Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 63.612
Filtrer
1.
J Biochem Mol Toxicol ; 38(8): e23799, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39132768

RÉSUMÉ

It is well established that pyruvate kinase M2 (PKM2) activity contributes to metabolic reprogramming in various cancers, including colorectal cancer (CRC). Estrogen or 17ß-estradiol (E2) signaling is also known to modulate glycolysis markers in cancer cells. However, whether the inhibition of PKM2 combined with E2 treatment could adversely affect glucose metabolism in CRC cells remains to be investigated. First, we confirmed the metabolic plasticity of CRC cells under varying environmental conditions. Next, we identified glycolysis markers that were upregulated in CRC patients and assessed in vitro mRNA levels following E2 treatment. We found that PKM2 expression, which is highly upregulated in CRC clinical samples, is not altered by E2 treatment in CRC cells. In this study, glucose uptake, generation of reactive oxygen species (ROS), lactate production, cell viability, and apoptosis were evaluated in CRC cells following E2 treatment, PKM2 silencing, or a combination of both. Compared to individual treatments, combination therapy resulted in a significant reduction in cell viability and enhanced apoptosis. Glucose uptake and ROS production were markedly reduced in PKM2-silenced E2-treated cells. The data presented here suggest that E2 signaling combined with PKM2 inhibition cumulatively targets glucose metabolism in a manner that negatively impacts CRC cell growth. These findings hold promise for novel therapeutic strategies targeting altered metabolic pathways in CRC.


Sujet(s)
Tumeurs colorectales , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Hormones thyroïdiennes/métabolisme , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Oestrogènes/pharmacologie , , Oestradiol/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Glucose/métabolisme , Protéines de transport/métabolisme , Pyruvate kinase/métabolisme , Pyruvate kinase/antagonistes et inhibiteurs , Pyruvate kinase/génétique , Glycolyse/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Femelle
2.
Cancer Med ; 13(15): e70105, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39140420

RÉSUMÉ

OBJECTIVE: Kidney renal clear cell carcinoma (KIRC, ccRCC) is the most common type of renal cancer with high recurrence and mortality. It has long been recognized that Antizyme inhibitor 1 (AZIN1) serves as a pro-oncogenic molecule in multiple cancers. However, the clinicopathological features of AZIN1 in KIRC remain unexplored. MATERIALS AND METHODS: The Cancer Genome Atlas (TCGA, TIMER, and GEPIA) were employed for pan-cancer expression and survival analysis of AZIN1, indicating the unique anti-tumor role of AZIN1 in KIRC. The expression and clinical characteristics of AZIN1 in KIRC were further proven via Human Protein Atlas and TCGA. single-sample GSEA was employed to investigate the immune infiltration of AZIN1. Then the downstream pathways were illustrated via the LinkedOmics, Metascape, and Cytoscape databases. The possible upper regulating noncoding RNAs (ncRNAs) were analyzed from five programs-TargetScan, StarBase, miRanda, PITA, and miRmap. RESULTS: AZIN1 is downregulated in KIRC patients. Lower levels of AZIN1 were linked with unfavorable outcomes in KIRC patients. The AZIN1 expression was positively related to immune cell infiltration in KIRC. We also elucidated a possible upstream regulatory ncRNA of AZIN1 in KIRC namely STK4-AS1/AC068338.2-miR-106b-5p-AZIN1 axis as well as the downstream signaling pathways. CONCLUSION: This study illustrated the unique anti-tumor role of AZIN1 in KIRC and provided potential value for guiding immunotherapy and targeted therapy.


Sujet(s)
Néphrocarcinome , Régulation de l'expression des gènes tumoraux , Tumeurs du rein , Humains , Néphrocarcinome/génétique , Néphrocarcinome/mortalité , Néphrocarcinome/anatomopathologie , Néphrocarcinome/métabolisme , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/mortalité , Tumeurs du rein/métabolisme , Pronostic , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Mâle , Protéines de transport/génétique , Protéines de transport/métabolisme , Femelle
3.
BMC Vet Res ; 20(1): 367, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39148099

RÉSUMÉ

BACKGROUND: Recurrent dehydration causes chronic kidney disease in humans and animal models. The dromedary camel kidney has remarkable capacity to preserve water and solute during long-term dehydration. In this study, we investigated the effects of dehydration and subsequent rehydration in the camel's kidney histology/ultrastructure and changes in aquaporin/solute carrier proteins along with gene expression. RESULTS: In light microscopy, dehydration induced few degenerative and necrotic changes in cells of the cortical tubules with unapparent or little effect on medullary cells. The ultrastructural changes encountered in the cortex were infrequent during dehydration and included nuclear chromatin condensation, cytoplasmic vacuolization, mitochondrial swelling, endoplasmic reticulum/ lysosomal degeneration and sometimes cell death. Some mRNA gene expressions involved in cell stability were upregulated by dehydration. Lesions in endothelial capillaries, glomerular membranes and podocyte tertiary processes in dehydrated camels indicated disruption of glomerular filtration barrier which were mostly corrected by rehydration. The changes in proximal tubules brush borders after dehydration, were accompanied by down regulation of ATP1A1 mRNA involved in Na + /K + pump that were corrected by rehydration. The increased serum Na, osmolality and vasopressin were paralleled by modulation in expression level for corresponding SLC genes with net Na retention in cortex which were corrected by rehydration. Medullary collecting ducts and interstitial connective tissue were mostly unaffected during dehydration. CKD, a chronic nephropathy induced by recurrent dehydration in human and animal models and characterized by interstitial fibrosis and glomerular sclerosis, were not observed in the dehydrated/rehydrated camel kidneys. The initiating factors, endogenous fructose, AVP/AVPR2 and uric acid levels were not much affected. TGF-ß1 protein and TGF-ß1gene expression showed no changes by dehydration in cortex/medulla to mediate fibrosis. KCNN4 gene expression level was hardly detected in the dehydrated camel's kidney; to encode for Ca + + -gated KCa3.1 channel for Ca + + influx to instigate TGF-ß1. Modulation of AQP 1, 2, 3, 4, 9 and SLC protein and/or mRNAs expression levels during dehydration/rehydration was reported. CONCLUSIONS: Long-term dehydration induces reversible or irreversible ultrastructural changes in kidney cortex with minor effects in medulla. Modulation of AQP channels, SLC and their mRNAs expression levels during dehydration/rehydration have a role in water conservation. Cortex and medulla respond differently to dehydration/rehydration.


Sujet(s)
Aquaporines , Chameaux , Déshydratation , Rein , Animaux , Déshydratation/médecine vétérinaire , Aquaporines/métabolisme , Aquaporines/génétique , Rein/anatomopathologie , Rein/métabolisme , Mâle , Traitement par apport liquidien/médecine vétérinaire , Régulation de l'expression des gènes , Protéines de transport/métabolisme , Protéines de transport/génétique
4.
Nat Commun ; 15(1): 7076, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152113

RÉSUMÉ

During the repair of interstrand crosslinks (ICLs) a DNA double-strand break (DSB) is generated. The Fanconi anemia (FA) core complex, which is recruited to ICLs, promotes high-fidelity repair of this DSB by homologous recombination (HR). However, whether the FA core complex also promotes HR at ICL-independent DSBs, for example induced by ionizing irradiation or nucleases, remains controversial. Here, we identified the FA core complex members FANCL and Ube2T as HR-promoting factors in a CRISPR/Cas9-based screen. Using isogenic cell line models, we further demonstrated an HR-promoting function of FANCL and Ube2T, and of their ubiquitination substrate FANCD2. We show that FANCL and Ube2T localize at DSBs in a FANCM-dependent manner, and are required for the DSB accumulation of FANCD2. Mechanistically, we demonstrate that FANCL ubiquitin ligase activity is required for the accumulation of CtIP at DSBs, thereby promoting end resection and Rad51 loading. Together, these data demonstrate a dual genome maintenance function of the FA core complex and FANCD2 in promoting repair of both ICLs and DSBs.


Sujet(s)
Cassures double-brin de l'ADN , Protéine du groupe de complémentation D2 de l'anémie de Fanconi , Protéine du groupe de complémentation L de l'anémie de Fanconi , Recombinaison homologue , Ubiquitin-conjugating enzymes , Humains , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation L de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation L de l'anémie de Fanconi/génétique , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Systèmes CRISPR-Cas , Ubiquitination , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Cellules HEK293 , Réparation de l'ADN par recombinaison , Réparation de l'ADN , Réparation de l'ADN par jonction d'extrémités , Helicase
5.
Sci Rep ; 14(1): 17987, 2024 08 03.
Article de Anglais | MEDLINE | ID: mdl-39097652

RÉSUMÉ

The genome is constantly exposed to DNA damage from endogenous and exogenous sources. Fine modulation of DNA repair, chromatin remodeling, and transcription factors is necessary for protecting genome integrity, but the precise mechanisms are still largely unclear. We found that after ionizing radiation (IR), global trimethylation of histone H3 at lysine 4 (H3K4me3) was decreased at an early (5 min) post-IR phase but increased at an intermediate (180 min) post-IR phase in both human and mouse hematopoietic cells. We demonstrated that PTIP, a component of the MLL histone methyltransferase complex, is required for H3K4me3 upregulation in the intermediate post-IR phase and promotes cell cycle arrest by epigenetically inducing a cell cycle inhibitor, PRDM1. In addition, we found that PTIP expression is specifically downregulated in acute myeloid leukemia patients. These findings collectively suggest that the PTIP-PRDM1 axis plays an essential role in proper DNA damage response and its deregulation contributes to leukemogenesis.


Sujet(s)
Points de contrôle du cycle cellulaire , Altération de l'ADN , Protéines de liaison à l'ADN , Facteur-1 liant le domaine de régulation positive I , Animaux , Humains , Souris , Protéines de transport/métabolisme , Protéines de transport/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Épigenèse génétique , Histone/métabolisme , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Facteur-1 liant le domaine de régulation positive I/métabolisme , Facteur-1 liant le domaine de régulation positive I/génétique , Rayonnement ionisant , Régulation positive
6.
J Phys Chem Lett ; 15(32): 8287-8295, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39143857

RÉSUMÉ

The human StAR-related lipid transfer domain protein 2 (STARD2), also known as phosphatidylcholine (PC) transfer protein, is a single-domain lipid transfer protein thought to transfer PC lipids between intracellular membranes. We performed extensive µs-long molecular dynamics simulations of STARD2 of its apo and holo forms in the presence or absence of complex lipid bilayers. The simulations in water reveal ligand-dependent conformational changes. In the 2 µs-long simulations of apo STARD2 in the presence of a lipid bilayer, we observed spontaneous reproducible PC lipid uptake into the protein hydrophobic cavity. We propose that the lipid extraction mechanism involves one to two metastable states stabilized by choline-tyrosine or choline-tryptophane cation-π interactions. Using free energy perturbation, we evaluate that PC-tyrosine cation-π interactions contribute 1.8 and 2.5 kcal/mol to the affinity of a PC-STARD2 metastable state, thus potentially providing a significant decrease of the energy barrier required for lipid desorption.


Sujet(s)
Double couche lipidique , Simulation de dynamique moléculaire , Phosphatidylcholines , Humains , Double couche lipidique/composition chimique , Double couche lipidique/métabolisme , Phosphatidylcholines/composition chimique , Protéines de transport/composition chimique , Protéines de transport/métabolisme , Thermodynamique , Interactions hydrophobes et hydrophiles , Tyrosine/composition chimique
7.
Cell Mol Biol Lett ; 29(1): 110, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39153986

RÉSUMÉ

BACKGROUND: Gastric cancer (GC) is a prevalent malignant tumor, and the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) has been identified as a crucial factor in various tumor types. Moreover, abnormal autophagy levels have been shown to significantly impact tumorigenesis and progression. Despite this, the precise regulatory mechanism of PTBP1 in autophagy regulation in GC remains poorly understood. METHODS: To assess the expression of PTBP1 in GC, we employed a comprehensive approach utilizing western blot, real-time quantitative polymerase chain reaction (RT-qPCR), and bioinformatics analysis. To further identify the downstream target genes that bind to PTBP1 in GC cells, we utilized RNA immunoprecipitation coupled with sequencing (si-PTBP1 RNA-seq). To evaluate the impact of PTBP1 on gastric carcinogenesis, we conducted CCK-8 assays, colony formation assays, and GC xenograft mouse model assays. Additionally, we utilized a transmission electron microscope, immunofluorescence, flow cytometry, western blot, RT-qPCR, and GC xenograft mouse model experiments to elucidate the specific mechanism underlying PTBP1's regulation of autophagy in GC. RESULTS: Our findings indicated that PTBP1 was significantly overexpressed in GC tissues compared with adjacent normal tissues. Silencing PTBP1 resulted in abnormal accumulation of autophagosomes, thereby inhibiting GC cell viability both in vitro and in vivo. Mechanistically, interference with PTBP1 promoted the stability of thioredoxin-interacting protein (TXNIP) mRNA, leading to increased TXNIP-mediated oxidative stress. Consequently, this impaired lysosomal function, ultimately resulting in blockage of autophagic flux. Furthermore, our results suggested that interference with PTBP1 enhanced the antitumor effects of chloroquine, both in vitro and in vivo. CONCLUSION: PTBP1 knockdown impairs GC progression by directly binding to TXNIP mRNA and promoting its expression. Based on these results, PTBP1 emerges as a promising therapeutic target for GC.


Sujet(s)
Autophagie , Protéines de transport , Ribonucléoprotéines nucléaires hétérogènes , Stress oxydatif , Protéine PTB , Tumeurs de l'estomac , Protéine PTB/métabolisme , Protéine PTB/génétique , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Autophagie/génétique , Humains , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Animaux , Protéines de transport/métabolisme , Protéines de transport/génétique , Stress oxydatif/génétique , Lignée cellulaire tumorale , Souris , Évolution de la maladie , Souris nude , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire/génétique , Techniques de knock-down de gènes , Souris de lignée BALB C , Mâle
8.
Nat Commun ; 15(1): 7092, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154037

RÉSUMÉ

Mammalian TIP60 is a multi-functional enzyme with histone acetylation and histone dimer exchange activities. It plays roles in diverse cellular processes including transcription, DNA repair, cell cycle control, and embryonic development. Here we report the cryo-electron microscopy structures of the human TIP60 complex with the core subcomplex and TRRAP module refined to 3.2-Å resolution. The structures show that EP400 acts as a backbone integrating the motor module, the ARP module, and the TRRAP module. The RUVBL1-RUVBL2 hexamer serves as a rigid core for the assembly of EP400 ATPase and YL1 in the motor module. In the ARP module, an ACTL6A-ACTB heterodimer and an extra ACTL6A make hydrophobic contacts with EP400 HSA helix, buttressed by network interactions among DMAP1, EPC1, and EP400. The ARP module stably associates with the motor module but is flexibly tethered to the TRRAP module, exhibiting a unique feature of human TIP60. The architecture of the nucleosome-bound human TIP60 reveals an unengaged nucleosome that is located between the core subcomplex and the TRRAP module. Our work illustrates the molecular architecture of human TIP60 and provides architectural insights into how this complex is bound by the nucleosome.


Sujet(s)
Cryomicroscopie électronique , Lysine acetyltransferase 5 , Humains , Lysine acetyltransferase 5/métabolisme , Lysine acetyltransferase 5/composition chimique , Lysine acetyltransferase 5/génétique , Nucléosomes/métabolisme , Nucléosomes/ultrastructure , Nucléosomes/composition chimique , Helicase/métabolisme , Helicase/composition chimique , Modèles moléculaires , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/composition chimique , ATPases associated with diverse cellular activities/génétique , Protéines de transport/métabolisme , Protéines de transport/composition chimique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/composition chimique , Liaison aux protéines , Multimérisation de protéines , Protéines contenant un bromodomaine , Protéines adaptatrices de la transduction du signal
9.
Mol Med ; 30(1): 124, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138413

RÉSUMÉ

BACKGROUND: Obesity is well-established as a significant contributor to the development of insulin resistance (IR) and diabetes, partially due to elevated plasma saturated free fatty acids like palmitic acid (PA). Grb10-interacting GYF Protein 2 (GIGYF2), an RNA-binding protein, is widely expressed in various tissues including the liver, and has been implicated in diabetes-induced cognitive impairment. Whereas, its role in obesity-related IR remains uninvestigated. METHODS: In this study, we employed palmitic acid (PA) exposure to establish an in vitro IR model in the human liver cancer cell line HepG2 with high-dose chronic PA treatment. The cells were stained with fluorescent dye 2-NBDG to evaluate cell glucose uptake. The mRNA expression levels of genes were determined by real-time qRT-PCR (RT-qPCR). Western blotting was employed to examine the protein expression levels. The RNA immunoprecipitation (RIP) was used to investigate the binding between protein and mRNA. Lentivirus-mediated gene knockdown and overexpression were employed for gene manipulation. In mice, an IR model induced by a high-fat diet (HFD) was established to validate the role and action mechanisms of GIGYF2 in the modulation of HFD-induced IR in vivo. RESULTS: In hepatocytes, high levels of PA exposure strongly trigger the occurrence of hepatic IR evidenced by reduced glucose uptake and elevated extracellular glucose content, which is remarkably accompanied by up-regulation of GIGYF2. Silencing GIGYF2 ameliorated PA-induced IR and enhanced glucose uptake. Conversely, GIGYF2 overexpression promoted IR, PTEN upregulation, and AKT inactivation. Additionally, PA-induced hepatic IR caused a notable increase in STAU1, which was prevented by depleting GIGYF2. Notably, silencing STAU1 prevented GIGYF2-induced PTEN upregulation, PI3K/AKT pathway inactivation, and IR. STAU1 was found to stabilize PTEN mRNA by binding to its 3'UTR. In liver cells, tocopherol treatment inhibits GIGYF2 expression and mitigates PA-induced IR. In the in vivo mice model, GIGYF2 knockdown and tocopherol administration alleviate high-fat diet (HFD)-induced glucose intolerance and IR, along with the suppression of STAU1/PTEN and restoration of PI3K/AKT signaling. CONCLUSIONS: Our study discloses that GIGYF2 mediates obesity-related IR by disrupting the PI3K/AKT signaling axis through the up-regulation of STAU1/PTEN. Targeting GIGYF2 may offer a potential strategy for treating obesity-related metabolic diseases, including type 2 diabetes.


Sujet(s)
Protéines de transport , Insulinorésistance , Foie , Phosphohydrolase PTEN , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Protéines de liaison à l'ARN , Transduction du signal , Humains , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Souris , Foie/métabolisme , Protéines de transport/métabolisme , Protéines de transport/génétique , Cellules HepG2 , Acide palmitique , Mâle , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/génétique , Souris de lignée C57BL , Alimentation riche en graisse/effets indésirables
10.
Cell Metab ; 36(8): 1637-1639, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39111282

RÉSUMÉ

In this issue of Cell Metabolism, Li et al. report that the highly expressed aldehyde dehydrogenase 1 family member A3 interacts with pyruvate kinase M2 (PKM2) in glioblastoma cells. Consequently, PKM2 tetramerization and activation promote lactate production, leading to the lactylation and nuclear translocation of XRCC1 for DNA damage repair and therapeutic resistance.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Humains , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , Effet Warburg en oncologie , Protéines de liaison à l'ADN/métabolisme , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X/métabolisme , , Hormones thyroïdiennes/métabolisme , Protéines membranaires/métabolisme , Protéines de transport/métabolisme , Aldehyde dehydrogenase/métabolisme , Aldehyde dehydrogenase/génétique
11.
Cell Metab ; 36(8): 1696-1710.e10, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39111285

RÉSUMÉ

Patients with high ALDH1A3-expressing glioblastoma (ALDH1A3hi GBM) show limited benefit from postoperative chemoradiotherapy. Understanding the mechanisms underlying such resistance in these patients is crucial for the development of new treatments. Here, we show that the interaction between ALDH1A3 and PKM2 enhances the latter's tetramerization and promotes lactate accumulation in glioblastoma stem cells (GSCs). By scanning the lactylated proteome in lactate-accumulating GSCs, we show that XRCC1 undergoes lactylation at lysine 247 (K247). Lactylated XRCC1 shows a stronger affinity for importin α, allowing for greater nuclear transposition of XRCC1 and enhanced DNA repair. Through high-throughput screening of a small-molecule library, we show that D34-919 potently disrupts the ALDH1A3-PKM2 interaction, preventing the ALDH1A3-mediated enhancement of PKM2 tetramerization. In vitro and in vivo treatment with D34-919 enhanced chemoradiotherapy-induced apoptosis of GBM cells. Together, our findings show that ALDH1A3-mediated PKM2 tetramerization is a potential therapeutic target to improve the response to chemoradiotherapy in ALDH1A3hi GBM.


Sujet(s)
Glioblastome , , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X , Glioblastome/métabolisme , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Humains , Animaux , Lignée cellulaire tumorale , Souris , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X/métabolisme , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Souris nude , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Protéines membranaires/métabolisme , Protéines de transport/métabolisme , Hormones thyroïdiennes/métabolisme , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Aldehyde oxidoreductases , Oxidoreductases acting on CH-NH group donors
12.
Methods Mol Biol ; 2841: 145-155, 2024.
Article de Anglais | MEDLINE | ID: mdl-39115773

RÉSUMÉ

Pollen tubes are typical polarized growth cells whose elongation occurs only in tip regions and is highly dependent on precise and ordered exocytosis/endocytosis in the top regions of the tubes. Although anionic phospholipids have been proven to be involved in regulating vesicle trafficking and the proper localization and functions of proteins in pollen tubes, the underlying cellular and molecular mechanisms remain poorly understood. To further understand how anionic phospholipids are involved in vesicle trafficking and in the control of protein localization and functions, assay methods to analyze the polar localization of anionic phospholipids and their binding proteins, and identifying phospholipid-protein interactions, should be developed. Here, we describe detailed protocols for analyzing anionic phospholipid polar localization and colocalization with their binding proteins in Arabidopsis pollen tubes and examining phospholipid-protein interactions in vitro.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Phospholipides , Tube pollinique , Arabidopsis/métabolisme , Arabidopsis/croissance et développement , Tube pollinique/métabolisme , Tube pollinique/croissance et développement , Phospholipides/métabolisme , Phospholipides/analyse , Protéines d'Arabidopsis/métabolisme , Liaison aux protéines , Protéines de transport/métabolisme , Anions/métabolisme
13.
J Cell Biol ; 223(11)2024 Nov 04.
Article de Anglais | MEDLINE | ID: mdl-39105757

RÉSUMÉ

The characterization of lipid binding to lipid transfer proteins (LTPs) is fundamental to understand their molecular mechanism. However, several structures of LTPs, and notably those proposed to act as bridges between membranes, do not provide the precise location of their endogenous lipid ligands. To address this limitation, computational approaches are a powerful alternative methodology, but they are often limited by the high flexibility of lipid substrates. Here, we develop a protocol based on unbiased coarse-grain molecular dynamics simulations in which lipids placed away from the protein can spontaneously bind to LTPs. This approach accurately determines binding pockets in LTPs and provides a working hypothesis for the lipid entry pathway. We apply this approach to characterize lipid binding to bridge LTPs of the Vps13-Atg2 family, for which the lipid localization inside the protein is currently unknown. Overall, our work paves the way to determine binding pockets and entry pathways for several LTPs in an inexpensive, fast, and accurate manner.


Sujet(s)
Protéines de transport , Simulation de dynamique moléculaire , Liaison aux protéines , Sites de fixation , Protéines de transport/métabolisme , Protéines de transport/composition chimique , Protéines de transport/génétique , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/composition chimique , Lipides/composition chimique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/composition chimique , Protéines du transport vésiculaire/métabolisme , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/composition chimique
14.
Proc Natl Acad Sci U S A ; 121(33): e2318601121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39116123

RÉSUMÉ

Serial capture affinity purification (SCAP) is a powerful method to isolate a specific protein complex. When combined with cross-linking mass spectrometry and computational approaches, one can build an integrated structural model of the isolated complex. Here, we applied SCAP to dissect a subpopulation of WDR76 in complex with SPIN1, a histone reader that recognizes trimethylated histone H3 lysine4 (H3K4me3). In contrast to a previous SCAP analysis of the SPIN1:SPINDOC complex, histones and the H3K4me3 mark were enriched with the WDR76:SPIN1 complex. Next, interaction network analysis of copurifying proteins and microscopy analysis revealed a potential role of the WDR76:SPIN1 complex in the DNA damage response. Since we detected 149 pairs of cross-links between WDR76, SPIN1, and histones, we then built an integrated structural model of the complex where SPIN1 recognized the H3K4me3 epigenetic mark while interacting with WDR76. Finally, we used the powerful Bayesian Integrative Modeling approach as implemented in the Integrative Modeling Platform to build a model of WDR76 and SPIN1 bound to the nucleosome.


Sujet(s)
Altération de l'ADN , Histone , Nucléosomes , Histone/métabolisme , Histone/composition chimique , Nucléosomes/métabolisme , Humains , Liaison aux protéines , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/génétique , Protéines de transport/métabolisme , Protéines de transport/composition chimique , Modèles moléculaires , ATPases associated with diverse cellular activities , Helicase
15.
Mol Cell ; 84(15): 2966-2983.e9, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39089251

RÉSUMÉ

Defects in organellar acidification indicate compromised or infected compartments. Recruitment of the autophagy-related ATG16L1 complex to pathologically neutralized organelles targets ubiquitin-like ATG8 molecules to perturbed membranes. How this process is coupled to proton gradient disruption is unclear. Here, we reveal that the V1H subunit of the vacuolar ATPase (V-ATPase) proton pump binds directly to ATG16L1. The V1H/ATG16L1 interaction only occurs within fully assembled V-ATPases, allowing ATG16L1 recruitment to be coupled to increased V-ATPase assembly following organelle neutralization. Cells lacking V1H fail to target ATG8s during influenza infection or after activation of the immune receptor stimulator of interferon genes (STING). We identify a loop within V1H that mediates ATG16L1 binding. A neuronal V1H isoform lacks this loop and is associated with attenuated ATG8 targeting in response to ionophores in primary murine and human iPSC-derived neurons. Thus, V1H controls ATG16L1 recruitment following proton gradient dissipation, suggesting that the V-ATPase acts as a cell-intrinsic damage sensor.


Sujet(s)
Protéines associées à l'autophagie , Vacuolar Proton-Translocating ATPases , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Humains , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Animaux , Souris , Liaison aux protéines , Neurones/métabolisme , Famille de la protéine-8 associée à l'autophagie/métabolisme , Famille de la protéine-8 associée à l'autophagie/génétique , Autophagie , Cellules HEK293 , Cellules souches pluripotentes induites/métabolisme , Grippe humaine/virologie , Grippe humaine/métabolisme , Grippe humaine/génétique , Souris de lignée C57BL , Transduction du signal , Protéines de transport/métabolisme , Protéines de transport/génétique , Souris knockout
16.
Elife ; 122024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39106189

RÉSUMÉ

Numerous lipids are heterogeneously distributed among organelles. Most lipid trafficking between organelles is achieved by a group of lipid transfer proteins (LTPs) that carry lipids using their hydrophobic cavities. The human genome encodes many intracellular LTPs responsible for lipid trafficking and the function of many LTPs in defining cellular lipid levels and distributions is unclear. Here, we created a gene knockout library targeting 90 intracellular LTPs and performed whole-cell lipidomics analysis. This analysis confirmed known lipid disturbances and identified new ones caused by the loss of LTPs. Among these, we found major sphingolipid imbalances in ORP9 and ORP11 knockout cells, two proteins of previously unknown function in sphingolipid metabolism. ORP9 and ORP11 form a heterodimer to localize at the ER-trans-Golgi membrane contact sites, where the dimer exchanges phosphatidylserine (PS) for phosphatidylinositol-4-phosphate (PI(4)P) between the two organelles. Consequently, loss of either protein causes phospholipid imbalances in the Golgi apparatus that result in lowered sphingomyelin synthesis at this organelle. Overall, our LTP knockout library toolbox identifies various proteins in control of cellular lipid levels, including the ORP9-ORP11 heterodimer, which exchanges PS and PI(4)P at the ER-Golgi membrane contact site as a critical step in sphingomyelin synthesis in the Golgi apparatus.


Sujet(s)
Réticulum endoplasmique , Sphingomyéline , Sphingomyéline/métabolisme , Sphingomyéline/biosynthèse , Humains , Réticulum endoplasmique/métabolisme , Appareil de Golgi/métabolisme , Protéines de transport/métabolisme , Protéines de transport/génétique , Multimérisation de protéines , Récepteurs aux stéroïdes/métabolisme , Récepteurs aux stéroïdes/génétique , Techniques de knock-out de gènes , Phosphates phosphatidylinositol/métabolisme , Phosphates phosphatidylinositol/biosynthèse
17.
J Biochem Mol Toxicol ; 38(8): e23780, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39056188

RÉSUMÉ

Sodium and potassium channels, especially Nav1.5 and Kir2.1, play key roles in the formation of action potentials in cardiomyocytes. These channels interact with, and are regulated by, synapse-associated protein 97 (SAP97). However, the regulatory role of SAP97 in myocyte remains incompletely understood. Here, we investigate the function of SAP97 phosphorylation in the regulation of Nav1.5 and Kir2.1 channel complexes and the upstream regulation of SAP97. We found that SAP97 is phosphorylated by casein kinase II (CK2) in vitro. In addition, transfection of casein kinase 2 interacting protein-1 (CKIP-1) into cardiomyocytes to drive CK2 from the nucleus to the cytoplasm, increased SAP97 phosphorylation and Nav1.5 and Kir2.1 current activity. These findings demonstrated that CKIP-1 modulates the subcellular translocation of CK2, which regulates Nav1.5 and Kir2.1 channel complex formation and activity in cardiomyocytes.


Sujet(s)
Casein Kinase II , Myocytes cardiaques , Canal sodique voltage-dépendant NAV1.5 , Canaux potassiques rectifiants entrants , Myocytes cardiaques/métabolisme , Casein Kinase II/métabolisme , Canaux potassiques rectifiants entrants/métabolisme , Canaux potassiques rectifiants entrants/génétique , Canal sodique voltage-dépendant NAV1.5/métabolisme , Canal sodique voltage-dépendant NAV1.5/génétique , Animaux , Rats , Phosphorylation , Transport des protéines , Humains , Protéines de transport/métabolisme , Rat Sprague-Dawley
18.
Sci Rep ; 14(1): 15422, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965264

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is an inherited disorder characterized by left ventricular hypertrophy and diastolic dysfunction, and increases the risk of arrhythmias and heart failure. Some patients with HCM develop a dilated phase of hypertrophic cardiomyopathy (D-HCM) and have poor prognosis; however, its pathogenesis is unclear and few pathological models exist. This study established disease-specific human induced pluripotent stem cells (iPSCs) from a patient with D-HCM harboring a mutation in MYBPC3 (c.1377delC), a common causative gene of HCM, and investigated the associated pathophysiological mechanisms using disease-specific iPSC-derived cardiomyocytes (iPSC-CMs). We confirmed the expression of pluripotent markers and the ability to differentiate into three germ layers in D-HCM patient-derived iPSCs (D-HCM iPSCs). D-HCM iPSC-CMs exhibited disrupted myocardial sarcomere structures and an increased number of damaged mitochondria. Ca2+ imaging showed increased abnormal Ca2+ signaling and prolonged decay time in D-HCM iPSC-CMs. Cell metabolic analysis revealed increased basal respiration, maximal respiration, and spare-respiratory capacity in D-HCM iPSC-CMs. RNA sequencing also showed an increased expression of mitochondrial electron transport system-related genes. D-HCM iPSC-CMs showed abnormal Ca2+ handling and hypermetabolic state, similar to that previously reported for HCM patient-derived iPSC-CMs. Although further studies are required, this is expected to be a useful pathological model for D-HCM.


Sujet(s)
Calcium , Cardiomyopathie hypertrophique , Protéines de transport , Mutation avec décalage du cadre de lecture , Cellules souches pluripotentes induites , Myocytes cardiaques , Cellules souches pluripotentes induites/métabolisme , Humains , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Cardiomyopathie hypertrophique/génétique , Cardiomyopathie hypertrophique/métabolisme , Cardiomyopathie hypertrophique/anatomopathologie , Calcium/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Signalisation calcique , Différenciation cellulaire , Mâle
19.
Gene ; 927: 148742, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38969244

RÉSUMÉ

Preeclampsia (PE) is clinically defined as a part of pregnancy characterized by hypertension and multiple organ failure. PE is broadly categorized into two types: "placental" and "maternal". Placental PE is associated with fetal growth restriction and adverse maternal and neonatal outcomes. STOX1 (Storkhead box 1), a transcription factor, discovered through a complete transcript analysis of the PE susceptibility locus of 70,000 bp on chromosome 10q22.1. So far, studies investigating the relationship between STOX1 and PE have focused on STOX1 overexpression, STOX1 isoform imbalance, and STOX1 variations that could have clinical consequence. Initially, the Y153H variation of STOX was associated with the placental form of PE. Additionally, studies focusing on the maternal and fetal interface have shown that NODAL and STOX1 variations play a role together in the unsuccessful remodeling of the spiral arteries. Research specifically addressing the overexpression of STOX1 has shown that its disruption of cellular hemoastasis, leading to impaired hypoxia response, disruption of the cellular antioxidant system, and nitroso/redox imbalance. Furthermore, functional studies have been conducted showing that the imbalance between STOX1 isoforms contributes to the pathogenesis of placental PE. Research indicates that STOX1B competes with STOX1A and that the overexpression of STOX1B reverses cellular changes that STOX1A induces to the pathogenesis of PE. In this review, we aimed at elucidating the relationship between STOX1 and PE as well as function of STOX1. In conclusion, based on a comprehensive literature review, numerous studies support the role of STOX1 in the pathogenesis of PE.


Sujet(s)
Pré-éclampsie , Humains , Pré-éclampsie/génétique , Pré-éclampsie/métabolisme , Femelle , Grossesse , Placenta/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme
20.
J Coll Physicians Surg Pak ; 34(7): 811-816, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38978246

RÉSUMÉ

OBJECTIVE: To investigate the expression level of pyruvate kinase M1 (PKM1) in patients with acute myeloid leukaemia (AML) as well as its clinical significance. STUDY DESIGN: A case-control study. Place and Duration of the Study: Department of Haematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China, from January 2013 to 2023. METHODOLOGY: The expression levels of PKM1 and pyruvate kinase m2 (PKM2) in the bone marrow of 65 AML patients (excluding M3) and 31 healthy volunteers were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), a method that measures fluorescence in real-time. The associations between PKM1, PKM2 expressions, clinical parameters, and the survival and prognosis of AML patients were analysed. RESULTS: AML patients showed higher PKM1 expression compared to controls. The area under the curve (AUC) of the receiver operating characteristics (ROC) was 0.65 (p = 0.017). PKM1 expression was correlated with peripheral blood leukocyte count (r = -0.276, p = 0.026), CCAAT enhancer-binding protein alpha CEBPA mutation (r = -0.306, p = 0.014), and chemotherapy-induced response (r = -0.292, p = 0.018). Patients with high PKM1 expression had a lower remission rate (p = 0.019) and long-term survival rate (p = 0.034) than those with low PKM1 expression. Patients with AML showed a rise in PKM2 levels; however, the variation was not statistically significant (p >0.05). CONCLUSION: PKM1 expression is upregulated in AML and patients with high PKM1 expression have a lower survival rate. KEY WORDS: PKM1, Acute myeloid leukaemia, Clinical prognosis.


Sujet(s)
Protéines de transport , Leucémie aigüe myéloïde , Protéines membranaires , , Hormones thyroïdiennes , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Études cas-témoins , Chine/épidémiologie , Leucémie aigüe myéloïde/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Pronostic , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme , Hormones thyroïdiennes/sang , Hormones thyroïdiennes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE