Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Commun ; 15(1): 1300, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38346942

RESUMEN

Osteoclasts are over-activated as we age, which results in bone loss. Src deficiency in mice leads to severe osteopetrosis due to a functional defect in osteoclasts, indicating that Src function is essential in osteoclasts. G-protein-coupled receptors (GPCRs) are the targets for ∼35% of approved drugs but it is still unclear how GPCRs regulate Src kinase activity. Here, we reveal that GPR54 activation by its natural ligand Kisspeptin-10 (Kp-10) causes Dusp18 to dephosphorylate Src at Tyr 416. Mechanistically, Gpr54 recruits both active Src and the Dusp18 phosphatase at its proline/arginine-rich motif in its C terminus. We show that Kp-10 binding to Gpr54 leads to the up-regulation of Dusp18. Kiss1, Gpr54 and Dusp18 knockout mice all exhibit osteoclast hyperactivation and bone loss, and Kp-10 abrogated bone loss by suppressing osteoclast activity in vivo. Therefore, Kp-10/Gpr54 is a promising therapeutic target to abrogate bone resorption by Dusp18-mediated Src dephosphorylation.


Asunto(s)
Resorción Ósea , Osteoclastos , Animales , Ratones , Osteoclastos/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Ratones Noqueados , Resorción Ósea/genética , Receptores de Kisspeptina-1
3.
Nat Biotechnol ; 41(5): 663-672, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36357717

RESUMEN

Cytosine base editors (CBEs) efficiently generate precise C·G-to-T·A base conversions, but the activation-induced cytidine deaminase/apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (AID/APOBEC) protein family deaminase component induces considerable off-target effects and indels. To explore unnatural cytosine deaminases, we repurpose the adenine deaminase TadA-8e for cytosine conversion. The introduction of an N46L variant in TadA-8e eliminates its adenine deaminase activity and results in a TadA-8e-derived C-to-G base editor (Td-CGBE) capable of highly efficient and precise C·G-to-G·C editing. Through fusion with uracil glycosylase inhibitors and further introduction of additional variants, a series of Td-CBEs was obtained either with a high activity similar to that of BE4max or with higher precision compared to other reported accurate CBEs. Td-CGBE/Td-CBEs show very low indel effects and a background level of Cas9-dependent or Cas9-independent DNA/RNA off-target editing. Moreover, Td-CGBE/Td-CBEs are more efficient in generating accurate edits in homopolymeric cytosine sites in cells or mouse embryos, suggesting their accuracy and safety for gene therapy and other applications.


Asunto(s)
Citosina , Edición Génica , Ratones , Animales , Edición Génica/métodos , Citosina/metabolismo , Aminohidrolasas/metabolismo , ARN , Sistemas CRISPR-Cas/genética , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo
4.
Clin Epigenetics ; 14(1): 81, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35765052

RESUMEN

Epigenetic abnormalities in DNA hydroxymethylation (5hmC) have been detected in patients with myeloid neoplasms, suggesting that 5hmC might act as a valuable epigenetic mark to reflect the disease status of myeloid neoplasms. Here, we report systematic genome-wide mapping of the DNA hydroxymethylomes in over 70 patients with myeloid neoplasms. Our integrative analysis leads to the identification of distinct 5hmC signatures that can sensitively discriminate patients from healthy individuals. At the molecular level, we unveiled dynamic 5hmC changes within key transcription factor (e.g., the CEBP family) binding motifs that are essential for hematopoiesis and myeloid lineage specification. 5hmC redistribution was found to alter the genome-wide binding of CEBP-α, thereby reprogramming transcriptional outputs to affect leukemia cell survival and stemness. Taken together, we provide a comprehensive 5hmC atlas representative of myeloid neoplasms, which sets the stage for future exploration on the epigenetic etiology of hematological malignancies. Mechanistically, our study further furnishes important insights into how abnormal 5hmC distribution in patients directly interrupts the binding of transcription factors to reshape transcriptional landscapes and aggravate leukemogenesis.


Asunto(s)
Neoplasias Hematológicas , Leucemia , Carcinogénesis , Supervivencia Celular , Metilación de ADN , Neoplasias Hematológicas/genética , Humanos , Leucemia/genética
5.
Cell Death Dis ; 13(2): 152, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-35165253

RESUMEN

Inflammatory diseases decrease the extracellular environmental pH. However, whether proton-activated G protein-coupled receptors (GPCRs) can regulate the development of osteoarthritis (OA) is largely unknown. In this study, we report that proton-activated GPR4 is essential for OA development. We found a marked increase in expression of the proton-activated GPR4 in human and mouse OA cartilage. Lentivirus-mediated overexpression of GPR4 in mouse joints accelerated the development of OA, including promotion of articular cartilage damage, synovial hyperplasia, and osteophyte formation, while Gpr4 knockout effectively attenuated the development of posttraumatic and aging-associated OA in mice. We also found that inhibition of GPR4 with the antagonist NE52-QQ57 ameliorated OA progression in mice, promoted extracellular matrix (ECM) production, and protected cartilage from degradation in human articular cartilage explants. Moreover, GPR4 overexpression upregulated matrix-degrading enzymes' expression and inflammation factors under pro-inflammatory and slightly acidic conditions. Mechanistically, GPR4 suppressed chondrocyte differentiation and upregulated cartilage homeostasis through NF-κB/MAPK signaling activation by regulating CXCR7/CXCL12 expression. Together, our results take the lead to illustrate that proton-activated GPCR acts as a key regulator for OA pathogenesis in vivo, and support that GPR4 could be a promising therapeutic target for OA treatment.


Asunto(s)
Cartílago Articular , Osteoartritis , Receptores Acoplados a Proteínas G , Transducción de Señal , Animales , Cartílago Articular/patología , Quimiocina CXCL12/metabolismo , Condrocitos/metabolismo , Ratones , Osteoartritis/metabolismo , Protones , Receptores CXCR/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
6.
Adv Sci (Weinh) ; 9(11): e2103940, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35076181

RESUMEN

Deregulated store-operated calcium entry (SOCE) mediated by aberrant STIM1-ORAI1 signaling is closely implicated in cancer initiation and progression. Here the authors report the identification of an alternatively spliced variant of STIM1, designated STIM1ß, that harbors an extra exon to encode 31 additional amino acids in the cytoplasmic domain. STIM1ß, highly conserved in mammals, is aberrantly upregulated in glioma tissues to perturb Ca2+ signaling. At the molecular level, the 31-residue insertion destabilizes STIM1ß by perturbing its cytosolic inhibitory domain and accelerating its activation kinetics to efficiently engage and gate ORAI calcium channels. Functionally, STIM1ß depletion affects SOCE in glioblastoma cells, suppresses tumor cell proliferation and growth both in vitro and in vivo. Collectively, their study establishes a splicing variant-specific tumor-promoting role of STIM1ß that can be potentially targeted for glioblastoma intervention.


Asunto(s)
Glioblastoma , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Glioblastoma/genética , Mamíferos/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
7.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34847079

RESUMEN

Therapeutics targeting osteoclasts are commonly used treatments for bone metastasis; however, whether and how osteoclasts regulate premetastatic niche and bone tropism are largely unknown. In this study, we report that osteoclast precursors (OPs) can function as a premetastatic niche component that facilitates breast cancer (BCa) bone metastasis at early stages. At the molecular level, unbiased GPCR ligand/agonist screening in BCa cells suggested that R-spondin 2 (RSPO2) and RANKL, through interaction with their receptor LGR4, promoted osteoclastic premetastatic niche formation and enhanced BCa bone metastasis. This was achieved by RSPO2/RANKL-LGR4 signal modulating the WNT inhibitor DKK1 through Gαq and ß-catenin signaling. DKK1 directly facilitated OP recruitment through suppression of its receptor LDL receptor-related protein 5 (LRP5) but not LRP6, upregulating Rnasek expression via inhibition of canonical WNT signaling. In clinical samples, RSPO2, LGR4, and DKK1 expression showed a positive correlation with BCa bone metastasis. Furthermore, soluble LGR4 extracellular domain (ECD) protein, acting as a decoy receptor for RSPO2 and RANKL, significantly alleviated bone metastasis and osteolytic lesions in a mouse bone metastasis model. These findings provide unique insights into the functional role of OPs as key components of the premetastatic niche for BCa bone metastasis and identify RSPO2/RANKL-LGR4 signaling as a promising target for inhibiting BCa bone metastasis.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/secundario , Línea Celular Tumoral , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Ligando RANK/genética , Receptores Acoplados a Proteínas G/genética
8.
Adv Sci (Weinh) ; 8(13): 2100424, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34540558

RESUMEN

Herein, a set of optogenetic tools (designated LiPOP) that enable photoswitchable necroptosis and pyroptosis in live cells with varying kinetics, is introduced. The LiPOP tools allow reconstruction of the key molecular steps involved in these two non-apoptotic cell death pathways by harnessing the power of light. Further, the use of LiPOPs coupled with upconversion nanoparticles or bioluminescence is demonstrated to achieve wireless optogenetic or chemo-optogenetic killing of cancer cells in multiple mouse tumor models. LiPOPs can trigger necroptotic and pyroptotic cell death in cultured prokaryotic or eukaryotic cells and in living animals, and set the stage for studying the role of non-apoptotic cell death pathways during microbial infection and anti-tumor immunity.


Asunto(s)
Muerte Celular , Optogenética , Animales , Escherichia coli , Células HeLa/metabolismo , Células HeLa/trasplante , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia , Luz , Ratones , Nanopartículas , Necroptosis , Optogenética/métodos , Proteínas de Unión a Fosfato/metabolismo , Proteínas Quinasas/metabolismo , Piroptosis
9.
Int J Mol Sci ; 22(8)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917049

RESUMEN

Hepatocellular carcinoma (HCC) is the sixth most common cancer and fourth leading cause of cancer-related death worldwide. The number of HCC cases continues to rise despite advances in screening and therapeutic inventions. More importantly, HCC poses two major health disparity issues. First, HCC occurs more commonly in men than women. Second, with the global increase in non-alcoholic fatty liver diseases (NAFLD), it has also become evident that HCC is more prevalent in some races and/or ethnic groups compared to others, depending on its predisposing etiology. Most studies on HCC in the past have been focused on genetic factors as the driving force for HCC development, and the results revealed that genetic mutations associated with HCC are often heterogeneous and involve multiple pathogenic pathways. An emerging new research field is epigenetics, in which gene expression is modified without altering DNA sequences. In this article, we focus on reviewing current knowledge on HCC-related DNA methylation changes that show disparities among different sexes or different racial/ethnic groups, in an effort to establish a point of departure for resolving the broader issue of health disparities in gastrointestinal malignancies using cutting-edge epigenetic approaches.


Asunto(s)
Carcinoma Hepatocelular/etnología , Carcinoma Hepatocelular/genética , Metilación de ADN , Epigénesis Genética , Neoplasias Hepáticas/etnología , Neoplasias Hepáticas/genética , Grupos Raciales/genética , Biomarcadores de Tumor , Carcinoma Hepatocelular/metabolismo , Epigenómica/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Mutación , Medición de Riesgo , Factores de Riesgo , Factores Sexuales , Transducción de Señal
10.
Liver Int ; 41(6): 1305-1319, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33529448

RESUMEN

BACKGROUND & AIMS: Pregnant women may transmit their metabolic phenotypes to their offspring, enhancing the risk for nonalcoholic fatty liver disease (NAFLD); however, the molecular mechanisms remain unclear. METHODS: Prior to pregnancy female mice were fed either a maternal normal-fat diet (NF-group, "no effectors"), or a maternal high-fat diet (HF-group, "persistent effectors"), or were transitioned from a HF to a NF diet before pregnancy (H9N-group, "effectors removal"), followed by pregnancy and lactation, and then offspring were fed high-fat diets after weaning. Offspring livers were analysed by functional studies, as well as next-generation sequencing for gene expression profiles and DNA methylation changes. RESULTS: The HF, but not the H9N offspring, displayed glucose intolerance and hepatic steatosis. The HF offspring also displayed a disruption of lipid homeostasis associated with an altered methionine cycle and abnormal one-carbon metabolism that caused DNA hypermethylation and L-carnitine depletion associated with deactivated AMPK signalling and decreased expression of PPAR-α and genes for fatty acid oxidation. These changes were not present in H9N offspring. In addition, we identified maternal HF diet-induced genes involved in one-carbon metabolism that were associated with DNA methylation modifications in HF offspring. Importantly, the DNA methylation modifications and their associated gene expression changes were reversed in H9N offspring livers. CONCLUSIONS: Our results demonstrate for the first time that maternal HF diet disrupted the methionine cycle and one-carbon metabolism in offspring livers which further altered lipid homeostasis. CpG islands of specific genes involved in one-carbon metabolism modified by different maternal diets were identified.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Efectos Tardíos de la Exposición Prenatal , Animales , Carbono/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Embarazo
11.
Nat Commun ; 12(1): 164, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431868

RESUMEN

The current optogenetic toolkit lacks a robust single-component Ca2+-selective ion channel tailored for remote control of Ca2+ signaling in mammals. Existing tools are either derived from engineered channelrhodopsin variants without strict Ca2+ selectivity or based on the stromal interaction molecule 1 (STIM1) that might crosstalk with other targets. Here, we describe the design of a light-operated Ca2+ channel (designated LOCa) by inserting a plant-derived photosensory module into the intracellular loop of an engineered ORAI1 channel. LOCa displays biophysical features reminiscent of the ORAI1 channel, which enables precise optical control over Ca2+ signals and hallmark Ca2+-dependent physiological responses. Furthermore, we demonstrate the use of LOCa to modulate aberrant hematopoietic stem cell self-renewal, transcriptional programming, cell suicide, as well as neurodegeneration in a Drosophila model of amyloidosis.


Asunto(s)
Canales de Calcio/metabolismo , Luz , Ingeniería de Proteínas , Animales , Fenómenos Biofísicos , Calcio/metabolismo , Drosophila/metabolismo , Células HEK293 , Células HeLa , Humanos , Degeneración Nerviosa/patología , Optogenética
12.
Pharmacol Res ; 164: 105406, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33359913

RESUMEN

It is well known that free fatty acids (FFAs) have beneficial effects on the skeletal system, however, which fatty acid sensing GPCR(s) and how the GPCR(s) regulating cartilage development and osteoarthritis (OA) pathogenesis is largely unknown. In this study, we found Gpr84, a receptor for medium-chain FFAs (MCFA), was the only FFA-sensing GPCR in human and mouse chondrocytes that exhibited elevated expression when stimulated by interleukin (IL)-1ß. Gpr84-deficiency upregulated cartilage catabolic regulator expression and downregulated anabolic factor expression in the IL-1ß-induced cell model and the destabilization of the medial meniscus (DMM)-induced OA mouse model. Gpr84-/- mice exhibited an aggravated OA phenotype characterized by severe cartilage degradation, osteophyte formation and subchondral bone sclerosis. Moreover, activating Gpr84 directly enhanced cartilage extracellular matrix (ECM) generation while knockout of Gpr84 suppressed ECM-related gene expression. Especially, the agonists of GPR84 protected human OA cartilage explants against degeneration by inducing cartilage anabolic factor expression. At the molecular level, GPR84 activation inhibited IL-1ß-induced NF-κB signaling pathway. Furthermore, deletion of Gpr84 had little effect on articular and spine cartilaginous tissues during skeletal growth. Together, all of our results demonstrated that fatty acid sensing GPCR (Gpr84) signaling played a critical role in OA pathogenesis, and activation of GPR84 or MCFA supplementation has potential in preventing the pathogenesis and progression of OA without severe cartilaginous side effect.


Asunto(s)
Osteoartritis/genética , Receptores Acoplados a Proteínas G/genética , Animales , Artralgia/genética , Artralgia/metabolismo , Artralgia/patología , Cartílago/metabolismo , Cartílago/patología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Ácidos Grasos/metabolismo , Homeostasis , Humanos , Interleucina-1beta/farmacología , Articulación de la Rodilla/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Columna Vertebral/patología , Tibia/patología
13.
EMBO Mol Med ; 13(1): e12798, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33283987

RESUMEN

Immune checkpoint blockade (ICB) has a limited effect on colorectal cancer, underlining the requirement of co-targeting the complementary mechanisms. Here, we identified prostaglandin E2 (PGE2 ) receptor 4 (EP4) as the master regulator of immunosuppressive myeloid cells (IMCs), which are the major driver of resistance to ICB therapy. PGE2 -bound EP4 promotes the differentiation of immunosuppressive M2 macrophages and myeloid-derived suppressor cells (MDSCs) and reduces the expansion of immunostimulated M1 macrophages. To explore the immunotherapeutic role of EP4 signaling, we developed a novel and selective EP4 antagonist TP-16. TP-16 effectively blocked the function of IMCs and enhanced cytotoxic T-cell-mediated tumor elimination in vivo. Cell co-culture experiments revealed that TP-16 promoted T-cell proliferation, which was impaired by tumor-derived CD11b+ myeloid cells. Notably, TP-16 and anti-PD-1 combination therapy significantly impeded tumor progression and prolonged mice survival. We further demonstrated that TP-16 increased responsiveness to anti-PD-1 therapy in an IMC-related spontaneous colorectal cancer mouse model. In summary, this study demonstrates that inhibition of EP4-expressing IMCs may offer a potential strategy for enhancing the efficacy of immunotherapy for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Células Supresoras de Origen Mieloide , Animales , Neoplasias Colorrectales/tratamiento farmacológico , Inmunoterapia , Ratones , Células Mieloides , Subtipo EP4 de Receptores de Prostaglandina E
14.
Curr Opin Biotechnol ; 66: 207-216, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32956902

RESUMEN

Synthetic biology, when combined with immunoengineering (designated synthetic immunology), has enabled the invention of an arsenal of genetically encoded synthetic devices and systems to reprogram cells for therapeutic purposes. The engineered intelligent cells can serve as 'living' drugs to treat a wide range of human diseases including cancer, disorders of the immune system, and infectious diseases. As the most successful example, cells of the immune system engineered with chimeric antigen receptors (CARs) have shown curative potentials for the treatment of hematological malignancies. We present herein emerging approaches of designing smart CARs to improve their safety, specificity and efficacy in cellular immunotherapy, and describe latest advances in applying CAR-engineered immune cells to target cancer and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Enfermedades Autoinmunes/terapia , Humanos , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T , Linfocitos T
15.
Mucosal Immunol ; 13(6): 931-945, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32518369

RESUMEN

Inflammasomes are essential for inflammation and pathogen elimination in response to microbial infection and endogenous danger signals. However, the mechanism of inflammasome activation by endogenous danger signals mediated posttranslational modification and the connection between inflammasomes and inflammatory diseases remains elusive. In this study, we found that ADP was highly released from injured colonic tissue as a danger signal during inflammatory bowel disease. Consequently, extracellular ADP activated the NLRP3 inflammasome through P2Y1 receptor-mediated calcium signaling, which led to the maturation and secretion of IL-1ß and further aggravation of experimental colitis. Genetic ablation or pharmacological blockade of the P2Y1 receptor significantly ameliorated DSS-induced colitis and endotoxic shock through reducing NLRP3 inflammasome activation. Moreover, ERK5-mediated tyrosine phosphorylation of ASC was essential for activation of the NLRP3 inflammasome. Thus, our study provides a novel theoretical basis for posttranslational modification of ASC in NLRP3 inflammasome activation and revealed that ADP/P2Y1 is a potential drug target for inflammatory bowel disease.


Asunto(s)
Colitis/inmunología , Inflamasomas/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Adenosina Difosfato/metabolismo , Animales , Señalización del Calcio , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Humanos , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Fosforilación
16.
Nat Biotechnol ; 38(7): 856-860, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32483363

RESUMEN

Although base editors are useful tools for precise genome editing, current base editors can only convert either adenines or cytosines. We developed a dual adenine and cytosine base editor (A&C-BEmax) by fusing both deaminases with a Cas9 nickase to achieve C-to-T and A-to-G conversions at the same target site. Compared to single base editors, A&C-BEmax's activity on adenines is slightly reduced, whereas activity on cytosines is higher and RNA off-target activity is substantially decreased.


Asunto(s)
Adenina , Sistemas CRISPR-Cas/genética , Citosina , Edición Génica/métodos , Proteína 9 Asociada a CRISPR/genética , Desoxirribonucleasa I/genética , Humanos , ARN/genética
17.
Int J Cancer ; 147(8): 2239-2252, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32372448

RESUMEN

Intestinal tumors mainly originate from transformed crypt stem cells supported by Wnt signaling, which functions through downstream critical factors enriched in the intestinal stem/progenitor compartment. Here, we show Uhrf2 is predominantly expressed in intestinal crypts and adenomas in mice and is transcriptionally regulated by Wnt signaling. Upregulated UHRF2 correlates with poor prognosis in colorectal cancer patients. Although loss of Uhrf2 did not affect intestinal homeostasis and regeneration, tumor initiation and progression were inhibited, leading to a markedly prolonged life span in Uhrf2 null mice on an ApcMin background. Uhrf2 deficiency also strongly reduced primary tumor organoid formation suggesting impairment of tumor stem cells. Moreover, ablation of Uhrf2 suppressed tumor cell proliferation through downregulation of the Wnt/ß-catenin pathway. Mechanistically, Uhrf2 directly interacts with and sumoylates Tcf4, a critical intranuclear effector of the Wnt pathway. Uhrf2 mediated SUMOylation stabilized Tcf4 and further sustained hyperactive Wnt signaling. Together, we demonstrate that Wnt-induced Uhrf2 expression promotes tumorigenesis through modulation of the stability of Tcf4 for maintaining oncogenic Wnt/ß-catenin signaling. This is a new reciprocal feedforward regulation between Uhrf2 and Wnt signaling in tumor initiation and progression.


Asunto(s)
Carcinogénesis/genética , Neoplasias Colorrectales/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Ubiquitina-Proteína Ligasas/genética , Vía de Señalización Wnt/genética , beta Catenina/genética , Adenoma/genética , Adenoma/patología , Animales , Carcinogénesis/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HCT116 , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/patología , Oncogenes/genética , Transcripción Genética/genética , Regulación hacia Arriba/genética
18.
Sci Adv ; 6(12): eaaz0368, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32219165

RESUMEN

Adhesion G protein-coupled receptor G6 (Adgrg6; also named GPR126) single-nucleotide polymorphisms are associated with human height in multiple populations. However, whether and how GPR126 regulates body height is unknown. In this study, we found that mouse body length was specifically decreased in Osx-Cre;Gpr126fl/fl mice. Deletion of Gpr126 in osteoblasts resulted in a remarkable delay in osteoblast differentiation and mineralization during embryonic bone formation. Postnatal bone formation, bone mass, and bone strength were also significantly affected in Gpr126 osteoblast deletion mice because of defects in osteoblast proliferation, differentiation, and ossification. Furthermore, type IV collagen functioned as an activating ligand of Gpr126 to regulate osteoblast differentiation and function by stimulating cAMP signaling. Moreover,the cAMP activator PTH(1-34), could partially restore the inhibition of osteoblast differentiation and the body length phenotype induced by Gpr126 deletion.Together, our results demonstrated that COLIV-Gpr126 regulated body length and bone mass through cAMP-CREB signaling pathway.


Asunto(s)
Estatura , Huesos/anatomía & histología , Huesos/metabolismo , Estudios de Asociación Genética , Receptores Acoplados a Proteínas G/genética , Esqueleto/anatomía & histología , Alelos , Animales , Biomarcadores , Calcificación Fisiológica , Diferenciación Celular , Proliferación Celular , Colágeno Tipo IV/metabolismo , Regulación de la Expresión Génica , Genotipo , Humanos , Ratones , Ratones Noqueados , Tamaño de los Órganos , Osteoblastos/metabolismo , Fenotipo , Polimorfismo de Nucleótido Simple , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
19.
Bone Res ; 7: 19, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31646011

RESUMEN

The superfamily of G protein-coupled receptors (GPCRs) contains immense structural and functional diversity and mediates a myriad of biological processes upon activation by various extracellular signals. Critical roles of GPCRs have been established in bone development, remodeling, and disease. Multiple human GPCR mutations impair bone development or metabolism, resulting in osteopathologies. Here we summarize the disease phenotypes and dysfunctions caused by GPCR gene mutations in humans as well as by deletion in animals. To date, 92 receptors (5 glutamate family, 67 rhodopsin family, 5 adhesion, 4 frizzled/taste2 family, 5 secretin family, and 6 other 7TM receptors) have been associated with bone diseases and dysfunctions (36 in humans and 72 in animals). By analyzing data from these 92 GPCRs, we found that mutation or deletion of different individual GPCRs could induce similar bone diseases or dysfunctions, and the same individual GPCR mutation or deletion could induce different bone diseases or dysfunctions in different populations or animal models. Data from human diseases or dysfunctions identified 19 genes whose mutation was associated with human BMD: 9 genes each for human height and osteoporosis; 4 genes each for human osteoarthritis (OA) and fracture risk; and 2 genes each for adolescent idiopathic scoliosis (AIS), periodontitis, osteosarcoma growth, and tooth development. Reports from gene knockout animals found 40 GPCRs whose deficiency reduced bone mass, while deficiency of 22 GPCRs increased bone mass and BMD; deficiency of 8 GPCRs reduced body length, while 5 mice had reduced femur size upon GPCR deletion. Furthermore, deficiency in 6 GPCRs induced osteoporosis; 4 induced osteoarthritis; 3 delayed fracture healing; 3 reduced arthritis severity; and reduced bone strength, increased bone strength, and increased cortical thickness were each observed in 2 GPCR-deficiency models. The ever-expanding number of GPCR mutation-associated diseases warrants accelerated molecular analysis, population studies, and investigation of phenotype correlation with SNPs to elucidate GPCR function in human diseases.

20.
Protein Cell ; 10(9): 700, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31087276

RESUMEN

In the original publication the grant number is incorrectly published. The correct grant number should be read as "17140901600". The corrected contents are provided in this correction article. This work was partially supported by grants from the National Natural Science Foundation of China (Nos. 81670470 and 81600149), a grant from the Shanghai Municipal Commission for Science and Technology (17140901600, 18411953500 and 15JC1400201) and a grant from National Key Research and Development Program (2016YFC0905100).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...