Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 83(7): 1093-1108.e8, 2023 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-36863348

RESUMO

The glucagon-PKA signal is generally believed to control hepatic gluconeogenesis via the CREB transcription factor. Here we uncovered a distinct function of this signal in directly stimulating histone phosphorylation for gluconeogenic gene regulation in mice. In the fasting state, CREB recruited activated PKA to regions near gluconeogenic genes, where PKA phosphorylated histone H3 serine 28 (H3S28ph). H3S28ph, recognized by 14-3-3ζ, promoted recruitment of RNA polymerase II and transcriptional stimulation of gluconeogenic genes. In contrast, in the fed state, more PP2A was found near gluconeogenic genes, which counteracted PKA by dephosphorylating H3S28ph and repressing transcription. Importantly, ectopic expression of phosphomimic H3S28 efficiently restored gluconeogenic gene expression when liver PKA or CREB was depleted. These results together highlight a different functional scheme in regulating gluconeogenesis by the glucagon-PKA-CREB-H3S28ph cascade, in which the hormone signal is transmitted to chromatin for rapid and efficient gluconeogenic gene activation.


Assuntos
Glucagon , Gluconeogênese , Animais , Camundongos , Gluconeogênese/genética , Glucagon/metabolismo , Histonas/metabolismo , Fosforilação , Proteínas 14-3-3/metabolismo , Fígado/metabolismo , Jejum/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo
2.
Nat Commun ; 14(1): 1521, 2023 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-36934083

RESUMO

The early phase lipid accumulation is essential for liver regeneration. However, whether this acute lipid accumulation can serve as signals to direct liver regeneration rather than simply providing building blocks for cell proliferation remains unclear. Through in vivo CRISPR screening, we identify MIER1 (mesoderm induction early response 1) as a key epigenetic regulator that bridges the acute lipid accumulation and cell cycle gene expression during liver regeneration in male animals. Physiologically, liver acute lipid accumulation induces the phosphorylation of EIF2S1(eukaryotic translation initiation factor 2), which consequently attenuated Mier1 translation. MIER1 downregulation in turn promotes cell cycle gene expression and regeneration through chromatin remodeling. Importantly, the lipids-EIF2S1-MIER1 pathway is impaired in animals with chronic liver steatosis; whereas MIER1 depletion significantly improves regeneration in these animals. Taken together, our studies identify an epigenetic mechanism by which the early phase lipid redistribution from adipose tissue to liver during regeneration impacts hepatocyte proliferation, and suggest a potential strategy to boost liver regeneration.


Assuntos
Proteínas de Ligação a DNA , Epigênese Genética , Fígado Gorduroso , Regeneração Hepática , Fatores de Transcrição , Animais , Masculino , Camundongos , Proliferação de Células/genética , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Hepatectomia , Hepatócitos/metabolismo , Lipídeos , Fígado/metabolismo , Regeneração Hepática/genética , Camundongos Endogâmicos C57BL , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
3.
Adv Sci (Weinh) ; 10(3): e2205462, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36453571

RESUMO

Acetylation of extracellular proteins has been observed in many independent studies where particular attention has been given to the dynamic change of the microenvironmental protein post-translational modifications. While extracellular proteins can be acetylated within the cells prior to their micro-environmental distribution, their deacetylation in a tumor microenvironment remains elusive. Here it is described that multiple acetyl-vWA domain-carrying proteins including integrin ß3 (ITGB3) and collagen 6A (COL6A) are deacetylated by Sirtuin family member SIRT2 in extracellular space. SIRT2 is secreted by macrophages following toll-like receptor (TLR) family member TLR4 or TLR2 activation. TLR-activated SIRT2 undergoes autophagosome translocation. TNF receptor associated factor 6 (TRAF6)-mediated autophagy flux in response to TLR2/4 activation can then pump SIRT2 into the microenvironment to function as extracellular SIRT2 (eSIRT2). In the extracellular space, eSIRT2 deacetylates ITGB3 on aK416 involved in cell attachment and migration, leading to a promotion of cancer cell metastasis. In lung cancer patients, significantly increased serum eSIRT2 level correlates with dramatically decreased ITGB3-K416 acetylation in cancer cells. Thus, the extracellular space is a subcellular organelle-like arena where eSIRT2 promotes cancer cell metastasis via catalyzing extracellular protein deacetylation.


Assuntos
Neoplasias Pulmonares , Sirtuína 2 , Humanos , Sirtuína 2/genética , Sirtuína 2/metabolismo , Receptor 2 Toll-Like/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Microambiente Tumoral
4.
J Hepatol ; 77(6): 1491-1503, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35985547

RESUMO

BACKGROUND & AIMS: How hepatic steatosis progresses to non-alcoholic steatohepatitis (NASH) is complicated and remains unclear. The mortality factor 4-like protein 1 (MORF4L1, also called MRG15) was previously identified as a master nuclear chromatin remodeler in the rhythmic regulation of lipid synthesis gene expression in the liver. Whether it also contributes to the progression from liver steatosis to NASH is unclear. METHODS: We adopted 2 different murine NASH models, liver biopsies from patients with NASH, and primary mouse and human hepatocyte cultures for functional examination of MRG15 in NASH progression. Immunoprecipitation-mass spectrometry was applied to identify protein partners of MRG15, and CRISPR targeting was used for gene depletion in liver cells in vivo. RESULTS: The MRG15 level is increased in the livers of humans and mice with NASH. The inflammatory cytokines in NASH livers stabilize MRG15 by increasing its acetylation. Considerable amounts of MRG15 associate with the outer mitochondrial membrane, where it interacts with and deacetylates the mitochondrial Tu translation elongation factor (TUFM). Deacetylated TUFM, especially at the K82 and K91 sites, is subjected to accelerated degradation by the mitochondrial ClpXP protease system. Reduced liver TUFM consequently results in impaired mitophagy, increased oxidative stress and activation of the NLRP3 inflammasome pathway. Blocking MRG15 expression protects the liver from NASH progression by increasing the stability of liver TUFM. Liver samples from patients with NASH also display a clear reduction in TUFM level, which correlates with increased MRG15 expression. CONCLUSION: Collectively, these findings uncover a mitochondrial MRG15-TUFM regulatory pathway that contributes significantly to progression from simple steatosis to NASH, and which could potentially be targeted to treat NASH. LAY SUMMARY: The incidence of non-alcoholic fatty liver disease and its progressive form non-alcoholic steatohepatitis (NASH) is increasing, posing a significant global health challenge. Herein, we have uncovered the importance of the MRG15-TUFM pathway in NASH development. This pathway is active in the mitochondria (energy powerhouse of the cell) and could be targeted for the treatment of NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Transativadores , Animais , Humanos , Camundongos , Proteínas Cromossômicas não Histona , Mitofagia , Peptídeo Hidrolases , Proteólise
5.
J Bone Oncol ; 35: 100436, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35733786

RESUMO

Background: Circular RNA (circRNA) is a newly-discovered endogenous transcript that has been reported to participate in osteosarcoma (OS) progression. However, the underlying mechanism of circ_0051079 modulating OS development remains unclear. Methods: RNA expressions of circ_0051079, miR-625-5p and tripartite motif containing 66 (TRIM66) were detected by quantitative real-time polymerase chain reaction. Protein expression was checked by Western blot analysis. The functional effects of circ_0051079 on OS cell malignancy were investigated by cell counting kit-8, clonogenicity, transwell, tube formation and flow cytometry assays. The interactions among circ_0051079, miR-625-5p and TRIM66 were identified by dual-luciferase reporter and RNA immunoprecipitation assays. Mouse xenograft model assay was performed to elucidate the effects of circ_0051079 knockdown on tumor formation in vivo. Results: Circ_0051079 and TRIM66 expressions were significantly upregulated, but miR-625-5p was downregulated in OS tissues and cells compared with control groups. Circ_0051079 expression was significantly associated with tumor-node-metastasis stage and tumor size of OS patients. Circ_0051079 knockdown inhibited OS cell proliferation, migration and invasion, repressed angiogenesis but induced cell apoptosis, accompanied by the decreases of PCNA and Bcl-2 production and an increase of Bax production. MiR-625-5p, a target miRNA of circ_0051079, participated in regulating circ_0051079-induced effects. Also, TRIM66 was identified as a target mRNA of miR-625-5p, and partially attenuated the inhibitory effects of miR-625-5p in OS cells. Circ_0051079 modulated the Wnt/ß-catenin pathway through TRIM66 in vitro. Importantly, circ_0051079 silencing reduced TRIM66 expression by interacting with miR-625-5p. Further, circ_0051079 depletion inhibited tumor formation in vivo. Conclusion: Circ_0051079 regulated OS development by the miR-625-5p/TRIM66/Wnt/ß-catenin pathway, providing a novel therapeutic target for OS.

6.
ACS Omega ; 7(14): 11839-11852, 2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35449961

RESUMO

Ubiquitination is a major posttranslational modification of proteins that affects their stability, and E3 ligases play a key role in ubiquitination by specifically recognizing their substrates. BTBD9, an adaptor of the Cullin-RING ligase complex, is responsible for substrate recognition and is associated with sleep homeostasis. However, the substrates of BTBD9-mediated ubiquitination remain unknown. Here, we generated an SH-SY5Y cell line stably expressing BTBD9 and performed proteomic analysis combined with ubiquitinome analysis to identify the downstream targets of BTBD9. Through this approach, we identified four potential BTBD9-mediated ubiquitination substrates that are targeted for degradation. Among these candidate substrates, inosine monophosphate dehydrogenase (IMPDH2), a novel target of BTBD9-mediated degradation, is a potential risk gene for sleep dysregulation. In conclusion, these findings not only demonstrate that proteomic analysis can be a useful general approach for the systematic identification of E3 ligase substrates but also identify novel substrates of BTBD9, providing a resource for future studies of sleep regulation mechanisms.

7.
Gene Ther ; 28(3-4): 155-161, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32994568

RESUMO

Follicle-stimulating hormone (FSH) is a pituitary glycoprotein that regulates follicle maturation through its binding to follicle-stimulating hormone receptor (FSHR). Endothelial cells express FSHR, but its exact role in endothelial cells remains unclear. Here we show that FSHR expression was detectable in human umbilical vein endothelial cells (HUVECs). FSH stimulation promoted HUVECs migration but not proliferation. Because FSHR is a GPCR, FSH treatment triggers the activation of cAMP-PKA signaling pathways, and the JAK-STAT, PI3K-AKT, and JNK-MAPK pathways. RNAi of FSHR dramatically attenuated the activation effect of FSH on HUVECs migration, as well as the related signaling pathways. Treatment of FSH in HUVECs also transcriptionally upregulated the expression of VAV3 and LAMA2, suppression either of VAV3 or LAMA2 by RNAi attenuated the FSH's effect on HUVECs migration. All of these results indicated a functional role of FSH in the regulation of endothelial cells.


Assuntos
Movimento Celular , Hormônio Foliculoestimulante Humano/metabolismo , Hormônio Foliculoestimulante , Receptores do FSH/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Receptores do FSH/genética , Transdução de Sinais
8.
iScience ; 23(9): 101446, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32829287

RESUMO

Identification of safe and effective compounds to increase or activate UCP1 expression in brown or white adipocytes remains a potent therapeutic strategy to combat obesity. Here we reported that, glyburide, one of the FDA-approved drugs currently used to treat type 2 diabetes, can significantly enhance UCP1 expression in both brown and white adipocytes. Glyburide-fed mice exhibited a clear resistance to high-fat diet-induced obesity, reduced blood triglyceride level, and increased UCP1 expression in brown adipose tissue. Moreover, in situ injection of glyburide to inguinal white adipose tissue remarkably enhanced UCP1 expression and increased thermogenesis. Further mechanistic studies indicated that the glyburide effect in UCP1 expression in adipocytes was KATP channel independent but may involve the regulation of the Ca2+-Calcineurin-NFAT signal pathway. Overall, our findings revealed the significant effects of glyburide in regulating UCP1 expression and thermogenesis in adipocytes, which can be potentially repurposed to treat obesity.

9.
Nat Metab ; 2(5): 447-460, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32694659

RESUMO

The rhythmic regulation of transcriptional processes is intimately linked to lipid homeostasis, to anticipate daily changes in energy access. The Rev-erbα-HDAC3 complex was previously discovered to execute the rhythmic repression of lipid genes; however, the epigenetic switch that turns on these genes is less clear. Here, we show that genomic recruitment of MRG15, which is encoded by the mortality factor on chromosome 4 (MORF4)-related gene on chromosome 15, displays a significant diurnal rhythm and activates lipid genes in the mouse liver. RNA polymerase II (Pol II) recruitment and histone acetylation correspond to MRG15 binding, and the rhythm is impaired upon MRG15 depletion, establishing MRG15 as a key modulator in global rhythmic transcriptional regulation. MRG15 interacts with the nuclear receptor LRH-1, rather than with known core clock proteins, and is recruited to genomic loci near lipid genes via LRH-1. Blocking of MRG15 by CRISPR targeting or by the FDA-approved drug argatroban, which is an antagonist to MRG15, attenuates liver steatosis. This work highlights MRG15 as a targetable master regulator in the rhythmic regulation of hepatic lipid metabolism.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética/genética , Epigênese Genética/fisiologia , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Transativadores/genética , Transativadores/metabolismo , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Arginina/uso terapêutico , Linhagem Celular , Ritmo Circadiano , Epigênese Genética/efeitos dos fármacos , Epigenômica , Fígado Gorduroso/tratamento farmacológico , Teste de Tolerância a Glucose , Histonas/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Pipecólicos/farmacologia , Ácidos Pipecólicos/uso terapêutico , RNA Polimerase II/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico
10.
Signal Transduct Target Ther ; 5(1): 52, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32355204

RESUMO

Wnt/ß-catenin signaling plays a critical role in colorectal cancer (CRC) tumorigenesis and the homeostasis of colorectal cancer stem cells (CSCs), but its molecular mechanism remains unclear. B-cell lymphoma 3 (Bcl-3), a member of the IκB family, is overexpressed in CRC and promotes tumorigenicity. Here, we report a novel function of Bcl-3 in maintaining colorectal CSC homeostasis by activating Wnt/ß-catenin signaling. Silencing Bcl-3 suppresses the self-renewal capacity of colorectal CSCs and sensitizes CRC cells to chemotherapeutic drugs through a decrease in Wnt/ß-catenin signaling. Moreover, our data show that Bcl-3 is a crucial component of Wnt/ß-catenin signaling and is essential for ß-catenin transcriptional activity in CRC cells. Interestingly, Wnt3a increases the level and nuclear translocation of Bcl-3, which binds directly to ß-catenin and enhances the acetylation of ß-catenin at lysine 49 (Ac-K49-ß-catenin) and transcriptional activity. Bcl-3 depletion decreases the Ac-K49-ß-catenin level by increasing the level of histone deacetylase 1 to remove acetyl groups from ß-catenin, thus interrupting Wnt/ß-catenin activity. In CRC clinical specimens, Bcl-3 expression negatively correlates with the overall survival of CRC patients. A significantly positive correlation was found between the expression of Bcl-3 and Ac-K49-ß-catenin. Collectively, our data reveal that Bcl-3 plays a crucial role in CRC chemoresistance and colorectal CSC maintenance via its modulation of the Ac-K49-ß-catenin, which serves as a promising therapeutic target for CRC.


Assuntos
Proteína 3 do Linfoma de Células B/metabolismo , Neoplasias Colorretais/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Acetilação , Proteína 3 do Linfoma de Células B/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Taxa de Sobrevida , beta Catenina/genética
11.
Cell Death Dis ; 11(1): 39, 2020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-31959745

RESUMO

Ginsenosides exhibit a large variety of biological activities in maintaining physical health; however, the molecule underpinnings underlining these biological activities remain to be defined. Here, we took a cellular condition that compound K (CK) induces autophagic cell death in HeLa cells, and setup a high-throughput genetic screening using CRISPR technology. We have identified a number of CK-resistant and CK-sensitive genes, and further validated PMAIP1 as a CK-resistant gene and WASH1 as a CK-sensitive gene. Compound K treatment reduces the expression of WASH1, which further accelerates the autophagic cell death, highlighting WASH1 as an interesting downstream mediator of CK effects. Overall, our study offers an easy-to-adopt platform to study the functional mediators of ginsenosides, and provides a candidate list of genes that are potential targets of CK.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Genoma , Ginsenosídeos/farmacologia , Autofagia/efeitos dos fármacos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células HEK293 , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
12.
Cell Rep ; 29(3): 781-784, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31618644

RESUMO

This Matters Arising Response paper addresses the Hoch et al. (2019) Matters Arising paper published concurrently in this issue of Cell Reports. The genetic study in humans revealed a strong association of DNA variants in the SLC16A11 coding region with type 2 diabetes mellitus (T2DM). However, how these T2D variants affect the function of SLC16A11 remains controversial. In Zhao et al. (2019), with studies using genetic knockout mouse models and in vivo gene reconstitution experiments, we demonstrated gain of aberrant functions of mutant SLC16A11-carrying T2D variants, which cause liver steatosis and insulin resistance. Hoch et al. (2019) raise concerns regarding the animal models and experimental settings used in the study. Here, we address their concerns and emphasize that discoveries from the physiological studies of SLC16A11 by using mouse models disagree with the previous proposal by Rusu et al. (2017) that "therapeutics that enhance SLC16A11 levels or activity may be beneficial for T2D."


Assuntos
Diabetes Mellitus Tipo 2/genética , Resistência à Insulina/genética , Animais , Mutação com Ganho de Função , Humanos , Camundongos , Transportadores de Ácidos Monocarboxílicos/genética , Fases de Leitura Aberta
13.
World J Stem Cells ; 11(8): 535-547, 2019 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-31523372

RESUMO

Human hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) promise a valuable source of cells with human genetic background, physiologically relevant liver functions, and unlimited supply. With over 10 years' efforts in this field, great achievements have been made. HLCs have been successfully derived and applied in disease modeling, toxicity testing and drug discovery. Large cohorts of induced pluripotent stem cells-derived HLCs have been recently applied in studying population genetics and functional outputs of common genetic variants in vitro. This has offered a new paradigm for genome-wide association studies and possibly in vitro pharmacogenomics in the nearly future. However, HLCs have not yet been successfully applied in bioartificial liver devices and have only displayed limited success in cell transplantation. HLCs still have an immature hepatocyte phenotype and exist as a population with great heterogeneity, and HLCs derived from different hPSC lines display variable differentiation efficiency. Therefore, continuous improvement to the quality of HLCs, deeper investigation of relevant biological processes, and proper adaptation of recent advances in cell culture platforms, genome editing technology, and bioengineering systems are required before HLCs can fulfill the needs in basic and translational research. In this review, we summarize the discoveries, achievements, and challenges in the derivation and applications of HLCs.

14.
Oral Dis ; 25(7): 1789-1797, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31283861

RESUMO

BACKGROUND: Porphyromonas gingivalis is the main pathogen of periodontal disease affecting over half of the worldwide adult population. Recent studies have shown that P. gingivalis is related to the development of non-alcoholic fatty liver disease (NAFLD), a global major chronic liver disease, especially in developed countries. However, how P. gingivalis contributes to the pathogenesis of NAFLD has not been fully clarified. We aimed to conduct a preliminary exploration of the underlying mechanism of P. gingivalis infection in the development of NAFLD. METHODS: Human hepatocellular cells HepG2 were incubated with/without oleic acid (OA) and tested for lipid accumulation upon stimulation by lipopolysaccharide (LPS) derived from P. gingivalis or Escherichia coli. Intracellular lipid droplet formation was analyzed and quantified by Oil Red O staining. The involvement of signaling pathway molecules and pro-inflammatory cytokines related to NF-κB and MAPKs were examined with Western blot and quantitative real-time PCR (qRT-PCR) analyses and further evaluated with inhibitor treatment and RNA interference. RESULTS: HepG2 cells accumulated more intracellular lipids when stimulated with P. gingivalis LPS, as compared to cells treated with E. coli LPS or control. Further pathway analysis demonstrated that after stimulation with P. gingivalis LPS, cells displayed significantly upregulated MyD88 expression, increased phosphorylation of p65 and JNK, and more release of pro-inflammatory cytokines, such as IL-1, IL-8, and TNF-α. In addition, suppression of phosphorylation of p65 and JNK by inhibitors and RNA interference resulted in a reduction in lipid accumulation upon P. gingivalis LPS treatment. CONCLUSIONS: These results suggest that P. gingivalis-derived LPS may contribute to intracellular lipid accumulation and inflammatory reaction of HepG2 cells via the activation of NF-κB and JNK signaling pathways. This study offers a possible explanation to the functional involvement of P. gingivalis infection in the pathological progression of NAFLD. These findings may help design new treatment strategies in NAFLD.


Assuntos
Lipopolissacarídeos , Sistema de Sinalização das MAP Quinases , NF-kappa B , Hepatopatia Gordurosa não Alcoólica/patologia , Periodontite/microbiologia , Porphyromonas gingivalis , Adulto , Infecções por Bacteroidaceae , Western Blotting , Humanos , Hepatopatia Gordurosa não Alcoólica/microbiologia , Porphyromonas gingivalis/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real
15.
Cell Rep ; 26(4): 884-892.e4, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30673611

RESUMO

DNA variants in the SLC16A11 coding region were identified to be strongly associated with type 2 diabetes (T2DM) in a Mexican population. Previous studies suggested that these variants disrupt SLC16A11 function and therefore proposed to revive SLC16A11 levels or activity to achieve therapeutic benefit. However, with knockout mouse models, here we show that Slc16a11 depletion has no significant metabolic defects. Further studies demonstrate that reconstitution of the mutant, but not the wild-type Slc16a11, in the liver of knockout mice causes more triglyceride accumulation and induction of insulin resistance via upregulation of lipin 1, suggesting gaining of aberrant functions of the mutant protein that affects lipid metabolism. Our findings offer a different explanation to the function of these diabetic variants, challenging the concept of enhancing SLC16A11 function to treat T2DM. The contradictory results by our and previous studies suggest that how the SLC16A11 locus contributes to human metabolism warrants further investigation.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Mutação com Ganho de Função , Resistência à Insulina/genética , Transportadores de Ácidos Monocarboxílicos , Triglicerídeos , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/metabolismo , Triglicerídeos/genética , Triglicerídeos/metabolismo
16.
Mol Cancer Res ; 17(2): 655-665, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30401720

RESUMO

The E-cadherin/ß-catenin signaling pathway plays a critical role in the maintenance of epithelial architecture and regulation of tumor progression. Normally, E-cadherin locates on the cell surface with its cytosolic domain linking to the actin cytoskeleton through interaction with catenins. Although the nuclear localization of E-cadherin has been frequently observed in various types of cancers, little is known regarding the functional consequences of its nuclear translocation. Here, we showed that in colorectal cancer samples and cell lines, E-cadherin localized in the nucleus; and the nuclear localization was mediated through protein interaction with CTNND1. In the nucleus, E-cadherin was acetylated by CREB-binding protein at Lysine870 and Lysine871 in its ß-catenin-binding domain, and the acetylation can be reversed by SIRT2. Acetylation of nuclear E-cadherin attenuated its interaction with ß-catenin, which therefore released ß-catenin from the complex, resulting in increased expression of its downstream genes and accelerated tumor growth and migration. Further study showed that acetylation level of nuclear E-cadherin had high prognostic significance in clinical colorectal samples. Taken together, our findings reveal a novel mechanism of tumor progression through posttranslational modification of E-cadherin, which may serve as a potential drug target of tumor therapy. IMPLICATIONS: This finding that acetylation of nuclear E-cadherin regulates ß-catenin activity expands our understanding of the acetylation of E-cadherin promotes colorectal cancer cell growth and suggests novel therapeutic approaches of targeting acetylation in tumors.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Neoplasias Colorretais/metabolismo , beta Catenina/metabolismo , Acetilação , Carcinogênese , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Neoplasias Colorretais/patologia , Células HCT116 , Células HEK293 , Humanos
17.
EBioMedicine ; 37: 344-355, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30348622

RESUMO

BACKGROUND: The pharmacological activation of thermogenesis in brown adipose tissue has long been considered promising strategies to treat obesity. However, identification of safe and effective agents remains a challenge. In this study, we addressed this challenge by developing a cellular system with a fluorescence readout, and applied in a high-throughput manner to screen for FDA-approved drugs that may activate endogenous UCP1 expression in adipocytes. METHODS: We have generated a Ucp1-2A-GFP reporter mouse, in which GFP intensity serves as a surrogate of the endogenous expression level of UCP1 protein; and immortalized brown adipocytes were derived from this mouse model and applied in drug screening. Candidate drugs were further tested in mouse models either fed with normal chow or high fat diet to induce obesity. FINDINGS: By using the cellular screening platform, we identified a group of FDA-approved drugs that can upregulate UCP1 expression in brown adipocyte, including previously known UCP1 activators and new candidate drugs. Further studies focusing on a previously unreported drug-sutent, revealed that sutent treatment could increase the energy expenditure and inhibit lipid synthesis in mouse adipose and liver tissues, resulting in improved metabolism and resistance to obesity. INTERPRETATION: This study offered an easy-to-use cellular screening system for UCP1 activators, and provided a candidate list of FDA-approved drugs that can potentially treat obesity. Further study of these candidates may shed new light on the drug discovery towards obesity. FUND: National Key Research and Development Program and the Strategic Priority Research Program of the Chinese Academy of Sciences, etc. (250 words).


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Desacopladora 1/biossíntese , Adipócitos Marrons/patologia , Tecido Adiposo Marrom/patologia , Animais , Linhagem Celular Transformada , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Camundongos , Camundongos Transgênicos , Proteína Desacopladora 1/genética , Estados Unidos , United States Food and Drug Administration
18.
Stem Cell Reports ; 11(1): 22-31, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29861165

RESUMO

Hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) offer a promising cell resource for disease modeling and transplantation. However, differentiated HLCs exhibit an immature phenotype and comprise a heterogeneous population. Thus, a better understanding of HLC differentiation will improve the likelihood of future application. Here, by taking advantage of CRISPR-Cas9-based genome-wide screening technology and a high-throughput hPSC screening platform with a reporter readout, we identified several potential genetic regulators of HLC differentiation. By using a chemical screening approach within our platform, we also identified compounds that can further promote HLC differentiation and preserve the characteristics of in vitro cultured primary hepatocytes. Remarkably, both screenings identified histone deacetylase 3 (HDAC3) as a key regulator in hepatic differentiation. Mechanistically, HDAC3 formed a complex with liver transcriptional factors, e.g., HNF4, and co-regulated the transcriptional program during hepatic differentiation. This study highlights a broadly useful approach for studying and optimizing hPSC differentiation.


Assuntos
Diferenciação Celular , Hepatócitos/citologia , Hepatócitos/metabolismo , Histona Desacetilases/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Benzamidas , Sistemas CRISPR-Cas , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Células Cultivadas , Citometria de Fluxo , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Genes Reporter , Genes abl , Fator 4 Nuclear de Hepatócito/metabolismo , Histona Desacetilases/genética , Humanos , Modelos Biológicos , Fenilenodiaminas/farmacologia
19.
Nat Commun ; 8(1): 1259, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29097654

RESUMO

Lysine acetylation is a post-translational modification known to regulate protein functions. Here we identify several acetylation sites of the influenza A virus nucleoprotein (NP), including the lysine residues K77, K113 and K229. Viral growth of mutant virus encoding K229R, mimicking a non-acetylated NP lysine residue, is severely impaired compared to wildtype or the mutant viruses encoding K77R or K113R. This attenuation is not the result of decreased polymerase activity, altered protein expression or disordered vRNP co-segregation but rather caused by impaired particle release. Interestingly, release deficiency is also observed mimicking constant acetylation at this site (K229Q), whereas virus encoding NP-K113Q could not be generated. However, mimicking NP hyper-acetylation at K77 and K229 severely diminishes viral polymerase activity, while mimicking NP hypo-acetylation at these sites has no effect on viral replication. These results suggest that NP acetylation at K77, K113 and K229 impacts multiple steps in viral replication of influenza A viruses.


Assuntos
Vírus da Influenza A/genética , Lisina/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas do Core Viral/genética , Replicação Viral/genética , Acetilação , Animais , Cães , Células HEK293 , Humanos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/metabolismo , Células Madin Darby de Rim Canino , Mutação , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo
20.
Cell Physiol Biochem ; 43(6): 2525-2534, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29130970

RESUMO

BACKGROUND/AIMS: The mammalian skull vault is a highly regulated structure and consists of several membrane bones of different tissue origins (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Although membrane bones form through intramembranous ossification, neural crest derived frontal bone has superior osteoblast activity and bone regeneration ability, triggering a novel conception for craniofacial reconstruction and bone regeneration called endogenous calvarial regeneration. However, a comprehensive landscape of the genes and signaling pathways involved in this process is not clear. METHODS: Transcriptome analysis within the two bone elements is firstly performed to determine the physiological signatures of differential gene expressions in mouse skull vault. RESULTS: Frontal bone tissues and parietal bone tissues maintain tissue origin through special gene expression similar to neural crest vs mesoderm tissue, and physiological functions between these two tissues are also found in differences related to proliferation, differentiation and extracellular matrix production and clustered signaling pathways. CONCLUSION: Our data provide novel insights into the potential gene regulatory network in regulating the development of neural crest-derived frontal bone and mesoderm-derived parietal bone.


Assuntos
Osso Parietal/metabolismo , Animais , Diferenciação Celular , Embrião de Mamíferos/metabolismo , Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , RNA/isolamento & purificação , RNA/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA