Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34561301

RESUMO

Nervous system malignancies are characterized by rapid progression and poor survival rates. These clinical observations underscore the need for novel therapeutic insights and pharmacological targets. To this end, here, we identify the orphan nuclear receptor NR5A2/LRH1 as a negative regulator of cancer cell proliferation and promising pharmacological target for nervous system-related tumors. In particular, clinical data from publicly available databases suggest that high expression levels of NR5A2 are associated with favorable prognosis in patients with glioblastoma and neuroblastoma tumors. Consistently, we experimentally show that NR5A2 is sufficient to strongly suppress proliferation of both human and mouse glioblastoma and neuroblastoma cells without inducing apoptosis. Moreover, short hairpin RNA-mediated knockdown of the basal expression levels of NR5A2 in glioblastoma cells promotes their cell cycle progression. The antiproliferative effect of NR5A2 is mediated by the transcriptional induction of negative regulators of the cell cycle, CDKN1A (encoding for p21cip1), CDKN1B (encoding for p27kip1) and Prox1 Interestingly, two well-established agonists of NR5A2, dilauroyl phosphatidylcholine (DLPC) and diundecanoyl phosphatidylcholine, are able to mimic the antiproliferative action of NR5A2 in human glioblastoma cells via the induction of the same critical genes. Most importantly, treatment with DLPC inhibits glioblastoma tumor growth in vivo in heterotopic and orthotopic xenograft mouse models. These data indicate a tumor suppressor role of NR5A2 in the nervous system and render this nuclear receptor a potential pharmacological target for the treatment of nervous tissue-related tumors.


Assuntos
Glioblastoma/patologia , Neoplasias do Sistema Nervoso/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Camundongos SCID , Neoplasias do Sistema Nervoso/tratamento farmacológico , Neoplasias do Sistema Nervoso/metabolismo , Neoplasias do Sistema Nervoso/mortalidade , Células-Tronco Neurais/efeitos dos fármacos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fosfatidilcolinas/farmacologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Cell Mol Med ; 27(9): 1192-1205, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37056054

RESUMO

Chronic kidney diseases affect a substantial percentage of the adult population worldwide. This observation emphasizes the need for novel insights into the molecular mechanisms that control the onset and progression of renal diseases. Recent advances in genomics have uncovered a previously unanticipated link between the non-coding genome and human kidney diseases. Here we screened and analysed long non-coding RNAs (lncRNAs) previously identified in mouse kidneys by genome-wide transcriptomic analysis, for conservation in humans and differential expression in renal tissue from healthy and diseased individuals. Our data suggest that LINC01187 is strongly down-regulated in human kidney tissues of patients with diabetic nephropathy and rapidly progressive glomerulonephritis, as well as in murine models of kidney diseases, including unilateral ureteral obstruction, nephrotoxic serum-induced glomerulonephritis and ischemia/reperfusion. Interestingly, LINC01187 overexpression in human kidney cells in vitro inhibits cell death indicating an anti-apoptotic function. Collectively, these data suggest a negative association of LINC01187 expression with renal diseases implying a potential protective role.


Assuntos
Nefropatias Diabéticas , Glomerulonefrite , RNA Longo não Codificante , Animais , Humanos , Camundongos , Nefropatias Diabéticas/metabolismo , Regulação para Baixo/genética , Glomerulonefrite/metabolismo , Rim/metabolismo , RNA Longo não Codificante/metabolismo
3.
Stem Cells ; 40(7): 678-690, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35429390

RESUMO

RNF113A (Ring Finger Protein 113A) is genetically associated with autism spectrum disorders and X-linked trichothiodystrophy (TTD) syndrome. Loss-of-function mutations in human RNF113A are causally linked to TTD, which is characterized by abnormal development of the central nervous system (CNS) and mental retardation. How the loss of RNF113A activity affects brain development is not known. Here we identify Rnf113a1 as a critical regulator of cell death and neurogenesis during mouse brain development. Rnf113a1 gene exhibits widespread expression in the embryonic CNS. Knockdown studies in embryonic cortical neural stem/progenitor cells (NSCs) and the mouse cortex suggest that Rnf113a1 controls the survival, proliferation, and differentiation properties of progenitor cells. Importantly, Rnf113a1 deficiency triggers cell apoptosis via a combined action on essential regulators of cell survival, including p53, Nupr1, and Rad51. Collectively, these observations establish Rnf113a1 as a regulatory factor in CNS development and provide insights into its role in neurodevelopmental defects associated with TTD and autism.


Assuntos
Células-Tronco Neurais , Transtornos do Neurodesenvolvimento , Animais , Apoptose/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Transtornos do Neurodesenvolvimento/genética , Neurogênese/fisiologia
4.
J Cell Mol Med ; 26(24): 5949-5954, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36440574

RESUMO

Fibrosis is a common denominator of several pathological conditions. Over the last decade, Calreticulin has emerged as a critical player in the fibrotic processes in many tissues and organs. Here we review the recent advances in our understanding of the regulatory roles of Calreticulin in renal fibrosis. In particular, a proteomic screen that we performed more than 15 years ago, for the identification of novel components involved in the mechanisms of renal fibrosis, led to the observation that Calreticulin is associated with the initiation and progression of kidney fibrosis in a rodent model. We also showed that altered expression levels of Calreticulin in vitro and in vivo are significantly affecting the fibrotic phenotype in cellular systems and animal models, respectively. We also identified an upstream regulatory mechanism that mediates the transcriptional control of Calreticulin expression during the progression of renal fibrosis, by showing that the druggable orphan nuclear receptor NR5A2 and its SUMOylation is involved in this action. These data provide novel targets for future pharmacological interventions against fibrosis. In addition, further proteomic analysis uncovered a correlation between the up-regulation of Calreticulin and that of 14-3-3σ protein. Collectively, our previous observations suggest that Calreticulin is a central node in a regulatory axis that controls the initiation and progression of renal fibrosis.


Assuntos
Calreticulina , Nefropatias , Animais , Calreticulina/genética , Calreticulina/metabolismo , Proteômica , Fibrose , Nefropatias/genética , Nefropatias/patologia , Regulação da Expressão Gênica , Rim/patologia
5.
Cell Mol Life Sci ; 78(7): 3443-3465, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33247761

RESUMO

During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.


Assuntos
Sinalização do Cálcio , Sistema Nervoso Central/patologia , Proteínas de Homeodomínio/metabolismo , Neuroblastoma/patologia , Crescimento Neuronal , Neurônios/patologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Células Cultivadas , Sistema Nervoso Central/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Neuroblastoma/metabolismo , Neurônios/metabolismo , Fosforilação , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
6.
J Cell Sci ; 132(11)2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31028178

RESUMO

A distinct combination of transcription factors elicits the acquisition of a specific fate and the initiation of a differentiation program. Multiciliated cells (MCCs) are a specialized type of epithelial cells that possess dozens of motile cilia on their apical surface. Defects in cilia function have been associated with ciliopathies that affect many organs, including brain and airway epithelium. Here we show that the geminin coiled-coil domain-containing protein 1 GemC1 (also known as Lynkeas) regulates the transcriptional activation of p73, a transcription factor central to multiciliogenesis. Moreover, we show that GemC1 acts in a trimeric complex with transcription factor E2F5 and tumor protein p73 (officially known as TP73), and that this complex is important for the activation of the p73 promoter. We also provide in vivo evidence that GemC1 is necessary for p73 expression in different multiciliated epithelia. We further show that GemC1 regulates multiciliogenesis through the control of chromatin organization, and the epigenetic marks/tags of p73 and Foxj1. Our results highlight novel signaling cues involved in the commitment program of MCCs across species and tissues.This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/genética , Proteína Tumoral p73/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Linhagem Celular , Cromatina/metabolismo , Células Epiteliais/citologia , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Ativação Transcricional/genética , Proteína Tumoral p73/genética
7.
Bioorg Chem ; 114: 105132, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34229198

RESUMO

Hydroxamic acid derivatives constitute an interesting novel class of antitumor agents. Three of them, including vorinostat, are approved drugs for the treatment of malignancies, while several others are currently under clinical trials. In this work, we present new vorinostat analogs containing the benzoxazole ring as the cap group and various linkers. The benzoxazole-based analogs were synthesized starting either from 2-aminobenzoxazole, through conventional coupling, or from benzoxazole, through a metal-free oxidative amination. All the synthesized compounds were evaluated for their antiproliferative activity on three diverse human cancer cell lines (A549, Caco-2 and SF268), in comparison to vorinostat. Compound 12 (GK601), carrying a benzoxazole ring replacement for the phenyl ring of vorinostat, was the most potent inhibitor of the growth of three cell lines (IC50 1.2-2.1 µΜ), similar in potency to vorinostat. Compound 12 also inhibited human HDAC1, HDAC2 and HDAC6 like vorinostat. This new analog also showed antiproliferative activity against two colon cancer cell lines genetically resembling pseudomyxoma peritonei (PMP), namely HCT116 GNAS R201C/+ and LS174T (IC50 0.6 and 1.4 µΜ, respectively) with potency comparable to vorinostat (IC50 1.1 and 2.1 µΜ, respectively).


Assuntos
Antineoplásicos/farmacologia , Benzoxazóis/farmacologia , Vorinostat/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Benzoxazóis/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas , Vorinostat/síntese química , Vorinostat/química
8.
Medicina (Kaunas) ; 57(10)2021 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-34684111

RESUMO

Background and Objectives: Calcium (Ca2+) signaling is critical for the normal functioning of various cellular activities. However, abnormal changes in cellular Ca2+ can contribute to pathological conditions, including various types of cancer. The maintenance of intracellular Ca2+ levels is achieved through tightly regulated processes that help maintain Ca2+ homeostasis. Several types of regulatory proteins are involved in controlling intracellular Ca2+ levels, including the sarco/endoplasmic reticulum (SR/ER) Ca2+ ATPase pump (SERCA), which maintains Ca2+ levels released from the SR/ER. In total, three ATPase SR/ER Ca2+-transporting (ATP2A) 1-3 genes exist, which encode for several isoforms whose expression profiles are tissue-specific. Recently, it has become clear that abnormal SERCA expression and activity are associated with various types of cancer, including breast cancer. Breast carcinomas represent 40% of all cancer types that affect women, with a wide variety of pathological and clinical conditions. Materials and methods: Using cBioPortal breast cancer patient data, Kaplan-Meier plots demonstrated that high ATP2A1 and ATP2A3 expression was associated with reduced patient survival. Results: The present study found significantly different SERCA specific-type expressions in a series of breast cancer cell lines. Moreover, bioinformatics analysis indicated that ATP2A1 and ATP2A3 expression was highly altered in patients with breast cancer. Conclusion: Overall, the present data suggest that SERCA gene-specific expressioncan possibly be considered as a crucial target for the control of breast cancer development and progression.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/genética , Cálcio , Feminino , Homeostase , Humanos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
9.
Proc Natl Acad Sci U S A ; 114(18): E3679-E3688, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28416701

RESUMO

α-Synuclein (αSyn) is the major gene linked to sporadic Parkinson's disease (PD), whereas the G209A (p.A53T) αSyn mutation causes a familial form of PD characterized by early onset and a generally severe phenotype, including nonmotor manifestations. Here we generated de novo induced pluripotent stem cells (iPSCs) from patients harboring the p.A53T mutation and developed a robust model that captures PD pathogenic processes under basal conditions. iPSC-derived mutant neurons displayed novel disease-relevant phenotypes, including protein aggregation, compromised neuritic outgrowth, and contorted or fragmented axons with swollen varicosities containing αSyn and Tau. The identified neuropathological features closely resembled those in brains of p.A53T patients. Small molecules targeting αSyn reverted the degenerative phenotype under both basal and induced stress conditions, indicating a treatment strategy for PD and other synucleinopathies. Furthermore, mutant neurons showed disrupted synaptic connectivity and widespread transcriptional alterations in genes involved in synaptic signaling, a number of which have been previously linked to mental disorders, raising intriguing implications for potentially converging disease mechanisms.


Assuntos
Axônios/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Mutação de Sentido Incorreto , Doença de Parkinson/metabolismo , Polineuropatias/metabolismo , Transmissão Sináptica , alfa-Sinucleína/metabolismo , Substituição de Aminoácidos , Axônios/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Polineuropatias/genética , Polineuropatias/patologia , alfa-Sinucleína/genética
10.
Cell Mol Life Sci ; 74(22): 4097-4120, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28638936

RESUMO

In the central nervous system, embryonic and adult neural stem/progenitor cells (NSCs) generate the enormous variety and huge numbers of neuronal and glial cells that provide structural and functional support in the brain and spinal cord. Over the last decades, nuclear receptors and their natural ligands have emerged as critical regulators of NSC homeostasis during embryonic development and adult life. Furthermore, substantial progress has been achieved towards elucidating the molecular mechanisms of nuclear receptors action in proliferative and differentiation capacities of NSCs. Aberrant expression or function of nuclear receptors in NSCs also contributes to the pathogenesis of various nervous system diseases. Here, we review recent advances in our understanding of the regulatory roles of steroid, non-steroid, and orphan nuclear receptors in NSC fate decisions. These studies establish nuclear receptors as key therapeutic targets in brain diseases.


Assuntos
Células-Tronco Neurais/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Diferenciação Celular , Humanos , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Células-Tronco Neurais/citologia , Neurogênese , Receptores de Calcitriol/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo
11.
J Biol Chem ; 291(16): 8756-72, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26907683

RESUMO

α-Synuclein, a presynaptic neuronal protein encoded by the SNCA gene, is strongly implicated in Parkinson disease (PD). PD pathogenesis is linked to increased SNCA levels; however, the transcriptional elements that control SNCA expression are still elusive. Previous experiments in PC12 cells demonstrated that the transcription factor zinc finger and SCAN domain containing 21 (ZSCAN21) plays an important regulatory role in SNCA transcription. Currently, we characterized the role of ZSCAN21 in SNCA transcription in primary neuronal cultures and in vivo We found that ZSCAN21 is developmentally expressed in neurons in different rat brain regions. We confirmed its binding in the intron 1 region of SNCA in rat cortical cultures. Lentivirus-mediated silencing of ZSCAN21 increased significantly SNCA promoter activity, mRNA, and protein levels in such cultures. In contrast, ZSCAN21 silencing reduced SNCA in neurosphere cultures. Interestingly, ZSCAN21 overexpression in cortical neurons led to robust mRNA but negligible protein expression, suggesting that ZSCAN21 protein levels are tightly regulated post-transcriptionally and/or post-translationally in primary neurons. Efficient adeno-associated virus-mediated knockdown of ZSCAN21 in the postnatal and adult hippocampus, an area linked with non-motor PD symptoms, revealed no significant alterations in SNCA levels. Overall, our study demonstrates that ZSCAN21 is involved in the transcriptional regulation of SNCA in primary neuronal cultures, but the direction of the effect is variable, likely depending on neuronal maturation. However, the unaltered SNCA levels observed following ZSCAN21 down-regulation in the rat brain, possibly due to compensatory mechanisms, imply that ZSCAN21 is not a master regulator of SNCA in vivo.


Assuntos
Córtex Cerebral/metabolismo , Regulação da Expressão Gênica , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Transativadores/metabolismo , alfa-Sinucleína/biossíntese , Animais , Células Cultivadas , Córtex Cerebral/citologia , Hipocampo/citologia , Neurônios/citologia , Ratos , Ratos Wistar
12.
Biochim Biophys Acta ; 1862(9): 1774-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27345266

RESUMO

BACKGROUND: Renal fibrosis is a common histological finding present in many pathologies; however, key signaling pathways and molecular determinants involved in the development of fibrosis are not fully known yet. Previous findings have established a causative role of calreticulin's up-regulation during the development of renal fibrosis while its down-regulation exhibited a protective effect against fibrosis. Therefore, the mechanism of its up-regulation needs to be explored. METHODS: Bioinformatics analyses of the calreticulin gene promoter combined with transcriptional assays and in vivo chromatin immunoprecipitation experiments in the Unilateral Ureteric Obstruction (UUO) model of renal fibrosis, indicated that NR5A2 is a critical regulator of calreticulin expression. To confirm this finding, and further study post-translational modifications of NR5A2, real time RT-qPCR, immunohistochemistry and Western blotting experiments were performed. RESULTS: NR5A2 is up-regulated at both mRNA and protein level during kidney fibrosis in the UUO model. The post-translational modification of SUMOylation was identified as a critical parameter in this phenomenon and SUMOylation was observed to be up-regulated during the development of renal fibrosis. The enzyme Ubc9, critical for the process of SUMOylation was also upregulated at mRNA and protein level during the process. CONCLUSION: These data establish for the first time a role for NR5A2 and its SUMOylation on the transcriptional regulation of the calreticulin gene in a rodent model of renal fibrosis and raise the possibility that NR5A2 might be a novel target for future anti-fibrotic interventions.


Assuntos
Calreticulina/genética , Rim/metabolismo , Rim/patologia , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Proteína SUMO-1/metabolismo , Sumoilação , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Regulação para Cima , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
13.
Stem Cells ; 34(8): 2115-29, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27068685

RESUMO

Adult neural stem cells with the ability to generate neurons and glia cells are active throughout life in both the dentate gyrus (DG) and the subventricular zone (SVZ). Differentiation of adult neural stem cells is induced by cell fate determinants like the transcription factor Prox1. Evidence has been provided for a function of Prox1 as an inducer of neuronal differentiation within the DG. We now show that within the SVZ Prox1 induces differentiation into oligodendrocytes. Moreover, we find that loss of Prox1 expression in vivo reduces cell migration into the corpus callosum, where the few Prox1 deficient SVZ-derived remaining cells fail to differentiate into oligodendrocytes. Thus, our work uncovers a novel function of Prox1 as a fate determinant for oligodendrocytes in the adult mammalian brain. These data indicate that the neurogenic and oligodendrogliogenic lineages in the two adult neurogenic niches exhibit a distinct requirement for Prox1, being important for neurogenesis in the DG but being indispensable for oligodendrogliogenesis in the SVZ. Stem Cells 2016;34:2115-2129.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Proteínas de Homeodomínio/metabolismo , Ventrículos Laterais/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Movimento Celular/genética , Células Cultivadas , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Neurogênese/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/genética , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores Notch/genética , Receptores Notch/metabolismo
14.
J Neurosci ; 34(47): 15816-31, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25411508

RESUMO

Specification of spinal cord neurons depends on gene regulation networks that impose distinct fates in neural progenitor cells (NPCs). Olig2 is a key transcription factor in these networks by inducing motor neuron (MN) specification and inhibiting interneuron identity. Despite the critical role of Olig2 in nervous system development and cancer progression, the upstream molecular mechanisms that control Olig2 gene transcription are not well understood. Here we demonstrate that Prox1, a transcription repressor and downstream target of proneural genes, suppresses Olig2 expression and therefore controls ventral spinal cord patterning. In particular, Prox1 is strongly expressed in V2 interneuron progenitors and largely excluded from Olig2+ MN progenitors (pMN). Gain- and loss-of-function studies in mouse NPCs and chick neural tube show that Prox1 is sufficient and necessary for the suppression of Olig2 expression and proper control of MN versus V2 interneuron identity. Mechanistically, Prox1 interacts with the regulatory elements of Olig2 gene locus in vivo and it is critical for proper Olig2 transcription regulation. Specifically, chromatin immunoprecipitation analysis in the mouse neural tube showed that endogenous Prox1 directly binds to the proximal promoter of the Olig2 gene locus, as well as to the K23 enhancer, which drives Olig2 expression in the pMN domain. Moreover, plasmid-based transcriptional assays in mouse NPCs suggest that Prox1 suppresses the activity of Olig2 gene promoter and K23 enhancer. These observations indicate that Prox1 controls binary fate decisions between MNs and V2 interneurons in NPCs via direct repression of Olig2 gene regulatory elements.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Proteínas de Homeodomínio/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Neurônios/fisiologia , Medula Espinal/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Embrião de Galinha , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Interneurônios/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios Motores/fisiologia , Células-Tronco Neurais/fisiologia , Fator de Transcrição 2 de Oligodendrócitos , Medula Espinal/citologia , Medula Espinal/crescimento & desenvolvimento
15.
Am J Pathol ; 183(5): 1474-1487, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24035512

RESUMO

Renal fibrosis is the common anatomical feature underlying the progression of chronic kidney disease, a leading cause of morbidity and mortality worldwide. In a previous study, we demonstrated that during development of renal fibrosis in a rat model of unilateral ureteric obstruction, calreticulin (CRT) is up-regulated in tubular epithelial cells (TECs). In the present study, we used in vitro and in vivo approaches to examine the role of CRT in TECs and its contribution to the progression of fibrosis. In cultured renal TECs, CRT overexpression induced acquisition of an altered, profibrotic cellular phenotype. Consistently, the opposite effects were observed for CRT knockdown. Subsequently, we confirmed that critical changes observed in vitro were also apparent in tubular cells in vivo in the animal model of unilateral ureteric obstruction. In agreement with these results, we demonstrate that substantial (50%) reduction in the expression of CRT reduced the development of tubulointerstitial fibrosis at a comparable level through regulation of inflammation, transcriptional activation, transforming growth factor ß1-associated effects, and apoptosis. In summary, our findings establish that CRT is critically involved in the molecular mechanisms that drive renal fibrosis progression and indicate that inhibition of CRT expression might be a therapeutic target for reduction of fibrosis and chronic kidney disease development.


Assuntos
Calreticulina/metabolismo , Células Epiteliais/metabolismo , Nefropatias/metabolismo , Nefropatias/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Regulação para Cima , Animais , Apoptose , Biomarcadores/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Estresse do Retículo Endoplasmático , Células Epiteliais/patologia , Fibrose , Técnicas de Silenciamento de Genes , Heterozigoto , Humanos , Masculino , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
16.
Biomolecules ; 14(1)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38254710

RESUMO

Hydroxy fatty acids (HFAs) constitute a class of lipids, distinguished by the presence of a hydroxyl on a long aliphatic chain. This study aims to expand our insights into HFA bioactivities, while also introducing new methods for asymmetrically synthesizing unsaturated and saturated HFAs. Simultaneously, a procedure previously established by us was adapted to generate new HFA regioisomers. An organocatalytic step was employed for the synthesis of chiral terminal epoxides, which either by alkynylation or by Grignard reagents resulted in unsaturated or saturated chiral secondary alcohols and, ultimately, HFAs. 7-(S)-Hydroxyoleic acid (7SHOA), 7-(S)-hydroxypalmitoleic acid (7SHPOA) and 7-(R)- and (S)-hydroxymargaric acids (7HMAs) were synthesized for the first time and, together with regioisomers of (R)- and (S)-hydroxypalmitic acids (HPAs) and hydroxystearic acids (HSAs), whose biological activity has not been tested so far, were studied for their antiproliferative activities. The unsaturation of the long chain, as well as an odd-numbered (C17) fatty acid chain, led to reduced activity, while the new 6-(S)-HPA regioisomer was identified as exhibiting potent antiproliferative activity in A549 cells. 6SHPA induced acetylation of histone 3 in A549 cells, without affecting acetylated α-tubulin levels, suggesting the selective inhibition of histone deacetylase (HDAC) class I enzymes, and was found to inhibit signal transducer and activator of transcription 3 (STAT3) expression.


Assuntos
Ácidos Graxos Insaturados , Ácidos Graxos , Humanos , Ácidos Graxos Insaturados/farmacologia , Ácidos Graxos/farmacologia , Células A549 , Acetilação , Fenômenos Químicos
17.
Transl Psychiatry ; 14(1): 104, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38378836

RESUMO

Fragile X syndrome (FXS) is the most common cause of inherited intellectual disabilities and the most prevalent monogenic cause of autism. Although the knockout (KO) of the Fmr1 gene homolog in mice is primarily used for elucidating the neurobiological substrate of FXS, there is limited association of the experimental data with the pathophysiological condition in humans. The use of Fmr1 KO rats offers additional translational validity in this regard. Therefore, we employed a multi-level approach to study the behavioral profile and the glutamatergic and GABAergic neurotransmission status in pathophysiology-associated brain structures of Fmr1 KO rats, including the recordings of evoked and spontaneous field potentials from hippocampal slices, paralleled with next-generation RNA sequencing (RNA-seq). We found that these rats exhibit hyperactivity and cognitive deficits, along with characteristic bidirectional glutamatergic and GABAergic alterations in the prefrontal cortex and the hippocampus. These results are coupled to affected excitability and local inhibitory processes in the hippocampus, along with a specific transcriptional profile, highlighting dysregulated hippocampal network activity in KO rats. Overall, our data provide novel insights concerning the biobehavioral profile of FmR1 KO rats and translationally upscales our understanding on pathophysiology and symptomatology of FXS syndrome.


Assuntos
Transtornos Cognitivos , Disfunção Cognitiva , Síndrome do Cromossomo X Frágil , Ratos , Camundongos , Animais , Humanos , Camundongos Knockout , Hipocampo/metabolismo , Encéfalo/metabolismo , Síndrome do Cromossomo X Frágil/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Modelos Animais de Doenças
18.
Arch Biochem Biophys ; 534(1-2): 27-37, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23044345

RESUMO

In the central nervous system (CNS) of vertebrates a large variety of cell types are specified from a pool of highly plastic neural stem/progenitor cells (NSCs) via a combined action of extrinsic morphogenetic cues and intrinsic transcriptional regulatory networks. Nuclear receptors and their ligands are key regulators of fate decisions in NSCs during development and adulthood, through their ability to control transcription of downstream genes. In the last few years considerable progress has been made towards the understanding of the actions of nuclear receptors in NSCs as well as elucidating the mechanistic basis for these actions. Here we summarize recent progress in the role of nuclear receptors in the biology of NSCs. These studies highlight the importance of this family of transcriptional regulators in CNS development and function in health and disease. Furthermore, they raise the intriguing possibility of using nuclear receptors as therapeutic targets for nervous system related diseases and traumas.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Neurais/citologia , Receptores dos Hormônios Tireóideos/metabolismo , Animais , Homeostase , Humanos , Ligantes , Receptores X do Fígado , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Fenótipo , Receptores dos Hormônios Tireóideos/genética , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Transcrição Gênica
19.
PLoS Biol ; 8(12): e1000565, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21203589

RESUMO

Activation of Notch1 signaling in neural progenitor cells (NPCs) induces self-renewal and inhibits neurogenesis. Upon neuronal differentiation, NPCs overcome this inhibition, express proneural genes to induce Notch ligands, and activate Notch1 in neighboring NPCs. The molecular mechanism that coordinates Notch1 inactivation with initiation of neurogenesis remains elusive. Here, we provide evidence that Prox1, a transcription repressor and downstream target of proneural genes, counteracts Notch1 signaling via direct suppression of Notch1 gene expression. By expression studies in the developing spinal cord of chick and mouse embryo, we showed that Prox1 is limited to neuronal precursors residing between the Notch1+ NPCs and post-mitotic neurons. Physiological levels of Prox1 in this tissue are sufficient to allow binding at Notch1 promoter and they are critical for proper Notch1 transcriptional regulation in vivo. Gain-of-function studies in the chick neural tube and mouse NPCs suggest that Prox1-mediated suppression of Notch1 relieves its inhibition on neurogenesis and allows NPCs to exit the cell cycle and differentiate. Moreover, loss-of-function in the chick neural tube shows that Prox1 is necessary for suppression of Notch1 outside the ventricular zone, inhibition of active Notch signaling, down-regulation of NPC markers, and completion of neuronal differentiation program. Together these data suggest that Prox1 inhibits Notch1 gene expression to control the balance between NPC self-renewal and neuronal differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/metabolismo , Tubo Neural/metabolismo , Receptor Notch1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , Embrião de Galinha , Proteínas de Homeodomínio/genética , Camundongos , Células-Tronco Neurais/citologia , Tubo Neural/citologia , Tubo Neural/embriologia , Neurogênese , Receptor Notch1/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
20.
Cells ; 12(14)2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37508533

RESUMO

Breast cancer is one of the most lethal malignancies in women worldwide and is characterized by rapid growth and low survival rates, despite advances in tumor biology and therapies. Novel therapeutic approaches require new insights into the molecular mechanisms of malignant transformation and progression. To this end, here, we identified Prox1 as a negative regulator of proliferation and tumor-related metabolism in breast cancer. In particular, we showed that breast tumors from human patients exhibited reduced levels of Prox1 expression, while high expression levels of Prox1 were associated with a favorable prognosis in breast cancer patients. Moreover, we experimentally demonstrated that Prox1 was sufficient to strongly suppress proliferation, migration, and the Warburg effect in human breast cancer cells without inducing apoptosis. Most importantly, over-expression of Prox1 inhibited breast tumor growth in vivo in both heterotopic and orthotopic xenograft mouse models. The anti-tumorigenic effect of Prox1 was mediated by the direct repression of c-Myc transcription and its downstream target genes. Consistently, c-Myc over-expression from an artificial promoter that was not targeted by Prox1 reversed Prox1's anti-tumor effects. These findings suggest that Prox1 has a tumor suppressive role via direct transcriptional regulation of c-Myc, making it a promising therapeutic gene for breast cancer.


Assuntos
Neoplasias da Mama , Proteínas de Homeodomínio , Humanos , Feminino , Camundongos , Animais , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas Supressoras de Tumor/metabolismo , Fatores de Transcrição/genética , Proliferação de Células , Expressão Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA