Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 557
Filtrar
1.
Nature ; 611(7936): 603-613, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36352230

RESUMEN

Around 30-40% of patients with colorectal cancer (CRC) undergoing curative resection of the primary tumour will develop metastases in the subsequent years1. Therapies to prevent disease relapse remain an unmet medical need. Here we uncover the identity and features of the residual tumour cells responsible for CRC relapse. An analysis of single-cell transcriptomes of samples from patients with CRC revealed that the majority of genes associated with a poor prognosis are expressed by a unique tumour cell population that we named high-relapse cells (HRCs). We established a human-like mouse model of microsatellite-stable CRC that undergoes metastatic relapse after surgical resection of the primary tumour. Residual HRCs occult in mouse livers after primary CRC surgery gave rise to multiple cell types over time, including LGR5+ stem-like tumour cells2-4, and caused overt metastatic disease. Using Emp1 (encoding epithelial membrane protein 1) as a marker gene for HRCs, we tracked and selectively eliminated this cell population. Genetic ablation of EMP1high cells prevented metastatic recurrence and mice remained disease-free after surgery. We also found that HRC-rich micrometastases were infiltrated with T cells, yet became progressively immune-excluded during outgrowth. Treatment with neoadjuvant immunotherapy eliminated residual metastatic cells and prevented mice from relapsing after surgery. Together, our findings reveal the cell-state dynamics of residual disease in CRC and anticipate that therapies targeting HRCs may help to avoid metastatic relapse.


Asunto(s)
Neoplasias Colorrectales , Metástasis de la Neoplasia , Proteínas de Neoplasias , Recurrencia Local de Neoplasia , Neoplasia Residual , Receptores de Superficie Celular , Animales , Humanos , Ratones , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Progresión de la Enfermedad , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Recurrencia Local de Neoplasia/terapia , Neoplasia Residual/genética , Neoplasia Residual/patología , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Metástasis de la Neoplasia/terapia , Modelos Animales de Enfermedad , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Terapia Neoadyuvante , Inmunoterapia
2.
Cell Mol Gastroenterol Hepatol ; 14(1): 193-217, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35367664

RESUMEN

BACKGROUND & AIMS: As an indispensable component of store-operated Ca2+ entry, stromal interaction molecule 1 (STIM1) is known to promote colorectal cancer and T-cell-mediated inflammatory diseases. However, whether the intestinal mucosal STIM1 is involved in inflammatory bowel diseases (IBDs) is unclear. This study aimed to investigate the role of intestinal epithelial STIM1 in IBD. METHODS: Inflammatory and matched normal intestinal tissues were collected from IBD patients to investigate the expression of STIM1. Intestinal epithelium-specific STIM1 conditional knockout mice (STIM1ΔIEC) were generated and induced to develop colitis and colitis-associated colorectal cancer. The mucosal barrier, including the epithelial barrier and mucus barrier, was analyzed. The mechanisms by which STIM1 regulate goblet cell endoplasmic reticulum stress and apoptosis were assessed. RESULTS: STIM1 could regulate intestinal epithelial homeostasis. STIM1 was augmented in the inflammatory intestinal tissues of IBD patients. In dextran sodium sulfate-induced colitis, STIM1 deficiency in intestinal epithelium reduced the loss of goblet cells through alleviating endoplasmic reticulum stress induced by disturbed Ca2+ homeostasis, resulting in the maintenance of the integrated mucus layer. These effects prevented commensal bacteria from contacting and stimulating the intestinal epithelium of STIM1ΔIEC mice and thereby rendered STIM1ΔIEC mice less susceptible to colitis and colitis-associated colorectal cancer. In addition, microbial diversity in dextran sodium sulfate-treated STIM1ΔIEC mice slightly shifted to an advantageous bacteria, which further protected the intestinal epithelium. CONCLUSIONS: Our results establish STIM1 as a crucial regulator for the maintenance of the intestinal barrier during colitis and provide a potential target for IBD treatment.


Asunto(s)
Neoplasias Asociadas a Colitis , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Carcinogénesis/metabolismo , Transformación Celular Neoplásica , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Células Caliciformes/metabolismo , Humanos , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
3.
Nature ; 600(7888): 324-328, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819670

RESUMEN

Activation-induced cytidine deaminase (AID) catalyses the deamination of deoxycytidines to deoxyuracils within immunoglobulin genes to induce somatic hypermutation and class-switch recombination1,2. AID-generated deoxyuracils are recognized and processed by subverted base-excision and mismatch repair pathways that ensure a mutagenic outcome in B cells3-6. However, why these DNA repair pathways do not accurately repair AID-induced lesions remains unknown. Here, using a genome-wide CRISPR screen, we show that FAM72A is a major determinant for the error-prone processing of deoxyuracils. Fam72a-deficient CH12F3-2 B cells and primary B cells from Fam72a-/- mice exhibit reduced class-switch recombination and somatic hypermutation frequencies at immunoglobulin and Bcl6 genes, and reduced genome-wide deoxyuracils. The somatic hypermutation spectrum in B cells from Fam72a-/- mice is opposite to that observed in mice deficient in uracil DNA glycosylase 2 (UNG2)7, which suggests that UNG2 is hyperactive in FAM72A-deficient cells. Indeed, FAM72A binds to UNG2, resulting in reduced levels of UNG2 protein in the G1 phase of the cell cycle, coinciding with peak AID activity. FAM72A therefore causes U·G mispairs to persist into S phase, leading to error-prone processing by mismatch repair. By disabling the DNA repair pathways that normally efficiently remove deoxyuracils from DNA, FAM72A enables AID to exert its full effects on antibody maturation. This work has implications in cancer, as the overexpression of FAM72A that is observed in many cancers8 could promote mutagenesis.


Asunto(s)
Linfocitos B , ADN Glicosilasas , Reparación de la Incompatibilidad de ADN , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana , Mutación , Proteínas de Neoplasias , Hipermutación Somática de Inmunoglobulina , Animales , Femenino , Humanos , Ratones , Linfocitos B/metabolismo , Sistemas CRISPR-Cas , ADN Glicosilasas/antagonistas & inhibidores , ADN Glicosilasas/metabolismo , Epistasis Genética , Células HEK293 , Cambio de Clase de Inmunoglobulina/genética , Región de Cambio de la Inmunoglobulina/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Hipermutación Somática de Inmunoglobulina/genética
4.
Bioengineered ; 12(2): 9806-9818, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34696661

RESUMEN

This study aimed to investigate the effects of carbonic anhydrase 12 (CA12)-siRNA on the paclitaxel sensitivity of breast cancer cells. Normal mammary glandular cell (MCF-10), breast cancer cell (MCF-7), and paclitaxel-resistant breast cancer cells (MCF-7 TaxR) were cultured in experimental control group. Western blot was adopted to detect the expressions of CA12 protein and apoptosis-related proteins in mitochondrial pathway of MCF-10, MCF-7, and MCF-7 TaxR cells. The methylthialazole tetrazolium (MTT) method was used to measure cell proliferation. The apoptosis of MCF-7 and MCF-7 TaxR cells was observed in phase contrast microscope, fluorescence inverted phase contrast microscope, and flow cytometry (FACS). The results showed that CA12 protein expression in MCF-7 and MCF-7 TaxR cells was significantly higher than that in MCF-10 cell. The growth rate of CA12-siRNA treated MCF-7 TaxR cells with paclitaxel (PTX) co-culture was markedly declined at 48 hours. Phase contrast microscope, fluorescence inverted phase contrast microscope, and FACS showed that apoptotic cells in the CA12-siRNA treated MCF-7 TaxR groups were significantly higher than that in CA12-siRNA treated MCF-7 cells. The expressions of pro-apoptotic proteins, Bax and Bid, were dramatically increased in CA12 siRNA treated MCF-7 TaxR cells. The expression quantity of the downstream effective molecules caspase-9, caspase-7, and the activated proteins of poly (ADP-ribose) polymerase (PARP), also were significantly increased. Our results indicated that the application of PTX combined silencing CA12 was able to activate the mitochondrial apoptosis pathway and promote MCF-7 TaxR apoptosis. CA12 silencing in the PTX-resistant breast cancer cell can reverse the sensitivity of PTX.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Anhidrasas Carbónicas , Resistencia a Antineoplásicos , Silenciador del Gen , Proteínas de Neoplasias , Paclitaxel/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Anhidrasas Carbónicas/deficiencia , Anhidrasas Carbónicas/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Células MCF-7 , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo
5.
Cell Rep ; 36(12): 109742, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34551310

RESUMEN

Cold-induced thermogenesis in endotherms demands adaptive thermogenesis fueled by mitochondrial respiration and Ucp1-mediated uncoupling in multilocular brown adipocytes (BAs). However, dietary regulation of thermogenesis in BAs isn't fully understood. Here, we describe that the deficiency of Leucine-rich pentatricopeptide repeat containing-protein (Lrpprc) in BAs reduces mtDNA-encoded ETC gene expression, causes ETC proteome imbalance, and abolishes the mitochondria-fueled thermogenesis. BA-specific Lrpprc knockout mice are cold resistant in a 4°C cold-tolerance test in the presence of food, which is accompanied by the activation of transcription factor 4 (ATF4) and proteome turnover in BAs. ATF4 activation genetically by BA-specific ATF4 overexpression or physiologically by a low-protein diet feeding can improve cold tolerance in wild-type and Ucp1 knockout mice. Furthermore, ATF4 activation in BAs improves systemic metabolism in obesogenic environment regardless of Ucp1's action. Therefore, our study reveals a diet-dependent but Ucp1-independent thermogenic mechanism in BAs that is relevant to systemic thermoregulation and energy homeostasis.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Tejido Adiposo Pardo/metabolismo , Termogénesis , Factor de Transcripción Activador 4/deficiencia , Factor de Transcripción Activador 4/genética , Animales , Peso Corporal , Frío , Dieta/veterinaria , Metabolismo Energético , Femenino , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Termogénesis/genética , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo , Yodotironina Deyodinasa Tipo II
6.
J Clin Immunol ; 41(8): 1781-1793, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34386911

RESUMEN

PURPOSE: Biallelic pathogenic NBAS variants manifest as a multisystem disorder with heterogeneous clinical phenotypes such as recurrent acute liver failure, growth retardation, and susceptibility to infections. This study explores how NBAS-associated disease affects cells of the innate and adaptive immune system. METHODS: Clinical and laboratory parameters were combined with functional multi-parametric immunophenotyping methods in fifteen NBAS-deficient patients to discover possible alterations in their immune system. RESULTS: Our study revealed reduced absolute numbers of mature CD56dim natural killer (NK) cells. Notably, the residual NK cell population in NBAS-deficient patients exerted a lower potential for activation and degranulation in response to K562 target cells, suggesting an NK cell-intrinsic role for NBAS in the release of cytotoxic granules. NBAS-deficient NK cell activation and degranulation was normalized upon pre-activation by IL-2 in vitro, suggesting that functional impairment was reversible. In addition, we observed a reduced number of naïve B cells in the peripheral blood associated with hypogammaglobulinemia. CONCLUSION: In summary, we demonstrate that pathogenic biallelic variants in NBAS are associated with dysfunctional NK cells as well as impaired adaptive humoral immunity.


Asunto(s)
Linfocitos B/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Células Asesinas Naturales/inmunología , Proteínas de Neoplasias/genética , Adolescente , Adulto , Niño , Preescolar , Citocinas/inmunología , Expresión Génica , Genotipo , Humanos , Lactante , Recuento de Leucocitos , Proteínas de Neoplasias/deficiencia , Fenotipo , Adulto Joven
8.
Cancer Med ; 10(15): 5256-5269, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34152094

RESUMEN

BACKGROUND: Human endogenous retrovirus-H long terminal repeat-associating protein 2 (HHLA2) is a member of B7 family, which is upregulated in multiple tumors. However, its exact functions in non-small cell lung cancer (NSCLC) have not been fully understood. This study aimed to investigate the biological roles of HHLA2 in human NSCLC and the relevant mechanisms. In addition, the effects of tumor cell-derived HHLA2 on tumor-associated macrophage (TAM) polarization were explored. METHODS: NSCLC cell growth, migration, and invasion were assessed by colony formation and modified Boyden chamber assays. Cell cycle and the CD163+ TAMs were examined by flow cytometry. A co-culture model of THP-1 macrophages and NSCLC cells was conducted to investigate the impacts of tumor cell-derived HHLA2 on THP-1 macrophage polarization. Moreover, a xenograft nude mouse model was established to explore the effects of HHLA2 on tumorigenesis in vivo. RESULTS: HHLA2 was upregulated in A549 and H1299 cells compared with the normal lung epithelial BEAS-2B cells. HHLA2 deficiency inhibited NSCLC cell proliferation, migration, invasion, and induced G0/G1 phase arrest partially via inhibiting EGFR/MAPK/ERK signaling pathway. Furthermore, HHLA2 knockdown inhibited M2 polarization of TAMs via downregulating IL-10. In addition, knockdown of HHLA2 inhibited tumor growth in vivo. CONCLUSION: HHLA2 downregulation inhibited NSCLC growth and TAM M2 polarization. HHLA2 may serve as a therapeutic target and promising prognostic biomarker in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Polaridad Celular , Progresión de la Enfermedad , Inmunoglobulinas/deficiencia , Neoplasias Pulmonares/patología , Macrófagos Asociados a Tumores/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclo Celular , Puntos de Control del Ciclo Celular , Movimiento Celular , Técnicas de Cocultivo , Regulación hacia Abajo , Xenoinjertos , Humanos , Inmunoglobulinas/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Células THP-1 , Regulación hacia Arriba
9.
Nat Commun ; 12(1): 2923, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011956

RESUMEN

Idiopathic pulmonary fibrosis (IPF) causes progressive fibrosis and worsening pulmonary function. Prognosis is poor and no effective therapies exist. We show that programmed cell death 5 (PDCD5) expression is increased in the lungs of patients with IPF and in mouse models of lung fibrosis. Lung fibrosis is significantly diminished by club cell-specific deletion of Pdcd5 gene. PDCD5 mediates ß-catenin/Smad3 complex formation, promoting TGF-ß-induced transcriptional activation of matricellular genes. Club cell Pdcd5 knockdown reduces matricellular protein secretion, inhibiting fibroblast proliferation and collagen synthesis. Here, we demonstrate the club cell-specific role of PDCD5 as a mediator of lung fibrosis and potential therapeutic target for IPF.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Proteínas de Neoplasias/genética , Anciano , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/metabolismo , Bronquiolos/metabolismo , Bronquiolos/patología , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Cancer Sci ; 112(7): 2714-2727, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33939216

RESUMEN

Breast cancer is one of the most commonly diagnosed malignancies worldwide, while the triple negative breast cancer (TNBC) is the most aggressive and virulent subtype in breast cancers. Compared with luminal type breast cancers, which could be well controlled by endocrine treatment, TNBC is worse in prognosis and lack of effective targeted therapy. Thus, it would be interesting and meaningful to identify novel therapeutic targets for TNBC treatments. Recent genomic data showed the activation of Hippo/YAP signaling in TNBC, indicating its critical roles in TNBC carcinogenesis and cancer progression. Hippo/YAP signaling could subject to several kinds of protein modifications, including ubiquitination and phosphorylation. Quite a few studies have demonstrated these modifications, which controlled YAP protein stability and turnover, played critical role in Hippo signaling activation In our current study, we identified ZNF213 as a negative modifier for Hippo/YAP axis. ZNF213 depletion promoted TNBC cell migration and invasion, which could be rescued by further YAP silencing. ZNF213 knocking down facilitated YAP protein stability and Hippo target gene expression, including CTGF and CYR61. Further mechanism studies demonstrated that ZNF213 associated with YAP and facilitated YAP K48-linked poly-ubiquitination at several YAP lysine sites (K252, K254, K321 and K497). Besides, the clinical data showed that ZNF213 negatively correlated with YAP protein level and Hippo target gene expression in TNBC samples. ZNF213 expression correlated with good prognosis in TNBC patients. Our data provided novel insights in YAP proteolytic regulation and TNBC progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Línea Celular Tumoral , Movimiento Celular , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Vía de Señalización Hippo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Fosforilación , Pronóstico , ARN Interferente Pequeño/genética , Transducción de Señal , Factores de Transcripción/deficiencia , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ubiquitinación , Proteínas Señalizadoras YAP
11.
Cancer Sci ; 112(7): 2625-2641, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33931921

RESUMEN

Bladder cancer (BLCA) remains the leading cause of cancer-related mortality among genitourinary malignancies worldwide. BLCA metastasis represents the primary reason for its poor prognosis. In this study, we report that decreased expression of partitioning defective 3 (Par3), a polarity protein (encoded by PARD3), is associated with tumor aggressive phenotypes and poor prognosis in BLCA patients. Consistently, ablation of Par3 promotes the metastasis and invasion of BLCA cells in vitro and in vivo. Further studies reveal that zinc finger protein Snail represses the expression of Par3 by binding to E2-box (CAGGTG) of PARD3 promoter-proximal. Inhibition of GSK-3ß promotes the expression and nuclear localization of Snail and then reduces the expression of Par3, resulting in the metastasis and invasion of BLCA cells. Moreover, we detected the interaction between Par3 (936-1356 aa) and ZO-1 (1372-1748 aa), which is involved in the maintenance of tight junction. Together, our results demonstrate that the GSK-3ß/Snail/Par3/ZO-1 axis regulates BLCA metastasis, and Snail is a major regulator for Par3 protein expression in BLCA.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Neoplasias Pulmonares/secundario , Factores de Transcripción de la Familia Snail/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Núcleo Celular/metabolismo , Polaridad Celular/fisiología , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fenotipo , Fosforilación , Pronóstico , Distribución Aleatoria , Factores de Transcripción de la Familia Snail/genética , Uniones Estrechas/fisiología , Proteína de la Zonula Occludens-1/metabolismo
12.
Cardiovasc Toxicol ; 21(8): 619-629, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33929718

RESUMEN

Fatty acid-binding protein 5 (FABP5) is an important member of the FABP family and plays a vital role in the metabolism of fatty acids. However, few studies have examined the role of FABP5 in pathological cardiac remodeling and heart failure. The aim of this study was to explore the role of FABP5 in transverse aortic constriction (TAC)-induced pathological cardiac remodeling and dysfunction in mice. Quantitative RT-PCR (qRT-PCR) and western blotting (WB) analysis showed that the levels of FABP5 mRNA and protein, respectively, were upregulated in hearts of the TAC model. Ten weeks after TAC in FABP5 knockout and wild type control mice, echocardiography, histopathology, qRT-PCR, and WB demonstrated that FABP5 deficiency aggravated cardiac injury (both cardiac hypertrophy and fibrosis) and dysfunction. In addition, transmission electron microscopy, ATP detection, and WB revealed that TAC caused severe impairment to mitochondria in the hearts of FABP5-deficient mice compared with that in control mice. When FABP5 was downregulated by siRNA in primary mouse cardiac fibroblasts, FABP5 silencing increased oxidative stress, reduced mitochondrial respiration, and increased the expression of myofibroblast activation marker genes in response to treatment with transforming growth factor-ß. Our findings demonstrate that FABP5 deficiency aggravates cardiac pathological remodeling and dysfunction by damaging cardiac mitochondrial function.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/deficiencia , Fibroblastos/metabolismo , Insuficiencia Cardíaca/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Neoplasias/deficiencia , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Fibroblastos/ultraestructura , Fibrosis , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/ultraestructura , Proteínas de Neoplasias/genética , Estrés Oxidativo , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
13.
Blood ; 137(20): 2827-2837, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33881523

RESUMEN

In patients with isocitrate dehydrogenase (IDH)-mutated acute myeloid leukemia (AML) treated by intensive chemotherapy (IC), prognostic significance of co-occurring genetic alterations and allogeneic hematopoietic stem cell transplantation (HSCT) are of particular interest with the advent of IDH1/2 mutant inhibitors. We retrospectively analyzed 319 patients with newly diagnosed AML (127 with IDH1, 135 with IDH2R140, and 57 with IDH2R172 mutations) treated with IC in 3 Acute Leukemia French Association prospective trials. In each IDH subgroup, we analyzed the prognostic impact of clinical and genetic covariates, and the role of HSCT. In patients with IDH1 mutations, the presence of NPM1 mutations was the only variable predicting improved overall survival (OS) in multivariate analysis (P < .0001). In IDH2R140-mutated AML, normal karyotype (P = .008) and NPM1 mutations (P = .01) predicted better OS. NPM1 mutations were associated with better disease-free survival (DFS; P = .0009), whereas the presence of DNMT3A mutations was associated with shorter DFS (P = .0006). In IDH2R172-mutated AML, platelet count was the only variable retained in the multivariate model for OS (P = .002). Among nonfavorable European LeukemiaNet 2010-eligible patients, 71 (36%) underwent HSCT in first complete remission (CR1) and had longer OS (P = .03) and DFS (P = .02) than nontransplanted patients. Future clinical trials testing frontline IDH inhibitors combined with IC may consider stratification on NPM1 mutational status, the primary prognostic factor in IDH1- or IDH2R140-mutated AML. HSCT improve OS of nonfavorable IDH1/2-mutated AML and should be fully integrated into the treatment strategy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Mutación Puntual , Cariotipo Anormal , Anciano , Aberraciones Cromosómicas , Ensayos Clínicos como Asunto/estadística & datos numéricos , ADN Metiltransferasa 3A/genética , Supervivencia sin Enfermedad , Femenino , Francia/epidemiología , Humanos , Hibridación Fluorescente in Situ , Isocitrato Deshidrogenasa/deficiencia , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/deficiencia , Nucleofosmina/genética , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Estudios Retrospectivos
14.
Nat Commun ; 12(1): 270, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431892

RESUMEN

Intestinal stem cells (ISCs) residing in the crypts are critical for the continual self-renewal and rapid recovery of the intestinal epithelium. The regulatory mechanism of ISCs is not fully understood. Here we report that CREPT, a recently identified tumor-promoting protein, is required for the maintenance of murine ISCs. CREPT is preferably expressed in the crypts but not in the villi. Deletion of CREPT in the intestinal epithelium of mice (Vil-CREPTKO) results in lower body weight and slow migration of epithelial cells in the intestine. Vil-CREPTKO intestine fails to regenerate after X-ray irradiation and dextran sulfate sodium (DSS) treatment. Accordingly, the deletion of CREPT decreases the expression of genes related to the proliferation and differentiation of ISCs and reduces Lgr5+ cell numbers at homeostasis. We identify that CREPT deficiency downregulates Wnt signaling by impairing ß-catenin accumulation in the nucleus of the crypt cells during regeneration. Our study provides a previously undefined regulator of ISCs.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Intestinos/fisiología , Proteínas de Neoplasias/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Animales , Recuento de Células , Proteínas de Ciclo Celular/deficiencia , Diferenciación Celular , Proliferación Celular , Epitelio/metabolismo , Eliminación de Gen , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas de Neoplasias/deficiencia , Organoides/metabolismo , Células Madre/citología , Vía de Señalización Wnt , Rayos X , beta Catenina/metabolismo
15.
Histochem Cell Biol ; 155(4): 439-449, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33398436

RESUMEN

In our previous study, fatty acid-binding protein 5 (FABP5) was expressed in septoclasts with long processes which are considered to resorb uncalcified matrix of the growth plate (GP) cartilage, and no apparent abnormalities were detected in the histo-architecture of the GP of FABP5-deficient (FABP5-/-) mice. Those finding lead us to hypothesize that another FABP can compensate the deletion of FABP5 in septoclasts of its gene-mutant mice. Based on the hypothesis, the present study examined the expression levels of several other FABPs in septoclasts and their morphology in FABP5-/- mouse tibiae. Processes of FABP5-/- septoclasts tend to be shorter than wild septoclasts. FABP4-positive septoclasts in FABP5-/- mice were more numerous than those cells in wild mice.Peroxisome proliferator-activated receptor (PPAR) γ was expressed in FABP4-positive septoclasts of FABP5-/- mice as well as mice administered with GW1929, a PPARγ agonist, suggesting that the occurrence of PPARγ induces an increase of FABP4-positive septoclasts. The present finding suggests that the functional exertion of FABP5 in septoclasts is supplemented by FABP4 in normal and FABP5-/- mice, and that the expression of FABP4 is up-regulated in accompany with PPARγ in FABP5-/- for maintenance of resorptive activity in the GP.


Asunto(s)
Condrocitos/metabolismo , Proteínas de Unión a Ácidos Grasos/biosíntesis , Proteínas de Unión a Ácidos Grasos/metabolismo , Placa de Crecimiento/metabolismo , Proteínas de Neoplasias/metabolismo , Tibia/metabolismo , Animales , Cartílago/metabolismo , Proteínas de Unión a Ácidos Grasos/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/deficiencia , Fenotipo
16.
Mol Med Rep ; 23(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33495826

RESUMEN

The formation of a hypertrophic scar (HS) may lead to failure of glaucoma surgery. Long non­coding RNAs (lncRNAs) are involved in the formation of HSs. Moreover, family with sequence similarity 225 member B (FAM225B) is upregulated in HS. However, the role of the lncRNA FAM225B in HS remains unknown. Thus, the present study aimed to investigate the function of FAM225B in HS. Scar fibroblasts were isolated from patients who had undergone glaucoma surgery. Western blotting was used to detect the expressions of Bax, Bcl­2, cleaved caspase 3, p62, ATG7 and Beclin 1, and reverse transcription­quantitative PCR (RT­qPCR) were conducted to determine the level of FAM225B in scar fibroblasts. Microtubule associated protein 1 light chain 3 α staining was performed to examine autophagosomes in scar fibroblasts. Furthermore, cell proliferation was evaluated via 5­ethynyl­2'­deoxyuridine staining. Flow cytometry was conducted to determine cell apoptosis and the levels of reactive oxygen species (ROS) in scar fibroblasts. The cell migratory ability was assessed using a Transwell assay. The results demonstrated that FAM225B knockdown significantly attenuated scar fibroblast proliferation and induced apoptosis. Additionally, transfection of scar fibroblasts with FAM225B small interfering RNA (siRNA) significantly increased the ROS levels and significantly decreased the migration of scar fibroblasts. The FAM225B overexpression­induced increase of scar fibroblast proliferation and migration was significantly reversed by 3­methyladenine administration. The results suggested that knockdown of FAM225B significantly inhibited the proliferation of scar fibroblasts by inhibiting autophagy. Therefore, knockdown of FAM225B could inhibit scar fibroblast proliferation after glaucoma surgery by inhibiting autophagy. These findings may provide a novel perspective of developing treatment strategy for the patients with HSs after glaucoma surgery.


Asunto(s)
Autofagia , Cicatriz Hipertrófica/metabolismo , Proteínas del Ojo , Fibroblastos/metabolismo , Glaucoma/metabolismo , Proteínas de Neoplasias/deficiencia , Células Cultivadas , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/patología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Femenino , Fibroblastos/patología , Técnicas de Inactivación de Genes , Glaucoma/genética , Glaucoma/patología , Glaucoma/cirugía , Humanos , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo
17.
J Med Genet ; 58(4): 247-253, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32487539

RESUMEN

INTRODUCTION: TMEM16A is a calcium-activated chloride channel expressed in various secretory epithelia. Two siblings presented in early infancy with reduced intestinal peristalsis and recurrent episodes of haemorrhagic diarrhoea. In one of them, the episodes were characterised by hepatic pneumatosis with gas bubbles in the portal vein similar to necrotising enterocolitis of the newborn. METHODS: Exome sequencing identified a homozygous truncating pathogenic variant in ANO1. Expression analysis was performed using reverse transcription PCR, western blot and immunohistochemistry. Electrophysiological and cell biological studies were employed to characterise the effects on ion transport both in patient respiratory epithelial cells and in transfected HEK293 cells. RESULTS: The identified variant led to TMEM16A dysfunction, which resulted in abolished calcium-activated Cl- currents. Secondarily, CFTR function is affected due to the close interplay between both channels without inducing cystic fibrosis (CF). CONCLUSION: TMEM16A deficiency is a potentially fatal disorder caused by abolished calcium-activated Cl- currents in secretory epithelia. Secondary impairment of CFTR function did not cause a CF phenotyp, which may have implications for CF treatment.


Asunto(s)
Anoctamina-1/genética , Canales de Cloruro/genética , Predisposición Genética a la Enfermedad , Enfermedades del Recién Nacido/genética , Proteínas de Neoplasias/genética , Anoctamina-1/deficiencia , Transporte Biológico/genética , Calcio/metabolismo , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/patología , Femenino , Células HEK293 , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/epidemiología , Enfermedades del Recién Nacido/patología , Masculino , Proteínas de Neoplasias/deficiencia
18.
Stem Cells ; 39(1): 78-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33022858

RESUMEN

Lyl1 encodes a hematopoietic- and endothelial-specific bHLH transcription factor. Lyl1-deficient mice are viable, but they display mild hematopoietic and vascular defects. Specifically, LYL1 is required for the maturation and stabilization of blood vessel endothelial adherens junctions. Here, we report that young adult Lyl1-/- mice exhibit transient overweight associated with general expansion of adipose tissue, without signs of metabolic disorder and unrelated to food intake. The increased fat tissue development in Lyl1-/- mice resulted from earlier differentiation of adipose stem cells (ASCs) into adipocytes through noncell autonomous mechanisms. Specifically, we found that in Lyl1-/- mice, the adipose tissue vascular structures are immature, as indicated by their high permeability, reduced coverage by pericytes, lower recruitment of VE-cadherin and ZO1 at cell junctions, and more prone to angiogenesis. Together, our data show that in Lyl1-/- mice, the impaired vascular compartment of the adipose niche promotes ASC differentiation, leading to early adipocyte expansion and premature ASC depletion. Our study highlights the major structural role of the adipose tissue vascular niche in coordinating stem cell self-renewal and differentiation into adipocytes.


Asunto(s)
Tejido Adiposo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Proteínas de Neoplasias/deficiencia , Neovascularización Patológica , Nicho de Células Madre , Células Madre/metabolismo , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Células Madre/patología
19.
Jpn J Clin Oncol ; 51(4): 639-645, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33244609

RESUMEN

BACKGROUND: The prevalence and molecular characteristics of deficient mismatch repair prostate cancer in the Japanese population have scarcely been investigated. METHODS: Immunohistochemistry for mismatch repair proteins (MLH1, MSH2, MSH6 and PMS2) was performed in formalin-fixed paraffin-embedded sections prepared from resected primary prostate cancers in patients who underwent prostatectomy at our institution between January 2001 and May 2016. Genetic and/or epigenetic alterations of mismatch repair genes were investigated in patients with any loss of mismatch repair protein expression in the tumour. RESULTS: Of the 337 patients, four (1.2%) showed loss of mismatch repair protein expression on immunohistochemistry. All four patients showed loss of both MSH2 and MSH6 protein expression. Genetic testing was performed in two of the four patients, demonstrating no pathogenic germline alterations were present. In each of these two patients, at least one somatic alteration inactivating MSH2 without MSH2 hypermethylation was identified, leading to the diagnosis of supposed 'Lynch-like syndrome'. Patients with deficient mismatch repair prostate cancer were at a significantly higher stage (pT2pN0 vs. pT3-4pN0/pTanypN1, P = 0.02) and had a greater Gleason score (<8 vs. ≥8, P < 0.01) than those with proficient mismatch repair prostate cancer. CONCLUSIONS: The prevalence of deficient mismatch repair prostate cancer in the Japanese hospital-based prostatectomized population was extremely low. To improve screening efficacy for deficient mismatch repair prostate cancer, screening candidates can be limited to patients with locally advanced, node-positive and/or Gleason score of 8 or greater prostate cancer. Universal tumour screening for Lynch syndrome seems ineffective in patients with prostate cancer.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Hospitales , Proteínas de Neoplasias/deficiencia , Neoplasias de la Próstata/cirugía , Anciano , Anciano de 80 o más Años , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Reparación de la Incompatibilidad de ADN/genética , Detección Precoz del Cáncer , Mutación de Línea Germinal/genética , Humanos , Inmunohistoquímica , Japón/epidemiología , Masculino , Persona de Mediana Edad , Proteína 2 Homóloga a MutS/genética , Prevalencia , Neoplasias de la Próstata/genética
20.
Mol Cell ; 80(5): 779-795.e10, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33207181

RESUMEN

Protein aggregates disrupt cellular homeostasis, causing toxicity linked to neurodegeneration. Selective autophagic elimination of aggregates is critical to protein quality control, but how aggregates are selectively targeted for degradation is unclear. We compared the requirements for autophagy receptor proteins: OPTN, NBR1, p62, NDP52, and TAX1BP1 in clearance of proteotoxic aggregates. Endogenous TAX1BP1 is recruited to and required for the clearance of stress-induced aggregates, whereas ectopic expression of TAX1BP1 increases clearance through autophagy, promoting viability of human induced pluripotent stem cell-derived neurons. In contrast, TAX1BP1 depletion sensitizes cells to several forms of aggregate-induced proteotoxicity. Furthermore, TAX1BP1 is more specifically expressed in the brain compared to other autophagy receptor proteins. In vivo, loss of TAX1BP1 results in accumulation of high molecular weight ubiquitin conjugates and premature lipofuscin accumulation in brains of young TAX1BP1 knockout mice. TAX1BP1 mediates clearance of a broad range of cytotoxic proteins indicating therapeutic potential in neurodegenerative diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/deficiencia , Autofagia , Encéfalo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas de Neoplasias/deficiencia , Enfermedades Neurodegenerativas/metabolismo , Agregación Patológica de Proteínas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Encéfalo/patología , Femenino , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipofuscina/genética , Lipofuscina/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Ratas , Ratas Sprague-Dawley , Ubiquitina/genética , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...