Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.535
Filtrer
1.
J Cell Biol ; 223(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-38980288

RÉSUMÉ

Autophagy is essential for maintaining glucose homeostasis. However, the mechanism by which cells sense and respond to glucose starvation to induce autophagy remains incomplete. Here, we show that calcium serves as a fundamental triggering signal that connects environmental sensing to the formation of the autophagy initiation complex during glucose starvation. Mechanistically, glucose starvation instigates the release of vacuolar calcium into the cytoplasm, thus triggering the activation of Rck2 kinase. In turn, Rck2-mediated Atg11 phosphorylation enhances Atg11 interactions with Bmh1/2 bound to the Snf1-Sip1-Snf4 complex, leading to recruitment of vacuolar membrane-localized Snf1 to the PAS and subsequent Atg1 activation, thereby initiating autophagy. We also identified Glc7, a protein phosphatase-1, as a critical regulator of the association between Bmh1/2 and the Snf1 complex. We thus propose that calcium-triggered Atg11-Bmh1/2-Snf1 complex assembly initiates autophagy by controlling Snf1-mediated Atg1 activation in response to glucose starvation.


Sujet(s)
Autophagie , Calcium , Glucose , Protein-Serine-Threonine Kinases , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Glucose/métabolisme , Calcium/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Phosphorylation , Vacuoles/métabolisme , Vacuoles/génétique
2.
Theranostics ; 14(10): 3984-3996, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994020

RÉSUMÉ

Rationale: Cataract is the leading cause of blindness and low vision worldwide, yet its pathological mechanism is not fully understood. Although macroautophagy/autophagy is recognized as essential for lens homeostasis and has shown potential in alleviating cataracts, its precise mechanism remains unclear. Uncovering the molecular details of autophagy in the lens could provide targeted therapeutic interventions alongside surgery. Methods: We monitored autophagic activities in the lens and identified the key autophagy protein ATG16L1 by immunofluorescence staining, Western blotting, and transmission electron microscopy. The regulatory mechanism of ATG16L1 ubiquitination was analyzed by co-immunoprecipitation and Western blotting. We used the crystal structure of E3 ligase gigaxonin and conducted the docking screening of a chemical library. The effect of the identified compound riboflavin was tested in vitro in cells and in vivo animal models. Results: We used HLE cells and connexin 50 (cx50)-deficient cataract zebrafish model and confirmed that ATG16L1 was crucial for lens autophagy. Stabilizing ATG16L1 by attenuating its ubiquitination-dependent degradation could promote autophagy activity and relieve cataract phenotype in cx50-deficient zebrafish. Mechanistically, the interaction between E3 ligase gigaxonin and ATG16L1 was weakened during this process. Leveraging these mechanisms, we identified riboflavin, an E3 ubiquitin ligase-targeting drug, which suppressed ATG16L1 ubiquitination, promoted autophagy, and ultimately alleviated the cataract phenotype in autophagy-related models. Conclusions: Our study identified an unrecognized mechanism of cataractogenesis involving ATG16L1 ubiquitination in autophagy regulation, offering new insights for treating cataracts.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Cataracte , Cristallin , Danio zébré , Animaux , Cataracte/métabolisme , Cataracte/traitement médicamenteux , Autophagie/effets des médicaments et des substances chimiques , Protéines associées à l'autophagie/métabolisme , Cristallin/métabolisme , Cristallin/effets des médicaments et des substances chimiques , Humains , Ubiquitination/effets des médicaments et des substances chimiques , Riboflavine/pharmacologie , Modèles animaux de maladie humaine , Lignée cellulaire
3.
J Cancer Res Clin Oncol ; 150(7): 335, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38969831

RÉSUMÉ

BACKGROUND: Ubiquilin-4 (UBQLN4), a member of the ubiquilin family, has received limited attention in cancer research to date. Here, we investigated for the first time the functional role and mechanism of UBQLN4 in non-small cell lung cancer (NSCLC). METHODS: The Cancer Genome Atlas (TCGA) database was employed to validate UBQLN4 as a differentially expressed gene. Expression differences of UBQLN4 in NSCLC cells and tissues were assessed using immunohistochemistry (IHC) experiment and western blotting (WB) experiment. Kaplan-Meier analysis was conducted to examine the association between UBQLN4 expression and NSCLC prognosis. Functional analyses of UBQLN4 were performed through cell counting kit-8 (CCK-8), colony formation, and transwell invasion assays. The impact of UBQLN4 on tumor-associated signaling pathways was assessed using the path scan intracellular signaling array. In vivo tumorigenesis experiments were conducted to further investigate the influence of UBQLN4 on tumor formation. RESULTS: UBQLN4 exhibited up-regulation in both NSCLC tissues and cells. Additionally, over-expression of UBQLN4 was associated with an unfavorable prognosis in NSCLC patients. Functional loss analyses demonstrated that inhibiting UBQLN4 could suppress the proliferation and invasion of NSCLC cells in both in vitro and in vivo settings. Conversely, functional gain experiments yielded opposite results. Path scan intracellular signaling array results suggested that the role of UBQLN4 is associated with the PI3K/AKT pathway, a correlation substantiated by in vitro and in vivo tumorigenesis experiments. CONCLUSION: We validated that UBQLN4 promotes proliferation and invasion of NSCLC cells by activating the PI3K/AKT pathway, thereby facilitating the progression of NSCLC. These findings underscore the potential of targeting UBQLN4 as a therapeutic strategy for NSCLC.


Sujet(s)
Protéines associées à l'autophagie , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Animaux , Souris , Femelle , Mâle , Pronostic , Lignée cellulaire tumorale , Souris nude , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Adulte d'âge moyen , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de transport , Protéines nucléaires
4.
Cell Death Dis ; 15(6): 410, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38866787

RÉSUMÉ

The role of circDHX8 in the interplay between autophagy and gastric cancer (GC) progression remains unclear. In this study, we investigated the mechanism underlying the role of hsa_circ_003899 (circDHX8) in the malignancy of GC. Differential expression of circRNAs between GC and normal tissues was determined using circle-seq and microarray datasets (GSE83521). These circRNAs were validated using qPCR and Sanger sequencing. The function of circDHX8 was investigated through interference with circDHX8 expression experiments using in vitro and in vivo functional assays. Western blotting, immunofluorescence, and transmission electron microscopy were used to establish whether circDHX8 promoted autophagy in GC cells. To elucidate the mechanism underlying the circDHX8-mediated regulation of autophagy, we performed bioinformatics analysis, RNA pull-down, mass spectrometry (MS), RNA immunoprecipitation (RIP), and other western Blot related experiments. Hsa_circ_0003899 (circDHX8) was identified as upregulated and shown to enhance the malignant progression in GC cells by promoting cellular autophagy. Mechanistically, circDHX8 increased ATG2B protein levels by preventing ubiquitin-mediated degradation, thereby facilitating cell proliferation and invasion in GC. Additionally, circDHX8 directly interacts with the E3 ubiquitin-protein ligase RNF5, inhibiting the RNF5-mediated degradation of ATG2B. Concurrently, ATG2B, an acetylated protein, is subjected to SIRT1-mediated deacetylation, enhancing its binding to RNF5. Consequently, we established a novel mechanism for the role of circDHX8 in the malignant progression of GC.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Évolution de la maladie , ARN circulaire , Tumeurs de l'estomac , Animaux , Femelle , Humains , Mâle , Souris , Autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Souris nude , Liaison aux protéines , ARN circulaire/génétique , ARN circulaire/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/anatomopathologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
5.
Stem Cell Res Ther ; 15(1): 180, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38902824

RÉSUMÉ

BACKGROUND: Telomeres consist of repetitive DNA sequences at the chromosome ends to protect chromosomal stability, and primarily maintained by telomerase or occasionally by alternative telomere lengthening of telomeres (ALT) through recombination-based mechanisms. Additional mechanisms that may regulate telomere maintenance remain to be explored. Simultaneous measurement of telomere length and transcriptome in the same human embryonic stem cell (hESC) revealed that mRNA expression levels of UBQLN1 exhibit linear relationship with telomere length. METHODS: In this study, we first generated UBQLN1-deficient hESCs and compared with the wild-type (WT) hESCs the telomere length and molecular change at RNA and protein level by RNA-seq and proteomics. Then we identified the potential interacting proteins with UBQLN1 using immunoprecipitation-mass spectrometry (IP-MS). Furthermore, the potential mechanisms underlying the shortened telomeres in UBQLN1-deficient hESCs were analyzed. RESULTS: We show that Ubiquilin1 (UBQLN1) is critical for telomere maintenance in human embryonic stem cells (hESCs) via promoting mitochondrial function. UBQLN1 deficiency leads to oxidative stress, loss of proteostasis, mitochondria dysfunction, DNA damage, and telomere attrition. Reducing oxidative damage and promoting mitochondria function by culture under hypoxia condition or supplementation with N-acetylcysteine partly attenuate the telomere attrition induced by UBQLN1 deficiency. Moreover, UBQLN1 deficiency/telomere shortening downregulates genes for neuro-ectoderm lineage differentiation. CONCLUSIONS: Altogether, UBQLN1 functions to scavenge ubiquitinated proteins, preventing their overloading mitochondria and elevated mitophagy. UBQLN1 maintains mitochondria and telomeres by regulating proteostasis and plays critical role in neuro-ectoderm differentiation.


Sujet(s)
Protéines associées à l'autophagie , Cellules souches embryonnaires humaines , Mitochondries , Homéostasie protéique , Homéostasie des télomères , Télomère , Humains , Cellules souches embryonnaires humaines/métabolisme , Cellules souches embryonnaires humaines/cytologie , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Mitochondries/métabolisme , Télomère/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Stress oxydatif , Altération de l'ADN
6.
BMC Cardiovasc Disord ; 24(1): 323, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38918713

RÉSUMÉ

BACKGROUND: Radiotherapy is a primary local treatment for tumors, yet it may lead to complications such as radiation-induced heart disease (RIHD). Currently, there is no standardized approach for preventing RIHD. Dexmedetomidine (Dex) is reported to have cardio-protection effects, while its role in radiation-induced myocardial injury is unknown. In the current study, we aimed to evaluate the radioprotective effect of dexmedetomidine in X-ray radiation-treated mice. METHODS: 18 male mice were randomized into 3 groups: control, 16 Gy, and 16 Gy + Dex. The 16 Gy group received a single dose of 16 Gy X-ray radiation. The 16 Gy + Dex group was pretreated with dexmedetomidine (30 µg/kg, intraperitoneal injection) 30 min before X-ray radiation. The control group was treated with saline and did not receive X-ray radiation. Myocardial tissues were collected 16 weeks after X-ray radiation. Hematoxylin-eosin staining was performed for histopathological examination. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was performed to assess the state of apoptotic cells. Immunohistochemistry staining was performed to examine the expression of CD34 molecule and von Willebrand factor. Besides, western blot assay was employed for the detection of apoptosis-related proteins (BCL2 apoptosis regulator and BCL2-associated X) as well as autophagy-related proteins (microtubule-associated protein 1 light chain 3, beclin 1, and sequestosome 1). RESULTS: The findings demonstrated that 16 Gy X-ray radiation resulted in significant changes in myocardial tissues, increased myocardial apoptosis, and activated autophagy. Pretreatment with dexmedetomidine significantly protects mice against 16 Gy X-ray radiation-induced myocardial injury by inhibiting apoptosis and autophagy. CONCLUSION: In summary, our study confirmed the radioprotective effect of dexmedetomidine in mitigating cardiomyocyte apoptosis and autophagy induced by 16 Gy X-ray radiation.


Sujet(s)
Apoptose , Autophagie , Dexmédétomidine , Myocytes cardiaques , Lésions radiques expérimentales , Animaux , Autophagie/effets des médicaments et des substances chimiques , Autophagie/effets des radiations , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/effets des radiations , Myocytes cardiaques/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Mâle , Dexmédétomidine/pharmacologie , Lésions radiques expérimentales/prévention et contrôle , Lésions radiques expérimentales/anatomopathologie , Lésions radiques expérimentales/métabolisme , Lésions radiques expérimentales/traitement médicamenteux , Radioprotecteurs/pharmacologie , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Protéines associées à l'autophagie/métabolisme , Souris de lignée C57BL , Protéines régulatrices de l'apoptose/métabolisme
7.
Cell Mol Biol Lett ; 29(1): 85, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38834954

RÉSUMÉ

The molecular basis for bulk autophagy activation due to a deficiency in essential nutrients such as carbohydrates, amino acids, and nitrogen is well understood. Given autophagy functions to reduce surplus to compensate for scarcity, it theoretically possesses the capability to selectively degrade specific substrates to meet distinct metabolic demands. However, direct evidence is still lacking that substantiates the idea that autophagy selectively targets specific substrates (known as selective autophagy) to address particular nutritional needs. Recently, Gross et al. found that during phosphate starvation (P-S), rather than nitrogen starvation (N-S), yeasts selectively eliminate peroxisomes by dynamically altering the composition of the Atg1/ULK kinase complex (AKC) to adapt to P-S. This study elucidates how the metabolite sensor Pho81 flexibly interacts with AKC and guides selective autophagic clearance of peroxisomes during P-S, providing novel insights into the metabolic contribution of autophagy to special nutritional needs.


Sujet(s)
Autophagie , Phosphates , Protéines de Saccharomyces cerevisiae , Phosphates/métabolisme , Phosphates/déficit , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Péroxysomes/métabolisme , Saccharomyces cerevisiae/métabolisme , Homologue de la protéine-1 associée à l'autophagie/métabolisme , Homologue de la protéine-1 associée à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein kinases
8.
Theriogenology ; 225: 9-15, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38781849

RÉSUMÉ

Autophagy is essential for oocyte maturation and preimplantation embryo development. ATG4C, a member of the ATG4 family, plays a crucial role in the autophagy process. The effect of ATG4C on the early embryonic development in pig has not been studied. In this study, the expression patterns of ATG4C were explored using qRT-PCR and immunofluorescence staining. Different concentrations of serum were added to in vitro maturation (IVM) medium to investigate its effects on oocyte maturation and embryonic development. Finally, the developmental potential of parthenogenetic embryos was detected by downregulating ATG4C in MII stage oocytes under 0 % serum condition. The results revealed that ATG4C was highly expressed in porcine oocytes matured in vitro and in parthenogenetic embryos. Compared with the 10 % serum group, the cumulus cell expansion, first polar body (PB1) extrusion rate, and subsequent developmental competence of embryos were reduced in the 0 % and 5 % serum groups. The mRNA levels of LC3, ATG5, BECLIN1, TFAM, PGC1α, and PINK1 were significantly increased (P < 0.05) in the 0 % serum group. ATG4C was significantly upregulated in the embryos at the 1-cell, 2-cell, 8-cell, and 16-cell stages in the 0 % serum group (P < 0.05). Compared with the negative control group, downregulation of ATG4C significantly decreased the 4-cell, 8-cell, and blastocyst rates (P < 0.05), and the expression of genes related to autophagy, mitochondria, and zygotic genome activation (ZGA) was significantly decreased (P < 0.05). The relative fluorescence intensity of LC3 and mitochondrial content in the ATG4C siRNA group was significantly reduced (P < 0.05). Collectively, the results indicate that ATG4C is highly expressed in porcine oocytes matured in vitro and in early embryos, and inhibition of ATG4C effects embryonic developmental competence by decreasing autophagy, mitochondrial content, and ZGA under serum-free condition.


Sujet(s)
Développement embryonnaire , Régulation de l'expression des gènes au cours du développement , Techniques de maturation in vitro des ovocytes , Ovocytes , Animaux , Suidae/embryologie , Ovocytes/métabolisme , Développement embryonnaire/physiologie , Techniques de maturation in vitro des ovocytes/médecine vétérinaire , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Techniques de culture d'embryons/médecine vétérinaire , Femelle , Autophagie , Parthénogenèse
9.
Int J Oncol ; 65(1)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38818827

RÉSUMÉ

Clear cell renal cell carcinoma (ccRCC), the most common type of renal cell carcinoma (RCC), is not sensitive to traditional radiotherapy and chemotherapy. The polyphenolic compound Gallic acid (GA) can be naturally found in a variety of fruits, vegetables and plants. Autophagy, an intracellular catabolic process, regulates the lysosomal degradation of organelles and portions in cytoplasm. It was reported that autophagy and GA could affect the development of several cancers. Therefore, the aim of the present study was to evaluate the effects of GA on ccRCC development and clarify the role of autophagy in this process. In the present study, the effects of GA on the proliferation, migration and invasion of ccRCC cells were investigated in vitro by Cell Counting Kit­8, colony formation, flow cytometry, wound healing and Transwell migration assays, respectively. Additionally, the effects of GA on ccRCC growth and metastasis were evaluated using hematoxylin­eosin and immunohistochemical staining in vivo. Moreover, it was sought to explore the underlying molecular mechanisms using transmission electron microscopy, western blotting and reverse transcription­quantitative PCR analyses. In the present study, it was revealed that GA had a more potent viability inhibitory effect on ccRCC cells (786­O and ACHN) than the effect on normal renal tubular epithelial cell (HK­2), which demonstrated that GA selectively inhibits the viability of cancer cells. Furthermore, it was identified that GA dose­dependently inhibited the proliferation, migration and invasion of ccRCC cells in vitro and in vivo. It was demonstrated that GA promoted the release of autophagy markers, which played a role in regulating the PI3K/Akt/Atg16L1 signaling pathway. All the aforementioned data provided evidence for the great potential of GA in the treatment of ccRCC.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Néphrocarcinome , Mouvement cellulaire , Prolifération cellulaire , Acide gallique , Tumeurs du rein , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/anatomopathologie , Néphrocarcinome/métabolisme , Humains , Acide gallique/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs du rein/anatomopathologie , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Phosphatidylinositol 3-kinases/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Animaux , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Évolution de la maladie , Mâle , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme
10.
Clin Exp Pharmacol Physiol ; 51(6): e13861, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38724488

RÉSUMÉ

Relevant studies have indicated the association of HCG18 with tumour occurrence and progression. In this study, we observed that PM2.5 can enhance the growth of lung adenocarcinoma cells by modulating the expression of HCG18. Further investigations, including overexpression and knockout experiments, elucidated that HCG18 suppresses miR-195, which in turn upregulates the expression of ATG14, resulting in the upregulation of autophagy. Consequently, exposure to PM2.5 leads to elevated HCG18 expression in lung tissues, which in turn increases Atg14 expression and activates autophagy pathways through inhibition of miR-195, thereby contributing to oncogenesis.


Sujet(s)
Adénocarcinome pulmonaire , Protéines associées à l'autophagie , Autophagie , Évolution de la maladie , Tumeurs du poumon , microARN , Matière particulaire , Humains , Cellules A549 , Protéines adaptatrices du transport vésiculaire/effets des médicaments et des substances chimiques , Protéines adaptatrices du transport vésiculaire/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Autophagie/génétique , Protéines associées à l'autophagie/effets des médicaments et des substances chimiques , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , microARN/génétique , microARN/métabolisme , Matière particulaire/effets indésirables , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme , Antigènes HLA/effets des médicaments et des substances chimiques , Antigènes HLA/métabolisme
11.
J Cell Biol ; 223(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38728007

RÉSUMÉ

Activation of PINK1 and Parkin in response to mitochondrial damage initiates a response that includes phosphorylation of RAB7A at Ser72. Rubicon is a RAB7A binding negative regulator of autophagy. The structure of the Rubicon:RAB7A complex suggests that phosphorylation of RAB7A at Ser72 would block Rubicon binding. Indeed, in vitro phosphorylation of RAB7A by TBK1 abrogates Rubicon:RAB7A binding. Pacer, a positive regulator of autophagy, has an RH domain with a basic triad predicted to bind an introduced phosphate. Consistent with this, Pacer-RH binds to phosho-RAB7A but not to unphosphorylated RAB7A. In cells, mitochondrial depolarization reduces Rubicon:RAB7A colocalization whilst recruiting Pacer to phospho-RAB7A-positive puncta. Pacer knockout reduces Parkin mitophagy with little effect on bulk autophagy or Parkin-independent mitophagy. Rescue of Parkin-dependent mitophagy requires the intact pRAB7A phosphate-binding basic triad of Pacer. Together these structural and functional data support a model in which the TBK1-dependent phosphorylation of RAB7A serves as a switch, promoting mitophagy by relieving Rubicon inhibition and favoring Pacer activation.


Sujet(s)
Protéines associées à l'autophagie , Mitophagie , Protein-Serine-Threonine Kinases , Ubiquitin-protein ligases , Protéines Rab7 liant le GTP , Humains , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Cellules HEK293 , Cellules HeLa , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Mitochondries/métabolisme , Mitochondries/génétique , Phosphorylation , Liaison aux protéines , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique
12.
BMC Cardiovasc Disord ; 24(1): 275, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38807081

RÉSUMÉ

BACKGROUND: Autophagy, as a regulator of cell survival, plays an important role in atherosclerosis (AS). Sperm associated antigen 5 (SPAG5) is closely associated with the classical autophagy pathway, PI3K/Akt/mTOR signaling pathway. This work attempted to investigate whether SPAG5 can affect AS development by regulating autophagy. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with oxidized-low density lipoprotein (ox-LDL) to induce cell damage. ApoE-/- mice were fed a Western diet to establish an AS mouse model. Haematoxylin and eosin (H&E) staining and Oil Red O staining evaluated the pathological changes and in lipid deposition in aortic tissues. CCK-8 and flow cytometry detected cell proliferation and apoptosis. Immunohistochemistry, Enzyme linked immunosorbent assay, qRT-PCR and western blotting assessed the levels of mRNA and proteins. RESULTS: Ox-LDL treatment elevated SPAG5 expression and the expression of autophagy-related proteins, LC3-I, LC3-II, Beclin-1, and p62, in HUVECs. GFP-LC3 dots were increased in ox-LDL-treated HUVECs and LPS-treated HUVECs. SPAG5 knockdown reversed both ox-LDL and LPS treatment-mediated inhibition of cell proliferation and promotion of apoptosis in HUVECs. SPAG5 silencing further elevated autophagy and repressed the expression of PI3K, p-Akt/Akt, and p-mTOR/mTOR in ox-LDL-treated HUVECs. 3-MA (autophagy inhibitor) treatment reversed SPAG5 silencing-mediated increase of cell proliferation and decrease of apoptosis in ox-LDL-treated HUVECs. In vivo, SPAG5 knockdown reduced atherosclerotic plaques in AS mice through activating autophagy and inhibiting PI3K/Akt/mTOR signaling pathway. CONCLUSION: This work demonstrated that SPAG5 knockdown alleviated AS development through activating autophagy. Thus, SPAG5 may be a potential target for AS therapy.


Sujet(s)
Apoptose , Athérosclérose , Autophagie , Prolifération cellulaire , Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Autophagie/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Humains , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/prévention et contrôle , Sérine-thréonine kinases TOR/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Souris de lignée C57BL , Lipoprotéines LDL/métabolisme , Mâle , Cellules cultivées , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Aorte/anatomopathologie , Aorte/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Souris , Apolipoprotéines E
13.
Exp Cell Res ; 439(1): 114094, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38750718

RÉSUMÉ

Pirarubicin (THP) is a new generation of cell cycle non-specific anthracycline-based anticancer drug. In the clinic, THP and THP combination therapies have been shown to be effective in hepatocellular carcinoma (HCC) patients with transcatheter arterial chemoembolization (TACE) without serious side effects. However, drug resistance limits its therapeutic efficacy. Berberine (BBR), an isoquinoline alkaloid, has been shown to possess antitumour properties against various malignancies. However, the synergistic effect of BBR and THP in the treatment of HCC is unknown. In the present study, we demonstrated for the first time that BBR sensitized HCC cells to THP, including enhancing THP-induced growth inhibition and apoptosis of HCC cells. Moreover, we found that BBR sensitized THP by reducing the expression of autophagy-related 4B (ATG4B). Mechanistically, the inhibition of HIF1α-mediated ATG4B transcription by BBR ultimately led to attenuation of THP-induced cytoprotective autophagy, accompanied by enhanced growth inhibition and apoptosis in THP-treated HCC cells. Tumor-bearing experiments in nude mice showed that the combination treatment with BBR and THP significantly suppressed the growth of HCC xenografts. These results reveal that BBR is able to strengthen the killing effect of THP on HCC cells by repressing the ATG4B-autophagy pathway, which may provide novel insights into the improvement of chemotherapeutic efficacy of THP, and may be conducive to the further clinical application of THP in HCC treatment.


Sujet(s)
Apoptose , Protéines associées à l'autophagie , Autophagie , Berbérine , Carcinome hépatocellulaire , Doxorubicine , Tumeurs du foie , Souris nude , Berbérine/pharmacologie , Berbérine/analogues et dérivés , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Animaux , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Souris , Apoptose/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Doxorubicine/analogues et dérivés , Tests d'activité antitumorale sur modèle de xénogreffe , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Souris de lignée BALB C , Antinéoplasiques/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Cysteine endopeptidases
14.
BMB Rep ; 57(5): 256-261, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38627949

RÉSUMÉ

In the context of aging, the susceptibility to infectious diseases increases, leading to heightened morbidity and mortality. This phenomenon, termed immunosenescence, is characterized by dysregulation in the aging immune system, including abnormal alterations in lymphocyte composition, elevated basal inflammation, and the accumulation of senescent T cells. Such changes contribute to increased autoimmune diseases, enhanced infection severity, and reduced responsiveness to vaccines. Utilizing aging animal models becomes imperative for a comprehensive understanding of immunosenescence, given the complexity of aging as a physiological process in living organisms. Our investigation focuses on Cisd2, a causative gene for Wolfram syndrome, to elucidate on immunosenescence. Cisd2 knockout (KO) mice, serving as a model for premature aging, exhibit a shortened lifespan with early onset of aging-related features, such as decreased bone density, hair loss, depigmentation, and optic nerve degeneration. Intriguingly, we found that the Cisd2 KO mice present a higher number of neutrophils in the blood; however, isolated neutrophils from these mice display functional defects. Through mass spectrometry analysis, we identified an interaction between Cisd2 and Calnexin, a protein known for its role in protein quality control. Beyond this function, Calnexin also regulates calcium homeostasis through interaction with sarcoendoplasmic reticulum calcium transport ATPase (SERCA). Our study proposes that Cisd2 modulates calcium homeostasis via its interaction with Calnexin and SERCA, consequently influencing neutrophil functions. [BMB Reports 2024; 57(5): 256-261].


Sujet(s)
Protéines associées à l'autophagie , Calcium , Homéostasie , Protéines de tissu nerveux , Granulocytes neutrophiles , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animaux , Souris , Calcium/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/déficit , Protéines membranaires/génétique , Souris knockout , Granulocytes neutrophiles/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme
15.
Cell Rep ; 43(5): 114131, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38656870

RÉSUMÉ

Atg8 paralogs, consisting of LC3A/B/C and GBRP/GBRPL1/GATE16, function in canonical autophagy; however, their function is controversial because of functional redundancy. In innate immunity, xenophagy and non-canonical single membranous autophagy called "conjugation of Atg8s to single membranes" (CASM) eliminate bacteria in various cells. Previously, we reported that intracellular Streptococcus pneumoniae can induce unique hierarchical autophagy comprised of CASM induction, shedding, and subsequent xenophagy. However, the molecular mechanisms underlying these processes and the biological significance of transient CASM induction remain unknown. Herein, we profile the relationship between Atg8s, autophagy receptors, poly-ubiquitin, and Atg4 paralogs during pneumococcal infection to understand the driving principles of hierarchical autophagy and find that GATE16 and GBRP sequentially play a pivotal role in CASM shedding and subsequent xenophagy induction, respectively, and LC3A and GBRPL1 are involved in CASM/xenophagy induction. Moreover, we reveal ingenious bacterial tactics to gain intracellular survival niches by manipulating CASM-xenophagy progression by generating intracellular pneumococci-derived H2O2.


Sujet(s)
Famille de la protéine-8 associée à l'autophagie , Streptococcus pneumoniae , Animaux , Souris , Autophagie , Famille de la protéine-8 associée à l'autophagie/métabolisme , Protéines associées à l'autophagie/métabolisme , Macroautophagie , Protéines associées aux microtubules/métabolisme , Infections à pneumocoques/microbiologie , Infections à pneumocoques/métabolisme , Infections à pneumocoques/immunologie , Streptococcus pneumoniae/métabolisme
16.
Int J Cancer ; 155(3): 569-581, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38630934

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is a heterogeneous disease with distinct molecular subtypes described as classical/progenitor and basal-like/squamous PDAC. We hypothesized that integrative transcriptome and metabolome approaches can identify candidate genes whose inactivation contributes to the development of the aggressive basal-like/squamous subtype. Using our integrated approach, we identified endosome-lysosome associated apoptosis and autophagy regulator 1 (ELAPOR1/KIAA1324) as a candidate tumor suppressor in both our NCI-UMD-German cohort and additional validation cohorts. Diminished ELAPOR1 expression was linked to high histological grade, advanced disease stage, the basal-like/squamous subtype, and reduced patient survival in PDAC. In vitro experiments demonstrated that ELAPOR1 transgene expression not only inhibited the migration and invasion of PDAC cells but also induced gene expression characteristics associated with the classical/progenitor subtype. Metabolome analysis of patient tumors and PDAC cells revealed a metabolic program associated with both upregulated ELAPOR1 and the classical/progenitor subtype, encompassing upregulated lipogenesis and downregulated amino acid metabolism. 1-Methylnicotinamide, a known oncometabolite derived from S-adenosylmethionine, was inversely associated with ELAPOR1 expression and promoted migration and invasion of PDAC cells in vitro. Taken together, our data suggest that enhanced ELAPOR1 expression promotes transcriptome and metabolome characteristics that are indicative of the classical/progenitor subtype, whereas its reduction associates with basal-like/squamous tumors with increased disease aggressiveness in PDAC patients. These findings position ELAPOR1 as a promising candidate for diagnostic and therapeutic targeting in PDAC.


Sujet(s)
Carcinome du canal pancréatique , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Lignée cellulaire tumorale , Mâle , Femelle , Métabolome , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Invasion tumorale , Transcriptome , Adulte d'âge moyen ,
17.
Mol Cell Endocrinol ; 589: 112252, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38649132

RÉSUMÉ

Pathological cardiac hypertrophy often precedes heart failure due to various stimuli, yet effective clinical interventions remain limited. Recently, microRNAs (miRNAs) have been identified as critical regulators of cardiovascular development. In this study, we investigated the role of miR-146b-5p and its underlying mechanisms of action in cardiac hypertrophy. Isoprenaline (ISO) treatment induced significant hypertrophy and markedly enhanced the expression of miR-146b-5p in cultured neonatal rat cardiomyocytes and hearts of C57BL/6 mice. Transfection with the miR-146b-5p mimic led to cardiomyocyte hypertrophy accompanied by autophagy inhibition. Conversely, miR-146b-5p inhibition significantly alleviated ISO-induced autophagy depression, thereby mitigating cardiac hypertrophy both in vitro and in vivo. Our results showed that the autophagy-related mediator double FYVE domain-containing protein 1 (DFCP1) is a target of miR-146b-5p. MiR-146b-5p blocked autophagic flux in cardiomyocytes by suppressing DFCP1, thus contributing to hypertrophy. These findings revealed that miR-146b-5p is a potential regulator of autophagy associated with the onset of cardiac hypertrophy, suggesting a possible therapeutic strategy involving the inhibition of miR-146b-5p.


Sujet(s)
Autophagie , Cardiomégalie , Isoprénaline , Souris de lignée C57BL , microARN , Myocytes cardiaques , Animaux , microARN/génétique , microARN/métabolisme , Isoprénaline/pharmacologie , Cardiomégalie/génétique , Cardiomégalie/induit chimiquement , Cardiomégalie/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Rat Sprague-Dawley , Rats , Mâle , Souris , Cellules cultivées , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Protéines associées à l'autophagie/génétique , Protéines associées à l'autophagie/métabolisme , Séquence nucléotidique
18.
Mol Microbiol ; 121(6): 1095-1111, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38574236

RÉSUMÉ

The protozoan parasite Plasmodium, the causative agent of malaria, undergoes an obligatory stage of intra-hepatic development before initiating a blood-stage infection. Productive invasion of hepatocytes involves the formation of a parasitophorous vacuole (PV) generated by the invagination of the host cell plasma membrane. Surrounded by the PV membrane (PVM), the parasite undergoes extensive replication. During intracellular development in the hepatocyte, the parasites provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterized by a long-lasting association of the autophagy marker protein, and ATG8 family member, LC3B with the PVM. LC3B localization at the PVM does not follow the canonical autophagy pathway since upstream events specific to canonical autophagy are dispensable. Here, we describe that LC3B localization at the PVM of Plasmodium parasites requires the V-ATPase and its interaction with ATG16L1. The WD40 domain of ATG16L1 is crucial for its recruitment to the PVM. Thus, we provide new mechanistic insight into the previously described PAAR response targeting Plasmodium liver stage parasites.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Hépatocytes , Foie , Protéines associées aux microtubules , Plasmodium berghei , Vacuolar Proton-Translocating ATPases , Vacuoles , Vacuoles/métabolisme , Vacuoles/parasitologie , Plasmodium berghei/génétique , Plasmodium berghei/croissance et développement , Plasmodium berghei/métabolisme , Plasmodium berghei/enzymologie , Animaux , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Foie/parasitologie , Souris , Hépatocytes/parasitologie , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Paludisme/parasitologie , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Humains
19.
Int J Mol Sci ; 25(8)2024 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-38674078

RÉSUMÉ

Canonical autophagy is an evolutionarily conserved process that forms double-membrane structures and mediates the degradation of long-lived proteins (LLPs). Noncanonical autophagy (NCA) is an important alternative pathway involving the formation of microtubule-associated protein 1 light chain 3 (LC3)-positive structures that are independent of partial core autophagy proteins. NCA has been defined by the conjugation of ATG8s to single membranes (CASM). During canonical autophagy and NCA/CASM, LC3 undergoes a lipidation modification, and ATG16L1 is a crucial protein in this process. Previous studies have reported that the WDR domain of ATG16L1 is not necessary for canonical autophagy. However, our study found that WDR domain deficiency significantly impaired LLP degradation in basal conditions and slowed down LC3-II accumulation in canonical autophagy. We further demonstrated that the observed effect was due to a reduced interaction between ATG16L1 and FIP200/WIPI2, without affecting lysosome function or fusion. Furthermore, we also found that the WDR domain of ATG16L1 is crucial for chemical-induced NCA/CASM. The results showed that removing the WDR domain or introducing the K490A mutation in ATG16L1 significantly inhibited the NCA/CASM, which interrupted the V-ATPase-ATG16L1 axis. In conclusion, this study highlights the significance of the WDR domain of ATG16L1 for both canonical autophagy and NCA functions, improving our understanding of its role in autophagy.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Protéines membranaires , Protéines associées aux microtubules , Protéines de liaison aux phosphates , Répétitions WD40 , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Autophagie/génétique , Humains , Répétitions WD40/génétique , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Lysosomes/métabolisme , Cellules HEK293 , Cellules HeLa
20.
Bioessays ; 46(6): e2300243, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38593284

RÉSUMÉ

The autophagy initiation complex is brought about via a highly ordered and stepwise assembly process. Two crucial signaling molecules, mTORC1 and AMPK, orchestrate this assembly by phosphorylating/dephosphorylating autophagy-related proteins. Activation of Atg1 followed by recruitment of both Atg9 vesicles and the PI3K complex I to the PAS (phagophore assembly site) are particularly crucial steps in its formation. Ypt1, a small Rab GTPase in yeast cells, also plays an essential role in the formation of the autophagy initiation complex through multiple regulatory pathways. In this review, our primary focus is to discuss how signaling molecules initiate the assembly of the autophagy initiation complex, and highlight the significant roles of Ypt1 in this process. We end by addressing issues that need future clarification.


Sujet(s)
Protéines associées à l'autophagie , Autophagie , Complexe-1 cible mécanistique de la rapamycine , Protéines de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Transduction du signal , Protéines G rab , Protéines de Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Protéines G rab/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Humains , Animaux , AMP-Activated Protein Kinases/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Complexes multiprotéiques/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...