Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.321
Filtrer
1.
Invest Ophthalmol Vis Sci ; 65(11): 4, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39230995

RÉSUMÉ

Purpose: We assessed whether NICD1 expression, c-MYC expression, and P63 expression by immunohistochemistry (IHC) correlate with prognosis and high-risk clinicopathological features in lacrimal gland adenoid cystic carcinoma (ACC). Methods: Records of patients with lacrimal gland ACC who underwent surgery between 1998 to 2018 were reviewed. Clinicopathologic and treatment data were collected. Tumor tissues were subjected to light microscopy and IHC. Results: Of 43 patients treated during the study period, 21 had archived tumor tissue available and were included. The median age at diagnosis was 47 years, and 13 patients (62%) were male. Thirteen patients (62%) had T2 disease, and none had nodal or distant metastasis at diagnosis. Tumors were positive for NICD1 expression in eight cases (38%), c-MYC expression in eight (38%), and P63 expression in 11 (52%). Positive NICD1 expression was associated with predominantly solid (vs. cribriform/tubular) pattern (P < 0.001), treatment with orbital exenteration (vs. eye-sparing surgery) (P = 0.008), local recurrence (P = 0.047), and death (P = 0.012). Negative P63 expression was associated with predominantly solid pattern (P = 0.001), local recurrence (P = 0.012), distant metastasis (P = 0.001), and death (P = 0.035). A higher percentage of tumor cells staining for c-MYC was associated with presence of perineural invasion (P = 0.036). Positive NICD1 expression was associated with worse disease-free survival (hazard ratio, 6.27; 95% CI, 1.29-30.46), whereas positive P63 expression was associated with better disease-free survival (hazard ratio, 0.03; 95% CI, 0.0002-0.26). Conclusions: IHC for NICD1 and P63 should be considered in lacrimal gland ACC because of their prognostic value and potential as treatment targets.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinome adénoïde kystique , Tumeurs de l'oeil , Maladies de l'appareil lacrymal , Protéines proto-oncogènes c-myc , Récepteur Notch1 , Humains , Carcinome adénoïde kystique/métabolisme , Carcinome adénoïde kystique/anatomopathologie , Carcinome adénoïde kystique/diagnostic , Mâle , Adulte d'âge moyen , Femelle , Adulte , Pronostic , Maladies de l'appareil lacrymal/métabolisme , Maladies de l'appareil lacrymal/anatomopathologie , Maladies de l'appareil lacrymal/diagnostic , Sujet âgé , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Tumeurs de l'oeil/métabolisme , Tumeurs de l'oeil/anatomopathologie , Tumeurs de l'oeil/diagnostic , Études rétrospectives , Marqueurs biologiques tumoraux/métabolisme , Récepteur Notch1/métabolisme , Immunohistochimie , Jeune adulte , Protéines membranaires , Facteurs de transcription , Protéines suppresseurs de tumeurs
2.
BMC Pharmacol Toxicol ; 25(1): 63, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39243105

RÉSUMÉ

The impact of Sodium Houttuyniae (SH) on lipopolysaccharide (LPS)-induced ALI has been investigated extensively. However, it remains ambiguous whether ferroptosis participates in this process. This study aimed to find out the impacts and probable mechanisms of SH on LPS-induced ferroptosis. A rat ALI model and type II alveolar epithelial (ATII) cell injury model were treated with LPS. Enzyme-linked immunosorbent assay (ELISA), hematoxylin-eosin (HE) staining, and Giemsa staining were executed to ascertain the effects of SH on LPS-induced ALI. Moreover, Transmission electron microscopy, Cell Counting Kit-8 (CCK8), ferrous iron colorimetric assay kit, Immunohistochemistry, Immunofluorescence, Reactive oxygen species assay kit, western blotting (Wb), and qRT-PCR examined the impacts of SH on LPS-induced ferroptosis and ferroptosis-related pathways. Theresults found that by using SH treatment, there was a remarkable attenuation of ALI by suppressing LPS-induced ferroptosis. Ferroptosis was demonstrated by a decline in the levels of glutathione peroxidase 4 (GPX4), FTH1, and glutathione (GSH) and a surge in the accumulation of malondialdehyde (MDA), reactive oxygen species (ROS), NOX1, NCOA4, and Fe2+, and disruption of mitochondrial structure, which were reversed by SH treatment. SH suppressed ferroptosis by regulating TRAF6-c-Myc in ALI rats and rat ATII cells. The results suggested that SH treatment attenuated LPS-induced ALI by repressing ferroptosis, and the mode of action can be linked to regulating the TRAF6-c-Myc signaling pathway in vivo and in vitro.


Sujet(s)
Lésion pulmonaire aigüe , Médicaments issus de plantes chinoises , Ferroptose , Lipopolysaccharides , Protéines proto-oncogènes c-myc , Transduction du signal , Facteur-6 associé aux récepteurs de TNF , Animaux , Mâle , Rats , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/anatomopathologie , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Ferroptose/effets des médicaments et des substances chimiques , Lipopolysaccharides/toxicité , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Rat Sprague-Dawley , Espèces réactives de l'oxygène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique
3.
Neoplasma ; 71(4): 347-358, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39267538

RÉSUMÉ

Breast cancer is the most common malignant tumor in women. Recurrence, metastasis, and chemotherapy resistance are the main causes of death in breast cancer patients. The inhibition of breast cancer metastasis is of great significance for prolonging its survival. Ribosome biogenesis regulatory protein homolog (RRS1) is overexpressed in breast cancer tissues and is involved in regulating the carcinogenic process of breast cancer cells. However, the exact signaling pathway and molecular mechanism of RRS1 promoting breast cancer metastasis are not fully understood. Hence, the primary objective of our study is to investigate the correlation between RRS1 and breast cancer metastasis. Bioinformatic analysis was used to identify the expression levels and prognostic significance of RRS1 in breast cancer. Lenti-sh RRS1 lentivirus was constructed and employed to downregulate the RRS1 expression in MDA-MB-231 and BT549 cells, which had a high-level expression of RRS1. Subsequently, we assessed the impact of RRS1 downregulation on the proliferation, migration, and invasion of breast cancer cells using CCK-8, apoptosis, and cell cycle by flow cytometry, wound healing test, Transwell migration, and invasion experiments. Moreover, we utilized an in vivo imaging system to examine the metastatic potential of breast cancer cells after RRS1 knockdown. Picrate staining and hematoxylin-eosin staining were employed to evaluate the presence of metastatic lesions. To gain a deeper understanding of the molecular mechanism, we conducted co-immunoprecipitation and western blot. The significant overexpression of RRS1 in breast cancer indicates a worse prognosis, as determined through TCGA databases (p<0.01). Additionally, RRS1 exhibits upregulation in breast cancer (p<0.001), which is tightly linked to the occurrence of lymph node metastasis (p<0.001). Clinical breast cancer tissues and breast cancer cell lines also demonstrated a noteworthy upregulation of RRS1 (p<0.05). Loss-of-function experiment illustrated that the inhibiting of RRS1 expression reduced the rapid proliferation capacity of MDA-MB-231 and BT549 cells and hindered their migration and invasion capabilities (p<0.05). Importantly, the suppression of RRS1 significantly diminished lung metastasis in Balb/c nude mice that were injected with MDA-MB-231 cells (p<0.01). Mechanistically, RRS1 may interact with the AEG-1 to modulate the phosphorylation of AKT at T308 and S473, consequently impeding the activity of c-Myc (p<0.05). To conclude, RRS1 functions as a potential oncogene in breast cancer by leveraging the AEG-1/AKT/c-Myc signaling.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Prolifération cellulaire , Protéines membranaires , Protéines proto-oncogènes c-akt , Protéines proto-oncogènes c-myc , Protéines de liaison à l'ARN , Transduction du signal , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Femelle , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines proto-oncogènes c-akt/métabolisme , Lignée cellulaire tumorale , Animaux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines membranaires/physiologie , Souris , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Molécules d'adhérence cellulaire/métabolisme , Molécules d'adhérence cellulaire/génétique , Apoptose , Pronostic , Régulation de l'expression des gènes tumoraux , Métastase tumorale , Souris nude , Invasion tumorale , Souris de lignée BALB C
4.
Cell Death Dis ; 15(9): 661, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39256367

RÉSUMÉ

De novo purine synthesis metabolism plays a crucial role in tumor cell survival and malignant progression. However, the specific impact of this metabolic pathway on chemoresistance in ovarian cancer remains unclear. This study aims to elucidate the influence of de novo purine synthesis on chemoresistance in ovarian cancer and its underlying regulatory mechanisms. We analyzed metabolic differences between chemosensitive and chemoresistant ovarian cancer tissues using mass spectrometry-based metabolomics. Cell growth, metabolism, chemoresistance, and DNA damage repair characteristics were assessed in vitro using cell line models. Tumor growth and chemoresistance were assessed in vivo using ovarian cancer xenograft tumors. Intervention of purines and NEK6-mediated purine metabolism on chemoresistance was investigated at multiple levels. Chemoresistant ovarian cancers exhibited higher purine abundance and NEK6 expression. Inhibiting NEK6 led to decreased de novo purine synthesis, resulting in diminished chemoresistance in ovarian cancer cells. Mechanistically, NEK6 directly interacted with FOXO3, contributing to the phosphorylation of FOXO3 at S7 through its kinase activity, thereby inhibiting its nuclear translocation. Nuclear FOXO3 promoted FBXW7 transcription, leading to c-MYC ubiquitination and suppression of de novo purine synthesis. Paeonol, by inhibiting NEK6, suppressed de novo purine synthesis and enhanced chemosensitivity. The NEK6-mediated reprogramming of de novo purine synthesis emerges as a critical pathway influencing chemoresistance in ovarian cancer. Paeonol exhibits the potential to interfere with NEK6, thereby inhibiting chemoresistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Protéine O3 à motif en tête de fourche , Kinases apparentées à NIMA , Tumeurs de l'ovaire , Protéines proto-oncogènes c-myc , Purines , Femelle , Humains , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Purines/pharmacologie , Purines/métabolisme , Lignée cellulaire tumorale , Animaux , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Kinases apparentées à NIMA/métabolisme , Kinases apparentées à NIMA/génétique , Souris , Souris nude , Noyau de la cellule/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
5.
J Hematol Oncol ; 17(1): 83, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39267119

RÉSUMÉ

BACKGROUND: Macrophage-based cell therapy is promising in solid tumors, but the efficient acquisition of macrophages remains a challenge. Induced pluripotent stem cell (iPSC)-induced macrophages are a valuable source, but time-consuming and costly. The application of reprogramming technologies allows for the generation of macrophages from somatic cells, thereby facilitating the advancement of cell-based therapies for numerous malignant diseases. METHODS: The composition of CD45+ myeloid-like cell complex (MCC) and induced macrophage (iMac) were analyzed by flow cytometry and single-cell RNA sequencing. The engraftment capacity of CD45+ MCC was evaluated by two transplantation assays. Regulation of c-Myc on MafB was evaluated by ChIP-qPCR and promoter reporter and dual luciferase assays. The phenotype and phagocytosis of iMac were explored by flow cytometry and immunofluorescence. Leukemia, breast cancer, and patient-derived tumor xenograft models were used to explore the anti-tumor function of iMac. RESULTS: Here we report on the establishment of a novel methodology allowing for reprogramming fibroblasts into functional macrophages with phagocytic activity by c-Myc overexpression. Fibroblasts with ectopic expression of c-Myc in iPSC medium rapidly generated CD45+ MCC intermediates with engraftment capacity as well as the repopulation of distinct hematopoietic compartments. MCC intermediates were stably maintained in iPSC medium and continuously generated functional and highly pure iMac just by M-CSF cytokine stimulation. Single-cell transcriptomic analysis of MCC intermediates revealed that c-Myc up-regulated the expression of MafB, a major regulator of macrophage differentiation, to promote macrophage differentiation. Characterization of the iMac activity showed NF-κB signaling activation and a pro-inflammatory phenotype. iMac cells displayed significantly increased in vivo persistence and inhibition of tumor progression in leukemia, breast cancer, and patient-derived tumor xenograft models. CONCLUSIONS: Our findings demonstrate that c-Myc alone is enough to reprogram fibroblasts into functional macrophages, supporting that c-Myc reprogramming strategy of fibroblasts can help circumvent long-standing obstacles to gaining "off-the-shelf" macrophages for anti-cancer immunotherapy.


Sujet(s)
Reprogrammation cellulaire , Fibroblastes , Macrophages , Protéines proto-oncogènes c-myc , Macrophages/métabolisme , Macrophages/cytologie , Animaux , Humains , Souris , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Fibroblastes/métabolisme , Fibroblastes/cytologie , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Femelle
6.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273365

RÉSUMÉ

Though Ginsenoside F2 (GF2), a protopanaxadiol saponin from Panax ginseng, is known to have an anticancer effect, its underlying mechanism still remains unclear. In our model, the anti-glycolytic mechanism of GF2 was investigated in human cervical cancer cells in association with miR193a-5p and the ß-catenin/c-Myc/Hexokinase 2 (HK2) signaling axis. Here, GF2 exerted significant cytotoxicity and antiproliferation activity, increased sub-G1, and attenuated the expression of pro-Poly (ADPribose) polymerase (pro-PARP) and pro-cysteine aspartyl-specific protease (procaspase3) in HeLa and SiHa cells. Consistently, GF2 attenuated the expression of Wnt, ß-catenin, and c-Myc and their downstream target genes such as HK2, pyruvate kinase isozymes M2 (PKM2), and lactate dehydrogenase A (LDHA), along with a decreased production of glucose and lactate in HeLa and SiHa cells. Moreover, GF2 suppressed ß-catenin and c-Myc stability in the presence and absence of cycloheximide in HeLa cells, respectively. Additionally, the depletion of ß-catenin reduced the expression of c-Myc and HK2 in HeLa cells, while pyruvate treatment reversed the ability of GF2 to inhibit ß-catenin, c-Myc, and PKM2 in GF2-treated HeLa cells. Notably, GF2 upregulated the expression of microRNA139a-5p (miR139a-5p) in HeLa cells. Consistently, the miR139a-5p mimic enhanced the suppression of ß-catenin, c-Myc, and HK2, while the miR193a-5p inhibitor reversed the ability of GF2 to attenuate the expression of ß-catenin, c-Myc, and HK2 in HeLa cells. Overall, these findings suggest that GF2 induces apoptosis via the activation of miR193a-5p and the inhibition of ß-catenin/c-Myc/HK signaling in cervical cancer cells.


Sujet(s)
Ginsénosides , Hexokinase , microARN , Protéines proto-oncogènes c-myc , Transduction du signal , Tumeurs du col de l'utérus , bêta-Caténine , Humains , Ginsénosides/pharmacologie , bêta-Caténine/métabolisme , bêta-Caténine/génétique , microARN/génétique , microARN/métabolisme , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/anatomopathologie , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Hexokinase/métabolisme , Hexokinase/génétique , Cellules HeLa , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Effet Warburg en oncologie/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
7.
Hereditas ; 161(1): 29, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39223679

RÉSUMÉ

BACKGROUND: Small cell lung carcinoma (SCLC) is characterized by -poor prognosis, -high predilection for -metastasis, -proliferation, and -absence of newer therapeutic options. Elucidation of newer pathways characterizing the disease may allow for development of targeted therapies and consequently favorable outcomes. METHODS: The current study explored the combinatorial action of arsenic trioxide (ATO) and apatinib (APA) in vitro and in vivo. In vitro models were tested using -H446 and -H196 SCLC cell lines. The ability of drugs to reduce -metastasis, -cell proliferation, and -migration were assessed. Using bioinformatic analysis, differentially expressed genes were determined. Gene regulation was assessed using gene knock down models and confirmed using Western blots. The in vivo models were used to confirm the resolution of pathognomic features in the presence of the drugs. Growth factor receptor bound protein (GRB) 10 expression levels of human small cell lung cancer tissues and adjacent tissues were detected by IHC. RESULTS: In combination, ATO and APA were found to significantly reduce -cell proliferation, -migration, and -metastasis in both the cell lines. Cell proliferation was found to be inhibited by activation of Caspase-3, -7 pathway. In the presence of drugs, it was found that expression of GRB10 was stabilized. The silencing of GRB10 was found to negatively regulate the VEGFR2/Akt/mTOR and Akt/GSK-3ß/c-Myc signaling pathway. Concurrently, absence of metastasis and reduction of tumor volume were confirmed in vivo. The immunohistochemical results confirmed that the expression level of GRB10 in adjacent tissues was significantly higher than that in human small cell lung cancer tissues. CONCLUSIONS: Synergistically, ATO and APA have a more significant impact on inhibiting cell proliferation than each drug independently. ATO and APA may be mediating its action through the stabilization of GRB10 thus acting as a tumor suppressor. We thus, preliminarily report the impact of GRB10 stability as a target for SCLC treatment.


Sujet(s)
Trioxyde d'arsenic , Prolifération cellulaire , Synergie des médicaments , Tumeurs du poumon , Protéines proto-oncogènes c-akt , Pyridines , Transduction du signal , Carcinome pulmonaire à petites cellules , Sérine-thréonine kinases TOR , Récepteur-2 au facteur croissance endothéliale vasculaire , Trioxyde d'arsenic/usage thérapeutique , Trioxyde d'arsenic/pharmacologie , Humains , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/génétique , Protéines proto-oncogènes c-akt/métabolisme , Lignée cellulaire tumorale , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéine adaptatrice GRB10/génétique , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation négative , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie
8.
Nat Commun ; 15(1): 7675, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39227594

RÉSUMÉ

Most prostate cancers express the androgen receptor (AR), and tumor growth and progression are facilitated by exceptionally low levels of systemic or intratumorally produced androgens. Thus, absolute inhibition of the androgen signaling axis remains the goal of current therapeutic approaches to treat prostate cancer (PCa). Paradoxically, high dose androgens also exhibit considerable efficacy as a treatment modality in patients with late-stage metastatic PCa. Here we show that low levels of androgens, functioning through an AR monomer, facilitate a non-genomic activation of the mTOR signaling pathway to drive proliferation. Conversely, high dose androgens facilitate the formation of AR dimers/oligomers to suppress c-MYC expression, inhibit proliferation and drive a transcriptional program associated with a differentiated phenotype. These findings highlight the inherent liabilities in current approaches used to inhibit AR action in PCa and are instructive as to strategies that can be used to develop new therapeutics for this disease and other androgenopathies.


Sujet(s)
Androgènes , Prolifération cellulaire , Tumeurs de la prostate , Récepteurs aux androgènes , Transduction du signal , Sérine-thréonine kinases TOR , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Mâle , Humains , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/génétique , Tumeurs de la prostate/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Androgènes/métabolisme , Androgènes/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Multimérisation de protéines/effets des médicaments et des substances chimiques , Animaux
9.
CNS Neurosci Ther ; 30(9): e70017, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39218810

RÉSUMÉ

OBJECTIVE: The E3 ubiquitin ligase is well recognized as a significant contributor to glioblastoma (GBM) progression and has promise as a prospective therapeutic target. This study explores the contribution of E3 ubiquitin ligase RNF122 in the GBM progression and the related molecular mechanisms. METHODS: RNF122 expression levels were evaluated using qRT-PCR, WB, and IHC, while functional assays besides animal experiments were used to assess RNF122's effect on GBM progression. We also tested the RNF122 impact on JAK2/STAT3/c-Myc signaling using WB. RESULTS: RNF122 was upregulated in GBM and correlated to the advanced stage and poor clinical outcomes, representing an independent prognostic factor. Based on functional assays, RNF122 promotes GBM growth and cell cycle, which was validated further in subsequent analyses by JAK2/STAT3/c-Myc pathway activation. Moreover, JAK2/STAT3 signaling pathway inhibitor WP1066 can weaken the effect of overexpression RNF122 on promoting GBM progression. CONCLUSION: Our results revealed that RNF122 caused an aggressive phenotype to GBM and was a poor prognosticator; thus, targeting RNF122 may be effectual in GBM treatment.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Kinase Janus-2 , Protéines proto-oncogènes c-myc , Facteur de transcription STAT-3 , Transduction du signal , Ubiquitin-protein ligases , Humains , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Kinase Janus-2/métabolisme , Facteur de transcription STAT-3/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Femelle , Animaux , Lignée cellulaire tumorale , Souris nude , Adulte d'âge moyen , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/physiologie , Souris de lignée BALB C , Protéines et peptides de signalisation intracellulaire
10.
PLoS One ; 19(9): e0310219, 2024.
Article de Anglais | MEDLINE | ID: mdl-39259742

RÉSUMÉ

Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPß. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.


Sujet(s)
Vieillissement , Protéines G , Régénération hépatique , Protéines nucléaires , Régions promotrices (génétique) , Protéines proto-oncogènes c-myc , Animaux , Régénération hépatique/génétique , Régénération hépatique/physiologie , Souris , Vieillissement/métabolisme , Vieillissement/physiologie , Vieillissement/génétique , Humains , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Protéines G/métabolisme , Protéines G/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/métabolisme , Protéine bêta de liaison aux séquences stimulatrices de type CCAAT/génétique , Facteur de transcription E2F1/métabolisme , Facteur de transcription E2F1/génétique , Hépatectomie , Sites de fixation , Foie/métabolisme , Protéine p300-E1A/métabolisme , Régulation de l'expression des gènes , Transcription génétique , Protéine alpha liant les séquences stimulatrices de type CCAAT/métabolisme , Protéine alpha liant les séquences stimulatrices de type CCAAT/génétique , Mâle , Protéines de transport/métabolisme , Protéines de transport/génétique , Souris de lignée C57BL , Lignée cellulaire tumorale , Protéines de liaison à l'ARN
11.
Cancer Res Commun ; 4(9): 2525-2538, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39225558

RÉSUMÉ

Genetically engineered mouse models (GEMM) have fundamentally changed how ovarian cancer etiology, early detection, and treatment are understood. MYC, an oncogene, is amongst the most amplified genes in high-grade serous ovarian cancer (HGSOC), but it has not previously been utilized to drive HGSOC GEMMs. We coupled Myc and dominant-negative mutant p53-R270H with a fallopian tube epithelium (FTE)-specific promoter Ovgp1 to generate a new GEMM of HGSOC. Female mice developed lethal cancer at an average of 14.5 months. Histopathologic examination of mice revealed HGSOC characteristics, including nuclear p53 and nuclear MYC in clusters of cells within the FTE and ovarian surface epithelium. Unexpectedly, nuclear p53 and MYC clustered cell expression was also identified in the uterine luminal epithelium, possibly from intraepithelial metastasis from the FTE. Extracted tumor cells exhibited strong loss of heterozygosity at the p53 locus, leaving the mutant allele. Copy-number alterations in these cancer cells were prevalent, disrupting a large fraction of genes. Transcriptome profiles most closely matched human HGSOC and serous endometrial cancer. Taken together, these results demonstrate that the Myc and Trp53-R270H transgenes were able to recapitulate many phenotypic hallmarks of HGSOC through the utilization of strictly human-mimetic genetic hallmarks of HGSOC. This new mouse model enables further exploration of ovarian cancer pathogenesis, particularly in the 50% of HGSOC which lack homology-directed repair mutations. Histologic and transcriptomic findings are consistent with the hypothesis that uterine serous cancer may originate from the FTE. SIGNIFICANCE: Mouse models using transgenes which generate spontaneous cancers are essential tools to examine the etiology of human diseases. Here, the first Myc-driven spontaneous model is described as a valid HGSOC model. Surprisingly, aspects of uterine serous carcinoma were also observed in this model.


Sujet(s)
Cystadénocarcinome séreux , Modèles animaux de maladie humaine , Tumeurs de l'ovaire , Protéines proto-oncogènes c-myc , Protéine p53 suppresseur de tumeur , Tumeurs de l'utérus , Femelle , Animaux , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Souris , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Cystadénocarcinome séreux/génétique , Cystadénocarcinome séreux/anatomopathologie , Tumeurs de l'utérus/génétique , Tumeurs de l'utérus/anatomopathologie , Souris transgéniques , Humains
12.
Med Sci Monit ; 30: e946584, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39290194

RÉSUMÉ

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Yihua Zhang, Yang Tan, Hao Wang, Minhui Xu, Lunshan Xu. Long Non-Coding RNA Plasmacytoma Variant Translocation 1 (PVT1) Enhances Proliferation, Migration, and Epithelial-Mesenchymal Transition (EMT) of Pituitary Adenoma Cells by Activating ß-Catenin, c-Myc, and Cyclin D1 Expression. Med Sci Monit, 2019; 25: 7652-7659. DOI: 10.12659/MSM.917110.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Cycline D1 , Transition épithélio-mésenchymateuse , Tumeurs de l'hypophyse , Protéines proto-oncogènes c-myc , ARN long non codant , bêta-Caténine , ARN long non codant/génétique , ARN long non codant/métabolisme , Humains , Transition épithélio-mésenchymateuse/génétique , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Tumeurs de l'hypophyse/génétique , Tumeurs de l'hypophyse/métabolisme , Tumeurs de l'hypophyse/anatomopathologie , Cycline D1/métabolisme , Cycline D1/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Lignée cellulaire tumorale , Adénomes/génétique , Adénomes/métabolisme , Adénomes/anatomopathologie , Régulation de l'expression des gènes tumoraux
13.
Nat Commun ; 15(1): 7875, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39285180

RÉSUMÉ

Dysregulation of master regulator c-MYC (MYC) plays a central role in hepatocellular carcinoma (HCC) and other cancers but remains an elusive target for therapeutic intervention. MYC expression is epigenetically modulated within naturally occurring DNA loop structures, Insulated Genomic Domains (IGDs). We present a therapeutic approach using an epigenomic controller (EC), a programmable epigenomic mRNA medicine, to precisely modify MYC IGD sub-elements, leading to methylation of MYC regulatory elements and durable downregulation of MYC mRNA transcription. Significant antitumor activity is observed in preclinical models of HCC treated with the MYC-targeted EC, as monotherapy or in combination with tyrosine kinase or immune checkpoint inhibitors. These findings pave the way for clinical development of MYC-targeting epigenomic controllers in HCC patients and provide a framework for programmable epigenomic mRNA therapeutics for cancer and other diseases.


Sujet(s)
Carcinome hépatocellulaire , Méthylation de l'ADN , Régulation négative , Épigénomique , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines proto-oncogènes c-myc , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Humains , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Régulation négative/génétique , Épigénomique/méthodes , Épigenèse génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Transcription génétique , ARN messager/génétique , ARN messager/métabolisme
15.
Biol Direct ; 19(1): 82, 2024 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-39285446

RÉSUMÉ

BACKGROUND: Armadillo Repeat Containing X-Linked 1 (ARMCX1), a member of the ARM Repeat X-linked protein family, exerts inhibitory function in various tumors. However, its biological role in lung adenocarcinoma (LUAD) and the underlying molecular mechanisms require further exploration. METHODS: LUAD tissue microarrays and bioinformatic databases were used to evaluate the relationship between ARMCX1 and clinicopathological features. The influence of ARMCX1 on LUAD cell proliferation, migration, and invasion in vitro was determined by colony formation, CCK-8, EdU incorporation, cell cycle, wound healing, and Transwell assays. The impact of ARMCX1 on LUAD cell growth and metastasis in vivo was determined by subcutaneously transplanted tumor and pulmonary metastasis assays. Western blot, immunoprecipitation, immunofluorescence, cycloheximide, and proteasome inhibitor assays were finally conducted to explore the potential underlying molecular mechanisms. RESULTS: ARMCX1 expression was downregulated in clinical LUAD samples due to which patient prognoses were poor. Functional experiments indicated that ARMCX1 overexpression inhibited the growth and metastasis of LUAD cells in vitro and in vivo. The molecular mechanism suggested that ARMCX1 recruits the E3 ubiquitin ligase FBXW7 for mediating ubiquitinated degradation of c-Myc, suppressing its nuclear accumulation, and ultimately inactivating cell cycle and epithelial-mesenchymal transition (EMT) signals. CONCLUSION: ARMCX1 inhibits LUAD cell proliferation and metastasis by interacting with c-Myc and enhancing its ubiquitination and degradation. Consequently, it can act as a tumor suppressor in this disease. These results suggest that ARMCX1 is a potential target in the treatment of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Protéine-7 contenant une boite F et des répétitions WD , Tumeurs du poumon , Protéines proto-oncogènes c-myc , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/anatomopathologie , Protéine-7 contenant une boite F et des répétitions WD/génétique , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Animaux , Souris , Lignée cellulaire tumorale , Prolifération cellulaire , Protéolyse , Évolution de la maladie , Mouvement cellulaire , Mâle , Souris nude , Femelle , Régulation de l'expression des gènes tumoraux
16.
Elife ; 132024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39298333

RÉSUMÉ

The Trp53 gene encodes several isoforms of elusive biological significance. Here, we show that mice lacking the Trp53 alternatively spliced (AS) exon, thereby expressing the canonical p53 protein but not isoforms with the AS C-terminus, have unexpectedly lost a male-specific protection against Myc-induced B-cell lymphomas. Lymphomagenesis was delayed in Trp53+/+Eµ-Myc males compared to Trp53ΔAS/ΔAS Eµ-Myc males, but also compared to Trp53+/+Eµ-Myc and Trp53ΔAS/ΔAS Eµ-Myc females. Pre-tumoral splenic cells from Trp53+/+Eµ-Myc males exhibited a higher expression of Ackr4, encoding an atypical chemokine receptor with tumor suppressive effects. We identified Ackr4 as a p53 target gene whose p53-mediated transactivation is inhibited by estrogens, and as a male-specific factor of good prognosis relevant for murine Eµ-Myc-induced and human Burkitt lymphomas. Furthermore, the knockout of ACKR4 increased the chemokine-guided migration of Burkitt lymphoma cells. These data demonstrate the functional relevance of alternatively spliced p53 isoforms and reveal sex disparities in Myc-driven lymphomagenesis.


Human cells divide many times during a lifetime, a process that requires careful regulation to avoid uncontrolled cell division, which can lead to various disorders, including cancer. For example, TP53, which encodes multiple proteins, is the most commonly mutated gene in cancers. TP53 carries the instructions to make a tumor suppressor protein, known as p53, which can stop cancers from forming and spreading. In humans and mice, TP53 (and the mouse analogue Trp53) can also be read by the cell to make several slightly different versions of the p53 protein, known as isoforms. The p53 isoforms are much less studied and their role in an organism is still unclear. To address this, Fajac et al. used genome editing to make mouse strains that were still able to express p53, but were only able to create a specific subset of p53 isoforms. In these mice, part of the Trp53 gene had been mutated to remove the cell's ability to make isoforms with an alternative C-terminal end. Fajac et al. then allowed these mice to breed with mice that were model organisms for a cancer called B-cell lymphoma. This revealed that male offspring that lacked alternative p53 isoforms were more susceptible to B-cell lymphoma and that they had decreased levels of the protein ACKR4, a receptor for signaling proteins that regulate cellular movement. Human datasets showed that having higher levels of ACKR4 could be linked to a better disease prognosis in male patients with Burkitt lymphoma, a rare but aggressive form of B-cell lymphoma. The same effect was not observed in females, suggesting that measuring ACKR4 gene expression in male patients with Burkitt lymphoma might be useful to identify the patients at higher risk. The study from Fajac et al. provides a new perspective on p53 ­ one of the most studied proteins. It highlights specific p53 isoforms and the ACKR4 protein as a potential way to identify male patients at higher risk from a type of B-cell lymphoma.


Sujet(s)
Épissage alternatif , Isoformes de protéines , Protéine p53 suppresseur de tumeur , Animaux , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Souris , Mâle , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Femelle , Lymphome B/génétique , Pronostic , Humains , Facteurs sexuels , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme
17.
J Cancer Res Clin Oncol ; 150(9): 426, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39299959

RÉSUMÉ

BACKGROUND: Double-hit lymphoma (DHL) with c-MYC gene translocation is highly aggressive and has a poor prognosis. In DHL cells, activation-induced cytidine deaminase (AID) promotes antibody class switch recombination (CSR), ultimately leading to c-MYC gene translocation caused by Myc/IgH DNA double-strand breaks. However, currently there is still no method to suppress the expression of AID. METHODS: In this study, we compared the clinical significance of AID expression in DHL, Additionally, two human double-hit lymphoma cell lines were used to analyze the effect of imatinib mesylate on c-MYC in vitro, and the therapeutic effect was also evaluated in xenograft mouse models. RESULTS: Imatinib mesylate downregulated the AID and c-MYC proteins in patients with chronic myelogenous leukemia associated with DHL. In addition, imatinib mesylate reduced AID and c-MYC expression in SU-DHL-4 and OCI-Ly18 DHL cells. Imatinib mesylate exerted significant inhibitory effects on the proliferation and metastasis of SU-DHL-4 and OCI-Ly18 cells. Finally, imatinib mesylate reduced not only tumor burden in DHL mouse models, but also AID and c-MYC expression in vivo. CONCLUSION: These findings reveal that imatinib mesylate effectively reduces the carcinogenic function of c-MYC in DHL, providing novel strategies for developing therapies targeting c-MYC-driven DHL.


Sujet(s)
Cytidine deaminase , Mésilate d'imatinib , Protéines proto-oncogènes c-myc , Tests d'activité antitumorale sur modèle de xénogreffe , Mésilate d'imatinib/pharmacologie , Animaux , Humains , Cytidine deaminase/génétique , Cytidine deaminase/métabolisme , Souris , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Lignée cellulaire tumorale , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Femelle , Antinéoplasiques/pharmacologie , Translocation génétique , Mâle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lymphomes/traitement médicamenteux , Lymphomes/anatomopathologie , Lymphomes/génétique , Lymphomes/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
18.
Discov Med ; 36(188): 1902-1910, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39327253

RÉSUMÉ

BACKGROUND: Cervical cancer (CC) is one of the major types of gynecological cancer, with a high global incidence and mortality rate. Methyltransferase-like 3 (METTL3), a key constituent of methyltransferase, plays a crucial role in various biological processes. Still, only a rare report has been made on its involvement in the progression of CC. Therefore, this study aims to investigate the impact of METTL3 in CC and its molecular mechanisms. METHODS: Gene expression datasets about CC were obtained from the Gene Expression Omnibus (GEO) database, and the expression of METTL3 and Myc was analyzed. Cell viability was detected after METTL3 knockdown in HeLa and SiHa cells, followed by cell counting Kit-8 (CCK-8) assays. The relative expression of METTL3 and Myc was detected via real-time quantitative PCR (qPCR) assays, and the protein expression was determined using Western blot. Meanwhile, cell invasion and migration capabilities were assessed utilizing transwell assays, and cell proliferation was detected using the EdU experiment. Furthermore, RNA methylation immunoprecipitation-qPCR detection was performed to determine the expression of Myc after N6-methyladenosine (m6A) modification. RESULTS: Analysis of the GEO database indicated elevated expression of METTL3 and Myc in CC tissues. Patients with high METTL3 expression had shorter disease-free survival, and patients with high Myc expression had shorter overall survival. Following the knockdown of METTL3, there was a significant reduction in the viability, proliferation, invasion, and migration abilities of HeLa and SiHa cells. Besides, the expression of METTL3 and Myc mRNAs and proteins was greatly reduced. The level of m6A Myc decreased significantly after METTL3 knockdown. CONCLUSIONS: METTL3 plays an important role in regulating cervical cancer cells. METTL3 promotes CC development through m6A modification to regulate the expression of the oncogene Myc.


Sujet(s)
Adénosine , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Methyltransferases , Protéines proto-oncogènes c-myc , Tumeurs du col de l'utérus , Humains , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Femelle , Methyltransferases/métabolisme , Methyltransferases/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Adénosine/analogues et dérivés , Adénosine/métabolisme , Cellules HeLa , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Biosynthèse des protéines
19.
Elife ; 132024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39264149

RÉSUMÉ

Familial exudative vitreoretinopathy (FEVR) is a severe genetic disorder characterized by incomplete vascularization of the peripheral retina and associated symptoms that can lead to vision loss. However, the underlying genetic causes of approximately 50% of FEVR cases remain unknown. Here, we report two heterozygous variants in calcyphosine-like gene (CAPSL) that is associated with FEVR. Both variants exhibited compromised CAPSL protein expression. Vascular endothelial cell (EC)-specific inactivation of Capsl resulted in delayed radial/vertical vascular progression, compromised endothelial proliferation/migration, recapitulating the human FEVR phenotypes. CAPSL-depleted human retinal microvascular endothelial cells (HRECs) exhibited impaired tube formation, decreased cell proliferation, disrupted cell polarity establishment, and filopodia/lamellipodia formation, as well as disrupted collective cell migration. Transcriptomic and proteomic profiling revealed that CAPSL abolition inhibited the MYC signaling axis, in which the expression of core MYC targeted genes were profoundly decreased. Furthermore, a combined analysis of CAPSL-depleted HRECs and c-MYC-depleted human umbilical vein endothelial cells uncovered similar transcription patterns. Collectively, this study reports a novel FEVR-associated candidate gene, CAPSL, which provides valuable information for genetic counseling of FEVR. This study also reveals that compromised CAPSL function may cause FEVR through MYC axis, shedding light on the potential involvement of MYC signaling in the pathogenesis of FEVR.


Sujet(s)
Vitréorétinopathies exsudatives familiales , Humains , Vitréorétinopathies exsudatives familiales/génétique , Cellules endothéliales/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Néovascularisation rétinienne/génétique , Néovascularisation rétinienne/métabolisme , Néovascularisation rétinienne/anatomopathologie , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Souris , Vaisseaux rétiniens/métabolisme , Vaisseaux rétiniens/anatomopathologie , Mâle , Néovascularisation pathologique/génétique , Transduction du signal , Femelle ,
20.
Cell Mol Life Sci ; 81(1): 404, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39277835

RÉSUMÉ

Proliferation of renal tubular epithelial cells (TEC) is essential for restoring tubular integrity and thereby to support renal functional recovery from kidney ischemia/reperfusion (KI/R) injury. Activation of transcriptional factor c-Myc promotes TEC proliferation following KI/R; however, the mechanism regarding c-Myc activation in TEC is incompletely known. Heat shock protein A12A (HSPA12A) is an atypic member of HSP70 family. In this study, we found that KI/R decreased HSPA12A expression in mouse kidneys and TEC, while ablation of HSPA12A in mice impaired TEC proliferation and renal functional recovery following KI/R. Gain-of-functional studies demonstrated that HSPA12A promoted TEC proliferation upon hypoxia/reoxygenation (H/R) through directly interacting with c-Myc and enhancing its nuclear localization to upregulate expression of its target genes related to TEC proliferation. Notably, c-Myc was lactylated in TEC after H/R, and this lactylation was enhanced by HSPA12A overexpression. Importantly, inhibition of c-Myc lactylation attenuated the HSPA12A-induced increases of c-Myc nuclear localization, proliferation-related gene expression, and TEC proliferation. Further experiments revealed that HSPA12A promoted c-Myc lactylation via increasing the glycolysis-derived lactate generation in a Hif1α-dependent manner. The results unraveled a role of HSPA12A in promoting TEC proliferation and facilitating renal recovery following KI/R, and this role of HSPA12A was achieved through increasing lactylation-mediated c-Myc activation. Therefore, targeting HSPA12A in TEC might be a viable strategy to promote renal functional recovery from KI/R injury in patients.


Sujet(s)
Prolifération cellulaire , Cellules épithéliales , Protéines du choc thermique HSP70 , Tubules rénaux , Souris de lignée C57BL , Protéines proto-oncogènes c-myc , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/génétique , Protéines du choc thermique HSP70/métabolisme , Protéines du choc thermique HSP70/génétique , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Souris , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Mâle , Humains , Rein/métabolisme , Rein/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE