Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.129
Filtrer
1.
Mol Cancer ; 23(1): 143, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992675

RÉSUMÉ

BACKGROUND: Emerging evidence indicates the pivotal involvement of circular RNAs (circRNAs) in cancer initiation and progression. Understanding the functions and underlying mechanisms of circRNAs in tumor development holds promise for uncovering novel diagnostic indicators and therapeutic targets. In this study, our focus was to elucidate the function and regulatory mechanism of hsa-circ-0003764 in hepatocellular carcinoma (HCC). METHODS: A newly discovered hsa-circ-0003764 (circPTPN12) was identified from the circbase database. QRT-PCR analysis was utilized to assess the expression levels of hsa-circ-0003764 in both HCC tissues and cells. We conducted in vitro and in vivo experiments to examine the impact of circPTPN12 on the proliferation and apoptosis of HCC cells. Additionally, RNA-sequencing, RNA immunoprecipitation, biotin-coupled probe pull-down assays, and FISH were employed to confirm and establish the relationship between hsa-circ-0003764, PDLIM2, OTUD6B, P65, and ESRP1. RESULTS: In HCC, the downregulation of circPTPN12 was associated with an unfavorable prognosis. CircPTPN12 exhibited suppressive effects on the proliferation of HCC cells both in vitro and in vivo. Mechanistically, RNA sequencing assays unveiled the NF-κB signaling pathway as a targeted pathway of circPTPN12. Functionally, circPTPN12 was found to interact with the PDZ domain of PDLIM2, facilitating the ubiquitination of P65. Furthermore, circPTPN12 bolstered the assembly of the PDLIM2/OTUD6B complex by promoting the deubiquitination of PDLIM2. ESRP1 was identified to bind to pre-PTPN12, thereby fostering the generation of circPTPN12. CONCLUSIONS: Collectively, our findings indicate the involvement of circPTPN12 in modulating PDLIM2 function, influencing HCC progression. The identified ESRP1/circPTPN12/PDLIM2/NF-κB axis shows promise as a novel therapeutic target in the context of HCC.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Protéines à domaine LIM , Tumeurs du foie , Facteur de transcription NF-kappa B , ARN circulaire , Protéines de liaison à l'ARN , Transduction du signal , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Humains , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , ARN circulaire/génétique , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Souris , Animaux , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Lignée cellulaire tumorale , Évolution de la maladie , Apoptose/génétique , Pronostic , Protéines des microfilaments/génétique , Protéines des microfilaments/métabolisme , Mâle , Femelle , Souris nude
2.
Oncol Rep ; 52(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38994754

RÉSUMÉ

Cancer constitutes a multifaceted ailment characterized by the dysregulation of numerous genes and pathways. Among these, LIM domain only 7 (LMO7) has emerged as a significant player in various cancer types, garnering substantial attention for its involvement in tumorigenesis and cancer progression. This review endeavors to furnish a comprehensive discourse on the functional intricacies and mechanisms of LMO7 in cancer, with a particular emphasis on its potential as both a therapeutic target and prognostic indicator. It delves into the molecular attributes of LMO7, its implications in cancer etiology and the underlying mechanisms propelling its oncogenic properties. Furthermore, it underscores the extant challenges and forthcoming prospects in targeting LMO7 for combating cancer.


Sujet(s)
Protéines à domaine LIM , Tumeurs , Facteurs de transcription , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Régulation de l'expression des gènes tumoraux , Pronostic , Carcinogenèse/génétique , Thérapie moléculaire ciblée/méthodes
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(4): 541-552, 2024 Apr 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-39019783

RÉSUMÉ

OBJECTIVES: Super-enhancer-associated genes may be closely related to the progression of osteosarcoma, curcumin exhibits a certain inhibitory effect on tumors such as osteosarcoma. This study aims to investigate the effects of curcumin on osteosarcoma in vitro and in vivo, and to determine whether curcumin can inhibit the progression of osteosarcoma by suppressing the expression of super-enhancer-associated genes LIM and senescent cell antigen-like-containing domain 1 (LIMS1), secreted protein acidic and rich in cysteine (SPARC), and sterile alpha motif domain containing 4A (SAMD4A). METHODS: Human osteosarcoma cell lines (MG63 cells or U2OS cells) were treated with 5 to 50 µmol/L curcumin for 24, 48, and 72 hours, followed by the methyl thiazolyl tetrazolium (MTT) assay to detect cell viability. Cells were incubated with dimethyl sulfoxide (DMSO) or curcumin (2.5, 5.0 µmol/L) for 7 days, and a colony formation assay was used to measure in vitro cell proliferation. After treatment with DMSO or curcumin (10, 15 µmol/L), a scratch healing assay and a transwell migration assay were performed to evaluate cell migration ability. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) and Western blotting were used to detect mRNA and protein expression levels of LIMS1, SPARC, and SAMD4A in the cells. An osteosarcoma-bearing nude mouse model was established, and curcumin was administered via gavage for 14 days to assess the impact of curcumin on tumor volume and weight in vivo. Real-time RT-PCR was used to measure mRNA expression levels of LIMS1, SPARC, and SAMD4A in the cancer and adjacent tissues from 12 osteosarcoma patients. RESULTS: After treating cells with different concentrations of curcumin for 24, 48, and 72 hours, cell viability were all significantly decreased. Compared with the DMSO group, the colony formation rates in the 2.5 µmol/L and 5.0 µmol/L curcumin groups significantly declined (both P<0.01). The scratch healing assay showed that, compared with the DMSO group, the migration rates of cells in the 10 µmol/L and 15 µmol/L curcumin groups were significantly reduced. The exception was the 10 µmol/L curcumin group at 24 h, where the migration rate of U2OS cells did not show a statistically significant difference (P>0.05), while all other differences were statistically significant (P<0.01 or P<0.001). The transwell migration assay results showed that the number of migrating cells in the 10 µmol/L and 15 µmol/L curcumin groups was significantly lower than that in the DMSO group (both P<0.001). In the in vivo tumor-bearing mouse experiment, the curcumin group showed a reduction in tumor mass (P<0.01) and a significant reduction in tumor volume (P<0.001) compared with the control group. Compared with the DMSO group, the mRNA expression levels of LIMS1, SPARC, and SAMD4A in the 10 µmol/L and 15 µmol/L curcumin groups were significantly down-regulated (all P<0.05). Additionally, the protein expression level of LIMS1 in U2OS cells in the 10 µmol/L curcumin group was significantly lower than that in the DMSO group (P<0.05). Compared with adjacent tissues, the mRNA expression level of SPARC in osteosarcoma tissues was significantly increased (P<0.001), while the mRNA expression levels of LIMS1 and SAMD4A did not show statistically significant differences (both P>0.05). CONCLUSIONS: Curcumin inhibits the proliferation and migration of osteosarcoma both in vitro and in vivo, which may be associated with the inactivation of super-enhancer-associated gene LIMS1.


Sujet(s)
Tumeurs osseuses , Mouvement cellulaire , Prolifération cellulaire , Curcumine , Souris nude , Ostéonectine , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/traitement médicamenteux , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Curcumine/pharmacologie , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Animaux , Tumeurs osseuses/génétique , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Lignée cellulaire tumorale , Souris , Ostéonectine/génétique , Ostéonectine/métabolisme , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Souris de lignée BALB C
4.
Nat Commun ; 15(1): 4945, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858386

RÉSUMÉ

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.


Sujet(s)
Modèles animaux de maladie humaine , Kétamine , Neurones , Cortex préfrontal , Synapses , Animaux , Kétamine/pharmacologie , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Synapses/effets des médicaments et des substances chimiques , Synapses/métabolisme , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Souris , Mâle , Humains , Polarité de la cellule/effets des médicaments et des substances chimiques , Trouble dépressif majeur/métabolisme , Trouble dépressif majeur/traitement médicamenteux , Souris knockout , Stress psychologique , Corticostérone , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Acide glutamique/métabolisme , Antidépresseurs/pharmacologie
5.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38838015

RÉSUMÉ

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.


Sujet(s)
Cytosquelette , Protéines à domaine LIM , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Cytosquelette/métabolisme , Acétyl-glucosamine/métabolisme , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Lignée cellulaire tumorale , Glycosylation , Hydrolyse , Mutation , Mouvement cellulaire , Antigènes néoplasiques , Hyaluronoglucosaminidase , Histone acetyltransferases
6.
BMC Oral Health ; 24(1): 729, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918827

RÉSUMÉ

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.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome épidermoïde , Protéines à domaine LIM , Tumeurs de l'oropharynx , Humains , Tumeurs de l'oropharynx/virologie , Tumeurs de l'oropharynx/métabolisme , Tumeurs de l'oropharynx/anatomopathologie , Tumeurs de l'oropharynx/mortalité , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Protéines à domaine LIM/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/virologie , Sujet âgé , Facteurs de transcription/métabolisme , Glycoprotéines membranaires/métabolisme , Adulte , Antigène KI-67/métabolisme , Antigènes CD44/métabolisme , Antigènes CD44/analyse , Protéine p53 suppresseur de tumeur/métabolisme , Infections à papillomavirus/complications , Immunohistochimie , Sujet âgé de 80 ans ou plus , Taux de survie
7.
Genes (Basel) ; 15(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38927709

RÉSUMÉ

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.


Sujet(s)
Génotype , Transplantation rénale , Tubules rénaux , Protéines à domaine LIM , Transplantation rénale/effets indésirables , Humains , Mâle , Femelle , Adulte d'âge moyen , Adulte , Protéines à domaine LIM/génétique , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Rejet du greffon/génétique , Rejet du greffon/anatomopathologie , Polymorphisme de nucléotide simple
8.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928252

RÉSUMÉ

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.


Sujet(s)
Protéines à domaine LIM , Myosite à inclusions , Pedigree , Humains , Myosite à inclusions/génétique , Myosite à inclusions/anatomopathologie , Mâle , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Adulte d'âge moyen , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Mutation , Adulte
9.
BMC Cancer ; 24(1): 772, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38937681

RÉSUMÉ

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.


Sujet(s)
Prédisposition génétique à une maladie , Tumeurs du rein , Protéines à domaine LIM , Polymorphisme de nucléotide simple , Tumeur de Wilms , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Protéines adaptatrices de la transduction du signal/génétique , Études cas-témoins , Chine/épidémiologie , Protéines de liaison à l'ADN/génétique , Peuples d'Asie de l'Est/génétique , Génotype , Tumeurs du rein/génétique , Protéines à domaine LIM/génétique , Protéines proto-oncogènes/génétique , Facteurs de transcription/génétique , Tumeur de Wilms/génétique , Famille multigénique
10.
Eur J Med Res ; 29(1): 309, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38831471

RÉSUMÉ

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.


Sujet(s)
Remodelage des voies aériennes , Apoptose , Prolifération cellulaire , microARN , Broncho-pneumopathie chronique obstructive , ARN long non codant , ARN long non codant/génétique , ARN long non codant/métabolisme , Humains , microARN/génétique , microARN/métabolisme , Apoptose/génétique , Remodelage des voies aériennes/génétique , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Broncho-pneumopathie chronique obstructive/anatomopathologie , Prolifération cellulaire/génétique , Animaux , Souris , Mâle , Système de signalisation des MAP kinases/génétique , Pneumocytes/métabolisme , Pneumocytes/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Femelle , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Adulte d'âge moyen , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Souris de lignée C57BL
11.
J Exp Clin Cancer Res ; 43(1): 169, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38880883

RÉSUMÉ

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.


Sujet(s)
Régulation négative , Sous-unité alpha du facteur-1 induit par l'hypoxie , Protéines à domaine LIM , Mitochondries , Espèces réactives de l'oxygène , Humains , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Animaux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Souris , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Lignée cellulaire tumorale , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Carcinome pulmonaire de Lewis/métabolisme , Carcinome pulmonaire de Lewis/anatomopathologie , Carcinome pulmonaire de Lewis/génétique , Régulation de l'expression des gènes tumoraux , Femelle , Mâle
12.
Arch Dermatol Res ; 316(7): 401, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878083

RÉSUMÉ

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.


Sujet(s)
Glucosides , Peroxyde d'hydrogène , Protéines à domaine LIM , Mélanocytes , Monoterpènes , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Transduction du signal , rho-Associated Kinases , Protéine G RhoA , Facteur-2 apparenté à NF-E2/métabolisme , rho-Associated Kinases/métabolisme , Mélanocytes/effets des médicaments et des substances chimiques , Mélanocytes/métabolisme , Humains , Glucosides/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Protéine G RhoA/métabolisme , Peroxyde d'hydrogène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Monoterpènes/pharmacologie , Lignée cellulaire
13.
Am J Physiol Gastrointest Liver Physiol ; 327(1): G25-G35, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38713618

RÉSUMÉ

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.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Cholestérol , Protéines du cytosquelette , Protéines à domaine LIM , Protéines de transport membranaire , Souris knockout , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Cellules Caco-2 , Humains , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines proto-oncogènes c-akt/métabolisme , Souris , Cholestérol/métabolisme , Cholestérol/sang , Protéines de transport membranaire/métabolisme , Protéines de transport membranaire/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Muqueuse intestinale/métabolisme , Entérocytes/métabolisme , Absorption intestinale , Alimentation riche en graisse , Protéines à homéodomaine
14.
Oncogene ; 43(28): 2184-2198, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38789663

RÉSUMÉ

Human papillomaviruses (HPV) are a major cause of malignancy, contributing to ~5% of all human cancers worldwide, including most cervical cancer cases and a growing number of anogenital and oral cancers. The major HPV viral oncogenes, E6 and E7, manipulate many host cellular pathways that promote cell proliferation and survival, predisposing infected cells to malignant transformation. Despite the availability of highly effective vaccines, there are still no specific anti-viral therapies targeting HPV or treatments for HPV-associated cancers. As such, a better understanding of viral-host interactions may allow the identification of novel therapeutic targets. Here, we demonstrate that the actin-binding protein LASP1 is upregulated in cervical cancer and significantly correlates with a poorer overall survival. In HPV positive cervical cancer, LASP1 depletion significantly inhibited the oncogenic phenotype in vitro, whilst having minimal effects in HPV negative cervical cancer cells. Furthermore, we demonstrate that the LASP1 SH3 domain is essential for LASP1-mediated oncogenicity in these cells. Mechanistically, we show that HPV E7 regulates LASP1 at the post-transcriptional level by repressing the expression of miR-203, which negatively regulates LASP1 mRNA levels by binding to its 3'UTR. Finally, we demonstrate that LASP1 expression is required for the growth of HPV positive cervical cancer cells in an in vivo tumourigenicity model. Together, these data demonstrate that HPV induces LASP1 expression to promote proliferation and survival in cervical cancer, thus identifying a potential therapeutic target in these cancers.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Prolifération cellulaire , Protéines du cytosquelette , Protéines à domaine LIM , microARN , Protéines E7 de papillomavirus , Infections à papillomavirus , Tumeurs du col de l'utérus , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/anatomopathologie , Protéines du cytosquelette/génétique , Protéines du cytosquelette/métabolisme , microARN/génétique , Humains , Femelle , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Prolifération cellulaire/génétique , Infections à papillomavirus/virologie , Infections à papillomavirus/génétique , Infections à papillomavirus/complications , Infections à papillomavirus/anatomopathologie , Protéines E7 de papillomavirus/génétique , Protéines E7 de papillomavirus/métabolisme , Animaux , Souris , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale
15.
Mol Med Rep ; 29(6)2024 06.
Article de Anglais | MEDLINE | ID: mdl-38695236

RÉSUMÉ

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.


Sujet(s)
Différenciation cellulaire , Cellules érythroïdes , Mégacaryocytes , Humains , Différenciation cellulaire/génétique , Lignée cellulaire , Cellules érythroïdes/métabolisme , Cellules érythroïdes/cytologie , Facteur de transcription GATA-1/métabolisme , Facteur de transcription GATA-1/génétique , Facteur de transcription GATA-2/métabolisme , Facteur de transcription GATA-2/génétique , Régulation de l'expression des gènes , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Mégacaryocytes/métabolisme , Mégacaryocytes/cytologie , Protéine proto-oncogène c-fli-1/métabolisme , Protéine proto-oncogène c-fli-1/génétique , Régulateur transcriptionnel ERG/métabolisme , Régulateur transcriptionnel ERG/génétique
16.
Life Sci Alliance ; 7(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38719752

RÉSUMÉ

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.


Sujet(s)
Adhérence cellulaire , Mouvement cellulaire , Fibroblastes , Contacts focaux , Protéines à domaine LIM , Septines , Humains , Septines/métabolisme , Septines/génétique , Mouvement cellulaire/génétique , Fibroblastes/métabolisme , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Contacts focaux/métabolisme , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Pseudopodes/métabolisme , Cytosquelette d'actine/métabolisme , Lignée cellulaire , Actines/métabolisme , Fibres de stress/métabolisme
17.
J Agric Food Chem ; 72(21): 12240-12250, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38764183

RÉSUMÉ

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.


Sujet(s)
Épissage alternatif , Poulets , Protéines à domaine LIM , Développement musculaire , Muscles squelettiques , Animaux , Poulets/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/composition chimique , Muscles squelettiques/croissance et développement , Développement musculaire/génétique , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Myoblastes/métabolisme , Protéines aviaires/génétique , Protéines aviaires/métabolisme , Protéines aviaires/composition chimique , Différenciation cellulaire , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/composition chimique
18.
J Mol Cell Cardiol ; 191: 40-49, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38604403

RÉSUMÉ

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.


Sujet(s)
Connectine , Mécanotransduction cellulaire , Souris knockout , Animaux , Souris , Connectine/métabolisme , Connectine/génétique , Hypertrophie ventriculaire gauche/métabolisme , Hypertrophie ventriculaire gauche/physiopathologie , Hypertrophie ventriculaire gauche/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Mâle , Conditionnement physique d'animal , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Modèles animaux de maladie humaine , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Protein kinases , Protéines et peptides de signalisation intracellulaire
19.
J Biol Chem ; 300(5): 107254, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569934

RÉSUMÉ

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.


Sujet(s)
Connectine , Protéines à domaine LIM , Protéines du muscle , Myocytes cardiaques , Protéines de tissu nerveux , Protéines nucléaires , Sarcomères , Animaux , Humains , Souris , Rats , Connectine/métabolisme , Connectine/génétique , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines à domaine LIM/métabolisme , Protéines à domaine LIM/génétique , Protéines à homéodomaine LIM , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Myocytes cardiaques/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Liaison aux protéines , Sarcomères/métabolisme , Facteurs de transcription
20.
Cells ; 13(8)2024 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-38667334

RÉSUMÉ

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.


Sujet(s)
Protéines à domaine LIM , Développement musculaire , Animaux , Développement musculaire/génétique , Protéines à domaine LIM/génétique , Protéines à domaine LIM/métabolisme , Suidae , Prolifération cellulaire/génétique , Différenciation cellulaire/génétique , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Muscles squelettiques/métabolisme , Muscles squelettiques/croissance et développement , Polymorphisme de nucléotide simple/génétique , Myoblastes/métabolisme , Myoblastes/cytologie , Régions promotrices (génétique)/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...