Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 103: 105093, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38569318

RESUMO

BACKGROUND: Human restricted genes contribute to human specific traits in the immune system. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR), the highest Ca2+ conductor of the ACh receptors implicated in innate immunity. Understanding the mechanism of how CHRFAM7A affects the immune system remains unexplored. METHODS: Two model systems are used, human induced pluripotent stem cells (iPSC) and human primary monocytes, to characterize α7 nAChR function, Ca2+ dynamics and decoders to elucidate the pathway from receptor to phenotype. FINDINGS: CHRFAM7A/α7 nAChR is identified as a hypomorphic receptor with mitigated Ca2+ influx and prolonged channel closed state. This shifts the Ca2+ reservoir from the extracellular space to the endoplasmic reticulum (ER) leading to Ca2+ dynamic changes. Ca2+ decoder small GTPase Rac1 is then activated, reorganizing the actin cytoskeleton. Observed actin mediated phenotypes include cellular adhesion, motility, phagocytosis and tissue mechanosensation. INTERPRETATION: CHRFAM7A introduces an additional, human specific, layer to Ca2+ regulation leading to an innate immune gain of function. Through the actin cytoskeleton it drives adaptation to the mechanical properties of the tissue environment leading to an ability to invade previously immune restricted niches. Human genetic diversity predicts profound translational significance as its understanding builds the foundation for successful treatments for infectious diseases, sepsis, and cancer metastasis. FUNDING: This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti) and in part by NIH grant R01HL163168 (Yongho Bae).


Assuntos
Citoesqueleto de Actina , Sinalização do Cálcio , Células-Tronco Pluripotentes Induzidas , Receptor Nicotínico de Acetilcolina alfa7 , Humanos , Citoesqueleto de Actina/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Cálcio/metabolismo , Monócitos/metabolismo , Imunidade Inata , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Fagocitose
2.
Mol Psychiatry ; 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38200291

RESUMO

Genes restricted to humans may contribute to human-specific traits and provide a different context for diseases. CHRFAM7A is a uniquely human fusion gene and a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR). The α7 nAChR has been a promising target for diseases affecting cognition and higher cortical functions, however, the treatment effect observed in animal models failed to translate into human clinical trials. As CHRFAM7A was not accounted for in preclinical drug screens it may have contributed to the translational gap. Understanding the complex genetic architecture of the locus, deciphering the functional impact of CHRFAM7A on α7 nAChR neurobiology and utilizing human-relevant models may offer novel approaches to explore α7 nAChR as a drug target.

3.
EBioMedicine ; 95: 104725, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37517100

RESUMO

BACKGROUND: While advancements in imaging techniques have led to major strides in deciphering the human brain, successful interventions are elusive and represent some of the most persistent translational gaps in medicine. Human restricted CHRFAM7A has been associated with neuropsychiatric disorders. METHODS: The physiological role of CHRFAM7A in human brain is explored using multiomics approach on 600 post mortem human brain tissue samples. The emerging pathways and mechanistic hypotheses are tested and validated in an isogenic hiPSC model of CHRFAM7A knock-in medial ganglionic eminence progenitors and neurons. FINDINGS: CHRFAM7A is identified as a modulator of intracellular calcium dynamics and an upstream regulator of Rac1. Rac1 activation re-designs the actin cytoskeleton leading to dynamic actin driven remodeling of membrane protrusion and a switch from filopodia to lamellipodia. The reinforced cytoskeleton leads to an advantage to tolerate stiffer mechanical properties of the extracellular environment. INTERPRETATION: CHRFAM7A modifies the actin cytoskeleton to a more dynamic and stiffness resistant state in an α7nAChR dependent manner. CHRFAM7A may facilitate neuronal adaptation to changes in the brain environment in physiological and pathological conditions contributing to risk or recovery. Understanding how CHRFAM7A affects human brain requires human studies in the areas of memory formation and erasure, cognitive reserve, and neuronal plasticity. FUNDING: This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti). Also, in part by the International Society for Neurochemistry (ISN) and The Company of Biologists (Nicolas Rosas). ROSMAP is supported by NIA grants P30AG10161, P30AG72975, R01AG15819, R01AG17917. U01AG46152, and U01AG61356.


Assuntos
Encéfalo , Mutação com Ganho de Função , Humanos , Encéfalo/metabolismo , Neurônios/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo
4.
Int J Mol Sci ; 21(24)2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33352944

RESUMO

Neuroinflammation in Alzheimer's disease (AD) has been the focus for identifying targetable pathways for drug development. The role of amyloid beta (Aß), a prototype of damage-associated molecular patterns (DAMPs), has been implicated in triggering an inflammatory response. As alpha7 nicotinic acetylcholine receptor (α7 nAChR) binds Aß with high affinity, α7 nAChR may play a role in Aß-induced neuroinflammation. The conundrum of how α7 nAChR as the mediator of the cholinergic anti-inflammatory response may trigger an inflammatory response has not been resolved. CHRFAM7A, the uniquely human fusion gene between ULK4 and CHRNA7, is a negative regulator of α7 nAChR ionotropic function. To provide the human context, isogenic induced pluripotent stem cell (iPSC) lines were developed from CHRFAM7A null and carrier individuals by genome-editing the null line using TALENs to knock-in CHRFAM7A. In iPSC-derived microglia-like cells, CHRFAM7A mitigated Aß uptake through the α7 nAChR. Despite the lower Aß uptake, the presence of CHRFAM7A was associated with an innate immune response that was characterized by NF-κB activation and NF-κB target transcription (TNFA, IL6, and IL1B). LPS, a prototype PAMP, induced a heightened immune response in CHRFAM7A carriers. CHRFAM7A modified the dynamics of NF-κB translocation by prolonging its nuclear presence. CHRFAM7A modified the α7 nAChR metabotropic function, resulting in a human-specific innate immune response. This iPSC model provided an opportunity to elucidate the mechanism and establish high throughput screens.


Assuntos
Alarminas/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Microglia/imunologia , Microglia/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Doença de Alzheimer/patologia , Movimento Celular , Suscetibilidade a Doenças , Expressão Gênica , Humanos , Imunidade Inata , Vigilância Imunológica , Microglia/citologia , NF-kappa B/metabolismo , Transdução de Sinais , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
5.
EBioMedicine ; 59: 102892, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32818803

RESUMO

BACKGROUND: Cholinergic neuronal loss is one of the hallmarks of AD related neurodegeneration; however, preclinical promise of α7 nAChR drugs failed to translate into humans. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of α7 nAChR and was unaccounted for in preclinical models. METHODS: Molecular methods: Function of CHRFAM7A alleles was studied in vitro in two disease relevant phenotypic readouts: electrophysiology and Aß uptake. Genome edited human induced pluripotent stem cells (iPSC) were used as a model system with the human context. Double blind pharmacogenetic study: We performed double-blind pharmacogenetic analysis on the effect of AChEI therapy based on CHRFAM7A carrier status in two paradigms: response to drug initiation and DMT effect. Mini Mental Status Examination (MMSE) was used as outcome measure. Change in MMSE score from baseline was compared by 2-tailed T-test. Longitudinal analysis of clinical outcome (MMSE) was performed using a fitted general linear model, based on an assumed autoregressive covariance structure. Model independent variables included age, sex, and medication regimen at the time of the first utilized outcome measure (AChEI alone or AChEI plus memantine), APOE4 carrier status (0, 1 or 2 alleles as categorical variables) and CHRFAM7A genotype. FINDINGS: The direct and inverted alleles have distinct phenotypes. Functional CHRFAM7A allele classifies the population as 25% non-carriers and 75% carriers. Induced pluripotent stem cell (iPSC) models α7 nAChR mediated Aß neurotoxicity. Pharmacological readout translates into both first exposure (p = 0.037) and disease modifying effect (p = 0.0048) in two double blind pharmacogenetic studies. INTERPRETATION: CHRFAM7A accounts for the translational gap in cholinergic strategies in AD. Clinical trials not accounting for this uniquely human genetic factor may have rejected drug candidates that would benefit 25% of AD. Reanalyses of the completed trials using this pharmacogenetic paradigm may identify effective therapy.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Neurônios Colinérgicos/metabolismo , Proteínas Recombinantes de Fusão , Receptor Nicotínico de Acetilcolina alfa7/genética , Alelos , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Biomarcadores , Linhagem Celular , Antagonistas Colinérgicos/farmacologia , Antagonistas Colinérgicos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Imunofluorescência , Dosagem de Genes , Frequência do Gene , Genótipo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fenótipo , Transmissão Sináptica , Pesquisa Translacional Biomédica , Resultado do Tratamento , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
6.
Transl Psychiatry ; 9(1): 59, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30710073

RESUMO

The α7 nicotinic acetylcholine receptor (α7nAChR) has been a promising target for diseases affecting cognition and higher cortical functions; however, the effect observed in animal models failed to translate into human clinical trials identifying a translational gap. CHRFAM7A is a human-specific fusion gene with properties that enable incorporation into the α7nAChR and, being human specific, CHRFAM7A effect was not accounted for in preclinical studies. We hypothesized that CHRFAM7A may account for this translational gap and understanding its function may offer novel insights when exploring α7nAChR as a drug target. CHRFAM7A is present in different copy number variations (CNV) in the human genome with high frequency. To study the functional consequences of the presence of the CHRFAM7A, two induced pluripotent stem cell (iPSC) lines (0 copy and 1 copy direct) were developed. The 0 copy line was rescued with CHRFAM7A transfection to control for genetic heterogeneity. As readouts for genotype-phenotype correlation, α7nAChR synaptic transmission and amyloid beta 1-42 (Aß1-42) uptake were tested. Synaptic transmission in the presence of CHRFAM7A demonstrated that PNU-modulated desensitization of α7nAChR currents increased as a function of CHRFAM7A dosage. CHRFAM7A mitigated the dose response of Aß1-42 uptake suggesting a protective effect beyond physiological concentrations. Furthermore, in the presence of CHRFAM7A Aß1-42 uptake activated neuronal interleukin 1ß (IL-1ß) and tumor necrosis factor α (TNF-α) without activating the canonical inflammasome pathway. Lead optimization may identify more potent molecules when the screen has a model harboring CHRFAM7A. Incorporating pharmacogenetics into clinical trials may enhance signals in efficacy measures.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Diferenciação Celular , Células Cultivadas , Expressão Gênica , Células HEK293 , Humanos , Inflamação/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/metabolismo , Transmissão Sináptica
7.
Stem Cells Int ; 2018: 3983090, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30050576

RESUMO

In vitro differentiation of human pluripotent stem cell into relevant cell types is a desirable model system that has the human biological context, is a renewable source, and is scalable. GABA interneurons and basal forebrain cholinergic neurons, derivates of the medial ganglionic eminence (MGE), are implicated in diverse neuropsychiatric diseases. Various protocols have been proposed to generate MGE progenitors: the embryoid body- (EB-) based rosette-derived (RD), the adherent (AdD), and the nonadherent (NAdD) approaches. While Wnt inhibition is frequently incorporated into the strategy, the timing varies between protocols and there is a lack of standardized outcome reporting, which precludes direct comparison. Here, we report a head-to-head comparison in three distinct experimental models to establish whether Wnt inhibition during neural stem cell, NSC (stage 1), or neural progenitor cell, NPC (stage 2), formation facilitates MGE differentiation. Wnt inhibition at both stages promotes MGE progenitor differentiation when compared to no inhibition. However, NSC (stage 1) Wnt inhibition markedly reduces the number of MGE progenitors available for downstream applications in the RD and the NAdD protocols due to early inhibition of proliferation. NPC (stage 2) Wnt inhibition in the adherent system is comparable to the EB-based methods offering a techically less challenging alternative.

8.
PLoS One ; 9(9): e108609, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25259793

RESUMO

Myosin IC is a single headed member of the myosin superfamily. We recently identified a novel isoform and showed that the MYOIC gene in mammalian cells encodes three isoforms (isoforms A, B, and C). Furthermore, we demonstrated that myosin IC isoform A but not isoform B exhibits a tissue specific expression pattern. In this study, we extended our analysis of myosin IC isoform expression patterns by analyzing the protein and mRNA expression in various mammalian cell lines and in various prostate specimens and tumor tissues from the transgenic mouse prostate (TRAMP) model by immunoblotting, qRT-PCR, and by indirect immunohistochemical staining of paraffin embedded prostate specimen. Analysis of a panel of mammalian cell lines showed an increased mRNA and protein expression of specifically myosin IC isoform A in a panel of human and mouse prostate cancer cell lines but not in non-cancer prostate or other (non-prostate-) cancer cell lines. Furthermore, we demonstrate that myosin IC isoform A expression is significantly increased in TRAMP mouse prostate samples with prostatic intraepithelial neoplasia (PIN) lesions and in distant site metastases in lung and liver when compared to matched normal tissues. Our observations demonstrate specific changes in the expression of myosin IC isoform A that are concurrent with the occurrence of prostate cancer in the TRAMP mouse prostate cancer model that closely mimics clinical prostate cancer. These data suggest that elevated levels of myosin IC isoform A may be a potential marker for the detection of prostate cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Miosina Tipo I/genética , Próstata/metabolismo , Neoplasia Prostática Intraepitelial/genética , Neoplasias da Próstata/genética , Isoformas de Proteínas/genética , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Miosina Tipo I/metabolismo , Próstata/patologia , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/metabolismo
9.
BMC Cell Biol ; 15: 8, 2014 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-24618359

RESUMO

BACKGROUND: Myosin IC is a single headed member of the myosin superfamily that localizes to the cytoplasm and the nucleus and is implicated in a variety of processes in both compartments. We recently identified a novel isoform of myosin IC and showed that the MYOIC gene in mammalian cells encodes three isoforms (isoforms A, B, and C) that differ only in the addition of short isoform-specific N-terminal peptides. The expression pattern of the isoforms and the mechanisms of expression regulation remain unknown. RESULTS: To determine the expression patterns of myosin IC isoforms, we performed a comprehensive expression analysis of the two myosin IC isoforms (isoform A and B) that contain isoform-specific sequences. By immunoblotting with isoform-specific antibodies and by qRT-PCR with isoform-specific primer we demonstrate that myosin IC isoforms A and B have distinct expression patterns in mouse tissues. Specifically, we show that myosin IC isoform A is expressed in a tissue specific pattern, while myosin IC isoform B is ubiquitously expressed at comparable levels in mouse tissues. CONCLUSIONS: The differences in the expression profile of the myosin IC isoforms indicate a tissue-specific MYOIC gene regulation and further suggest that the myosin IC isoforms, despite their high sequence homology, might have tissue-specific and isoform-specific functions.


Assuntos
Regulação da Expressão Gênica , Miosinas/genética , Miosinas/metabolismo , Animais , Western Blotting , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
10.
Exp Cell Res ; 319(8): 1111-23, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23438938

RESUMO

Myosin IC is a single headed member of the myosin superfamily that localizes to the cytoplasm and the nucleus, where it is involved in transcription by RNA polymerases I and II, intranuclear transport, and nuclear export. In mammalian cells, three isoforms of myosin IC are expressed that differ only in the addition of short isoform-specific N-terminal peptides. Despite the high sequence homology, the isoforms show differences in cellular distribution, in localization to nuclear substructures, and in their interaction with nuclear proteins through yet unknown mechanisms. In this study, we used EGFP-fusion constructs that express truncated or mutated versions of myosin IC isoforms to detect regions that are involved in isoform-specific localization. We identified two nucleolar localization signals (NoLS). One NoLS is located in the myosin IC isoform B specific N-terminal peptide, the second NoLS is located upstream of the neck region within the head domain. We demonstrate that both NoLS are functional and necessary for nucleolar localization of specifically myosin IC isoform B. Our data provide a first mechanistic explanation for the observed functional differences between the myosin IC isoforms and are an important step toward our understanding of the underlying mechanisms that regulate the various and distinct functions of myosin IC isoforms.


Assuntos
Nucléolo Celular/metabolismo , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Sinais Direcionadores de Proteínas/genética , Animais , Células COS , Chlorocebus aethiops , Clonagem Molecular , Humanos , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Miosina Tipo I/química , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/genética , Especificidade por Substrato , Transfecção
11.
Cytoskeleton (Hoboken) ; 69(8): 555-65, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22736583

RESUMO

In vertebrates, two myosin Ic isoforms that localize to the cytoplasm and to the nucleus have been characterized. The isoform that predominantly localizes to the nucleus is called nuclear myosin I (NMI). NMI has been identified as a key factor involved in nuclear processes such as transcription by RNA polymerases I and II and intranuclear transport processes. We report here the identification of a previously uncharacterized third MYOIC gene product that is called isoform A. Similar to NMI, this isoform contains a unique N-terminal peptide sequence, localizes to the nucleus and colocalizes with RNA polymerase II. However, unlike NMI, upon exposure to inhibitors of RNA polymerase II transcription the newly identified isoform translocates to nuclear speckles. Furthermore, in contrast to NMI, this new isoform is absent from nucleoli and does not colocalize with RNA polymerase I. Our results suggest an unexpected diversity among nuclear myosin Ic isoforms in respect to their intranuclear localization and interaction with nuclear binding partners that could provide new insights into the regulation of myosin-dependent nuclear processes.


Assuntos
Núcleo Celular/metabolismo , Miosina Tipo I/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , Camundongos , Dados de Sequência Molecular , Miosina Tipo I/química , Células NIH 3T3 , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Transporte Proteico , RNA Polimerase I/metabolismo , RNA Polimerase II/antagonistas & inibidores , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Transcrição Gênica
12.
Reprod Sci ; 18(10): 1014-24, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21693774

RESUMO

Regulation of the actin cytoskeleton is essential for epithelial cell polarity and protein trafficking within human uterine epithelium. The actin-binding protein cofilin is involved in regulation of actin dynamics by promoting actin branching and cytoskeleton reorganization. Dual immunohistochemical staining of cofilin and G-actin (represented by DNAse I staining) revealed cofilin-G-actin colocalization in the apical side of luminal epithelial cells of human uterine endometrium during the proliferative phase of the menstrual cycle. Interestingly, during the secretory phase of the menstrual cycle, cofilin was only present on the basolateral side. To determine whether the disease endometriosis causes a different pattern of actin remodeling, we investigated an established baboon model of induced endometriosis. The cofilin pattern in the secretory phase of baboons with endometriosis was similar to the proliferative phase in normal animals; cofilin was observed in the apical parts of luminal and glandular epithelium. A phosphatase regulating the activity of cofilin, slingshot (SSH1), revealed a similar staining pattern within these tissues. These patterns were confirmed through quantitative image analysis. Quantification of messenger RNA (mRNA) detected upregulated SSH1 and suggested a progesterone resistance-related pattern of nuclear steroid hormone receptors, but no change in membrane progesterone receptors (mPR alpha or mPR beta) was observed in endometriosis. Our data indicate that the severe dyssynchrony during menstrual cycle phases in endometriosis is connected with improper cytoskeleton rearrangements. We suggest that cofilin-mediated actin reorganization in uterine epithelial cells might be important in preparation for blastocyst implantation; dysregulation of this reorganization may lead to decreased fertility in endometriosis.


Assuntos
Actinas/metabolismo , Cofilina 1/metabolismo , Citoesqueleto/metabolismo , Endometriose/metabolismo , Ciclo Menstrual/fisiologia , Animais , Citoesqueleto/patologia , Feminino , Imuno-Histoquímica , Papio
13.
Biol Reprod ; 82(6): 1112-8, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20164440

RESUMO

Transcriptional coactivator with PDZ-binding motif (TAZ) is known to bind to a variety of transcription factors to control cell differentiation and organ development. However, its role in uterine physiology has not yet been described. To study its regulation during the unique process of differentiation of fibroblasts into decidual cells (decidualization), we utilized the human uterine fibroblast (HuF) in vitro cell model. Immunocytochemistry data demonstrated that the majority of the TAZ protein is localized in the nucleus. Treatment of HuF cells with the embryonic stimulus cytokine interleukin 1 beta in the presence of steroid hormones (estradiol-17 beta and medroxyprogesterone acetate) for 13 days did not cause any apparent TAZ mRNA changes but resulted in a significant TAZ protein decline (approximately 62%) in total cell lysates. Analysis of cytosolic and nuclear extracts revealed that the decline of total TAZ was caused primarily by a drop of TAZ protein levels in the nucleus. TAZ was localized on the peroxisome proliferator-activated receptor response element site (located at position -1200 bp relative to the transcription start site) of the genomic region of decidualization marker insulin-like growth factor-binding protein 1 (IGFBP1) in HuF cells as detected by chromatin immunoprecipitation. TAZ is also present in human endometrium tissue as confirmed by immunohistochemistry. During the secretory phase of the menstrual cycle, specific TAZ staining particularly diminishes in the stroma, suggesting its participation during the decidualization process, as well as implantation. During early baboon pregnancy, TAZ protein expression remains minimal in the endometrium close to the implantation site. In summary, the presented evidence shows for the first time to date TAZ protein in the human uterine tract, its downregulation during in vitro decidualization, and its localization on the IGFBP1 promoter region, all of which indicate its presence in the uterine differentiation program during pregnancy.


Assuntos
Diferenciação Celular , Decídua/citologia , Decídua/crescimento & desenvolvimento , Fibroblastos/citologia , Domínios PDZ , Fatores de Transcrição/metabolismo , Aciltransferases , Animais , Decídua/efeitos dos fármacos , Regulação para Baixo , Implantação do Embrião , Estradiol/administração & dosagem , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Interleucina-1beta/administração & dosagem , Medroxiprogesterona/administração & dosagem , Papio , Receptores Ativados por Proliferador de Peroxissomo/genética , Gravidez , Fatores de Transcrição/análise , Fatores de Transcrição/genética
14.
Biol Reprod ; 81(1): 222-30, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19339710

RESUMO

The differentiation of uterine stromal fibroblasts into decidual cells is critical for establishing pregnancy. This process, called decidualization, requires the reorganization of the actin cytoskeleton, which mainly depends on actin dynamics and the phosphorylation status of the myosin light chain. We manipulated actin dynamics with jasplakinolide (100 nM) and latrunculin B (1 microM), both of which significantly inhibited the synthesis of decidualization markers induced by 6 days of treatment with embryo-mimicking stimulus interleukin 1beta (IL1B) and steroid hormones (SHs; 17beta-estradiol and medroxyprogesterone acetate) in the human uterine fibroblast (HuF) in vitro model. However, only jasplakinolide had long-lasting effects on the G-actin:F-actin ratio and prevented decidualization induced by the artificial stimulus cAMP (and SHs). Actin-binding protein cofilin mainly colocalized with G-actin in the nucleus as well as the cytoplasm. Only some spots of colocalization between cofilin and F-actin were detected in the cytoplasm. Brief extraction of cytosolic proteins from living cells revealed that in cells treated with IL1B or cAMP (and SHs) for 6 days, cofilin was mainly detected in the nucleus. The translocation of cofilin from cytosol to nucleus was also detected in HuFs treated for 12 days with SHs, IL1B and SHs, and cAMP and SHs. The same significant translocation was confirmed in primary baboon stromal uterine fibroblasts. We conclude that changes in actin dynamics, particularly the stabilization of F-actin, have a significant negative impact on decidualization, and the translocation of cofilin to the nucleus is a key feature of this process in the primate.


Assuntos
Actinas/metabolismo , Decídua/metabolismo , Decídua/fisiologia , Animais , Células Cultivadas , Cofilina 1/metabolismo , AMP Cíclico/farmacologia , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/fisiologia , Feminino , Hormônios Esteroides Gonadais/farmacologia , Humanos , Interleucina-1beta/farmacologia , Cinética , Papio , Gravidez , Multimerização Proteica/efeitos dos fármacos , Distribuição Tecidual/efeitos dos fármacos
15.
Cell Tissue Res ; 332(3): 479-88, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18401596

RESUMO

Human uterine fibroblasts (HuF) isolated from the maternal part (decidua parietalis) of a term placenta provide a useful model of in vitro cell differentiation into decidual cells (decidualization, a critical process for successful pregnancy). After isolation, the cells adhere to plastic and have either a small round or spindle-shaped morphology that later changes into a flattened pattern in culture. HuF robustly proliferate in culture until passage 20 and form colonies when plated at low densities. The cells express the mesenchymal cell markers fibronectin, integrin-beta1, ICAM-1 (CD54), and collagen I. Flow cytometry of HuF has detected the presence of CD34, a marker of the hematopoietic stem cell lineage, and an absence of CD10, CD11b/Mac, CD14, CD45, and HLA type II. Furthermore, they also express the pluripotency markers SSEA-1, SSEA-4, Oct-4, Stro-1, and TRA-1-81 as detected by confocal microscopy. Treatment for 14-21 days with differentiation-inducing media leads to the differentiation of HuF into osteoblasts, adipocytes, and chondrocytes. The presence of alpha-smooth muscle actin, calponin, and myosin light-chain kinase in cultured HuF implies their similarity to myofibroblasts. Treatment of the HuF with dimethyl sufoxide causes reversion to the spindle-shaped morphology and a loss of myofibroblast characteristics, suggesting a switch into a less differentiated phenotype. The unique abilities of HuF to exhibit multipotency, even with myofibroblast characteristics, and their ready availability and low maintenance requirements make them an interesting cell model for further exploration as a possible tool for regenerative medicine.


Assuntos
Fibroblastos/citologia , Células-Tronco Multipotentes/citologia , Placenta/citologia , Adipogenia , Biomarcadores/análise , Diferenciação Celular , Condrogênese , Proteínas do Citoesqueleto/metabolismo , Dimetil Sulfóxido/farmacologia , Feminino , Fibroblastos/química , Fibroblastos/metabolismo , Humanos , Mesoderma/citologia , Células-Tronco Multipotentes/química , Células-Tronco Multipotentes/metabolismo , Mioblastos/citologia , Osteogênese , Fenótipo , Células-Tronco Pluripotentes/química
16.
Endocrinology ; 148(7): 3176-84, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17412815

RESUMO

Differentiation of stromal cells into decidual cells, which is critical to successful pregnancy, represents a complex transformation requiring changes in cytoskeletal architecture. We demonstrate that in vitro differentiation of human uterine fibroblasts into decidual cells includes down-regulation of alpha-smooth muscle actin and beta-tubulin, phosphorylation of focal adhesion kinase, and redistribution of vinculin. This is accompanied by varied adhesion to fibronectin and a modified ability to migrate. Cytoskeletal organization is determined primarily by actin-myosin II interactions governed by the phosphorylation of myosin light chain (MLC20). Decidualization induced by cAMP [with estradiol-17beta (E) and medroxyprogesterone acetate (P)] results in a 40% decrease in MLC20 phosphorylation and a 55% decline in the long (214 kDa) form of myosin light-chain kinase (MLCK). Destabilization of the cytoskeleton by inhibitors of MLCK (ML-7) or myosin II ATPase (blebbistatin) accelerates decidualization induced by cAMP (with E and P) but inhibits decidualization induced by IL-1beta (with E and P). Adenoviral infection of human uterine fibroblast cells with a constitutively active form of MLCK followed by decidualization stimuli leads to a 30% increase in MLC20 phosphorylation and prevents decidualization. These data provide evidence that the regulation of cytoskeletal dynamics by MLC20 phosphorylation is critical for decidualization.


Assuntos
Endométrio/metabolismo , Cadeias Leves de Miosina/metabolismo , Actinas/metabolismo , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/farmacologia , Citoesqueleto/metabolismo , Endométrio/citologia , Endométrio/efeitos dos fármacos , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/farmacologia , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Gravidez , Tubulina (Proteína)/metabolismo , Útero/citologia , Útero/efeitos dos fármacos , Útero/metabolismo
17.
Mol Cell Biol ; 25(14): 6259-66, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15988034

RESUMO

Previous short-term studies have correlated an increase in the phosphorylation of the 20-kDa light chain of myosin II (MLC20) with blebbing in apoptotic cells. We have found that this increase in MLC20 phosphorylation is rapidly followed by MLC20 dephosphorylation when cells are stimulated with various apoptotic agents. MLC20 dephosphorylation is not a consequence of apoptosis because MLC20 dephosphorylation precedes caspase activation when cells are stimulated with a proapoptotic agent or when myosin light chain kinase (MLCK) is inhibited pharmacologically or by microinjecting an inhibitory antibody to MLCK. Moreover, blocking caspase activation increased cell survival when MLCK is inhibited or when cells are treated with tumor necrosis factor alpha. Depolymerizing actin filaments or detaching cells, processes that destabilize the cytoskeleton, or inhibiting myosin ATPase activity also resulted in MLC20 dephosphorylation and cell death. In vivo experiments showed that inhibiting MLCK increased the number of apoptotic cells and retarded the growth of mammary cancer cells in mice. Thus, MLC20 dephosphorylation occurs during physiological cell death and prolonged MLC20 dephosphorylation can trigger apoptosis.


Assuntos
Apoptose , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Humanos , Camundongos , Quinase de Cadeia Leve de Miosina/imunologia , Fosforilação
18.
Brain Res Dev Brain Res ; 151(1-2): 67-73, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15246693

RESUMO

The number and affinity of GABA(B) receptors (assayed by the specific antagonist [(3)H]CGP54626A) was unchanged when compared in carefully washed cerebrocortical membranes from young (12-day-old) and adult (90-day-old) rats. In contrast, high-affinity GTPase activity, both basal and baclofen-stimulated was significantly higher (by 45% and 56%, respectively) in adult than in young rats. Similar results were obtained by concomitant determination of agonist (baclofen)-stimulated GTP gamma S binding. Under standard conditions, baclofen-stimulated GTPase activity was further considerably enhanced by exogenously added regulator of G protein function, RGS1, but not by RGS16. RGS16 was able to affect agonist-stimulated GTPase activity only in the presence of markedly increase substrate (GTP) concentrations. RGS1 alone slightly increased GTPase activity in adult rats, but neither RGS1 nor RGS16 influenced GTPase activity in membrane preparations isolated from young animals. These findings indicate increasing functional activity of trimeric G protein(s) involved in GABAergic transmission in the developing rat brain cortex and suggest a high potential of RGS1 in regulation of high-affinity GTPase activity.


Assuntos
Baclofeno/farmacologia , Córtex Cerebral/efeitos dos fármacos , Proteínas de Ligação ao GTP/fisiologia , Envelhecimento/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Córtex Cerebral/crescimento & desenvolvimento , Relação Dose-Resposta a Droga , Interações Medicamentosas , Agonistas GABAérgicos/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Masculino , Compostos Organofosforados/farmacocinética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteínas/farmacologia , Proteínas RGS/farmacologia , Ensaio Radioligante/métodos , Ratos , Trítio/farmacocinética
19.
Neurosci Lett ; 330(1): 9-12, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12213622

RESUMO

Ontogenetic changes in the levels of GABA(B) receptors and their ability to modulate adenylyl cyclase (AC) activity were analyzed in rat cortex, thalamus and hippocampus. The relative numbers of GABA(B) receptors (measured as saturable, high-affinity [(3)H](-)baclofen binding sites) in cortex and thalamus were high already at postnatal day 1 (PD 1) and they reached a maximum at PD 25 and PD 12, respectively. There were no detectable high-affinity [(3)H](-)baclofen binding sites in hippocampus between birth and PD 12 and low-affinity [(3)H](-)baclofen binding attained at PD 12 did not change in adulthood (PD 90). Whereas GTP-stimulated AC activity in cortex and thalamus was depressed by baclofen, it was enhanced in hippocampus. These data indicate that the inhibitory effect of baclofen on AC in cortex and thalamus is primarily mediated through the alpha subunits of G(i)/G(o) proteins. The stimulatory effect of baclofen in hippocampus may be explained by engagement of Gbetagamma subunits.


Assuntos
Adenilil Ciclases/metabolismo , Baclofeno/farmacologia , Encéfalo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Hipocampo/crescimento & desenvolvimento , Masculino , Ratos , Ratos Wistar , Receptores de GABA-B/metabolismo , Tálamo/efeitos dos fármacos , Tálamo/enzimologia , Tálamo/crescimento & desenvolvimento
20.
Brain Res Dev Brain Res ; 133(1): 57-67, 2002 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-11850064

RESUMO

Developmental changes in the distribution of guanine nucleotide-binding regulatory proteins (G proteins) were investigated in the rat brain during postnatal development. Using a standard or high-resolution urea-SDS-PAGE and specific polyclonal antipeptide antibodies oriented against G(i)alpha1/G(i)alpha2, G(i)alpha3, G(s)alpha, G(o)alpha1/G(o)alpha2, G(q)alpha/G(11)alpha and Gbeta subunit, all these proteins were determined by quantitative immunoblotting in homogenates prepared from cortex, thalamus, hippocampus and pituitary of 1-, 7-, 12-, 18-, 25- and 90-day-old animals. The levels of the majority of G protein alpha subunits, namely G(i)alpha1, G(i)alpha2, G(i)alpha3, G(o)alpha1, G(o)alpha2, G(q)alpha, G(11)alpha and Gbeta, were high already at birth. Whereas the short variant of G(s)alpha, G(s)alphaS, rose sharply in all tested brain regions between postnatal day (PD) 1 and 90, the long variant of G(s)alpha, G(s)alphaL, was unchanged in cortex and thalamus and slightly increased in hippocampus. An increase was observed also in expression of G(i)alpha1/G(i)alpha2 and G(o)alpha1 protein, while G(o)alpha2 remained constant. Minority protein G(o)alpha* dramatically increased in cortex and thalamus, was unchanged in hippocampus and not detectable in pituitary. By contrast, the highest levels of G(i)alpha3 and G(q)alpha/G(11)alpha were detected as early as at PD 1. During the next 90 days, the immunological signal of G(i)alpha3 almost disappeared and G(q)alpha/G(11)alpha continuously declined to the levels corresponding to 50% of the levels determined at birth. Expression of Gbeta subunit was basically unchanged during postnatal development. Our present analysis indicates that G(s)alpha, G(i)alpha/G(o)alpha and G(q)alpha/G(11)alpha proteins are differently expressed in the course of brain development. Differential expression of the individual alpha subunits of trimeric G proteins during postnatal development suggests their different roles in maturation of the brain tissue.


Assuntos
Envelhecimento/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Regulação para Baixo/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Regulação para Cima/fisiologia , Animais , Animais Recém-Nascidos , Subunidade alfa Gi2 de Proteína de Ligação ao GTP , Subunidades alfa de Proteínas de Ligação ao GTP , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Masculino , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...