Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 385
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Metallomics ; 15(12)2023 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-37994650

RESUMO

The molecular mechanism of aluminum toxicity in biological systems is not completely understood. Saccharomyces cerevisiae is one of the most used model organisms in the study of environmental metal toxicity. Using an unbiased metallomic approach in yeast, we found that aluminum treatment caused phosphorus deprivation, and the lack of phosphorus increased as the pH of the environment decreased compared to the control strain. By screening the phosphate signaling and response pathway (PHO pathway) in yeast with the synthetic lethality of a new phosphorus-restricted aluminum-sensitive gene, we observed that pho84Δ mutation conferred severe growth defect to aluminum under low-phosphorus conditions, and the addition of phosphate alleviated this sensitivity. Subsequently, the data showed that PHO84 determined the intracellular aluminum-induced phosphorus deficiency, and the expression of PHO84 was positively correlated with aluminum stress, which was mediated by phosphorus through the coordinated regulation of PHO4/PHO2. Moreover, aluminum reduced phosphorus absorption and inhibited tobacco plant growth in acidic media. In addition, the high-affinity phosphate transporter NtPT1 in tobacco exhibited similar effects to PHO84, and overexpression of NtPT1 conferred aluminum resistance in yeast cells. Taken together, positive feedback regulation of the PHO pathway centered on the high-affinity phosphate transporters is a highly conservative mechanism in response to aluminum toxicity. The results may provide a basis for aluminum-resistant microorganisms or plant engineering and acidic soil treatment.


Assuntos
Fósforo na Dieta , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Nicotiana/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Alumínio/toxicidade , Alumínio/metabolismo , Fósforo na Dieta/metabolismo , Fósforo , Simportadores de Próton-Fosfato/genética , Simportadores de Próton-Fosfato/metabolismo , Fosfatos/metabolismo , Proteínas de Homeodomínio/metabolismo
2.
Oxid Med Cell Longev ; 2022: 8488269, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36199421

RESUMO

Population aging has led to increased sick sinus syndrome (SSS) incidence; however, no effective and safe medical therapy has been reported thus far. Yixin-Fumai granules (YXFMs), a Chinese medicine granule designed for bradyarrhythmia treatment, can effectively increase SSS patients' heart rate. Senescence-induced sinoatrial node (SAN) degeneration is an important part of SSS pathogenesis, and older people often show high levels of oxidative stress; reactive oxygen species (ROS) accumulation in the SAN causes abnormal SAN pacing or conduction functions. The current study observed the protective effects of YXFMs on senescent SAN and explored the relationship between the NRF-2/HO-1 pathway, SHOX2, and T-type calcium channels. We selected naturally senescent C57BL/6 mice with bradycardia to simulate SSS; electrocardiography, Masson's trichrome staining, and DHE staining were used to assess SAN function and tissue damage. Immunofluorescence staining and Western blotting were used to assay related proteins. In vitro, we treated human-induced pluripotent stem cell-derived atrial myocytes (hiPSC-AMs) and mouse atrial myocyte-derived cell line HL-1 with D-galactose to simulate senescent SAN-pacemaker cells. CardioExcyte96 was used to evaluate the pulsatile function of the hiPSC-AMs, and the mechanism was verified by DCFH-DA, immunofluorescence staining, RT-qPCR, and Western blotting. The results demonstrated that YXFMs effectively inhibited senescence-induced SAN hypofunction, and this effect possibly originated from scavenging of ROS and promotion of NRF-2, SHOX2, and T-type calcium channel expression. In vitro experiment results indicated that ML385, si-SHOX2, LDN193189, and Mibefradil reversed YXFMs' effects. Moreover, we, for the first time, found that ROS accumulation may hinder SHOX2 expression; YXFMs can activate SHOX2 through the NRF-2/HO-1 pathway-mediated ROS scavenging and then regulate CACNA1G through the SHOX2/BMP4/GATA4/NKX2-5 axis, improve T-type calcium channel function, and ameliorate the SAN dysfunction. Finally, through network pharmacology and molecular docking, we screened for the most stable YXFMs compound that docks to NRF-2, laying the foundation for future studies.


Assuntos
Canais de Cálcio Tipo T , Heme Oxigenase-1/metabolismo , Proteínas de Homeodomínio , Fator 2 Relacionado a NF-E2/metabolismo , Aceleração , Idoso , Animais , Galactose , Frequência Cardíaca , Proteínas de Homeodomínio/metabolismo , Humanos , Medicina Tradicional Chinesa , Mibefradil , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo
3.
Int J Obes (Lond) ; 46(11): 2029-2039, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36115924

RESUMO

OBJECTIVE: Obesity, a leading cause of several metabolic abnormalities, is mainly caused by imbalanced energy homeostasis. IRX3 and IRX5 have been suggested as genetic determinants of obesity in connection with the intronic variants of the FTO gene, the strongest genetic risk factor of polygenic obesity in humans. Although the causal effects of Irx3 and its cooperation with Irx5 in obesity and associated metabolic abnormalities have been demonstrated in vivo, the function of Irx5 in energy homeostasis remains unclear. Here we aim to decipher the actions of Irx5 in the regulation of obesity and metabolic abnormalities. METHODS: We employed a mouse model homozygous for an Irx5-knockout (Irx5KO) allele and determined its metabolic phenotype in the presence or absence of a high-fat diet challenge. To investigate the function of Irx5 in the regulation of energy homeostasis, adipose thermogenesis and hypothalamic leptin response were assessed, and single-cell RNA sequencing (scRNA-seq) in the hypothalamic arcuate-median eminence (ARC-ME) was conducted. RESULTS: Irx5KO mice were leaner and resistant to diet-induced obesity as well as associated metabolic abnormalities, primarily through loss of adiposity. Assessments of energy expenditure and long-term dietary intake revealed that an increase in basal metabolic rate with adipose thermogenesis and a reduction of food intake with improved hypothalamic leptin response in Irx5KO mice may contribute to the anti-obesity effects. Utilizing scRNA-seq and marker gene analyses, we demonstrated the number of ARC-ME neurons was elevated in Irx5KO mice, suggesting a direct role for Irx5 in hypothalamic feeding control. CONCLUSIONS: Our study demonstrates that Irx5 is a genetic factor determining body mass/composition and obesity and regulates both energy expenditure and intake.


Assuntos
Leptina , Obesidade , Humanos , Animais , Camundongos , Leptina/metabolismo , Obesidade/genética , Obesidade/metabolismo , Dieta Hiperlipídica , Hipotálamo/metabolismo , Metabolismo Energético/genética , Camundongos Knockout , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
4.
J Neurosci ; 42(42): 7885-7899, 2022 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-36028316

RESUMO

Anterolateral system (AS) neurons transmit pain signals from the spinal cord to the brain. Their morphology, anatomy, and physiological properties have been extensively characterized and suggest that specific AS neurons and their brain targets are concerned with the discriminatory aspects of noxious stimuli, such as their location or intensity, and their motivational/emotive dimension. Among the recently unraveled molecular markers of AS neurons is the developmentally expressed transcription factor Phox2a, providing us with the opportunity to selectively disrupt the embryonic wiring of AS neurons to gain insights into the logic of their adult function. As mice with a spinal-cord-specific loss of the netrin-1 receptor deleted in colorectal carcinoma (DCC) have increased AS neuron innervation of ipsilateral brain targets and defective noxious stimulus localization or topognosis, we generated mice of either sex carrying a deletion of Dcc in Phox2a neurons. Such DccPhox2a mice displayed impaired topognosis along the rostrocaudal axis but with little effect on left-right discrimination and normal aversive responses. Anatomical tracing experiments in DccPhox2a mice revealed defective targeting of cervical and lumbar AS axons within the thalamus. Furthermore, genetic labeling of AS axons revealed their expression of DCC on their arrival in the brain, at a time when many of their target neurons are being born and express Ntn1 Our experiments suggest a postcommissural crossing function for netrin-1:DCC signaling during the formation of somatotopically ordered maps and are consistent with a discriminatory function of some of the Phox2a AS neurons.SIGNIFICANCE STATEMENT How nociceptive (pain) signals are relayed from the body to the brain remains an important question relevant to our understanding of the basic physiology of pain perception. Previous studies have demonstrated that the AS is a main effector of this function. It is composed of AS neurons located in the spinal cord that receive signals from nociceptive sensory neurons that detect noxious stimuli. In this study, we generate a genetic miswiring of mouse AS neurons that results in a decreased ability to perceive the location of a painful stimulus. The precise nature of this defect sheds light on the function of different kinds of AS neurons and how pain information may be organized.


Assuntos
Neoplasias Colorretais , Fatores de Crescimento Neural , Animais , Camundongos , Neoplasias Colorretais/metabolismo , Receptor DCC/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Crescimento Neural/metabolismo , Receptores de Netrina/metabolismo , Netrina-1 , Neurônios/fisiologia , Dor/metabolismo , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Tálamo
5.
Proc Natl Acad Sci U S A ; 119(36): e2119854119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037364

RESUMO

Clear cell renal cell carcinoma (ccRCC) is characterized by the loss of tumor suppressor Von Hippel Lindau (VHL) function. VHL is the component of an E3 ligase complex that promotes the ubiquitination and degradation of hypoxia inducible factor α (HIF-α) (including HIF1α and HIF2α) and Zinc Fingers And Homeoboxes 2 (ZHX2). Our recent research showed that ZHX2 contributed to ccRCC tumorigenesis in a HIF-independent manner. However, it is still unknown whether ZHX2 could be modified through deubiquitination even in the absence of pVHL. Here, we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and identified USP13 as a DUB that bound ZHX2 and promoted ZHX2 deubiquitination. As a result, USP13 promoted ZHX2 protein stability in an enzymatically dependent manner, and depletion of USP13 led to ZHX2 down-regulation in ccRCC. Functionally, USP13 depletion led to decreased cell proliferation measured by two-dimensional (2D) colony formation and three-dimensional (3D) anchorage-independent growth. Furthermore, USP13 was essential for ccRCC tumor growth in vivo, and the effect was partially mediated by its regulation on ZHX2. Our findings support that USP13 may be a key effector in ccRCC tumorigenesis.


Assuntos
Carcinoma de Células Renais , Proteínas de Homeodomínio , Neoplasias Renais , Fatores de Transcrição , Proteases Específicas de Ubiquitina , Carcinogênese/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
6.
Cells ; 11(7)2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35406651

RESUMO

H6 family homeobox 1 (HMX1) regulates multiple aspects of craniofacial development, and mutations in HMX1 are linked to an ocular defect termed oculoauricular syndrome of Schorderet-Munier-Franceschetti (OAS) (MIM #612109). Recently, additional altered orofacial features have been reported, including short mandibular rami, asymmetry of the jaws, and altered premaxilla. We found that in two mutant zebrafish lines termed hmx1mut10 and hmx1mut150, precocious mineralization of the proximal vertebrae occurred. Zebrafish hmx1mut10 and hmx1mut150 report mutations in the SD1 and HD domains, which are essential for dimerization and activity of hmx1. In hmx1mut10, the bone morphogenetic protein (BMP) antagonists chordin and noggin1 were downregulated, while bmp2b and bmp4 were highly expressed and specifically localized to the dorsal region prior to the initiation of the osteogenic process. The osteogenic promoters runx2b and spp1 were also upregulated. Supplementation with DMH1-an inhibitor of the BMP signaling pathway-at the specific stage in which bmp2b and bmp4 are highly expressed resulted in reduced vertebral mineralization, resembling the wildtype mineralization progress of the axial skeleton. These results point to a possible role of hmx1 as part of a complex gene network that inhibits bmp2b and bmp4 in the dorsal region, thus regulating early axial skeleton development.


Assuntos
Doenças Ósseas , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Doenças Ósseas/genética , Calcificação Fisiológica , Genes Homeobox , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
7.
Bioengineered ; 13(3): 4911-4922, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35152841

RESUMO

Increasing evidence has shown that traditional Chinese medicines and their bioactive components exert an anti-tumor effect, representing a novel treatment strategy. Actinidia chinensis Planch Root extracts (acRoots) have been reported to repress cancer cell proliferation and metastasis. The effect of acRoots on hypopharyngeal carcinoma progression was explored in this study. Firstly, data from MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and colony formation assays showed that incubation with accRoots reduced cell proliferation of hypopharyngeal carcinoma cells. Moreover, acRoots promoted the cell apoptosis of hypopharyngeal carcinoma. Secondly, cell migration and invasion of hypopharyngeal carcinoma cells were suppressed by acRoots. Thirdly, E2F1 (E2F Transcription Factor 1) and lncRNA MNX1-AS1 (MNX1 antisense RNA 1) were up-regulated in hypopharyngeal carcinoma tissues, and reduced in hypopharyngeal carcinoma cells post acRoots incubation. Overexpression of E2F1 attenuated acRoots-induced decrease in MNX1-AS1 in hypopharyngeal carcinoma cells. Lastly, administration with acRoots retarded in vivo hypopharyngeal carcinoma growth through down-regulation of E2F1-mediated MNX1-AS1. In conclusion, acRoots exerted tumor-suppressive role in hypopharyngeal carcinoma through inhibition of E2F1-mediated MNX1-AS1.


Assuntos
Actinidia , Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , Extratos Vegetais , Actinidia/química , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Fatores de Transcrição E2F/genética , Fator de Transcrição E2F1 , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Neoplasias Hepáticas/genética , MicroRNAs/genética , Extratos Vegetais/farmacologia , RNA Antissenso/genética , Fatores de Transcrição/metabolismo
8.
J Biochem Mol Toxicol ; 36(4): e23004, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35191145

RESUMO

Saponin gypenoside A (GP) has shown its potential to handle diabetes mellitus. MicroRNA-150-3p (miR-150-3p) is closely related to the dysfunction of pancreatic ß cells by targeting PDX1. Given the function of GP is related to its regulation on different miRs, the current study assessed the role of miR-150-3p as a therapeutic target for the hypoglycemic effects of GP. Pancreatic ß cell dysfunction was induced in mice using the high-fatty diet (HFD) method and then handled with GP. Changes in insulin release and resistance and the activity of the miR-150-3p/PDX1 axis were detected. The expression of miR-150-3p was induced to confirm its central in the effects of GP. The results of in vivo tests were then validated with in vitro assays. HFD administration suppressed glucose tolerance, delayed insulin release, and induced insulin resistance and pancreas apoptosis in mice, which was indicative of the dysfunction of ß pancreatic cells. Changes in pancreatic ß function were associated with the increased expression of miR-150-3p and suppressed expression of PDX1. After the administration of GP, the impairments of the pancreas were alleviated and the expression of miR-150-3p was inhibited, contributing to the restored level of PDX1. The injection of miR-150-3p agomir counteracted the protective effects of GP. In in vitro assays, the pretransfection of miR-150-3p mimetics also counteracted the protective effects of GP on pancreatic ß cells against palmitic acid. Collectively, miR-150-3p played a key role in the protective effects of GP against pancreatic ß cell dysfunction by inhibiting PDX1 expression.


Assuntos
Células Secretoras de Insulina , MicroRNAs , Animais , Gynostemma , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Extratos Vegetais , Transdução de Sinais , Transativadores/metabolismo
9.
Cell Mol Gastroenterol Hepatol ; 13(4): 1201-1221, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34920145

RESUMO

BACKGROUND & AIMS: TOB1 is an anti-proliferative protein of Tob/BTG family and typically involved in the tumorigenesis and T cell activation. Although TOB1 is associated with T helper 17 cell-related autoimmunity, its role in modulating T cell-mediated immune responses in IBD remains poorly understood. Here, we explored its expression and the underlying mechanisms involved in the pathogenesis of inflammatory bowel disease (IBD). METHODS: TOB1 and ID2 expression in IBD patients was examined by quantitative real time polymerase chain reaction and immunohistochemistry. IBD CD4+ T cells were transfected with lentivirus expressing TOB1, ID2, TOB1 short hairpin RNA and ID2 short hairpin RNA, respectively, and Tob1-/-CD4+ T cells were transfected with lentivirus expressing Id2. Experimental colitis was established in Tob1-/- mice by trinitrobenzene sulfonic acid enema and in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells to further explore the role of Tob1 in intestinal mucosal inflammation. Splenic CD4+ T cells of Tob1-/- mice were sorted to determine transcriptome differences by RNA sequencing. RESULTS: TOB1 expression was decreased in inflamed mucosa and peripheral blood CD4+ T cells of IBD patients compared with healthy subjects. Overexpression of TOB1 downregulated IBD CD4+ T cells to differentiate into Th1/Th17 cells compared with control subjects. Severe colitis was observed in Tob1-/- mice through trinitrobenzene sulfonic acid enema or in Rag1-/- mice reconstituted with Tob1-/-CD45RBhighCD4+ T cells, compared with control animals. RNA sequencing analysis revealed ID2 as functional target of TOB1 to inhibit IBD CD4+ T cell differentiation into Th1/Th17 cells. Mechanistically, TOB1 was associated with Smad4/5 to induce ID2 expression and restrain Th1/Th17 cell differentiation. CONCLUSIONS: TOB1 restrains intestinal mucosal inflammation through suppressing Th1/Th17 cell-mediated immune responses via the Smad4/5-ID2 pathway. It may serve as a novel therapeutic target for treatment of human IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Proteínas de Homeodomínio/metabolismo , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Mucosa Intestinal/metabolismo , Ativação Linfocitária , Camundongos , RNA Interferente Pequeno/metabolismo , Ácidos Sulfônicos/metabolismo , Ácidos Sulfônicos/uso terapêutico , Células Th1 , Células Th17/metabolismo , Células Th17/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Cell Death Dis ; 12(10): 879, 2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34564702

RESUMO

Dopaminergic (DA) neurons in the arcuate nucleus (ARC) of the hypothalamus play essential roles in the secretion of prolactin and the regulation of energy homeostasis. However, the gene regulatory network responsible for the development of the DA neurons remains poorly understood. Here we report that the transcription factor special AT-rich binding protein 2 (Satb2) is required for the development of ARC DA neurons. Satb2 is expressed in a large proportion of DA neurons without colocalization with proopiomelanocortin (POMC), orexigenic agouti-related peptide (AgRP), neuropeptide-Y (NPY), somatostatin (Sst), growth hormone-releasing hormone (GHRH), or galanin in the ARC. Nestin-Cre;Satb2flox/flox (Satb2 CKO) mice show a reduced number of ARC DA neurons with unchanged numbers of the other types of ARC neurons, and exhibit an increase of serum prolactin level and an elevated metabolic rate. The reduction of ARC DA neurons in the CKO mice is observed at an embryonic stage and Dlx1 is identified as a potential downstream gene of Satb2 in regulating the development of ARC DA neurons. Together, our study demonstrates that Satb2 plays a critical role in the gene regulatory network directing the development of DA neurons in ARC.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Fatores de Transcrição/metabolismo , Envelhecimento/metabolismo , Proteína Relacionada com Agouti/metabolismo , Animais , Metabolismo Basal , Diferenciação Celular , Feminino , Proteínas de Fluorescência Verde/metabolismo , Hipotálamo/metabolismo , Lactação , Proteínas de Ligação à Região de Interação com a Matriz/deficiência , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeo Y/metabolismo , Fosforilação , Pró-Opiomelanocortina/metabolismo , Prolactina/sangue , Fator de Transcrição STAT5/metabolismo , Fatores de Transcrição/deficiência , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Int J Biol Macromol ; 190: 940-959, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34478798

RESUMO

Among the diverse nanomaterials, polymer-based nanocomposites are gained more attention due to their high efficacy, target biological activities, biodegradability and biocompatibility-gum acacia (GA) - a polymer obtained from acacia trees-is considering the multifunctional nanocomposite synthesis. Distinctive Physico-chemical and biocompatibility properties of gum acacia are utilised to prepare a highly stable, biologically active, eco-friendly Nanocomposite. In this current investigation, gum acacia - poly ethylene glycol grafted iron oxide nanocomposite (GA-PEG-IONC) was synthesised by in situ green science principles. The synthesised Nanocomposite was evaluated against the molecular mechanism of urinary tract pathogenic bacterial strains and prostate cancer cells (Pc 3). Nanocomposite prepared in this examination exhibited notable structural, functional stability with nanoarchitecture which was affirmed by Fourier transform infrared spectroscopy (FTIR), electron microscopic studies, atomic force microscopy (AFM), vibrating sample magnetometric analysis (VSM) and X-ray diffraction (XRD), Synthesised Nanocomposite brought about notable antibacterial activity against urinary tract pathogenic strains by recording potential inhibitory effect on the expression of Las R gene. Inhibition of Las R gene expression reduced notable effect on biofilm development. Anticancer activity against prostate cancer cells (Pc3) was investigated by measurement of HOXB13 gene expression level. Inhibition of HOXB13 gene expression by the IONC brought about structural, functional changes. HOXB13 gene expression inhibition reveals a remarkable cytotoxic effect by recording decreased cell viability. Morphometric analysis by phase-contrast and DAPI fluorescence staining demonstrates that the Nanocomposite prompted cell morphology anomalies or apoptotic changes. Nanocomposite treatment brought about a good sign of Apoptosis by recording enhanced caspase 3 and 9 activities, DNA fragmentation and elevated reactive oxygen species generation (ROS). Hemocompatibility studies were carried out to determine the biocompatibility of the Nanocomposite. Spectrophotometric estimation of plasma haemoglobin, microscopic examination of whole blood cells shows the Nanocomposite was not inciting any indication of toxicity. These findings infer that IONC synthesised in the present study is the promising contender for a broad scope of biomedical applications, especially as an antibacterial and anticancer agent.


Assuntos
Compostos Férricos/química , Genes vpr , Goma Arábica/química , Proteínas de Homeodomínio/genética , Nanocompostos/química , Polietilenoglicóis/química , Neoplasias da Próstata/genética , Pseudomonas aeruginosa/genética , Antibacterianos/farmacologia , Antineoplásicos/farmacologia , Biofilmes/efeitos dos fármacos , Caspase 3/metabolismo , Catéteres , Fragmentação do DNA/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Química Verde , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Testes de Sensibilidade Microbiana , Nanocompostos/ultraestrutura , Células PC-3 , Filogenia , Pseudomonas aeruginosa/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
12.
Sci Rep ; 11(1): 17912, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34504158

RESUMO

A recently developed Phox2a::Cre mouse line has been shown to capture anterolateral system (ALS) projection neurons. Here, we used this line to test whether Phox2a-positive cells represent a distinct subpopulation among lamina I ALS neurons. We show that virtually all lamina I Phox2a cells can be retrogradely labelled from injections targeted on the lateral parabrachial area (LPb), and that most of those in the cervical cord also belong to the spinothalamic tract. Phox2a cells accounted for ~ 50-60% of the lamina I cells retrogradely labelled from LPb or thalamus. Phox2a was preferentially associated with smaller ALS neurons, and with those showing relatively weak neurokinin 1 receptor expression. The Phox2a cells were also less likely to project to the ipsilateral LPb. Although most Phox2a cells phosphorylated extracellular signal-regulated kinases following noxious heat stimulation, ~ 20% did not, and these were significantly smaller than the activated cells. This suggests that those ALS neurons that respond selectively to skin cooling, which have small cell bodies, may be included among the Phox2a population. Previous studies have defined neurochemical populations among the ALS cells, based on expression of Tac1 or Gpr83. However, we found that the proportions of Phox2a cells that expressed these genes were similar to the proportions reported for all lamina I ALS neurons, suggesting that Phox2a is not differentially expressed among cells belonging to these populations. Finally, we used a mouse line that resulted in membrane labelling of the Phox2a cells and showed that they all possess dendritic spines, although at a relatively low density. However, the distribution of the postsynaptic protein Homer revealed that dendritic spines accounted for a minority of the excitatory synapses on these cells. Our results confirm that Phox2a-positive cells in lamina I are ALS neurons, but show that the Phox2a::Cre line preferentially captures specific types of ALS cells.


Assuntos
Proteínas de Homeodomínio/metabolismo , Neurônios , Corno Dorsal da Medula Espinal , Animais , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Corno Dorsal da Medula Espinal/citologia , Corno Dorsal da Medula Espinal/metabolismo , Sinapses , Tálamo/citologia
13.
Int J Mol Sci ; 22(17)2021 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-34502163

RESUMO

Garlic (Allium sativum L.) is an important vegetable and is cultivated and consumed worldwide for its economic and medicinal values. Garlic cloves, the major reproductive and edible organs, are derived from the axillary meristems. KNOTTED-like homeobox (KNOX) proteins, such as SHOOT MERISTEM-LESS (STM), play important roles in axillary meristem formation and development. However, the KNOX proteins in garlic are still poorly known. Here, 10 AsKNOX genes, scattered on 5 of the 8 chromosomes, were genome-wide identified and characterized based on the newly released garlic genome. The typical conserved domains of KNOX proteins were owned by all these 10 AsKNOX homologs, which were divided into two Classes (Class I and Class II) based on the phylogenetic analysis. Prediction and verification of the subcellular localizations revealed the diverse subcellular localization of these 10 AsKNOX proteins. Cis-element prediction, tissue expression analysis, and expression profilings in responding to exogenous GA3 and 6-BA showed the potential involvement of AsKNOX genes in the gibberellin and cytokinin signaling pathways. Overall, the results of this work provided a better understanding of AsKNOX genes in garlic and laid an important foundation for their further functional studies.


Assuntos
Citocininas/farmacologia , Alho/genética , Giberelinas/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Plantas/genética , Alho/efeitos dos fármacos , Alho/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Plantas/metabolismo , Transdução de Sinais
14.
Mol Metab ; 53: 101312, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34329773

RESUMO

OBJECTIVE: Proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus are essential regulators of energy balance. Selective loss of POMC production in these cells results in extreme obesity and metabolic comorbidities. Neurogenesis occurs in the adult hypothalamus, but it remains uncertain whether functional POMC neurons emerge in physiologically significant numbers during adulthood. Here, we tested whether Rax-expressing precursors generate POMC neurons in adult mice and rescue the metabolic phenotype caused by congenital hypothalamic POMC deficiency. METHODS: Initially, we identified hypothalamic Rax-expressing cell types using wild-type and Rax-CreERT2:Ai34D mice. Then we generated compound Rax-CreERT2:ArcPomcloxTB/loxTB mice in which endogenous hypothalamic Pomc expression is silenced, but can be restored by tamoxifen administration selectively in neurons derived from Rax+ progenitors. The number of POMC neurons generated by Rax+ progenitors in adult mice and their axonal projections was determined. The metabolic effects of these neurons were assessed by measuring food intake, bodyweight, and body composition, along with glucose and insulin levels. RESULTS: We found that Rax is expressed by tanycytes and a previously unrecognized cell type in the hypothalamic parenchyma of adult mice. Rax+ progenitors generated ~10% of the normal adult hypothalamic POMC neuron population within two weeks of tamoxifen treatment. The same rate and steady state of POMC neurogenesis persisted from young adult to aged mice. These new POMC neurons established terminal projections to brain regions that were involved in energy homeostasis. Mice with Rax+ progenitor-derived POMC neurons had reduced body fat mass, improved glucose tolerance, increased insulin sensitivity, and decreased bodyweight in proportion to the number of new POMC neurons. CONCLUSIONS: These data demonstrate that Rax+ progenitors generate POMC neurons in sufficient numbers during adulthood to mitigate the metabolic abnormalities of hypothalamic POMC-deficient mice. The findings suggest that adult hypothalamic neurogenesis is a robust phenomenon in mice that can significantly impact energy homeostasis.


Assuntos
Insuficiência Adrenal/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/deficiência , Pró-Opiomelanocortina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição/genética
15.
Circulation ; 144(8): 638-654, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34098726

RESUMO

BACKGROUND: Cardiac hypertrophy is an important prepathology of, and will ultimately lead to, heart failure. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. This study aims to elucidate the effects and mechanisms of HINT1 (histidine triad nucleotide-binding protein 1) in cardiac hypertrophy and heart failure. METHODS: HINT1 was downregulated in human hypertrophic heart samples compared with nonhypertrophic samples by mass spectrometry analysis. Hint1 knockout mice were challenged with transverse aortic constriction surgery. Cardiac-specific overexpression of HINT1 mice by intravenous injection of adeno-associated virus 9 (AAV9)-encoding Hint1 under the cTnT (cardiac troponin T) promoter were subjected to transverse aortic construction. Unbiased transcriptional analyses were used to identify the downstream targets of HINT1. AAV9 bearing shRNA against Hoxa5 (homeobox A5) was administrated to investigate whether the effects of HINT1 on cardiac hypertrophy were HOXA5-dependent. RNA sequencing analysis was performed to recapitulate possible changes in transcriptome profile.Coimmunoprecipitation assays and cellular fractionation analyses were conducted to examine the mechanism by which HINT1 regulates the expression of HOXA5. RESULTS: The reduction of HINT1 expression was observed in the hearts of hypertrophic patients and pressure overloaded-induced hypertrophic mice, respectively. In Hint1-deficient mice, cardiac hypertrophy deteriorated after transverse aortic construction. Conversely, cardiac-specific overexpression of HINT1 alleviated cardiac hypertrophy and dysfunction. Unbiased profiler polymerase chain reaction array showed HOXA5 is 1 target for HINT1, and the cardioprotective role of HINT1 was abolished by HOXA5 knockdown in vivo. Hoxa5 was identified to affect hypertrophy through the TGF-ß (transforming growth factor ß) signal pathway. Mechanically, HINT1 inhibited PKCß1 (protein kinase C ß type 1) membrane translocation and phosphorylation via direct interaction, attenuating the MEK/ERK/YY1 (mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/yin yang 1) signal pathway, downregulating HOXA5 expression, and eventually attenuating cardiac hypertrophy. CONCLUSIONS: HINT1 protects against cardiac hypertrophy through suppressing HOXA5 expression. These findings indicate that HINT1 may be a potential target for therapeutic interventions in cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/metabolismo , Animais , Biomarcadores , Cardiomegalia/diagnóstico , Células Cultivadas , Bases de Dados Genéticas , Modelos Animais de Doenças , Suscetibilidade a Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
16.
J Cardiovasc Pharmacol ; 78(3): 411-421, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34132687

RESUMO

ABSTRACT: Calcific aortic valve disease is a common heart disease that contributes to increased cardiovascular morbidity and mortality. There is a lack of effective pharmaceutical therapy because its mechanisms are not yet fully known. Ginkgo biloba extract (EGB761) is reported to alleviate vascular calcification. However, whether EGB761 protects against aortic valve calcification, a disease whose pathogenesis shares many similarities with vascular calcification, and potential molecular mechanisms remain unknown. In this study, porcine aortic valve interstitial cell (pAVIC) calcification was induced by warfarin with or without the presence of EGB761. Immunostaining was performed to establish and characterize the pAVIC phenotype. Calcium deposition and calcium content were examined by Alizarin Red S staining and an intracellular calcium content assay. Alkaline phosphatase activity was detected by the p-nitrophenyl phosphate method. The expression levels of bone morphogenetic protein-2 (BMP2), Runt-related transcription factor 2 (Runx2), homeobox protein MSX-2, and phosphorylated (p)-Smad1/5 were detected by reverse transcription-quantitative polymerase chain reaction (PCR) and Western blot analysis. Consistent with these in vitro data, we also confirmed the suppression of in vivo calcification by EGB761 in the warfarin-induced C57/Bl6 mice. The results indicated that both pAVICs and aortic valves tissue of mice stimulated with warfarin showed increased calcium deposition and expression of osteogenic markers (alkaline phosphatase, BMP2, homeobox protein MSX-2, and Runx2) and promoted p-Smad1/5 translocation from the cytoplasm to the nucleus. The addition of EGB761 significantly inhibited p-Smad1/5 translocation from the cytoplasm to the nucleus, thus suppressing calcification. In conclusion, EGB761 could ameliorate warfarin-induced aortic valve calcification through the inhibition of the BMP2-medicated Smad1/5/Runx2 signaling pathway.


Assuntos
Valva Aórtica/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Calcinose/prevenção & controle , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Doenças das Valvas Cardíacas/prevenção & controle , Extratos Vegetais/farmacologia , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Calcinose/induzido quimicamente , Calcinose/metabolismo , Calcinose/patologia , Cálcio/metabolismo , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Ginkgo biloba , Doenças das Valvas Cardíacas/induzido quimicamente , Doenças das Valvas Cardíacas/metabolismo , Doenças das Valvas Cardíacas/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Osteogênese/efeitos dos fármacos , Fosforilação , Transdução de Sinais , Sus scrofa , Varfarina
17.
Mol Psychiatry ; 26(11): 6550-6561, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33967268

RESUMO

Activity-dependent neuroprotective protein (ADNP) is essential for brain formation and function. As such, de novo mutations in ADNP lead to the autistic ADNP syndrome and somatic ADNP mutations may drive Alzheimer's disease (AD) tauopathy. Sirtuin 1 (SIRT1) is positively associated with aging, the major risk for AD. Here, we revealed two key interaction sites for ADNP and SIRT1. One, at the microtubule end-binding protein (EB1 and EB3) Tau level, with EB1/EB3 serving as amplifiers for microtubule dynamics, synapse formation, axonal transport, and protection against tauopathy. Two, on the DNA/chromatin site, with yin yang 1, histone deacetylase 2, and ADNP, sharing a DNA binding motif and regulating SIRT1, ADNP, and EB1 (MAPRE1). This interaction was linked to sex- and age-dependent altered histone modification, associated with ADNP/SIRT1/WD repeat-containing protein 5, which mediates the assembly of histone modification complexes. Single-cell RNA and protein expression analyses as well as gene expression correlations placed SIRT1-ADNP and either MAPRE1 (EB1), MAPRE3 (EB3), or both in the same mouse and human cell; however, while MAPRE1 seemed to be similarly regulated to ADNP and SIRT1, MAPRE3 seemed to deviate. Finally, we demonstrated an extremely tight correlation for the gene transcripts described above, including related gene products. This correlation was specifically abolished in affected postmortem AD and Parkinson's disease brain select areas compared to matched controls, while being maintained in blood samples. Thus, we identified an ADNP-SIRT1 complex that may serve as a new target for the understanding of brain degeneration.


Assuntos
Histonas , Sirtuína 1 , Animais , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Metilação , Camundongos , Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo
18.
J Immunother Cancer ; 9(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33883257

RESUMO

BACKGROUND: In the tumor microenvironment, tumor cells are able to suppress antitumor immunity by competing for essential nutrients, including amino acids. However, whether amino acid depletion modulates the activity of CD8+ tumor-infiltrating lymphocytes (TILs) is unclear. METHOD: In this study, we evaluated the roles of amino acids and the Rag complex in regulating mammalian target of rapamycin complex 1 (mTORC1) signaling in CD8+ TILs. RESULTS: We discovered that the Rag complex, particularly RagD, was crucial for CD8+ T-cell antitumor immunity. RagD expression was positively correlated with the antitumor response of CD8+ TILs in both murine syngeneic tumor xenografts and clinical human colon cancer samples. On RagD deficiency, CD8+ T cells were rendered more dysfunctional, as demonstrated by attenuation of mTORC1 signaling and reductions in proliferation and cytokine secretion. Amino acids maintained RagD-mediated mTORC1 translocation to the lysosome, thereby achieving maximal mTORC1 activity in CD8+ T cells. Moreover, the limited T-cell access to leucine (LEU), overshadowed by tumor cell amino acid consumption, led to impaired RagD-dependent mTORC1 activity. Finally, combined with antiprogrammed cell death protein 1 antibody, LEU supplementation improved T-cell immunity in MC38 tumor-bearing mice in vivo. CONCLUSION: Our results revealed that robust signaling of amino acids by RagD and downstream mTORC1 signaling were crucial for T-cell receptor-initiated antitumor immunity. The characterization the role of RagD and LEU in nutrient mTORC1 signaling in TILs might suggest potential therapeutic strategies based on the manipulation of RagD and its upstream pathway.


Assuntos
Linfócitos T CD8-Positivos/enzimologia , Leucina/metabolismo , Linfócitos do Interstício Tumoral/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Melanoma Experimental/enzimologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neoplasias Cutâneas/enzimologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Ativação Enzimática , Células HEK293 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ativação Linfocitária , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Evasão Tumoral , Microambiente Tumoral
19.
Dokl Biochem Biophys ; 497(1): 130-136, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33895929

RESUMO

The present study investigated Punica granatum extract (PGE) as potential proliferation inhibitory agent for bladder cancer cells and elucidated the possible mechanism. PGE reduced viabilities of HT-1197 and RT4 cells in concentration-based manner at 72 h. Colony forming potential of HT-1197 and RT4 cells was also significantly (p < 0.05) inhibited on exposure to 2 and 12 mg/mL PGE. Exposure to 12 mg/mL PGE for 72 h significantly (p < 0.05) decreased miR­10b expression and suppressed migration potential of HT-1197 and RT4 cells. In PGE exposed HT-1197 and RT4 cells, invasiveness was reduced to 30.25 and 33.47%, respectively. PGE treatment of HT-1197 and RT4 cells caused a significant (p < 0.05) elevation in HOXD10 protein and mRNA levels compared to control. The miR­10b mimic transfection in HT-1197 and RT4 cells reversed inhibitory effect of PGE on cell viability. Thus, PGE exhibited cytotoxicity and anti-invasive effect on HT-1197 and RT4 cells through targeting miR­10b and up-regulation of HOXD10 expression. Thus, PGE may be developed as therapeutic agent for treatment of bladder cancer.


Assuntos
Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Extratos Vegetais/farmacologia , Punica granatum/química , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Neoplasias da Bexiga Urinária/patologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Humanos , Invasividade Neoplásica , Fatores de Tempo
20.
Cell Rep ; 35(3): 109016, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33882317

RESUMO

The mammalian cochlea cannot regenerate functional hair cells (HCs) spontaneously. Atoh1 overexpression as well as other strategies are unable to generate functional HCs. Here, we simultaneously upregulated the expression of Gfi1, Pou4f3, and Atoh1 in postnatal cochlear supporting cells (SCs) in vivo, which efficiently converted SCs into HCs. The newly regenerated HCs expressed HC markers Myo7a, Calbindin, Parvalbumin, and Ctbp2 and were innervated by neurites. Importantly, many new HCs expressed the mature and terminal marker Prestin or vesicular glutamate transporter 3 (vGlut3), depending on the subtypes of the source SCs. Finally, our patch-clamp analysis showed that the new HCs in the medial region acquired a large K+ current, fired spikes transiently, and exhibited signature refinement of ribbon synapse functions, in close resemblance to native wild-type inner HCs. We demonstrated that co-upregulating Gfi1, Pou4f3, and Atoh1 enhances the efficiency of HC generation and promotes the functional maturation of new HCs.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ligação a DNA/genética , Células Ciliadas Auditivas/metabolismo , Proteínas de Homeodomínio/genética , Células Labirínticas de Suporte/metabolismo , Organogênese/genética , Fator de Transcrição Brn-3C/genética , Fatores de Transcrição/genética , Potenciais de Ação/fisiologia , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Calbindinas/genética , Calbindinas/metabolismo , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/citologia , Proteínas de Homeodomínio/metabolismo , Transporte de Íons , Células Labirínticas de Suporte/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/metabolismo , Miosina VIIa/genética , Miosina VIIa/metabolismo , Neuritos/metabolismo , Neuritos/ultraestrutura , Parvalbuminas/genética , Parvalbuminas/metabolismo , Técnicas de Patch-Clamp , Potássio/metabolismo , Transdução de Sinais , Fator de Transcrição Brn-3C/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA