Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
1.
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
2.
Cancer Biol Ther ; 25(1): 2343450, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38742566

RÉSUMÉ

The potential function and mechanism of circRNAs in regulating malignant performances of Osteosarcoma (OS) cells have not been well investigated. The expression level of CircLMO7, miR-21-5p and ARHGAP24 were detected by RT-qPCR. The relationship between miR-21-5p and circ-LMO7, as well as between miR-21-5p and ARHGAP24, was predicted and examined through bioinformatics analysis and luciferase reporter gene experiments. Moreover, OS cell growth, invasion, migration, and apoptosis were detected using the cell counting kit-8 (CCK-8), transwell and flow cytometry assays, respectively. ARHGAP24 protein level was measured using western blotting. In present study, we choose to investigate the role and mechanism of circ-LOM7 on OS cell proliferation, migration and invasion. circ-LOM7 was found to be down-regulated in OS tissues and cell lines. Enforced expression of circ-LOM7 suppressed the growth, invasion, and migration of OS cells. In contrast, decreasing circ-LMO7 expression had opposite effects. Furthermore, miR-21-5p was predicted to be sponged by circ-LMO7, and had an opposite role of circ-LMO7 in OS. Moreover, ARHGAP24 served as miR-21-5p's downstream target. Mechanistically, circ-LMO7 was packed in exosomes and acted as a cancer-suppresser on OS by sponging miR-21-5p and upregulating the expression of ARHGAP24. The exosomal circ-LMO7 expression was significantly decreased in OS cell exosomes, and co-culture experiments showed that exosomal circ-LMO7 suppressed the proliferation ability of OS cells. Circ-LMO7 exerts as a tumor suppressor in OS, and the circ-LMO7/miR-21-5P/ARHGAP24 axis is involved in OS progression.


Sujet(s)
Évolution de la maladie , Exosomes , Protéines d'activation de la GTPase , microARN , Ostéosarcome , ARN circulaire , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , microARN/génétique , microARN/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme , Exosomes/métabolisme , Exosomes/génétique , Protéines d'activation de la GTPase/génétique , Protéines d'activation de la GTPase/métabolisme , Prolifération cellulaire , Souris , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Apoptose/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Régulation de l'expression des gènes tumoraux , Mâle , Femelle
3.
Acta Pharm Sin B ; 13(12): 4785-4800, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38045056

RÉSUMÉ

Inflammatory bowel disease (IBD) is a formidable disease due to its complex pathogenesis. Macrophages, as a major immune cell population in IBD, are crucial for gut homeostasis. However, it is still unveiled how macrophages modulate IBD. Here, we found that LIM domain only 7 (LMO7) was downregulated in pro-inflammatory macrophages, and that LMO7 directly degraded 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) through K48-mediated ubiquitination in macrophages. As an enzyme that regulates glycolysis, PFKFB3 degradation led to the glycolytic process inhibition in macrophages, which in turn inhibited macrophage activation and ultimately attenuated murine colitis. Moreover, we demonstrated that PFKFB3 was required for histone demethylase Jumonji domain-containing protein 3 (JMJD3) expression, thereby inhibiting the protein level of trimethylation of histone H3 on lysine 27 (H3K27me3). Overall, our results indicated the LMO7/PFKFB3/JMJD3 axis is essential for modulating macrophage function and IBD pathogenesis. Targeting LMO7 or macrophage metabolism could potentially be an effective strategy for treating inflammatory diseases.

4.
Int J Mol Sci ; 24(18)2023 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-37762205

RÉSUMÉ

The role of long non-coding RNA (lncRNAs) in biological processes remains poorly understood, despite their significant impact. Our previous research discovered that the expression of AL137782.1, a long transcript of the novel lncRNA ENSG00000261553, is upregulated in lung epithelial cells upon exposure to microbes. Furthermore, the expression of AL137782.1 exhibits variability between para-cancerous and lung adenocarcinoma samples. These findings imply that this lncRNA may play a role in both normal lung epithelial cellular processes and pathophysiology. To elucidate the function of AL137782.1 in lung epithelial cells, we utilized bioinformatics retrieval and analysis to examine its expression. We then analyzed its subcellular localization using fluorescence in situ hybridization (FISH) and subcellular fractionation. Through rapid amplification of cDNA ends (RACE), we confirmed the presence of a 4401 nt lncRNA AL137782.1 in lung epithelial cells. Moreover, we discovered that this lncRNA positively regulates both mRNA and the protein expression of LMO7, a protein that may regulate the cell migration of normal lung epithelial cells. Although the overexpression of AL137782.1 has been shown to enhance the migration of both normal lung epithelial cells and lung adenocarcinoma cells in vitro, our study revealed that the expression of this lncRNA was significantly decreased in lung cancers compared to adjacent tissues. This suggests that the cell migration pattern regulated by the AL137782.1-LMO7 axis is more likely to occur in normal lung epithelial cells, rather than being a pathway that promotes lung cancer cell migration. Therefore, our study provides new insights into the mechanism underlying cell migration in human lung epithelial cells. This finding may offer a potential strategy to enhance normal lung epithelial cell migration after lung injury.


Sujet(s)
Adénocarcinome , Tumeurs du poumon , microARN , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Lignée cellulaire tumorale , Hybridation fluorescente in situ , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/métabolisme , Cellules épithéliales/métabolisme , Poumon/anatomopathologie , Adénocarcinome/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , microARN/génétique , Facteurs de transcription/métabolisme , Protéines à domaine LIM/génétique
5.
Acta Pharmaceutica Sinica B ; (6): 4785-4800, 2023.
Article de Anglais | WPRIM (Pacifique Occidental) | ID: wpr-1011216

RÉSUMÉ

Inflammatory bowel disease (IBD) is a formidable disease due to its complex pathogenesis. Macrophages, as a major immune cell population in IBD, are crucial for gut homeostasis. However, it is still unveiled how macrophages modulate IBD. Here, we found that LIM domain only 7 (LMO7) was downregulated in pro-inflammatory macrophages, and that LMO7 directly degraded 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) through K48-mediated ubiquitination in macrophages. As an enzyme that regulates glycolysis, PFKFB3 degradation led to the glycolytic process inhibition in macrophages, which in turn inhibited macrophage activation and ultimately attenuated murine colitis. Moreover, we demonstrated that PFKFB3 was required for histone demethylase Jumonji domain-containing protein 3 (JMJD3) expression, thereby inhibiting the protein level of trimethylation of histone H3 on lysine 27 (H3K27me3). Overall, our results indicated the LMO7/PFKFB3/JMJD3 axis is essential for modulating macrophage function and IBD pathogenesis. Targeting LMO7 or macrophage metabolism could potentially be an effective strategy for treating inflammatory diseases.

6.
Cells ; 11(23)2022 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-36497072

RÉSUMÉ

The kidney epithelial barrier has multifaceted functions in body fluids, electrolyte homeostasis, and urine production. The renal epithelial barrier (REB) frequently faces and challenges osmotic dynamics, which gives rise to osmotic pressure (a physical force). Osmotic pressure overloading can crack epithelial integrity and damage the REB. The endurance of REB to osmotic pressure forces remains obscure. LMO7 (LIM domain only 7) is a protein associated with the cell-cell junctional complex and cortical F-actin. Its upregulation was observed in cells cultured under hypertonic conditions. LMO7 is predominantly distributed in renal tubule epithelial cells. Hypertonic stimulation leads to LMO7 and F-actin assembly in the cortical stress fibers of renal epithelial cells. Hypertonic-isotonic alternation, as a pressure force pushing the plasma membrane inward/outward, was set as osmotic disturbance and was applied to test FAK signaling and LMO7 functioning in maintaining junctional integrity. LMO7 depletion in cells resulted in junctional integrity loss in the epithelial sheet-cultured hypertonic medium or hypertonic-isotonic alternation. Conversely, FAK inhibition by PF-573228 led to failure in robust cortical F-actin assembly and LMO7 association with cortical F-actin in epithelial cells responding to hypertonic stress. Epithelial integrity against osmotic stress and LMO7 and FAK signaling are involved in assembling robust cortical F-actin and maintaining junctional integrity. LMO7 elaborately manages FAK activation in renal epithelial cells, which was demonstrated excessive FAK activation present in LMO7 depleted NRK-52E cells and epithelial integrity loss when cells with LMO7 depletion were exposed to a hypertonic environment. Our data suggests that LMO7 regulates FAK activation and is responsible for maintaining REB under osmotic disturbance.


Sujet(s)
Actines , Podocytes , Pression osmotique , Actines/métabolisme , Podocytes/métabolisme , Cytosquelette d'actine/métabolisme , Transduction du signal
7.
Front Oncol ; 12: 841493, 2022.
Article de Anglais | MEDLINE | ID: mdl-35664754

RÉSUMÉ

Background: Rearrangements of the anaplastic lymphoma kinase (ALK) gene comprise a small subset of non-small cell lung cancer (NSCLC). Patients with NSCLC harboring ALK fusion proteins are sensitive to ALK tyrosine kinase inhibitors (TKIs). Various fusion partners of ALK are being discovered with the application of next-generation sequencing (NGS). Case presentation: Here, we report a female patient with metastatic lung adenocarcinoma harboring LMO7-ALK (L15, A20) rearrangement revealed by NGS. The patient received crizotinib as first-line treatment and has achieved partial response with a progression-free survival over 1 year. Conclusions: We firstly found that the satisfactory response to crizotinib verified the oncogenic activity of LMO7-ALK fusion. Great progression and wide application of NGS facilitate the findings of rare fusion types.

8.
Development ; 149(10)2022 05 15.
Article de Anglais | MEDLINE | ID: mdl-35451459

RÉSUMÉ

Apical constriction, or a reduction in size of the apical domain, underlies many morphogenetic events during development. Actomyosin complexes play an essential role in apical constriction; however, the detailed analysis of molecular mechanisms is still pending. Here, we show that Lim domain only protein 7 (Lmo7), a multidomain adaptor at apical junctions, promotes apical constriction in the Xenopus superficial ectoderm, whereas apical domain size increases in Lmo7-depleted cells. Lmo7 is primarily localized at apical junctions and promotes the formation of the dense circumferential actomyosin belt. Strikingly, Lmo7 binds non-muscle myosin II (NMII) and recruits it to apical junctions and the apical cortex. This NMII recruitment is essential for Lmo7-mediated apical constriction. Lmo7 knockdown decreases NMIIA localization at apical junctions and delays neural tube closure in Xenopus embryos. Our findings suggest that Lmo7 serves as a scaffold that regulates actomyosin contractility and apical domain size.


Sujet(s)
Actomyosine , Ectoderme , Actomyosine/métabolisme , Animaux , Ectoderme/métabolisme , Morphogenèse/physiologie , Chaînes lourdes de myosine , Myosine de type II/génétique , Myosine de type II/métabolisme , Xenopus laevis/métabolisme
9.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-34884689

RÉSUMÉ

LMO7 is a multifunctional PDZ-LIM protein that can interact with different molecular partners and is found in several intracellular locations. The aim of this work was to shed light on LMO7 evolution, alternative transcripts, protein structure and gene regulation through multiple in silico analyses. We also explored the intracellular distribution of the LMO7 protein in chicken and zebrafish embryonic skeletal muscle cells by means of confocal fluorescence microscopy. Our results revealed a single LMO7 gene in mammals, sauropsids, Xenopus and in the holostean fish spotted gar while two lmo7 genes (lmo7a and lmo7b) were identified in teleost fishes. In addition, several different transcripts were predicted for LMO7 in human and in major vertebrate model organisms (mouse, chicken, Xenopus and zebrafish). Bioinformatics tools revealed several structural features of the LMO7 protein including intrinsically disordered regions. We found the LMO7 protein in multiple intracellular compartments in chicken and zebrafish skeletal muscle cells, such as membrane adhesion sites and the perinuclear region. Curiously, the LMO7 protein was detected within the nuclei of muscle cells in chicken but not in zebrafish. Our data showed that a conserved regulatory element may be related to muscle-specific LMO7 expression. Our findings uncover new and important information about LMO7 and open new challenges to understanding how the diverse regulation, structure and distribution of this protein are integrated into highly complex vertebrate cellular milieux, such as skeletal muscle cells.


Sujet(s)
Évolution moléculaire , Protéines à domaine LIM/métabolisme , Modèles moléculaires , Fibres musculaires squelettiques/métabolisme , Facteurs de transcription/métabolisme , Épissage alternatif , Séquence d'acides aminés , Animaux , Poulets , Simulation numérique , Humains , Protéines à domaine LIM/génétique , Souris , Modèles animaux , Conformation des protéines , Facteurs de transcription/génétique , Danio zébré
10.
Front Cell Dev Biol ; 9: 748844, 2021.
Article de Anglais | MEDLINE | ID: mdl-34692701

RÉSUMÉ

Circular RNA (circRNA) is a class of endogenous non-coding RNAs without 5' and 3' ends; an increasing number of studies show that circRNA is involved in skeletal muscle development. From our previous sequencing data, the circRNAome in breast muscle of two chicken lines with a distinct rate of muscle development, which included a fast muscle growing broiler (FMGB) and a slow muscle growing layer (SMGL), we found a novel differentially expressed circRNA generated by intersectin 2 (ITSN2) gene (named circITSN2). We verified that circITSN2 is a skeletal muscle-enriched circRNA that promotes chicken primary myoblast (CPM) proliferation and differentiation. Further molecular mechanism analysis of circITSN2 in chicken myogenesis was performed, and we found circITSN2 directly targeting miR-218-5p. Besides, miR-218-5p inhibits CPM proliferation and differentiation, which is contrary to circITSN2. Commonly, circRNAs act as a miRNA sponge to alleviate the inhibition of miRNAs on mRNAs. Thus, we also identified that a downstream gene LIM domain 7 (LMO7) was inhibited by miR-218-5p, while circITSN2 could block the inhibitory effect of miR-218-5p by targeting it. Functional analysis revealed that LMO7 also accelerates CPM proliferation and differentiation, which was similar to circITSN2 but contrary to miR-218-5p. Taken together, these results suggested that circITSN2 promotes chicken embryonic skeletal muscle development via relieving the inhibition of miR-218-5p on LMO7. Our findings revealed a novel circITSN2/miR-218-5p/LMO7 axis in chicken embryonic skeletal muscle development, which expands our understanding of the complex muscle development regulatory network.

11.
Trends Parasitol ; 37(11): 937-939, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34598896

RÉSUMÉ

Host cytoskeletal rearrangements are an essential yet poorly understood component of Cryptosporidium invasion. Guérin et al. demonstrate that actin rearrangements occur immediately during adherence and capture a unique mechanism of invasion using live-cell imaging. The authors identify a parasite-secreted effector, ROP1, recruited by a host protein, LMO7, involved in pathogenesis.


Sujet(s)
Cryptosporidiose , Cryptosporidium , Parasites , Animaux , Cryptosporidiose/parasitologie , Interactions hôte-parasite , Parasites/métabolisme , Protéines de protozoaire/métabolisme
12.
J Cell Mol Med ; 25(19): 9476-9481, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34541785

RÉSUMÉ

Anaplastic lymphoma kinase (ALK) fusion is a well-defined biomarker for ALK tyrosine kinase inhibitors (TKIs) treatment in non-small cell lung cancer (NSCLC). Alectinib, a second-generation ALK-TKI, has been shown to have significantly longer progression-free survival (PFS) than first-generation ALK inhibitors in untreated ALK-rearranged NSCLC patients. However, its clinical efficacy on rare ALK fusions remains unclear. Herein, two advanced NSCLC patients received first-line alectinib treatment, given their positive ALK fusion status as determined by immunohistochemistry (IHC) testing results. Patients showed limited clinical response (PFS: 4 months) and primary resistance to alectinib respectively. Molecular profiling using next-generation sequencing (NGS) further revealed a striatin (STRN)-ALK fusion in the first patient accompanied by MET amplification, and a LIM domain only protein 7 (LMO7)-ALK fusion in another patient without any other known oncogenic alterations. Both patients demonstrated improved survival after they switched to second-line crizotinib (PFS: 11 months) and ensartinib (PFS: 18 months), respectively, up till the last follow-up assessment. In conclusion, the clinical efficacy of ALK-TKIs including alectinib for lung cancer with uncommon ALK gene fusions is still under evaluation. This study and literature review results showed mixed responses to alectinib in NSCLC patients who harboured rare ALK fusions. Comprehensive molecular profiling of tumour is thus strongly warranted for precise treatment strategies.


Sujet(s)
Kinase du lymphome anaplasique/génétique , Carbazoles/usage thérapeutique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Protéines de fusion oncogènes/génétique , Pipéridines/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Adulte , Marqueurs biologiques tumoraux , Carbazoles/administration et posologie , Carbazoles/effets indésirables , Carcinome pulmonaire non à petites cellules/diagnostic , Carcinome pulmonaire non à petites cellules/mortalité , Prise en charge de la maladie , Femelle , Dépistage génétique , Humains , Tumeurs du poumon/diagnostic , Tumeurs du poumon/mortalité , Mâle , Adulte d'âge moyen , Thérapie moléculaire ciblée , Stadification tumorale , Pipéridines/administration et posologie , Pipéridines/effets indésirables , Pronostic , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/effets indésirables , Tomodensitométrie , Résultat thérapeutique
13.
Cell Host Microbe ; 29(9): 1407-1420.e5, 2021 09 08.
Article de Anglais | MEDLINE | ID: mdl-34348092

RÉSUMÉ

The parasite Cryptosporidium invades and replicates in intestinal epithelial cells and is a leading cause of diarrheal disease and early childhood mortality. The molecular mechanisms that underlie infection and pathogenesis are largely unknown. Here, we delineate the events of host cell invasion and uncover a mechanism unique to Cryptosporidium. We developed a screen to identify parasite effectors, finding the injection of multiple parasite proteins into the host from the rhoptry organelle. These factors are targeted to diverse locations within the host cell and its interface with the parasite. One identified effector, rhoptry protein 1 (ROP1), accumulates in the terminal web of enterocytes through direct interaction with the host protein LIM domain only 7 (LMO7) an organizer of epithelial cell polarity and cell-cell adhesion. Genetic ablation of LMO7 or ROP1 in mice or parasites, respectively, impacts parasite burden in vivo in opposite ways. Taken together, these data provide molecular insight into how Cryptosporidium manipulates its intestinal host niche.


Sujet(s)
Cryptosporidiose/anatomopathologie , Cryptosporidium parvum/pathogénicité , Entérocytes/parasitologie , Protéines à domaine LIM/métabolisme , Protéines membranaires/métabolisme , Protéines de protozoaire/métabolisme , Facteurs de transcription/métabolisme , Animaux , Cellules Caco-2 , Adhérence cellulaire/physiologie , Lignée cellulaire , Modèles animaux de maladie humaine , Entérocytes/cytologie , Cellules épithéliales/parasitologie , Cellules HEK293 , Interactions hôte-parasite/physiologie , Humains , Protéines à domaine LIM/génétique , Souris , Souris de lignée C57BL , Souris knockout , Organites/métabolisme , Facteurs de transcription/génétique
14.
Pathol Res Pract ; 223: 153475, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-33991849

RÉSUMÉ

In our previous study, we reported that the long noncoding RNA, LMO7 downstream neighbor (LMO7DN), has a strong prognostic value in lung adenocarcinoma (LUAD). In this study, we further investigated the role of LMO7DN in LUAD progression. LMO7DN was found to be expressed at low levels in LUAD tissues, and its high expression predicted good prognosis. Bioinformatics analysis indicated that LMO7DN was closely associated with the cell cycle. Furthermore, we found that cell proliferation was significantly enhanced following knockdown of LMO7DN, and the number of cells in the G2/M phase was markedly decreased, whereas there was no change in apoptosis. Thus, LMO7DN inhibits cell proliferation by affecting the cell cycle and is of significant prognostic value in LUAD.


Sujet(s)
Adénocarcinome pulmonaire/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Points de contrôle du cycle cellulaire , Prolifération cellulaire , Tumeurs du poumon/métabolisme , ARN long non codant/métabolisme , Cellules A549 , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Bases de données génétiques , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , ARN long non codant/génétique
15.
Front Cell Dev Biol ; 9: 647387, 2021.
Article de Anglais | MEDLINE | ID: mdl-33763427

RÉSUMÉ

Pancreatic cancer (PC) is one of the most lethal human malignancies without effective treatment. In an effort to discover key genes and molecular pathways underlying PC growth, we have identified LIM domain only 7 (LMO7) as an under-investigated molecule, which highly expresses in primary and metastatic human and mouse PC with the potential of impacting PC tumorigenesis and metastasis. Using genetic methods with siRNA, shRNA, and CRISPR-Cas9, we have successfully generated stable mouse PC cells with LMO7 knockdown or knockout. Using these cells with loss of LMO7 function, we have demonstrated that intrinsic LMO7 defect significantly suppresses PC cell proliferation, anchorage-free colony formation, and mobility in vitro and slows orthotopic PC tumor growth and metastasis in vivo. Mechanistic studies demonstrated that loss of LMO7 function causes PC cell-cycle arrest and apoptosis. These data indicate that LMO7 functions as an independent and unrecognized druggable factor significantly impacting PC growth and metastasis, which could be harnessed for developing a new targeted therapy for PC.

16.
Gene ; 684: 10-19, 2019 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-30359736

RÉSUMÉ

Smallish (Smash), the Drosophila homologue of human LIM domain only 7 (LMO7), is a key regulator of Drosophila embryogenesis associated with planner cell polarity and actomyosin contractility at the zonula adherence. Although smash mRNA is expressed in several tissues during Drosophila development, only Smash function at the adherence junction in the embryonic epithelial cells has been reported. We herein demonstrated that the knockdown of smash in eye imaginal discs induced morphological aberrations in adult compound eyes that were associated with increased apoptosis. Furthermore, immunohistochemical analyses revealed that Smash localized to the nucleus in several tissues, including eye imaginal discs. The knockdown of smash in eye imaginal discs down-regulated the expression of the ote and bocks genes as well as the Drosophila homologue of the emerin gene, which is a target of LMO7. Collectively, these results indicate that Smash functions in proper Drosophila eye development mediated by the regulation of ote and bocks gene expression.


Sujet(s)
Oeil/embryologie , Protéines à domaine LIM/métabolisme , Jonctions adhérentes/métabolisme , Animaux , Apoptose , Noyau de la cellule/métabolisme , Drosophila/embryologie , Drosophila/génétique , Protéines de Drosophila/génétique , Oeil/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Disques imaginaux/métabolisme , Organogenèse , Transduction du signal , Transcription génétique/génétique
17.
Lung Cancer ; 125: 174-184, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30429017

RÉSUMÉ

OBJECTIVES: The human leucine-rich repeats and immunoglobulin-like domains (LRIG) protein family comprises the integral membrane proteins LRIG1, LRIG2 and LRIG3. LRIG1 is frequently down-regulated in human cancer, and high levels of LRIG1 in tumor tissue are associated with favorable clinical outcomes in several tumor types including non-small cell lung cancer (NSCLC). Mechanistically, LRIG1 negatively regulates receptor tyrosine kinases and functions as a tumor suppressor. However, the details of the molecular mechanisms involved are poorly understood, and even less is known about the functions of LRIG2 and LRIG3. The aim of this study was to further elucidate the functions and molecular interactions of the LRIG proteins. MATERIALS AND METHODS: A yeast two-hybrid screen was performed using a cytosolic LRIG3 peptide as bait. In transfected human cells, co-immunoprecipitation and co-localization experiments were performed. Proximity ligation assay was performed to investigate interactions between endogenously expressed proteins. Expression levels of LMO7 and LIMCH1 in normal and malignant lung tissue were investigated using qRT-PCR and through in silico analyses of public data sets. Finally, a clinical cohort comprising 355 surgically treated NSCLC cases was immunostained for LMO7. RESULTS: In the yeast two-hybrid screen, the two paralogous proteins LMO7 and LIMCH1 were identified as interaction partners to LRIG3. LMO7 and LIMCH1 co-localized and co-immunoprecipitated with both LRIG1 and LRIG3. Endogenously expressed LMO7 was in close proximity of both LRIG1 and LRIG3. LMO7 and LIMCH1 were highly expressed in normal lung tissue and down-regulated in malignant lung tissue. LMO7 immunoreactivity was shown to be a negative prognostic factor in LRIG1 positive tumors, predicting poor patient survival. CONCLUSION: These findings suggest that LMO7 and LIMCH1 physically interact with LRIG proteins and that expression of LMO7 is of clinical importance in NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules/métabolisme , Protéines à domaine LIM/métabolisme , Tumeurs du poumon/métabolisme , Glycoprotéines membranaires/métabolisme , Facteurs de transcription/métabolisme , Animaux , Cellules COS , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire , Chlorocebus aethiops , Régulation négative/physiologie , Gènes suppresseurs de tumeur/physiologie , Cellules HEK293 , Humains , Tumeurs du poumon/anatomopathologie , Pronostic
18.
Thyroid ; 28(6): 748-754, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29768105

RÉSUMÉ

BACKGROUND: The BRAFV600E mutation is the most common driver in papillary thyroid carcinoma (PTC) tumors. In recent years, gene fusions have also been recognized as important drivers of cancer in PTC. Previous studies have suggested that thyroid tumors with fusion genes frequently display an aggressive course. These observations prompted further exploration of gene fusions in PTC tumors. The aim was to search for previously unrecognized gene fusions using thyroid tissue samples from PTC patients. METHODS: Gene fusions were analyzed in RNA sequencing data obtained from 12 PTC tumors and paired unaffected thyroid tissue samples. Candidate fusions were further filtered and validated using reverse transcriptase polymerase chain reaction, Sanger sequencing, and fluorescence in situ hybridization. An Ohio cohort of 148 PTC tumor samples was screened for a LMO7-BRAF fusion and the BRAFV600E mutation. Functional assays were performed to assess the LMO7-BRAF fusion. RESULTS: Two coding fusions (CCDC6-RET and LMO7-BRAF) were found in one tumor sample each. The novel LMO7-BRAF fusion was validated by reverse transcriptase polymerase chain reaction and fluorescence in situ hybridization. The LMO7-BRAF fusion was a recurrent somatic alteration with a frequency of 2.0% (3/148) in PTC tumors, while the BRAFV600E point mutation was found in 63.5% (94/148) of tumors. Enforced expression of LMO7-BRAF fusion protein stimulated endogenous ERK1/2 phosphorylation and promoted anchorage independent cell growth to an extent similar to BRAFV600E. CONCLUSIONS: A novel fusion gene, LMO7-BRAF, was identified in PTC tumors. The results indicate that the LMO7-BRAF fusion behaves as an oncogenic alteration. This observation expands the spectrum of fusion genes involving kinases in thyroid cancer.


Sujet(s)
Protéines à domaine LIM/génétique , Protéines de fusion oncogènes/génétique , Protéines proto-oncogènes B-raf/génétique , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/anatomopathologie , Facteurs de transcription/génétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Carcinome papillaire/génétique , Carcinome papillaire/anatomopathologie , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Mutation , Cellules NIH 3T3 , Récidive tumorale locale , Phosphorylation , Mutation ponctuelle , ARN/analyse , Analyse de séquence d'ARN , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/anatomopathologie , Jeune adulte
19.
Int J Biochem Cell Biol ; 94: 22-30, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29158164

RÉSUMÉ

LMO7 (LIM domain only 7) is a transcription regulator for expression of many Emery-Dreifuss muscular dystrophy-relevant genes, and binds to α-actinin and AF6/afadin at adherens junctions for epithelial cell-cell adhesion. In this study, we found that human LMO7 interacted with the spindle assembly checkpoint (SAC) protein MAD1. LMO7 colocalized with actin filaments at the cell membrane but did not colocalize with MAD1 at kinetochores in prometaphase. Our observations reveal that overexpression but not depletion of LMO7 caused a SAC defect, and that the LIM domain of LMO7 was a determinant of its ability to interfere with kinetochore localization of the SAC proteins MAD2 and BUBR1 and cause a SAC defect though the LIM peptide itself did neither bind to MAD1, MAD2 and BUBR1 nor localize to the actin filaments. However, overexpression of LMO7 or the LIM peptide did not interfere with kinetochore localization of MAD1. Additionally, overexpression of the LIM peptide prolonged mitotic timing and interfered with chromosome congression whereas that of LMO7b did not. Taken together, we conclude that LMO7 via its LIM domain acts to control mitosis progression and exerts an effect on the SAC.


Sujet(s)
Cytosquelette d'actine/métabolisme , Protéines du cycle cellulaire/métabolisme , Membrane cellulaire/métabolisme , Protéines à domaine LIM/métabolisme , Points de contrôle de la phase M du cycle cellulaire , Mitose , Protéines nucléaires/métabolisme , Facteurs de transcription/métabolisme , Animaux , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Humains , Interphase , Kinétochores/métabolisme , Protéines à domaine LIM/antagonistes et inhibiteurs , Protéines à domaine LIM/composition chimique , Protéines à domaine LIM/génétique , Protéines luminescentes/composition chimique , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Métaphase , Protéines nucléaires/composition chimique , Protéines nucléaires/génétique , Fragments peptidiques/composition chimique , Fragments peptidiques/génétique , Fragments peptidiques/métabolisme , Prométaphase , Domaines protéiques , Multimérisation de protéines , Transport des protéines , Interférence par ARN , Rats , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/métabolisme , Pôles du fuseau/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/composition chimique , Facteurs de transcription/génétique
20.
J Cell Mol Med ; 21(6): 1228-1236, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28026121

RÉSUMÉ

Detection and treatment of lung cancer still remain a clinical challenge. This study aims to validate exosomal microRNA-96 (miR-96) as a serum biomarker for lung cancer and understand the underlying mechanism in lung cancer progression. MiR-96 expressions in normal and lung cancer patients were characterized by qPCR analysis. Changes in cell viability, migration and cisplatin resistance were monitored after incubation with isolated miR-96-containing exosomes, anti-miR-96 and anti-miR negative control (anti-miR-NC) transfections. Dual-luciferase reporter assay was used to study interaction between miR-96 and LIM-domain only protein 7 (LMO7). Changes induced by miR-96 transfection and LMO7 overexpression were also evaluated. MiR-96 expression was positively correlated with high-grade and metastatic lung cancers. While anti-miR-96 transfection exhibited a tumour-suppressing function, exosomes isolated from H1299 enhanced cell viability, migration and cisplatin resistance. Potential miR-96 binding sites were found within the 3'-UTR of wild-type LMO7 gene, but not of mutant LMO7 gene. LMO7 expression was inversely correlated with lung cancer grades, and LMO7 overexpression reversed promoting effect of miR-96. We have identified exosomal miR-96 as a serum biomarker of malignant lung cancer. MiR-96 promotes lung cancer progression by targeting LMO7. The miR-96-LMO7 axis may be a therapeutic target for lung cancer patients, and new diagnostic or therapeutic strategies could be developed by targeting the miR-96-LMO7 axis.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Protéines à domaine LIM/génétique , Tumeurs du poumon/génétique , microARN/sang , Facteurs de transcription/génétique , Cellules A549 , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Cisplatine/administration et posologie , Résistance aux médicaments antinéoplasiques/génétique , Exosomes/génétique , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Tumeurs du poumon/sang , Tumeurs du poumon/anatomopathologie , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE