Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 785
Filtrer
1.
FASEB J ; 38(16): e70024, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39190024

RÉSUMÉ

The role of programmed cell death 4 (PDCD4) in multiple myeloma (MM) development remains unknown. Here, we investigated its role and action mechanism in MM. Bioinformatic analysis indicated that patients with MM and high PDCD4 expression had higher overall survival than those with low PDCD4 expression. PDCD4 expression promoted MM cell apoptosis and inhibited their viability in vitro and tumor growth in vivo. RNA-binding protein immunoprecipitation sequencing analysis showed that PDCD4 is bound to the 5' UTR of the apoptosis-related genes PIK3CB, Cathepsin Z (CTSZ), and X-chromosome-linked apoptosis inhibitor (XIAP). PDCD4 knockdown reduced the cell apoptosis rate, which was rescued by adding PIK3CB, CTSZ, or XIAP inhibitors. Dual luciferase reporter assays confirmed the internal ribosome entry site (IRES) activity of the 5' UTRs of PIK3CB and CTSZ. An RNA pull-down assay confirmed binding of the 5' UTR of PIK3CB and CTSZ to PDCD4, identifying the specific binding fragments. PDCD4 is expected to promote MM cell apoptosis by binding to the IRES domain in the 5' UTR of PIK3CB and CTSZ and inhibiting their translation. Our findings suggest that PDCD4 plays an important role in MM development by regulating the expression of PIK3CB, CTSZ, and XIAP, and highlight new potential molecular targets for MM treatment.


Sujet(s)
Protéines régulatrices de l'apoptose , Apoptose , Myélome multiple , Protéines de liaison à l'ARN , Animaux , Humains , Mâle , Souris , Protéines régulatrices de l'apoptose/génétique , Protéines régulatrices de l'apoptose/métabolisme , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases de classe I/métabolisme , Phosphatidylinositol 3-kinases de classe I/génétique , Régulation de l'expression des gènes tumoraux , Myélome multiple/métabolisme , Myélome multiple/génétique , Myélome multiple/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique
2.
Cancer Lett ; 598: 217126, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39053726

RÉSUMÉ

The MDM2 oncogene is amplified and/or overexpressed in various human cancers and elevated expression of MDM2 protein acts as a survival factor promoting cancer progression through both p53-dependent and -independent pathways. Here, we report a novel small-molecule chemical compound (MX69-102) that we identified to induce MDM2 protein degradation, resulting in reactivation of p53, inhibition of XIAP, and potent cell growth inhibition and apoptosis in MDM2-overexpressing acute lymphoblastic leukemia (ALL) in vitro and in vivo. We have previously identified a compound (MX69) that binds to the MDM2 C-terminal RING domain and induces MDM2 protein degradation. In the present study, we performed structural modifications of MX69 and selected analog MX69-102, showing increased MDM2-targeting activity. MX69-102 exhibited significantly enhanced inhibitory and apoptotic effects on a group of MDM2-overexpressing ALL cell lines in vitro with IC50 values of about 0.2 µM, representing an approximately 38-fold increase in activity compared to MX69. MX69-102 also showed effective inhibition on xenografted human MDM2-overexpressing ALL in SCID mice. Importantly, MX69-102 had minimal or no inhibitory effect on normal human hematopoiesis in vitro and was very well tolerated in vivo in animal models. Based on the strong inhibitory and apoptotic activity against MDM2-overexpressing ALL, along with minimal or no toxicity to normal cells/tissues, MX69-102 is a candidate for further development as a novel MDM2-targeted therapeutic drug for refractory/MDM2-overexpressing ALL.


Sujet(s)
Apoptose , Souris SCID , Leucémie-lymphome lymphoblastique à précurseurs B et T , Protéines proto-oncogènes c-mdm2 , Protéine p53 suppresseur de tumeur , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines proto-oncogènes c-mdm2/métabolisme , Protéines proto-oncogènes c-mdm2/génétique , Humains , Animaux , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Relation dose-effet des médicaments , Souris , Femelle , Relation structure-activité
3.
Blood ; 144(11): 1183-1192, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-38820590

RÉSUMÉ

ABSTRACT: Deficiency of X-linked inhibitor of apoptosis protein (XIAP) is a rare genetic condition that can present with recurrent episodes of hemophagocytic lymphohistiocytosis (HLH), though the exact mechanisms leading to this hyperinflammatory disorder are unclear. Understanding its biology is critical to developing targeted therapies for this potentially fatal disease. Here, we report on a novel multiexonic intragenic duplication leading to XIAP deficiency with recurrent HLH that demonstrated complete response to interleukin (IL)-1ß blockade. We further demonstrate using both primary patient cells and genetically modified THP-1 monocyte cell lines that, contrary to what has previously been shown in mouse cells, XIAP-deficient human macrophages do not produce excess IL-1ß when stimulated under standard conditions. Instead, nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated hyperproduction of IL-1ß is observed only when the XIAP-deficient cells are stimulated under autophagy-promoting conditions and this correlates with defective autophagic flux as measured by decreased accumulation of the early autophagy marker LC3-II. This work, therefore, highlights IL-1ß blockade as a therapeutic option for patients with XIAP deficiency experiencing recurrent HLH and identifies a critical role for XIAP in promoting autophagy as a means of limiting IL-1ß-mediated hyperinflammation during periods of cellular stress.


Sujet(s)
Autophagie , Interleukine-1 bêta , Protéine inhibitrice de l'apoptose liée au chromosome X , Humains , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/déficit , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Interleukine-1 bêta/métabolisme , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/anatomopathologie , Lymphohistiocytose hémophagocytaire/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Inflammasomes/métabolisme , Mâle , Macrophages/métabolisme , Macrophages/anatomopathologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/déficit , Femelle
4.
Apoptosis ; 29(7-8): 1145-1160, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38684550

RÉSUMÉ

Mutations resulting in decreased activity of p53 tumor suppressor protein promote tumorigenesis. P53 protein levels are tightly regulated through the Ubiquitin Proteasome System (UPS). Several E3 ligases were shown to regulate p53 stability, including MDM2. Here we report that the ubiquitin E3 ligase XIAP (X-linked Inhibitors of Apoptosis) is a direct ligase for p53 and describe a novel approach for modulating the levels of p53 by targeting the XIAP pathway. Using in vivo (live-cell) and in vitro (cell-free reconstituted system) ubiquitylation assays, we show that the XIAP-antagonist ARTS regulates the levels of p53 by promoting the degradation of XIAP. XIAP directly binds and ubiquitylates p53. In apoptotic cells, ARTS inhibits the ubiquitylation of p53 by antagonizing XIAP. XIAP knockout MEFs express higher p53 protein levels compared to wild-type MEFs. Computational screen for small molecules with high affinity to the ARTS-binding site within XIAP identified a small-molecule ARTS-mimetic, B3. This compound stimulates apoptosis in a wide range of cancer cells but not normal PBMC (Peripheral Blood Mononuclear Cells). Like ARTS, the B3 compound binds to XIAP and promotes its degradation via the UPS. B3 binding to XIAP stabilizes p53 by disrupting its interaction with XIAP. These results reveal a novel mechanism by which ARTS and p53 regulate each other through an amplification loop to promote apoptosis. Finally, these data suggest that targeting the ARTS binding pocket in XIAP can be used to increase p53 levels as a new strategy for developing anti-cancer therapeutics.


Sujet(s)
Apoptose , Protéolyse , Protéine p53 suppresseur de tumeur , Ubiquitination , Protéine inhibitrice de l'apoptose liée au chromosome X , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Apoptose/effets des médicaments et des substances chimiques , Ubiquitination/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Animaux , Souris , Lignée cellulaire tumorale , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Régulation positive/effets des médicaments et des substances chimiques , Liaison aux protéines
5.
Gene ; 912: 148355, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38467314

RÉSUMÉ

BACKGROUND: Breast cancer (BC) is the most prevalent malignant disease affecting women globally. PANoptosis, a novel form of cell death combining features of pyroptosis, apoptosis, and necroptosis, has recently gained attention. However, its precise function in BC and the predictive values of PANoptosis-related genes remain unclear. METHODS: We used the expression data and clinical information of BC tissues or normal breast tissues from public databases, and then successfully developed and verified a BC PANoptosis-related risk model through a combination of univariate Cox regression, least absolute shrinkage and selection operator (LASSO) regression, and Kaplan-Meier (KM) analysis. A nomogram was constructed to estimate survival probability, and its accuracy was assessed using calibration curves. RESULTS: Among 37 PANoptosis-related genes, we identified 4 differentially expressed genes related to overall survival (OS). Next, a risk model incorporating these four PANoptosis-related genes was established. Patients were stratified into low/high-risk groups based on the median risk score, with the low-risk group showing better prognoses and higher levels of immune infiltration. Utilizing the risk score and clinical features, we developed a nomogram to predict 1-, 3- and 5-year survival probability. X-linked inhibitor of apoptosis protein (XIAP) emerged as a potentially risky factor with the highest hazard ratio. In vitro experiments demonstrated that XIAP inhibition enhances the antitumor effect of doxorubicin through the PANoptosis pathway. CONCLUSION: PANoptosis holds an important role in BC prognosis and treatment.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Tumeurs du sein/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Oncogènes/génétique , Doxorubicine , Apoptose/génétique
6.
Exp Cell Res ; 437(1): 113995, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38490621

RÉSUMÉ

PURPOSE: Oral Squamous Cell Carcinoma (OSCC) is the 6th most common cancer worldwide. It is generally aggressive and closely associated with chemoresistance and poor survival. There is accumulating evidence for the involvement of inhibitors of apoptosis proteins (IAPs), including IAP1 and XIAP, in mediating chemotherapy resistance in OSCC. Various strategies for targeting IAPs have been designed and tested in recent years and several small molecule IAP inhibitors are in clinical trials as monotherapies as well as in combination with radiotherapy and chemotherapy. The purpose of this study was to evaluate and compare the efficacy and biological activity of three IAP inhibitors both as stand-alone and sensitising agents to cisplatin in a preclinical model of squamous cell carcinoma of the tongue. METHODS: Cisplatin-sensitive SCC4 and -resistant SCC4cisR cells were utilised in this study. Apoptosis was evaluated by flow cytometric analysis of Annexin V/Propidium Iodide-stained cells. Expression of IAP proteins was determined by western blotting and knockdown of cIAP1, livin and XIAP was conducted by transfection of cells with siRNA. RESULTS: We establish for the first time the therapeutic efficacy of the Smac mimetic, BV6 and the XIAP inhibitor Embelin, for OSCC. Both of these IAP targeting agents synergistically enhanced cisplatin-mediated apoptotic cell death in resistant cells which was mediated in part by depletion of XIAP. In addition, knockdown of XIAP using siRNA enhanced cisplatin-mediated cell death, demonstrating the importance of targeting XIAP in this sensitisation. CONCLUSION: These findings provide pre-clinical evidence that IAP inhibition may be a valuable therapeutic option in OSCC.


Sujet(s)
Carcinome épidermoïde , Tumeurs de la bouche , Humains , Cisplatine/pharmacologie , Carcinome épidermoïde/traitement médicamenteux , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Lignée cellulaire tumorale , Tumeurs de la bouche/traitement médicamenteux , Protéines IAP/génétique , Protéines IAP/métabolisme , Apoptose/physiologie , Protéines de transport , Petit ARN interférent
7.
FEBS J ; 291(12): 2636-2655, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38317520

RÉSUMÉ

Parkinson's disease (PD) is a chronic neurodegenerative disease characterized by progressive loss of dopamine-producing neurons from the substantia nigra region of the brain. Mitochondrial dysfunction is one of the major causes of oxidative stress and neuronal cell death in PD. E3 ubiquitin ligases such as Parkin (PRKN) modulate mitochondrial quality control in PD; however, the role of other E3 ligases associated with mitochondria in the regulation of neuronal cell death in PD has not been explored. The current study investigated the role of TRIM32, RING E3 ligase, in sensitization to oxidative stress-induced neuronal apoptosis. The expression of TRIM32 sensitizes SH-SY5Y dopaminergic cells to rotenone and 6-OHDA-induced neuronal death, whereas the knockdown increased cell viability under PD stress conditions. The turnover of TRIM32 is enhanced under PD stress conditions and is mediated by autophagy. TRIM32 translocation to mitochondria is enhanced under PD stress conditions and localizes on the outer mitochondrial membrane. TRIM32 decreases complex-I assembly and activity as well as mitochondrial reactive oxygen species (ROS) and ATP levels under PD stress. Deletion of the RING domain of TRIM32 enhanced complex I activity and rescued ROS levels and neuronal viability under PD stress conditions. TRIM32 decreases the level of XIAP, and co-expression of XIAP with TRIM32 rescued the PD stress-induced cell death and mitochondrial ROS level. In conclusion, turnover of TRIM32 increases during stress conditions and translocation to mitochondria is enhanced, regulating mitochondrial functions and neuronal apoptosis by modulating the level of XIAP in PD.


Sujet(s)
Apoptose , Neurones dopaminergiques , Mitochondries , Stress oxydatif , Maladie de Parkinson , Espèces réactives de l'oxygène , Protéines à motif tripartite , Ubiquitin-protein ligases , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Humains , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Mitochondries/génétique , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , Maladie de Parkinson/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Espèces réactives de l'oxygène/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Roténone/pharmacologie , Transport des protéines , Complexe I de la chaîne respiratoire/métabolisme , Complexe I de la chaîne respiratoire/génétique , Lignée cellulaire tumorale , Oxidopamine/pharmacologie , Autophagie , Adénosine triphosphate/métabolisme , Survie cellulaire/génétique
8.
J Pediatr Hematol Oncol ; 46(2): e191-e194, 2024 03 01.
Article de Anglais | MEDLINE | ID: mdl-38277621

RÉSUMÉ

X-linked inhibitor of apoptosis protein (XIAP) deficiency is an inborn error of immunity (IEI). Allogeneic hematopoietic cell transplantation (HCT) is currently the only curative therapy available for XIAP deficiency. Granulomatous and lymphocytic interstitial lung disease (GLILD) is a common immune-related lung complication of IEIs. We present a 6-year-old boy with XIAP deficiency and GLILD. Computed tomography showed lung nodes but no symptoms. Before HCT, GLILD was not managed with immunosuppressive therapy, because he was asymptomatic. The HCT procedure was subsequently performed. The post-HCT course was uneventful; follow-up computed tomography on day 46 showed nodules had disappeared. HCT could potentially ameliorate GLILD like other inflammatory processes associated with the underlying IEIs.


Sujet(s)
Déficit immunitaire commun variable , Maladies génétiques liées au chromosome X , Transplantation de cellules souches hématopoïétiques , Pneumopathies interstitielles , Syndromes lymphoprolifératifs , Mâle , Humains , Enfant , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Pneumopathies interstitielles/étiologie , Pneumopathies interstitielles/thérapie , Syndromes lymphoprolifératifs/thérapie , Syndromes lymphoprolifératifs/complications , Transplantation de cellules souches hématopoïétiques/méthodes , Déficit immunitaire commun variable/complications
9.
Sci Rep ; 14(1): 853, 2024 01 09.
Article de Anglais | MEDLINE | ID: mdl-38191507

RÉSUMÉ

X-linked inhibitor of apoptosis protein (XIAP) deficiency causes refractory inflammatory bowel disease. The XIAP protein plays a pivotal role in the pro-inflammatory response through the nucleotide-binding oligomerization domain-containing signaling pathway that is important in mucosal homeostasis. We analyzed the molecular mechanism of non-synonymous pathogenic variants (PVs) of XIAP BIR2 domain. We generated N-terminally green fluorescent protein-tagged XIAP constructs of representative non-synonymous PVs. Co-immunoprecipitation and fluorescence cross-correlation spectroscopy showed that wild-type XIAP and RIP2 preferentially interacted in live cells, whereas all non-synonymous PV XIAPs failed to interact properly with RIP2. Structural analysis showed that various structural changes by mutations, such as hydrophobic core collapse, Zn-finger loss, and spatial rearrangement, destabilized the two loop structures (174-182 and 205-215) that critically interact with RIP2. Subsequently, it caused a failure of RIP2 ubiquitination and loss of protein deficiency by the auto-ubiquitination of all XIAP mutants. These findings could enhance our understanding of the role of XIAP mutations in XIAP-deficient inflammatory bowel disease and may benefit future therapeutic strategies.


Sujet(s)
Maladies inflammatoires intestinales , Protéine inhibitrice de l'apoptose liée au chromosome X , Humains , Protéines à fluorescence verte , Homéostasie , Maladies inflammatoires intestinales/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique
10.
J Allergy Clin Immunol ; 153(1): 256-264, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37678575

RÉSUMÉ

BACKGROUND: The contribution of genetic factors to the severity of adult hemophagocytic lymphohistiocytosis (HLHa) remains unclear. OBJECTIVE: We sought to assess a potential link between HLHa outcomes and HLH-related gene variants. METHODS: Clinical characteristics of 130 HLHa patients (age ≥ 18 years and HScore ≥ 169) and genotype of 8 HLH-related genes (LYST, PRF1, UNC13-D, STX11, STXBP2, RAB27A, XIAP, and SAP) were collected. A total of 34 variants found in only 6 genes were selected on the basis of their frequency and criteria predicted to impair protein function. Severity was defined by refractory disease to HLH treatment, death, or transfer to an intensive care unit. RESULTS: HLHa-associated diseases (ADs) were neoplasia (n = 49 [37.7%]), autoimmune/inflammatory disease (n = 33 [25.4%]), or idiopathic when no AD was identified (n = 48 [36.9%]). Infectious events occurred in 76 (58.5%) patients and were equally distributed in all ADs. Severe and refractory HLHa were observed in 80 (61.5%) and 64 (49.2%) patients, respectively. HScore, age, sex ratio, AD, and infectious events showed no significant association with HLHa severity. Variants were identified in 71 alleles and were present in 56 (43.1%) patients. They were distributed as follows: 44 (34.4%), 9 (6.9%), and 3 (2.3%) patients carrying 1, 2, and 3 variant alleles, respectively. In a logistic regression model, only the number of variants was significantly associated with HLHa severity (1 vs 0: 3.86 [1.73-9.14], P = .0008; 2-3 vs 0: 29.4 [3.62-3810], P = .0002) and refractoriness (1 vs 0: 2.47 [1.17-5.34], P = .018; 2-3 vs 0: 13.2 [2.91-126.8], P = .0003). CONCLUSIONS: HLH-related gene variants may be key components to the severity and refractoriness of HLHa.


Sujet(s)
Lymphohistiocytose hémophagocytaire , Adulte , Humains , Adolescent , Lymphohistiocytose hémophagocytaire/génétique , Lymphohistiocytose hémophagocytaire/thérapie , Allèles , Génotype , Protéine associée aux molécules de signalisation de l'activation des lymphocytes/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique
11.
J Cell Mol Med ; 28(2): e18071, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38044583

RÉSUMÉ

Oral squamous cell carcinoma (OSCC), which accounts for 90% of all oral cancers, has become a public health crisis worldwide. despite advances in therapeutic interventions, the prognosis remains poor for advanced-stage OSCC. In this study, we investigate the anticancer activity and the mode of action of hellebrigenin in human OSCC. The findings demonstrated that hellebrigenin exerted cytotoxic effects in OSCC cells through cell cycle arrest at the G2/M phase and downregulation of cell cycle-related proteins (cyclins A2, B1 and D3, Cdc2, CDK4 and CDK6). Moreover, hellebrigenin caused activation of PARP and caspase 3, 8 and 9, followed by downregulation of antiapoptotic proteins (Bcl-2 and Bcl-xL) and upregulation of pro-apoptotic proteins (Bax and Bak). The hellebrigenin treatment also increased Fas, DR5, DcR2 and DcR3 expressions in oral cancer cells, indicating the compound causes oral cancer cell apoptosis through both intrinsic and extrinsic pathways. Regarding upstream signalling, hellebrigenin was found to reduce the phosphorylation of ERK, p38, and JNK, indicating that hellebrigenin triggers caspase-mediated apoptosis by downregulating MAPK signalling pathway. Finally, the human apoptosis array findings revealed that hellebrigenin specifically suppressed the expression of XIAP to execute its pro-apoptotic activities. Taken together, the study suggests that hellebrigenin can act as a potent anticancer compound in human OSCC.


Sujet(s)
Bufanolide , Carcinome épidermoïde , Tumeurs de la bouche , Humains , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/génétique , Tumeurs de la bouche/anatomopathologie , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Transduction du signal , Apoptose/physiologie , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme
12.
Toxicol Lett ; 390: 25-32, 2023 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-37944651

RÉSUMÉ

Triptolide (TP) is extracted from the traditional Chinese medicine Tripterygium wilfordii Hook. F. (TWHF). Its severe toxic side effects, especially hepatotoxicity, have limited the clinical application of TP-related drugs. In this study, we investigated the mechanism of the hepatotoxic effects of TP from the perspective that TP inhibited the expression of the pro-survival protein X-linked inhibitor of apoptosis protein (XIAP) and enhanced FasL-mediated apoptosis of hepatocytes. TP and CD95/Fas antibody (Jo-2) were administered by gavage to C57BL/6 mice for 7 consecutive days. After co-administration of TP and Jo-2, mouse livers showed large areas of necrosis and apoptosis and significantly increased Caspase-3 activity. KEGG pathway enrichment analysis indicated that TP may cause the development of liver injury through the apoptotic signaling pathway. Proteinprotein interaction networks showed that XIAP played an essential role in this process. TP reduced the protein expression of XIAP after combination treatment with Jo-2/FasL in vivo/in vitro. TP and FasL co-stimulation significantly increased microRNA-137 (miR-137) levels in AML12 cells, while inhibition of miR-137 expression induced a rebound in XIAP protein expression. In conclusion, TP presensitizes hepatocytes and enhances the sensitivity of hepatocytes to the Fas/FasL pathway by inhibiting the protein expression of XIAP, leading to hepatocyte apoptosis.


Sujet(s)
microARN , Protéine inhibitrice de l'apoptose liée au chromosome X , Souris , Animaux , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/pharmacologie , Souris de lignée C57BL , Foie/métabolisme , Hépatocytes , Apoptose , microARN/métabolisme
13.
Cancer Res Commun ; 3(11): 2386-2399, 2023 11 22.
Article de Anglais | MEDLINE | ID: mdl-37874199

RÉSUMÉ

XIAP, the most potent mammalian inhibitor of apoptosis protein (IAP), critically restricts developmental culling of sympathetic neuronal progenitors, and is correspondingly overexpressed in most MYCN-amplified neuroblastoma tumors. Because apoptosis-related protein in the TGFß signaling pathway (ARTS) is the only XIAP antagonist that directly binds and degrades XIAP, we evaluated the preclinical effectiveness and tolerability of XIAP antagonism as a novel targeting strategy for neuroblastoma. We found that antagonism of XIAP, but not other IAPs, triggered apoptotic death in neuroblastoma cells. XIAP silencing induced apoptosis while overexpression conferred protection from drug-induced apoptosis. From a screen of IAP inhibitors, first-in-class ARTS mimetic A4 was most effective against high-risk and high XIAP-expressing neuroblastoma cells, and least toxic toward normal liver- and bone marrow-derived cells, compared with pan-IAP antagonists. On target engagement assays and nuclear magnetic resonance spectroscopy, A4 was observed to degrade rather than inhibit XIAP, catalyzing rapid degradation of XIAP through the ubiquitin-proteasome pathway. In MYCN-amplified neuroblastoma patient-derived xenografts, A4 significantly prolonged survival as a single agent, and demonstrated synergism with standard-of-care agents to reduce their effective required doses 3- to 6-fold. Engagement and degradation of XIAP by ARTS mimetics is a novel targeting strategy for neuroblastoma that may be especially effective against MYCN-amplified disease with intrinsically high XIAP expression. First-in-class ARTS mimetic A4 demonstrates preclinical efficacy and warrants further development and study. SIGNIFICANCE: XIAP degradation is sufficient to kill MYCN-amplified neuroblastoma which overexpresses and relies on XIAP as a brake against cell death, without affecting normal cells.


Sujet(s)
Neuroblastome , Protéine inhibitrice de l'apoptose liée au chromosome X , Animaux , Humains , Protéine du proto-oncogène N-Myc/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Apoptose , Neuroblastome/traitement médicamenteux , Protéines IAP/métabolisme , Mammifères/métabolisme
14.
Infect Immun ; 91(9): e0000223, 2023 09 14.
Article de Anglais | MEDLINE | ID: mdl-37594275

RÉSUMÉ

Ehrlichia chaffeensis has evolved multiple strategies to evade innate defenses of the mononuclear phagocyte. Recently, we reported the E. chaffeensis tandem repeat protein (TRP)120 effector functions as a Notch ligand mimetic and a ubiquitin ligase that degrades the nuclear tumor suppressor, F-box and WD repeat domain-containing 7, a negative regulator of Notch. The Notch intracellular domain (NICD) is known to inhibit apoptosis primarily by interacting with X-linked inhibitor of apoptosis protein (XIAP) to prevent degradation. In this study, we determined that E. chaffeensis activation of Notch signaling increases XIAP levels, thereby inhibiting apoptosis through both the intrinsic and executioner pathways. Increased NICD and XIAP levels were detected during E. chaffeensis infection and after TRP120 Notch ligand mimetic peptide treatment. Conversely, XIAP levels were reduced in the presence of Notch inhibitor DAPT. Cytoplasmic and nuclear colocalization of NICD and XIAP was observed during infection and a direct interaction was confirmed by co-immunoprecipitation. Procaspase levels increased temporally during infection, consistent with increased XIAP levels; however, knockdown (KD) of XIAP during infection significantly increased apoptosis and Caspase-3, -7, and -9 levels. Furthermore, treatment with SM-164, a second mitochondrial activator of caspases (Smac/DIABLO) antagonist, resulted in decreased procaspase levels and increased caspase activation, induced apoptosis, and significantly decreased infection. In addition, RNAi KD of XIAP also decreased infection and significantly increased apoptosis. Moreover, ectopic expression of TRP120 HECT Ub ligase catalytically defective mutant in HeLa cells decreased NICD and XIAP levels and increased caspase activation compared to HeLa cells with functional HECT Ub ligase catalytic activity (TRP120-WT). This investigation reveals a mechanism whereby E. chaffeensis modulates Notch signaling to stabilize XIAP and inhibit apoptosis.


Sujet(s)
Ehrlichia chaffeensis , Ehrlichiose , Humains , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Cellules HeLa , Ligands , Apoptose , Caspases , Ehrlichia chaffeensis/génétique
15.
BMC Cardiovasc Disord ; 23(1): 368, 2023 07 21.
Article de Anglais | MEDLINE | ID: mdl-37479963

RÉSUMÉ

BACKGROUND: The X-linked inhibitor of apoptosis (XIAP) protein is encoded by the XIAP gene and is critical for multiple cell responses and plays a role in preventing cell death. XIAP mutations are associated with several diseases, primarily including hemophagocytic lymphohistiocytosis and inflammatory bowel disease (IBD). We report the clinical features and results associated with hemizygous mutation of the XIAP gene in a young male with Crohn's disease complicated with acute heart failure.This 16-year-old patient ultimately died of heart failure. CASE PRESENTATION: A young male of 16 years of age was initially diagnosed with Crohn's disease based on evidences from endoscopic and histological findings. Although supportive care, anti-infective drugs and biologics were administered consecutively for 11 months, his clinical manifestations and laboratory indices (patient's condition) did not improved. Additionally, the patient exhibited a poor nutritional status and sustained weight loss. Subsequently, acute heart failure led to the exacerbation of the patient's condition. He was diagnosed with wet beriberi according to thiamine deficiency, but the standard medical therapy for heart failure and thiamine supplementation did not reverse the adverse outcomes. Comprehensive genetic analysis of peripheral blood-derived DNA revealed a novel hemizygous mutation of the XIAP gene (c.1259_1262 delACAG), which was inherited from his mother. CONCLUSION: A novel XIAP mutation (c.1259_1262 delACAG) was identified in this study. It may be one of the potential pathogenic factors in Crohn's disease and plays an important role in the progression of heart failure. Additionally, thiamine deficiency triggers a vicious cycle.


Sujet(s)
Maladie de Crohn , Défaillance cardiaque , Carence en thiamine , Mâle , Humains , Adolescent , Maladie de Crohn/complications , Maladie de Crohn/diagnostic , Maladie de Crohn/génétique , Défaillance cardiaque/étiologie , Défaillance cardiaque/génétique , Perte de poids , Apoptose , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique
16.
Chemotherapy ; 68(4): 210-218, 2023.
Article de Anglais | MEDLINE | ID: mdl-37429260

RÉSUMÉ

INTRODUCTION: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered to be an effective apoptosis inducer due to its selectivity for tumor cells. However, many cancer cells, especially metastatic cancer cells, often exhibit resistance to TRAIL because their apoptotic pathway is impaired or their pro-survival pathway is overactivated. TRAIL resistance is the main obstacle to current TRAIL therapy. Nowadays, ceramide analogs represent a new class of potential anticancer agents. Therefore, we hypothesized that disrupting pro-survival signaling with ceramide analogs would increase TRAIL-mediated apoptosis. METHODS: MTT assay and flow cytometry were conducted to evaluate the synergistic effect of ceramide analog 5cc on TRAIL in metastatic colon cancer cells. Western blot was used to detect signaling proteins affected by 5cc. RNA interference was performed to analyze the effects of specific gene on 5cc-enhanced apoptosis. RESULTS: Ceramide analog 5cc markedly enhanced TRAIL-induced apoptosis evidenced by increased propidium iodide/annexin V double-positive cells and PARP cleavage in SW620 and LS411N cells. At the molecular level, 5cc significantly reduced the expression of anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through the activation of the c-Jun n-terminal kinase (JNK) pathway which is critically involved in sensitizing tumor cells to TRAIL/5cc combination. JNK-silenced cells exhibited a significant reversal of TRAIL/5cc-mediated apoptosis. CONCLUSION: Our data demonstrated that ceramide analog 5cc overcomes TRAIL resistance by enhancing JNK activation and repressing XIAP expression in metastatic colon cancer cells.


Sujet(s)
Tumeurs du côlon , Protéine inhibitrice de l'apoptose liée au chromosome X , Humains , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/pharmacologie , Céramides/pharmacologie , Ligands , Lignée cellulaire tumorale , Apoptose , Tumeurs du côlon/traitement médicamenteux , Facteur de nécrose tumorale alpha/pharmacologie , Ligand TRAIL/pharmacologie
17.
J Biol Inorg Chem ; 28(5): 485-494, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37268744

RÉSUMÉ

Modulation of metalloprotein structure and function via metal ion substitution may constitute a molecular basis for metal ion toxicity and/or metal-mediated functional control. The X-linked Inhibitor of Apoptosis Protein (XIAP) is a metalloprotein that requires zinc for proper structure and function. In addition to its role as a modulator of apoptosis, XIAP has been implicated in copper homeostasis. Given the similar coordination preferences of copper and zinc, investigation of XIAP structure and function upon interaction with copper is relevant. The Really Interesting New Gene (RING) domain of XIAP is representative of a class of zinc finger proteins that utilize a bi-nuclear zinc-binding motif to maintain proper structure and ubiquitin ligase function. Herein, we report the characterization of copper (I) binding to the Zn2-RING domain of XIAP. Electronic absorption studies that monitor copper-thiolate interactions demonstrate that the RING domain of XIAP binds 5-6 Cu(I) ions and that copper is thermodynamically preferred relative to zinc. Repetition of the experiments in the presence of the Zn(II)-specific dye Mag-Fura2 shows that Cu(I) addition results in Zn(II) ejection from the protein, even in the presence of glutathione. Loss of dimeric structure of the RING domain, which is a requirement for its ubiquitin ligase activity, upon copper substitution at the zinc-binding sites, was readily observed via size exclusion chromatography. These results provide a molecular basis for the modulation of RING function by copper and add to the growing body of literature that describe the impact of Cu(I) on zinc metalloprotein structure and function.


Sujet(s)
Cuivre , Métalloprotéines , Cuivre/composition chimique , Métalloprotéines/métabolisme , Liaison aux protéines , Ubiquitines/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/composition chimique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Zinc/métabolisme , Domaines à doigts de zinc de type RING/physiologie
19.
PLoS Pathog ; 19(6): e1011455, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37347786

RÉSUMÉ

XIAP is an endogenous inhibitor of cell death and inactivating mutations of XIAP are responsible for X-linked lymphoproliferative disease (XLP-2) and primary immunodeficiency, but the mechanism(s) behind these contradictory outcomes have been unclear. We report that during infection of macrophages and dendritic cells with various intracellular bacteria, XIAP restricts cell death and secretion of IL-1ß but promotes increased activation of NFκB and JNK which results in elevated secretion of IL-6 and IL-10. Poor secretion of IL-6 by Xiap-deficient antigen presenting cells leads to poor expansion of recently activated CD8 T cells during the priming phase of the response. On the other hand, Xiap-deficient CD8 T cells displayed increased proliferation and effector function during the priming phase but underwent enhanced contraction subsequently. Xiap-deficient CD8 T cells underwent skewed differentiation towards short lived effectors which resulted in poor generation of memory. Consequently Xiap-deficient CD8 T cells failed to provide effective control of bacterial infection during re-challenge. These results reveal the temporal impact of XIAP in promoting the fitness of activated CD8 T cells through cell extrinsic and intrinsic mechanisms and provide a mechanistic explanation of the phenotype observed in XLP-2 patients.


Sujet(s)
Interleukine-6 , Syndromes lymphoprolifératifs , Humains , Mort cellulaire , Syndromes lymphoprolifératifs/génétique , Macrophages/métabolisme , Lymphocytes T CD8+/métabolisme , Mémoire immunologique , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme
20.
J Theor Biol ; 572: 111562, 2023 09 07.
Article de Anglais | MEDLINE | ID: mdl-37348784

RÉSUMÉ

Chemotherapeutic drugs are used to treat almost all types of cancer, but the intended response, i.e., elimination, is often incomplete, with a subset of cancer cells resisting treatment. Two critical factors play a role in chemoresistance: the p53 tumour suppressor gene and the X-linked inhibitor of apoptosis (XIAP). These proteins have been shown to act synergistically to elicit cellular responses upon DNA damage induced by chemotherapy, yet, the mechanism is poorly understood. This study introduces a mathematical model characterising the apoptosis pathway activation by p53 before and after mitochondrial outer membrane permeabilisation upon treatment with the chemotherapy Doxorubicin (Dox). "In-silico" simulations show that the p53 dynamics change dose-dependently. Under medium to high doses of Dox, p53 concentration ultimately stabilises to a high level regardless of XIAP concentrations. However, caspase-3 activation may be triggered or not depending on the XIAP induction rate, ultimately determining whether the cell will perish or resist. Consequently, the model predicts that failure to activate apoptosis in some cancer cells expressing wild-type p53 might be due to heterogeneity between cells in upregulating the XIAP protein, rather than due to the p53 protein concentration. Our model suggests that the interplay of the p53 dynamics and the XIAP induction rate is critical to determine the cancer cells' therapeutic response.


Sujet(s)
Protéine p53 suppresseur de tumeur , Protéine inhibitrice de l'apoptose liée au chromosome X , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Apoptose/physiologie , Mort cellulaire , Doxorubicine/pharmacologie , Lignée cellulaire tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE