs identified at each stage of differentiation are of potential use for drug discovery and prospective clinical applications.

16.
Stem Cell Res Ther ; 9(1): 71, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29562912

RESUMO

BACKGROUND: The regulatory role of long noncoding RNAs (lncRNAs) have been partially proved in embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). METHODS: In the current study, we investigated mouse ESC (mESC) self-renewal, differentiation, and proliferation in vitro by knocking down a lncRNA, growth arrest specific 5 (Gas5). A series of related indicators were examined by cell counting kit-8 (CCK-8) assay, quantitative reverse-transcription polymerase chain reaction (qRT-PCR), Western blot, alkaline phosphatase staining, propidium iodide (PI) staining, Annexin V staining, competition growth assay, immunofluorescence, and chromatin immunoprecipitation (ChIP)-qPCR. An in vivo teratoma formation assay was also performed to validate the in vitro results. qRT-PCR, fluorescence-activated cell sorting (FACS), alkaline phosphatase staining, and immunofluorescence were used to evaluate the role of Gas5 during mouse iPSC reprogramming. The regulatory axis of Dicer-miR291a-cMyc-Gas5 and the relationship between Gas5 and Tet/5hmC in mESCs was examined by qRT-PCR, Dot blot, and Western blot. RESULTS: We identified that Gas5 was required for self-renewal and pluripotency of mESCs and iPSCs. Gas5 formed a positive feedback network with a group of key pluripotent modulators (Sox2, Oct4, Nanog, Tcl1, Esrrb, and Tet1) in mESCs. Knockdown of Gas5 promoted endodermal differentiation of mESCs and impaired the efficiency of iPSC reprogramming. In addition, Gas5 was regulated by the Dicer-miR291a-cMyc axis and was involved in the DNA demethylation process in mESCs. CONCLUSIONS: Taken together, our results suggest that the lncRNA Gas5 plays an important role in modulating self-renewal and pluripotency of mESCs as well as iPSC reprogramming.


Assuntos
Reprogramação Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , RNA Longo não Codificante/metabolismo
17.
Mol Neurobiol ; 54(6): 4507-4523, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27356918

RESUMO

Autism spectrum disorder is a complex neurodevelopmental disorder whose pathophysiology remains elusive as a consequence of the unavailability for study of patient brain neurons; this deficit may potentially be circumvented by neural differentiation of induced pluripotent stem cells. Rare syndromes with single gene mutations and autistic symptoms have significantly advanced the molecular and cellular understanding of autism spectrum disorders; however, in aggregate, they only represent a fraction of all cases of autism. In an effort to define the cellular and molecular phenotypes in human neurons of non-syndromic autism, we generated induced pluripotent stem cells (iPSCs) from three male autism spectrum disorder patients who had no identifiable clinical syndromes, and their unaffected male siblings and subsequently differentiated these patient-specific stem cells into electrophysiologically active neurons. iPSC-derived neurons from these autistic patients displayed decreases in the frequency and kinetics of spontaneous excitatory postsynaptic currents relative to controls, as well as significant decreases in Na+ and inactivating K+ voltage-gated currents. Moreover, whole-genome microarray analysis of gene expression identified 161 unique genes that were significantly differentially expressed in autistic patient iPSC-derived neurons (>twofold, FDR < 0.05). These genes were significantly enriched for processes related to synaptic transmission, such as neuroactive ligand-receptor signaling and extracellular matrix interactions, and were enriched for genes previously associated with autism spectrum disorder. Our data demonstrate aberrant voltage-gated currents and underlying molecular changes related to synaptic function in iPSC-derived neurons from individuals with idiopathic autism as compared to unaffected siblings controls.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Adolescente , Diferenciação Celular , Linhagem Celular , Criança , Potenciais Pós-Sinápticos Excitadores , Perfilação da Expressão Gênica , Ontologia Genética , Humanos , Ativação do Canal Iônico , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Canais de Potássio/metabolismo , Canais de Sódio/metabolismo
18.
Oncotarget ; 7(33): 54010-54027, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27304053

RESUMO

The unfolded protein response (UPR) is an evolutionarily conserved adaptive mechanism to increase cell survival under endoplasmic reticulum (ER) stress conditions. The UPR is critical for maintaining cell homeostasis under physiological and pathological conditions. The vital functions of the UPR in development, metabolism and immunity have been demonstrated in several cell types. UPR dysfunction activates a variety of pathologies, including cancer, inflammation, neurodegenerative disease, metabolic disease and immune disease. Stem cells with the special ability to self-renew and differentiate into various somatic cells have been demonstrated to be present in multiple tissues. These cells are involved in development, tissue renewal and certain disease processes. Although the role and regulation of the UPR in somatic cells has been widely reported, the function of the UPR in stem cells is not fully known, and the roles and functions of the UPR are dependent on the stem cell type. Therefore, in this article, the potential significances of the UPR in stem cells, including embryonic stem cells, tissue stem cells, cancer stem cells and induced pluripotent cells, are comprehensively reviewed. This review aims to provide novel insights regarding the mechanisms associated with stem cell differentiation and cancer pathology.


Assuntos
Células-Tronco , Resposta a Proteínas não Dobradas , Animais , Humanos
19.
Biol Reprod ; 94(6): 137, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26911431

RESUMO

Ovarian cryopreservation by vitrification is a highly useful method for preserving female fertility during radiotherapy and chemotherapy. However, cryoinjury, osmotic stress during vitrification, and ischemia/reperfusion during transplantation lead to loss of ovarian follicles. Ovarian follicle loss may be partially reduced by several methods; however, studies regarding the mechanism of ovarian follicle loss have only investigated cell apoptosis, which consists of type I programmed cell death. Autophagy is type II programmed cell death, and cell homeostasis is maintained by autophagy during conditions of stress. The role of autophagy during cryopreservation by vitrification has rarely been reported. The potential role of autophagy during ovarian cryopreservation by vitrification is reviewed in this article.


Assuntos
Autofagia , Criopreservação , Ovário , Vitrificação , Feminino , Humanos , Ovário/transplante
20.
Br J Cancer ; 114(2): 230-6, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26625006

RESUMO

BACKGROUND: Testicular embryonal carcinoma (EC) is a major subtype of non-seminomatous germ cell tumours in males. Embryonal carcinomas are pluripotent, undifferentiated germ cell tumours believed to originate from primordial germ cells. Epigenetic changes during testicular EC tumorigenesis require better elucidation. METHODS: To identify epigenetic changes during testicular neoplastic transformation, we profiled DNA methylation of six ECs. These samples represent different stages (stage I and stage III) of divergent invasiveness. Non-cancerous testicular tissues were included. Expression of a number of hypermethylated genes were examined by quantitative RT-PCR and immunohistochemistry (IHC). RESULTS: A total of 1167 tumour-hypermethylated differentially methylated regions (DMRs) were identified across the genome. Among them, 40 genes/ncRNAs were found to have hypermethylated promoters. Quantitative RT-PCR confirmed downregulation of 8 out of 9 of the genes. Among the confirmed genes, five were sex-linked genes, including X-linked genes STAG2, SPANXD/E and MIR1184, and Y-linked genes RBMY1A1/1B/1D and FAM197Y2P. RBMY1A is a testis-specific gene for spermatogenesis. RNF168 and USP13 are potential tumour suppressors. Expression of RBMY1A was lost in EC and seminoma as documented in the Protein Atlas. We confirmed downregulation of USP13 in EC by IHC. CONCLUSIONS: Our genome-wide analysis of testicular EC identified methylation changes in several previously unknown genes. This may provide insight of crosstalk between normal germ cell development and carcinogenesis.


Assuntos
Carcinoma Embrionário/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Neoplasias Testiculares/genética , Aciltransferases/genética , Adolescente , Adulto , Carcinoma Embrionário/patologia , Estudos de Casos e Controles , Estudos de Coortes , Endopeptidases/genética , Epigênese Genética , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Testiculares/patologia , Análise Serial de Tecidos , Ubiquitina-Proteína Ligases/genética , Proteases Específicas de Ubiquitina , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...