Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.089
Filtrar
1.
BMC Cancer ; 24(1): 772, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937681

RESUMEN

BACKGROUND: Wilms tumor is the most prevalent embryonal kidney malignancy in children worldwide. Previous genome-wide association study (GWAS) identified that LIM domain only 1 (LMO1) gene polymorphisms affected the susceptibility to develop certain tumor types. Apart from LMO1, the LMO gene family members also include LMO2-4, each of which has oncogenic potential. METHODS: We conducted this five-center case‒control study to assess the correlations between single nucleotide polymorphisms in LMO family genes and Wilms tumor susceptibility. Odds ratios and 95% confidence intervals were calculated to evaluate the strength of the association. RESULTS: We found LMO1 rs2168101 G > T and rs11603024 C > T as well as LMO2 rs7933499 G > A were significantly associated with Wilms tumor risk. Stratified analysis demonstrated a protective role of rs2168101 GT/TT genotypes against Wilms tumor in the subgroups of age ≤ 18 months, males and clinical stages I/II compared to the rs2168101 GG genotype. Nevertheless, carriers with the rs11603024 TT genotype were more likely to have an increased risk of Wilms tumor than those with rs11603024 CC/CT genotypes in age > 18 months. And the rs11603024 was identified as a protective polymorphism for reducing the risk of Wilms tumor in the sex- and gender- subgroup. Likewise, carriers with the rs7933499 GA/AA genotypes were at significantly elevated risk of Wilms tumor in age ≤ 18 months and clinical stages I/II. CONCLUSION: Overall, our study identified the importance of LMO family gene polymorphisms on Wilms tumor susceptibility in Chinese children. Further investigations are needed to validate our conclusions.


Asunto(s)
Predisposición Genética a la Enfermedad , Neoplasias Renales , Proteínas con Dominio LIM , Polimorfismo de Nucleótido Simple , Tumor de Wilms , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Proteínas Adaptadoras Transductoras de Señales/genética , Estudios de Casos y Controles , China/epidemiología , Proteínas de Unión al ADN/genética , Pueblos del Este de Asia/genética , Genotipo , Neoplasias Renales/genética , Proteínas con Dominio LIM/genética , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/genética , Tumor de Wilms/genética , Familia de Multigenes
2.
BMC Oral Health ; 24(1): 729, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918827

RESUMEN

BACKGROUND: Despite the better prognosis associated with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC), some patients experience relapse and succumb to the disease; thus, there is a need for biomarkers identifying these patients for intensified treatment. Leucine-rich repeats and immunoglobulin-like domain (LRIG) protein 1 is a negative regulator of receptor tyrosine kinase signaling and a positive prognostic factor in OPSCC. Studies indicate that LRIG1 interacts with the LIM domain 7 protein (LMO7), a stabilizer of adherence junctions. Its role in OPSCC has not been studied before. METHODS: A total of 145 patients diagnosed with OPSCC were enrolled. Immunohistochemical LMO7 expression and staining intensity were evaluated in the tumors and correlated with known clinical and pathological prognostic factors, such as HPV status and LRIG1, CD44, Ki67, and p53 expression. RESULTS: Our results show that high LMO7 expression is associated with significantly longer overall survival (OS) (p = 0.044). LMO7 was a positive prognostic factor for OS in univariate analysis (HR 0.515, 95% CI: 0.267-0.994, p = 0.048) but not in multivariate analysis. The LMO7 expression correlated with LRIG1 expression (p = 0.048), consistent with previous findings. Interestingly, strong LRIG1 staining intensity was an independent negative prognostic factor in the HPV-driven group of tumors (HR 2.847, 95% Cl: 1.036-7.825, p = 0.043). CONCLUSIONS: We show for the first time that high LMO7 expression is a positive prognostic factor in OPSCC, and we propose that LMO7 should be further explored as a biomarker. In contrast to previous reports, LRIG1 expression was shown to be an independent negative prognostic factor in HPV-driven OPSCC.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Escamosas , Proteínas con Dominio LIM , Neoplasias Orofaríngeas , Humanos , Neoplasias Orofaríngeas/virología , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/patología , Neoplasias Orofaríngeas/mortalidad , Masculino , Femenino , Persona de Mediana Edad , Pronóstico , Proteínas con Dominio LIM/metabolismo , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Anciano , Factores de Transcripción/metabolismo , Glicoproteínas de Membrana/metabolismo , Adulto , Antígeno Ki-67/metabolismo , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/análisis , Proteína p53 Supresora de Tumor/metabolismo , Infecciones por Papillomavirus/complicaciones , Inmunohistoquímica , Anciano de 80 o más Años , Tasa de Supervivencia
3.
Arch Dermatol Res ; 316(7): 401, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38878083

RESUMEN

BACKGROUND: The adhesive properties of vitiligo melanocytes have decreased under oxidative stress., cytoskeleton proteins can control cell adhesion. Paeoniflorin (PF) was proved to resist hydrogen peroxide (H2O2)-induced oxidative stress in melanocytes via nuclear factorE2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. OBJECTIVES: This study was to investigate whether PF exerts anti-oxidative effect through influencing cytoskeleton markers or potential signaling pathway. METHODS: Human Oxidative Stress Plus array was used to identify the differentially expressed genes between H2O2 + PF group and H2O2 only group, in PIG1 and PIG3V melanocyte cell lines respectively. Western blotting was used to verify the PCR array results and to test the protein expression levels of cytoskeleton markers including Ras homolog family member A (RhoA), Rho-associated kinase 1 (ROCK1) and antioxidative marker Nrf2. Small interfering RNA was used to knock down PDZ and LIM domain 1 (PDLIM1). RESULTS: PF increased the expressions of PDLIM1, RhoA and ROCK1 in H2O2-induced PIG1, in contrast, decreased the expressions of PDLIM1 and ROCK1 in H2O2-induced PIG3V. Knockdown of PDLIM1 increased the expressions of RhoA and Nrf2 in PF-pretreated H2O2-induced PIG1, and ROCK1 and Nrf2 in PF-pretreated H2O2-induced PIG3V. CONCLUSIONS: PF regulates RhoA/ROCK1 and Nrf2 pathways in PDLIM1-dependent or independent manners in H2O2-induced melanocytes. In PIG1, PF promotes PDLIM1 to inhibit RhoA/ROCK1 pathway or activates Nrf2/HO-1 pathway, separately. In PIG3V, PF directly downregulates ROCK1 in PDLIM1-independent manner or upregulates Nrf2 dependent of PDLIM1.


Asunto(s)
Glucósidos , Peróxido de Hidrógeno , Proteínas con Dominio LIM , Melanocitos , Monoterpenos , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Transducción de Señal , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA , Factor 2 Relacionado con NF-E2/metabolismo , Quinasas Asociadas a rho/metabolismo , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Humanos , Glucósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Peróxido de Hidrógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Monoterpenos/farmacología , Línea Celular
4.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928252

RESUMEN

Inclusion body myositis (IBM) is a slowly progressive disorder belonging to the idiopathic inflammatory myopathies, and it represents the most common adult-onset acquired myopathy. The main clinical features include proximal or distal muscular asymmetric weakness, with major involvement of long finger flexors and knee extensors. The main histological findings are the presence of fiber infiltrations, rimmed vacuoles, and amyloid inclusions. The etiopathogenesis is a challenge because both environmental and genetic factors are implicated in muscle degeneration and a distinction has been made previously between sporadic and hereditary forms. Here, we describe an Italian patient affected with a hereditary form of IBM with onset in his mid-forties. Next-generation sequencing analysis disclosed a heterozygous mutation c.76C>T (p.Pro26Ser) in the PDZ motif of the LDB3/ZASP gene, a mutation already described in a family with a late-onset myopathy and highly heterogenous degree of skeletal muscle weakness. In the proband's muscle biopsy, the expression of ZASP, myotilin, and desmin were increased. In our family, in addition to the earlier age of onset, the clinical picture is even more peculiar given the evidence, in one of the affected family members, of complete ophthalmoplegia in the vertical gaze. These findings help extend our knowledge of the clinical and genetic background associated with inclusion body myopathic disorders.


Asunto(s)
Proteínas con Dominio LIM , Miositis por Cuerpos de Inclusión , Linaje , Humanos , Miositis por Cuerpos de Inclusión/genética , Miositis por Cuerpos de Inclusión/patología , Masculino , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Persona de Mediana Edad , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Mutación , Adulto
5.
Genes (Basel) ; 15(6)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38927709

RESUMEN

BACKGROUND: Homozygosity for LIMS1 rs893403-GG genotype is linked to an increased risk of allograft rejection after kidney transplantation. Ischemia-reperfusion of the kidney allograft leads to long term infiltration of activated and effector-memory T lymphocytes and resulting in rejection and long-term fibrosis. However, the genotype, LIMS1 expression under ischemic conditions and the long-term histopathological relationships remain ill-defined. METHODS: We examined the impact of the recipient's LIMS1-rs893403 genotype with transplant kidney histopathology. The association of the LIMS1-rs893403 genotype and LIMS1 and GCC2 mRNA expression in ischemic donor kidneys were also examined. Recipients who underwent transplant kidney biopsy were genotyped for the LIMS1-rs893403 variant and associated deletion. Histopathological findings were compared between recipients with LIMS1 risk and non-risk genotypes. Real-time PCR and immunofluorescence staining for LIMS1 and GCC2 expression were performed in non-utilized donor kidneys. RESULTS: Demographic, clinical, and treatment characteristics and the histopathological diagnosis were similar between recipients with rs893403 GG and AA/AG genotype. The Banff tubulitis score was higher in GG recipients (n = 24) compared to AA/AG (n = 86) recipients (1.42 ± 0.65 vs. 1.12 ± 0.66, p = 0.03). Ischemic kidneys with GG showed higher LIMS1 and GCC2 mRNA expression than kidneys with AG. Kidneys with rs893403-GG had higher tubular LIMS1 and GCC2 immunohistochemical staining compared to kidneys with rs893403-AG. CONCLUSIONS: Our data supports the role of the LIMS1 locus in kidney transplant rejection, particularly in lymphocyte infiltration into the internal aspect of the tubular basement membranes. Increased LIMS1 and GCC2 expression in ischemic donor kidneys with the GG genotype require further studies.


Asunto(s)
Genotipo , Trasplante de Riñón , Túbulos Renales , Proteínas con Dominio LIM , Trasplante de Riñón/efectos adversos , Humanos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Proteínas con Dominio LIM/genética , Túbulos Renales/patología , Túbulos Renales/metabolismo , Inflamación/genética , Inflamación/patología , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Polimorfismo de Nucleótido Simple
6.
Eur J Med Res ; 29(1): 309, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38831471

RESUMEN

The long non-coding RNA (lncRNA) Small Nucleolar RNA Host Gene 4 (SNHG4) has been demonstrated to be significantly downregulated in various inflammatory conditions, yet its role in chronic obstructive pulmonary disease (COPD) remains elusive. This study aims to elucidate the biological function of SNHG4 in COPD and to unveil its potential molecular targets. Our findings reveal that both SNHG4 and Four and a Half LIM Domains 1 (FHL1) were markedly downregulated in COPD, whereas microRNA-409-3p (miR-409-3p) was upregulated. Importantly, SNHG4 exhibited a negative correlation with inflammatory markers in patients with COPD, but a positive correlation with forced expiratory volume in 1s percentage (FEV1%). SNHG4 distinguished COPD patients from non-smokers with high sensitivity, specificity, and accuracy. Overexpression of SNHG4 ameliorated cigarette smoke extract (CSE)-mediated inflammation, apoptosis, oxidative stress, and airway remodeling in 16HBE bronchial epithelial cells. These beneficial effects of SNHG4 overexpression were reversed by the overexpression of miR-409-3p or the silencing of FHL1. Mechanistically, SNHG4 competitively bound to miR-409-3p, mediating the expression of FHL1, and consequently improving inflammation, apoptosis, oxidative stress, and airway remodeling in 16HBE cells. Additionally, SNHG4 regulated the miR-409-3p/FHL1 axis to inhibit the activation of the mitogen-activated protein kinase (MAPK) pathway induced by CSE. In a murine model of COPD, knockdown of SNHG4 exacerbated CSE-induced pulmonary inflammation, apoptosis, and oxidative stress. In summary, our data affirm that SNHG4 mitigates pulmonary inflammation, apoptosis, and oxidative damage mediated by COPD through the regulation of the miR-409-3p/FHL1 axis.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Apoptosis , Proliferación Celular , MicroARNs , Enfermedad Pulmonar Obstructiva Crónica , ARN Largo no Codificante , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Apoptosis/genética , Remodelación de las Vías Aéreas (Respiratorias)/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Proliferación Celular/genética , Animales , Ratones , Masculino , Sistema de Señalización de MAP Quinasas/genética , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Inflamación/metabolismo , Inflamación/genética , Femenino , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Persona de Mediana Edad , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones Endogámicos C57BL
7.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38838015

RESUMEN

O-GlcNAcase (OGA) is the only human enzyme that catalyzes the hydrolysis (deglycosylation) of O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation) from numerous protein substrates. OGA has broad implications in many challenging diseases including cancer. However, its role in cell malignancy remains mostly unclear. Here, we report that a cancer-derived point mutation on the OGA's noncatalytic stalk domain aberrantly modulates OGA interactome and substrate deglycosylation toward a specific set of proteins. Interestingly, our quantitative proteomic studies uncovered that the OGA stalk domain mutant preferentially deglycosylated protein substrates with +2 proline in the sequence relative to the O-GlcNAcylation site. One of the most dysregulated substrates is PDZ and LIM domain protein 7 (PDLIM7), which is associated with the tumor suppressor p53. We found that the aberrantly deglycosylated PDLIM7 suppressed p53 gene expression and accelerated p53 protein degradation by promoting the complex formation with E3 ubiquitin ligase MDM2. Moreover, deglycosylated PDLIM7 significantly up-regulated the actin-rich membrane protrusions on the cell surface, augmenting the cancer cell motility and aggressiveness. These findings revealed an important but previously unappreciated role of OGA's stalk domain in protein substrate recognition and functional modulation during malignant cell progression.


Asunto(s)
Citoesqueleto , Proteínas con Dominio LIM , Proteína p53 Supresora de Tumor , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Citoesqueleto/metabolismo , Acetilglucosamina/metabolismo , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Línea Celular Tumoral , Glicosilación , Hidrólisis , Mutación , Movimiento Celular , Antígenos de Neoplasias , Hialuronoglucosaminidasa , Histona Acetiltransferasas
8.
Nat Commun ; 15(1): 4945, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858386

RESUMEN

Single administration of low-dose ketamine has both acute and sustained anti-depressant effects. Sustained effect is associated with restoration of glutamatergic synapses in medial prefrontal cortic (mFPC) neurons. Ketamine induced profound changes in a number of molecular pathways in a mouse model for chronic stress. Cell-cell communication analyses predicted that planar-cell-polarity (PCP) signaling was decreased after chronic administration of corticosterone but increased following ketamine administration in most of the excitatory neurons. Similar decrease of PCP signaling in excitatory neurons was predicted in dorsolateral prefrontal cortical (dl-PFC) neurons of patients with major depressive disorder (MDD). We showed that the basolateral amygdala (BLA)-projecting infralimbic prefrontal cortex (IL PFC) neurons regulate immobility time in the tail suspension test and food consumption. Conditionally knocking out Celsr2 and Celsr3 or Prickle2 in the BLA-projecting IL PFC neurons abolished ketamine-induced synapse restoration and behavioral remission. Therefore, PCP proteins in IL PFC-BLA neurons mediate synapse restoration induced by of low-dose ketamine.


Asunto(s)
Modelos Animales de Enfermedad , Ketamina , Neuronas , Corteza Prefrontal , Sinapsis , Animales , Ketamina/farmacología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones , Masculino , Humanos , Polaridad Celular/efectos de los fármacos , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/tratamiento farmacológico , Ratones Noqueados , Estrés Psicológico , Corticosterona , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/efectos de los fármacos , Ratones Endogámicos C57BL , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Ácido Glutámico/metabolismo , Antidepresivos/farmacología
9.
J Exp Clin Cancer Res ; 43(1): 169, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38880883

RESUMEN

BACKGROUND: Cancer is characterized by dysregulated cellular metabolism. Thus, understanding the mechanisms underlying these metabolic alterations is important for developing targeted therapies. In this study, we investigated the pro-tumoral effect of PDZ and LIM domain 2 (PDLIM2) downregulation in lung cancer growth and its association with the accumulation of mitochondrial ROS, oncometabolites and the activation of hypoxia-inducible factor-1 (HIF-1) α in the process. METHODS: Databases and human cancer tissue samples were analyzed to investigate the roles of PDLIM2 and HIF-1α in cancer growth. DNA microarray and gene ontology enrichment analyses were performed to determine the cellular functions of PDLIM2. Seahorse assay, flow cytometric analysis, and confocal microscopic analysis were employed to study mitochondrial functions. Oncometabolites were analyzed using liquid chromatography-mass spectrometry (LC-MS). A Lewis lung carcinoma (LLC) mouse model was established to assess the in vivo function of PDLIM2 and HIF-1α. RESULTS: The expression of PDLIM2 was downregulated in lung cancer, and this downregulation correlated with poor prognosis in patients. PDLIM2 highly regulated genes associated with mitochondrial functions. Mechanistically, PDLIM2 downregulation resulted in NF-κB activation, impaired expression of tricarboxylic acid (TCA) cycle genes particularly the succinate dehydrogenase (SDH) genes, and mitochondrial dysfunction. This disturbance contributed to the accumulation of succinate and other oncometabolites, as well as the buildup of mitochondrial reactive oxygen species (mtROS), leading to the activation of hypoxia-inducible factor 1α (HIF-1α). Furthermore, the expression of HIF-1α was increased in all stages of lung cancer. The expression of PDLIM2 and HIF-1α was reversely correlated in lung cancer patients. In the animal study, the orally administered HIF-1α inhibitor, PX-478, significantly reduces PDLIM2 knockdown-promoted tumor growth. CONCLUSION: These findings shed light on the complex action of PDLIM2 on mitochondria and HIF-1α activities in lung cancer, emphasizing the role of HIF-1α in the tumor-promoting effect of PDLIM2 downregulation. Additionally, they provide new insights into a strategy for precise targeted treatment by suggesting that HIF-1α inhibitors may serve as therapy for lung cancer patients with PDLIM2 downregulation.


Asunto(s)
Regulación hacia Abajo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas con Dominio LIM , Mitocondrias , Especies Reactivas de Oxígeno , Humanos , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Línea Celular Tumoral , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/genética , Regulación Neoplásica de la Expresión Génica , Femenino , Masculino
10.
Am J Physiol Gastrointest Liver Physiol ; 327(1): G25-G35, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38713618

RESUMEN

Cholesterol is essential for the stability and architecture of the plasma membrane and a precursor of bile acids and steroid hormones in mammals. Excess dietary cholesterol uptake leads to hypercholesterolemia and atherosclerosis and plays a role in cancer development. The role of actin-binding scaffolding protein LIM and SH3 protein 1 (LASP1) in cholesterol trafficking has not been investigated previously. Cholesterol levels, its uptake, and excretion were studied in mice deficient for low-density lipoprotein receptor and Lasp1 (Ldlr-/-Lasp1-/- mice) upon feeding a high-fat diet, and in LASP1-knockdown, differentiated human intestinal epithelial CaCo-2 cells. When compared with diet-fed Ldlr-/- control mice, Ldlr-/-Lasp1-/- mice displayed a reduction in serum cholesterol levels. Mechanistically, we identified a new role of LASP1 in controlling the translocation of the intestinal cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1) to the apical cell surface, which was limited in LASP1-knockdown human CaCo-2 enterocytes and in the intestine of Ldlr-/- Lasp1-/- compared with Ldlr-/- mice, linked to LASP1-pAKT signaling but not CDC42 activation. In line, a reduction in cholesterol reabsorption was noted in LASP1-knockdown CaCo-2 cells in vitro, and an enhanced cholesterol excretion via the feces was observed in Ldlr-/- Lasp1-/- mice. These data uncover a novel function of Lasp1 in cholesterol trafficking, promoting cholesterol reabsorption in the intestine. Targeting LASP1 locally could thus represent a novel targeting strategy to ameliorate hypercholesterolemia and associated diseases.NEW & NOTEWORTHY We here uncovered LASP1 as a novel regulator of the shuttling of the sterol transporter NPC1L1 to the cell surface in enterocytes to control cholesterol absorption. Accordingly, LASP1-deficient mice displayed lowered serum cholesterol levels under dietary cholesterol supplementation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Colesterol , Proteínas del Citoesqueleto , Proteínas con Dominio LIM , Proteínas de Transporte de Membrana , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Animales , Células CACO-2 , Humanos , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones , Colesterol/metabolismo , Colesterol/sangre , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Receptores de LDL/metabolismo , Receptores de LDL/genética , Mucosa Intestinal/metabolismo , Enterocitos/metabolismo , Absorción Intestinal , Dieta Alta en Grasa , Proteínas de Homeodominio
11.
J Agric Food Chem ; 72(21): 12240-12250, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38764183

RESUMEN

LIM domain binding 3 (LDB3) serves as a striated muscle-specific Z-band alternatively spliced protein that plays an important role in mammalian skeletal muscle development, but its regulatory role and molecular mechanism in avian muscle development are still unclear. In this study, we reanalyzed RNA sequencing data sets of 1415 samples from 21 chicken tissues published in the NCBI GEO database. First, three variants (LDB3-X, LDB3-XN1, and LDB3-XN2) generated by alternative splicing of the LDB3 gene were identified in chicken skeletal muscle, among which LDB3-XN1 and LDB3-XN2 are novel variants. LDB3-X and LDB3-XN1 are derived from exon skipping in chicken skeletal muscle at the E18-D7 stage and share three LIM domains, but LDB3-XN2 lacks a LIM domain. Our results preliminarily suggest that the formation of three variants of LDB3 is regulated by RBM20. The three splice isomers have divergent functions in skeletal muscle according to in vitro and in vivo assays. Finally, we identified the mechanism by which different variants play different roles through interactions with IGF2BP1 and MYHC, which promote the proliferation and differentiation of chicken myoblasts, in turn regulating chicken myogenesis. In conclusion, this study revealed the divergent roles of three LDB3 variants in chicken myogenesis and muscle remodeling and demonstrated their regulatory mechanism through protein-protein interactions.


Asunto(s)
Empalme Alternativo , Pollos , Proteínas con Dominio LIM , Desarrollo de Músculos , Músculo Esquelético , Animales , Pollos/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/química , Músculo Esquelético/crecimiento & desarrollo , Desarrollo de Músculos/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Mioblastos/metabolismo , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Proteínas Aviares/química , Diferenciación Celular , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química
12.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719752

RESUMEN

Septins are cytoskeletal proteins that participate in cell adhesion, migration, and polarity establishment. The septin subunit SEPT9 directly interacts with the single LIM domain of epithelial protein lost in neoplasm (EPLIN), an actin-bundling protein. Using a human SEPT9 KO fibroblast cell line, we show that cell adhesion and migration are regulated by the interplay between both proteins. The low motility of SEPT9-depleted cells could be partly rescued by increased levels of EPLIN. The normal organization of actin-related filopodia and stress fibers was directly dependent on the expression level of SEPT9 and EPLIN. Increased levels of SEPT9 and EPLIN enhanced the size of focal adhesions in cell protrusions, correlating with stabilization of actin bundles. Conversely, decreased levels had the opposite effect. Our work thus establishes the interaction between SEPT9 and EPLIN as an important link between the septin and the actin cytoskeleton, influencing cell adhesion, motility, and migration.


Asunto(s)
Adhesión Celular , Movimiento Celular , Fibroblastos , Adhesiones Focales , Proteínas con Dominio LIM , Septinas , Humanos , Septinas/metabolismo , Septinas/genética , Movimiento Celular/genética , Fibroblastos/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Adhesiones Focales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Seudópodos/metabolismo , Citoesqueleto de Actina/metabolismo , Línea Celular , Actinas/metabolismo , Fibras de Estrés/metabolismo
13.
Mol Med Rep ; 29(6)2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38695236

RESUMEN

During hematopoiesis, megakaryocytic erythroid progenitors (MEPs) differentiate into megakaryocytic or erythroid lineages in response to specific transcriptional factors, yet the regulatory mechanism remains to be elucidated. Using the MEP­like cell line HEL western blotting, RT­qPCR, lentivirus­mediated downregulation, flow cytometry as well as chromatin immunoprecipitation (ChIp) assay demonstrated that the E26 transformation­specific (ETS) transcription factor friend leukemia integration factor 1 (Fli­1) inhibits erythroid differentiation. The present study using these methods showed that while FLI1­mediated downregulation of GATA binding protein 1 (GATA1) suppresses erythropoiesis, its direct transcriptional induction of GATA2 promotes megakaryocytic differentiation. GATA1 is also involved in megakaryocytic differentiation through regulation of GATA2. By contrast to FLI1, the ETS member erythroblast transformation­specific­related gene (ERG) negatively controls GATA2 and its overexpression through exogenous transfection blocks megakaryocytic differentiation. In addition, FLI1 regulates expression of LIM Domain Binding 1 (LDB1) during erythroid and megakaryocytic commitment, whereas shRNA­mediated depletion of LDB1 downregulates FLI1 and GATA2 but increases GATA1 expression. In agreement, LDB1 ablation using shRNA lentivirus expression blocks megakaryocytic differentiation and modestly suppresses erythroid maturation. These results suggested that a certain threshold level of LDB1 expression enables FLI1 to block erythroid differentiation. Overall, FLI1 controlled the commitment of MEP to either erythroid or megakaryocytic lineage through an intricate regulation of GATA1/GATA2, LDB1 and ERG, exposing multiple targets for cell fate commitment and therapeutic intervention.


Asunto(s)
Diferenciación Celular , Células Eritroides , Megacariocitos , Humanos , Diferenciación Celular/genética , Línea Celular , Células Eritroides/metabolismo , Células Eritroides/citología , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA2/genética , Regulación de la Expresión Génica , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Megacariocitos/metabolismo , Megacariocitos/citología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Regulador Transcripcional ERG/metabolismo , Regulador Transcripcional ERG/genética
14.
J Biol Chem ; 300(5): 107254, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38569934

RESUMEN

Nesprins comprise a family of multi-isomeric scaffolding proteins, forming the linker of nucleoskeleton-and-cytoskeleton complex with lamin A/C, emerin and SUN1/2 at the nuclear envelope. Mutations in nesprin-1/-2 are associated with Emery-Dreifuss muscular dystrophy (EDMD) with conduction defects and dilated cardiomyopathy (DCM). We have previously observed sarcomeric staining of nesprin-1/-2 in cardiac and skeletal muscle, but nesprin function in this compartment remains unknown. In this study, we show that specific nesprin-2 isoforms are highly expressed in cardiac muscle and localize to the Z-disc and I band of the sarcomere. Expression of GFP-tagged nesprin-2 giant spectrin repeats 52 to 53, localized to the sarcomere of neonatal rat cardiomyocytes. Yeast two-hybrid screening of a cardiac muscle cDNA library identified telethonin and four-and-half LIM domain (FHL)-2 as potential nesprin-2 binding partners. GST pull-down and immunoprecipitation confirmed the individual interactions between nesprin-2/telethonin and nesprin-2/FHL-2, and showed that nesprin-2 and telethonin binding was dependent on telethonin phosphorylation status. Importantly, the interactions between these binding partners were impaired by mutations in nesprin-2, telethonin, and FHL-2 identified in EDMD with DCM and hypertrophic cardiomyopathy patients. These data suggest that nesprin-2 is a novel sarcomeric scaffold protein that may potentially participate in the maintenance and/or regulation of sarcomeric organization and function.


Asunto(s)
Conectina , Proteínas con Dominio LIM , Proteínas Musculares , Miocitos Cardíacos , Proteínas del Tejido Nervioso , Proteínas Nucleares , Sarcómeros , Animales , Humanos , Ratones , Ratas , Conectina/metabolismo , Conectina/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Sarcómeros/metabolismo , Factores de Transcripción
15.
Cells ; 13(8)2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38667334

RESUMEN

Meat yield, determined by muscle growth and development, is an important economic trait for the swine industry and a focus of research in animal genetics and breeding. PDZ and LIM domain 5 (PDLIM5) are cytoskeleton-related proteins that play key roles in various tissues and cells. These proteins have multiple isoforms, primarily categorized as short (PDLIM5-short) and long (PDLIM5-long) types, distinguished by the absence and presence of an LIM domain, respectively. However, the expression patterns of swine PDLIM5 isoforms and their regulation during porcine skeletal muscle development remain largely unexplored. We observed that PDLIM5-long was expressed at very low levels in pig muscles and that PDLIM5-short and total PDLIM5 were highly expressed in the muscles of slow-growing pigs, suggesting that PDLIM5-short, the dominant transcript in pigs, is associated with a slow rate of muscle growth. PDLIM5-short suppressed myoblast proliferation and myogenic differentiation in vitro. We also identified two single nucleotide polymorphisms (-258 A > T and -191 T > G) in the 5' flanking region of PDLIM5, which influenced the activity of the promoter and were associated with muscle growth rate in pigs. In summary, we demonstrated that PDLIM5-short negatively regulates myoblast proliferation and differentiation, providing a theoretical basis for improving pig breeding programs.


Asunto(s)
Proteínas con Dominio LIM , Desarrollo de Músculos , Animales , Desarrollo de Músculos/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Porcinos , Proliferación Celular/genética , Diferenciación Celular/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Polimorfismo de Nucleótido Simple/genética , Mioblastos/metabolismo , Mioblastos/citología , Regiones Promotoras Genéticas/genética
16.
Invest Ophthalmol Vis Sci ; 65(4): 43, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38683564

RESUMEN

Purpose: Complement dysregulation is a key component in the pathogenesis of age-related macular degeneration (AMD) and related diseases such as early-onset macular drusen (EOMD). Although genetic variants of complement factor H (CFH) are associated with AMD risk, the impact of CFH and factor H-like protein 1 (FHL-1) expression on local complement activity in human retinal pigment epithelium (RPE) remains unclear. Methods: We identified a novel CFH variant in a family with EOMD and generated patient induced pluripotent stem cell (iPSC)-derived RPE cells. We assessed CFH and FHL-1 co-factor activity through C3b breakdown assays and measured complement activation by immunostaining for membrane attack complex (MAC) formation. Expression of CFH, FHL-1, local alternative pathway (AP) components, and regulators of complement activation (RCA) in EOMD RPE cells was determined by quantitative PCR, western blot, and immunostaining. Isogenic EOMD (cEOMD) RPE was generated using CRISPR/Cas9 gene editing. Results: The CFH variant (c.351-2A>G) resulted in loss of CFH and FHL-1 expression and significantly reduced CFH and FHL-1 protein expression (∼50%) in EOMD iPSC RPE cells. These cells exhibited increased MAC deposition upon exposure to normal human serum. Under inflammatory or oxidative stress conditions, CFH and FHL-1 expression in EOMD RPE cells paralleled that of controls, whereas RCA expression, including MAC formation inhibitors, was elevated. CRISPR/Cas9 correction restored CFH/FHL-1 expression and mitigated alternative pathway complement activity in cEOMD RPE cells. Conclusions: Identification of a novel CFH variant in patients with EOMD resulting in reduced CFH and FHL-1 and increased local complement activity in EOMD iPSC RPE supports the involvement of CFH haploinsufficiency in EOMD pathogenesis.


Asunto(s)
Factor H de Complemento , Haploinsuficiencia , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Degeneración Macular , Proteínas Musculares , Epitelio Pigmentado de la Retina , Humanos , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/metabolismo , Activación de Complemento/genética , Linaje , Western Blotting , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/genética , Drusas Retinianas/genética , Drusas Retinianas/metabolismo , Persona de Mediana Edad
17.
J Mol Cell Cardiol ; 191: 40-49, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38604403

RESUMEN

The heart has the ability to detect and respond to changes in mechanical load through a process called mechanotransduction. In this study, we focused on investigating the role of the cardiac-specific N2B element within the spring region of titin, which has been proposed to function as a mechanosensor. To assess its significance, we conducted experiments using N2B knockout (KO) mice and wildtype (WT) mice, subjecting them to three different conditions: 1) cardiac pressure overload induced by transverse aortic constriction (TAC), 2) volume overload caused by aortocaval fistula (ACF), and 3) exercise-induced hypertrophy through swimming. Under conditions of pressure overload (TAC), both genotypes exhibited similar hypertrophic responses. In contrast, WT mice displayed robust left ventricular hypertrophy after one week of volume overload (ACF), while the KO mice failed to undergo hypertrophy and experienced a high mortality rate. Similarly, swim exercise-induced hypertrophy was significantly reduced in the KO mice. RNA-Seq analysis revealed an abnormal ß-adrenergic response to volume overload in the KO mice, as well as a diminished response to isoproterenol-induced hypertrophy. Because it is known that the N2B element interacts with the four-and-a-half LIM domains 1 and 2 (FHL1 and FHL2) proteins, both of which have been associated with mechanotransduction, we evaluated these proteins. Interestingly, while volume-overload resulted in FHL1 protein expression levels that were comparable between KO and WT mice, FHL2 protein levels were reduced by over 90% in the KO mice compared to WT. This suggests that in response to volume overload, FHL2 might act as a signaling mediator between the N2B element and downstream signaling pathways. Overall, our study highlights the importance of the N2B element in mechanosensing during volume overload, both in physiological and pathological settings.


Asunto(s)
Conectina , Mecanotransducción Celular , Ratones Noqueados , Animales , Ratones , Conectina/metabolismo , Conectina/genética , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Hipertrofia Ventricular Izquierda/genética , Miocardio/metabolismo , Miocardio/patología , Masculino , Condicionamiento Físico Animal , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Modelos Animales de Enfermedad , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Proteínas Quinasas , Péptidos y Proteínas de Señalización Intracelular
18.
Cell Signal ; 119: 111155, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38565413

RESUMEN

BACKGROUND: Esophageal cancer (EC) is highly ranked among all cancers in terms of its incidence and mortality rates. MicroRNAs (miRNAs) are considered to play key regulatory parts in EC. Multiple research studies have indicated the involvement of miR-3682-3p and four and a half LIM domain protein 1 (FHL1) in the achievement of tumors. The aim of this research was to clarify the significance of these genes and their possible molecular mechanism in EC. METHODS: Data from a database and the tissue microarray were made to analyze the expression and clinical significance of miR-3682-3p or FHL1 in EC. Reverse transcription quantitative PCR and Western blotting were used to detect the expression levels of miR-3682-3p and FHL1 in EC cells. CCK8, EdU, wound healing, Transwell, flow cytometry, and Western blotting assays were performed to ascertain the biological roles of miR-3682-3p and FHL1 in EC cells. To confirm the impact of miR-3682-3p in vivo, a subcutaneous tumor model was created in nude mice. The direct interaction between miR-3682-3p and FHL1 was demonstrated through a luciferase assay, and the western blotting technique was employed to assess the levels of crucial proteins within the Wnt/ß-catenin pathway. RESULTS: The noticeable increase in the expression of miR-3682-3p and the decrease in the expression of FHL1 were observed, which correlated with a negative impact on the patients' overall survival. Upregulation of miR-3682-3p expression promoted the growth and metastasis of EC, while overexpression of FHL1 partially reversed these effects. Finally, miR-3682-3p motivates the Wnt/ß-catenin signal transduction by directly targeting FHL1. CONCLUSION: MiR-3682-3p along the FHL1 axis activated the Wnt/ß-catenin signaling pathway and thus promoted EC malignancy.


Asunto(s)
Proliferación Celular , Neoplasias Esofágicas , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Ratones Desnudos , MicroARNs , Proteínas Musculares , Vía de Señalización Wnt , Humanos , MicroARNs/metabolismo , MicroARNs/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Animales , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Línea Celular Tumoral , Ratones , Masculino , Femenino , Progresión de la Enfermedad , Persona de Mediana Edad , beta Catenina/metabolismo , Ratones Endogámicos BALB C , Movimiento Celular/genética
20.
Leukemia ; 38(5): 951-962, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38553571

RESUMEN

Relapse in T-cell acute lymphoblastic leukemia (T-ALL) may signify the persistence of leukemia-initiating cells (L-ICs). Ectopic TAL1/LMO expression defines the largest subset of T-ALL, but its role in leukemic transformation and its impact on relapse-driving L-ICs remain poorly understood. In TAL1/LMO mouse models, double negative-3 (DN3; CD4-CD8-CD25+CD44-) thymic progenitors harbored L-ICs. However, only a subset of DN3 leukemic cells exhibited L-IC activity, and studies linking L-ICs and chemotolerance are needed. To investigate L-IC heterogeneity, we used mouse models and applied single-cell RNA-sequencing and nucleosome labeling techniques in vivo. We identified a DN3 subpopulation with a cell cycle-restricted profile and heightened TAL1/LMO2 activity, that expressed genes associated with stemness and quiescence. This dormant DN3 subset progressively expanded throughout leukemogenesis, displaying intrinsic chemotolerance and enrichment in genes linked to minimal residual disease. Examination of TAL/LMO patient samples revealed a similar pattern in CD7+CD1a- thymic progenitors, previously recognized for their L-IC activity, demonstrating cell cycle restriction and chemotolerance. Our findings substantiate the emergence of dormant, chemotolerant L-ICs during leukemogenesis, and demonstrate that Tal1 and Lmo2 cooperate to promote DN3 quiescence during the transformation process. This study provides a deeper understanding of TAL1/LMO-induced T-ALL and its clinical implications in therapy failure.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas con Dominio LIM , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Proteína 1 de la Leucemia Linfocítica T Aguda , Animales , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/genética , Timo/metabolismo , Timo/patología , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...