Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 578
Filtrer
1.
Appl Microbiol Biotechnol ; 108(1): 459, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39230729

RÉSUMÉ

The recombinant adeno-associated virus (rAAV) vector is among the most promising viral vectors in gene therapy. However, the limited manufacturing capacity in human embryonic kidney (HEK) cells is a barrier to rAAV commercialization. We investigated the impact of endoplasmic reticulum (ER) protein processing and apoptotic genes on transient rAAV production in HEK293 cells. We selected four candidate genes based on prior transcriptomic studies: XBP1, GADD34 / PPP1R15A, HSPA6, and BCL2. These genes were stably integrated into HEK293 host cells. Traditional triple-plasmid transient transfection was used to assess the vector production capability and the quality of both the overexpressed stable pools and the parental cells. We show that the overexpression of XBP1, HSPA6, and GADD34 increases rAAV productivity by up to 100% and increases specific rAAV productivity by up to 78% in HEK293T cells. Additionally, more prominent improvement associated with ER protein processing gene overexpression was observed when parental cell productivity was high, but no substantial variation was detected under low-producing conditions. We also confirmed genome titer improvement across different serotypes (AAV2 and AAV8) and different cell lines (HEK293T and HEK293); however, the extent of improvement may vary. This study unveiled the importance of ER protein processing pathways in viral particle synthesis, capsid assembly, and vector production. KEY POINTS: • Upregulation of endoplasmic reticulum (ER) protein processing (XBP1, HSPA6, and GADD34) leads to a maximum 100% increase in rAAV productivity and a maximum 78% boost in specific rAAV productivity in HEK293T cells • The enhancement in productivity can be validated across different HEK293 cell lines and can be used for the production of various AAV serotypes, although the extent of the enhancement might vary slightly • The more pronounced improvements linked to overexpressing ER protein processing genes were observed when parental cell productivity was high, with minimal variation noted under low-producing conditions.


Sujet(s)
Dependovirus , Réticulum endoplasmique , Vecteurs génétiques , Protéine-1 liant la boite X , Humains , Cellules HEK293 , Dependovirus/génétique , Protéine-1 liant la boite X/génétique , Protéine-1 liant la boite X/métabolisme , Réticulum endoplasmique/métabolisme , Vecteurs génétiques/génétique , Expression des gènes , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Capside/métabolisme
2.
Nat Commun ; 15(1): 6810, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122682

RÉSUMÉ

Multiple myeloma is a hematological malignancy arising from immunoglobulin-secreting plasma cells. It remains poorly understood how chromatin rewiring of regulatory elements contributes to tumorigenesis and therapy resistance in myeloma. Here we generate a high-resolution contact map of myeloma-associated super-enhancers by integrating H3K27ac ChIP-seq and HiChIP from myeloma cell lines, patient-derived myeloma cells and normal plasma cells. Our comprehensive transcriptomic and phenomic analyses prioritize candidate genes with biological and clinical implications in myeloma. We show that myeloma cells frequently acquire SE that transcriptionally activate an oncogene PPP1R15B, which encodes a regulatory subunit of the holophosphatase complex that dephosphorylates translation initiation factor eIF2α. Epigenetic silencing or knockdown of PPP1R15B activates pro-apoptotic eIF2α-ATF4-CHOP pathway, while inhibiting protein synthesis and immunoglobulin production. Pharmacological inhibition of PPP1R15B using Raphin1 potentiates the anti-myeloma effect of bortezomib. Our study reveals that myeloma cells are vulnerable to perturbation of PPP1R15B-dependent protein homeostasis, highlighting a promising therapeutic strategy.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Myélome multiple , Protein Phosphatase 1 , Homéostasie protéique , , Facteur de transcription CHOP , Animaux , Humains , Facteur de transcription ATF-4/métabolisme , Facteur de transcription ATF-4/génétique , Bortézomib/pharmacologie , Lignée cellulaire tumorale , Facteur-2 d'initiation eucaryote/métabolisme , Facteur-2 d'initiation eucaryote/génétique , Myélome multiple/génétique , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , /génétique , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique
3.
Commun Biol ; 7(1): 1073, 2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39217250

RÉSUMÉ

The coordination between kinases and phosphatases is crucial for regulating the phosphorylation levels of essential signaling molecules. Methods enabling precise control of kinase activities are valuable for understanding the kinase functions and for developing targeted therapies. Here, we use the abscisic acid (ABA)-induced proximity system to reversibly control kinase signaling by recruiting phosphatases. Using this method, we found that the oncogenic tyrosine kinase BCR::ABL1 can be inhibited by recruiting various cytoplasmic phosphatases. We also discovered that the oncogenic serine/threonine kinase BRAF(V600E), which has been reported to bypass phosphorylation regulation, can be positively regulated by protein phosphatase 1 (PP1) and negatively regulated by PP5. Additionally, we observed that the dual-specificity kinase MEK1 can be inhibited by recruiting PP5. This suggests that bifunctional molecules capable of recruiting PP5 to MEK or RAF kinases could be promising anticancer drug candidates. Thus, the ABA-induced dephosphorylation method enables rapid screening of phosphatases to precisely control kinase signaling.


Sujet(s)
Acide abscissique , Transduction du signal , Phosphorylation , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Acide abscissique/métabolisme , Acide abscissique/pharmacologie , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes B-raf/génétique , MAP Kinase Kinase 1/métabolisme , Protéines de fusion bcr-abl/métabolisme , Protéines de fusion bcr-abl/génétique , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique
4.
CNS Neurosci Ther ; 30(7): e14830, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39046182

RÉSUMÉ

N6-methyladenosine (m6A) methylation is a vital epigenetic mechanism associated with drug addiction. However, the relationship between m6A modification and oxycodone rewarding is less well explored. Based on an open field test, the present study evaluated oxycodone rewarding using chromatin immunoprecipitation PCR, immunofluorescence, and RNA sequencing. A marked increase in METTL14 protein and a decrease in PP1α protein due to oxycodone abundance in the striatal neurons were observed in a dose- and time-dependent manner. Oxycodone markedly increased LSD1 expression, and decreased H3K4me1 expression in the striatum. In the open field test, intra-striatal injection of METTL14 siRNA, HOTAIR siRNA, or LSD1 shRNA blocked oxycodone-induced increase in locomotor activity. The downregulation of PP1α was also inhibited after treatment with METTL14/HOTAIR siRNA and LSD1 shRNA. Enhanced binding of LSD1 with CoRest and of CoRest with the PP1α gene induced by oxycodone was also reversed by LSD1 shRNA. In addition, H3K4me1 demethylation was also blocked by the treatment. In summary, the investigation confirmed that METTL14-mediated upregulation of HOTAIR resulted in the repression of PP1α, which in turn facilitated the recruitment of LSD1, thus catalyzing H3K4me1 demethylation and promoting oxycodone addiction.


Sujet(s)
Methyltransferases , Oxycodone , ARN long non codant , Animaux , Mâle , Souris , Corps strié/métabolisme , Corps strié/effets des médicaments et des substances chimiques , Déméthylation , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Histone/métabolisme , Lysine/analogues et dérivés , Methyltransferases/métabolisme , Methyltransferases/génétique , Souris de lignée C57BL , Oxycodone/pharmacologie , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , ARN long non codant/métabolisme , ARN long non codant/génétique , Régulation positive
5.
Ann Hematol ; 103(8): 2853-2863, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38842564

RÉSUMÉ

Acute Myeloid Leukemia (AML) is a life-threatening disease whose induction treatment consists of combination chemotherapy with Idarubicin and Cytarabine for fit patients. Treatment failures are frequent, urging the need for novel treatments for this disease. The DNA Damage Response Mechanism (DDR) comprises numerous molecules and pathways intended to arrest the cell cycle until DNA damage is repaired or else drive the cell to apoptosis. AML-derived cell lines after treatment with Idarubicin and Cytarabine were used for studying the expression profile of 84 DDR genes, through PCR arrays. Utilizing de novo AML patient and control samples we studied the expression of PPP1R15A, CDKN1A, GADD45A, GADD45G, and EXO1. Next, we performed PPP1R15A silencing in AML cell lines in two separate experiments using siRNA and CRISPR-cas9, respectively. Our findings highlight that DDR regulators demonstrate increased expression in patients with high cytogenetic risk possibly reflecting increased genotoxic stress. Especially, PPP1R15A is mainly involved in the recovery of the cells from stress and it was the only DDR gene upregulated in AML patients. The PPP1R15A silencing resulted in decreased viability of Idarubicin and Cytarabine-treated cell lines, in contrast to untreated cells. These findings shed light on new strategies to enhance chemotherapy efficacy and demonstrate that PPP1R15A is an important DDR regulator in AML and its downregulation might be a safe and effective way to increase sensitivity to chemotherapy in this disease.


Sujet(s)
Cytarabine , Altération de l'ADN , Extinction de l'expression des gènes , Leucémie aigüe myéloïde , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Cytarabine/pharmacologie , Lignée cellulaire tumorale , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Idarubicine/pharmacologie , Idarubicine/administration et posologie , Mâle , Femelle , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Réparation de l'ADN/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Adulte , Sujet âgé , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques
6.
J Med Case Rep ; 18(1): 269, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38835078

RÉSUMÉ

BACKGROUND: Acute myeloid leukemia (AML) is the second most common type of leukemia in children. Although prognostic and diagnostic tests of AML patients have improved, there is still a great demand for new reliable clinical biomarkers for AML. Read-through fusion transcripts (RTFTs) are complex transcripts of adjacent genes whose molecular mechanisms are poorly understood. This is the first report of the presence of the PPP1R1B::STARD3 fusion transcript in an AML patient. Here, we investigated the presence of PPP1R1B::STARD3 RTFT in a case of AML using paired-end RNA sequencing (RNA-seq). CASE PRESENTATION: A Persian 12-year-old male was admitted to Dr. Sheikh Hospital of Mashhad, Iran, in September 2019 with the following symptoms, including fever, convulsions, hemorrhage, and bone pain. The patient was diagnosed with AML (non-M3-FAB subtype) based on cell morphologies and immunophenotypical features. Chromosomal analysis using the G-banding technique revealed t (9;22) (q34;q13). CONCLUSIONS: Single-cell RNA sequencing (scRNA-seq) analysis suggested that the PPP1R1B promoter may be responsible for the PPP1R1B::STARD3 expression. Alterations in the level of lipid metabolites implicate cancer development, and this fusion can play a crucial role in the cholesterol movement in cancer cells. PPP1R1B::STARD3 may be considered a candidate for targeted therapies of the cholesterol metabolic and the PI3K/AKT signaling pathways involved in cancer development and progression.


Sujet(s)
Leucémie aigüe myéloïde , Humains , Mâle , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/diagnostic , Enfant , Protein Phosphatase 1/génétique , Protéines de fusion oncogènes/génétique
7.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38928151

RÉSUMÉ

Valosin-containing protein (VCP), an ATPase-associated protein, is emerging as a crucial regulator in cardiac pathologies. However, the pivotal role of VCP in the heart under physiological conditions remains undetermined. In this study, we tested a hypothesis that sufficient VCP expression is required for cardiac development and physiological cardiac function. Thus, we generated a cardiac-specific VCP knockout (KO) mouse model and assessed the consequences of VCP suppression on the heart through physiological and molecular studies at baseline. Our results reveal that homozygous KO mice are embryonically lethal, whereas heterozygous KO mice with a reduction in VCP by ~40% in the heart are viable at birth but progressively develop heart failure and succumb to mortality at the age of 10 to 12 months. The suppression of VCP induced a selective activation of the mammalian target of rapamycin complex 1 (mTORC1) but not mTORC2 at the early age of 12 weeks. The prolonged suppression of VCP increased the expression (by ~2 folds) and nuclear translocation (by >4 folds) of protein phosphatase 1 (PP1), a key mediator of protein dephosphorylation, accompanied by a remarked reduction (~80%) in AKTSer473 phosphorylation in VCP KO mouse hearts at a later age but not the early stage. These temporal molecular alterations were highly associated with the progressive decline in cardiac function. Overall, our findings shed light on the essential role of VCP in the heart under physiological conditions, providing new insights into molecular mechanisms in the development of heart failure.


Sujet(s)
Défaillance cardiaque , Complexe-2 cible mécanistique de la rapamycine , Souris knockout , Protein Phosphatase 1 , Protéine contenant la valosine , Animaux , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Protéine contenant la valosine/métabolisme , Protéine contenant la valosine/génétique , Souris , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/génétique , Myocarde/métabolisme , Myocarde/anatomopathologie , Mâle , Modèles animaux de maladie humaine
8.
Nat Commun ; 15(1): 5359, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918402

RÉSUMÉ

SDS22 forms an inactive complex with nascent protein phosphatase PP1 and Inhibitor-3. SDS22:PP1:Inhibitor-3 is a substrate for the ATPase p97/VCP, which liberates PP1 for binding to canonical regulatory subunits. The exact role of SDS22 in PP1-holoenzyme assembly remains elusive. Here, we show that SDS22 stabilizes nascent PP1. In the absence of SDS22, PP1 is gradually lost, resulting in substrate hyperphosphorylation and a proliferation arrest. Similarly, we identify a female individual with a severe neurodevelopmental disorder bearing an unstable SDS22 mutant, associated with decreased PP1 levels. We furthermore find that SDS22 directly binds to Inhibitor-3 and that this is essential for the stable assembly of SDS22:PP1: Inhibitor-3, the recruitment of p97/VCP, and the extraction of SDS22 during holoenzyme assembly. SDS22 with a disabled Inhibitor-3 binding site co-transfers with PP1 to canonical regulatory subunits, thereby forming non-functional holoenzymes. Our data show that SDS22, through simultaneous interaction with PP1 and Inhibitor-3, integrates the major steps of PP1 holoenzyme assembly.


Sujet(s)
Protein Phosphatase 1 , Femelle , Humains , Cellules HEK293 , Holoenzymes/métabolisme , Phosphorylation , Liaison aux protéines , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Protéine contenant la valosine/métabolisme , Protéine contenant la valosine/génétique
9.
Open Biol ; 14(5): 230460, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38806145

RÉSUMÉ

The precise spatial and temporal control of histone phosphorylations is important for the ordered progression through the different phases of mitosis. The phosphorylation of H2B at S6 (H2B S6ph), which is crucial for chromosome segregation, reaches its maximum level during metaphase and is limited to the inner centromere. We discovered that the temporal and spatial regulation of this modification, as well as its intensity, are governed by the scaffold protein RepoMan and its associated catalytically active phosphatases, PP1α and PP1γ. Phosphatase activity is inhibited at the area of maximal H2B S6 phosphorylation at the inner centromere by site-specific Aurora B-mediated inactivation of the PP1/RepoMan complex. The motor protein Mklp2 contributes to the relocalization of Aurora B from chromatin to the mitotic spindle during anaphase, thus alleviating Aurora B-dependent repression of the PP1/RepoMan complex and enabling dephosphorylation of H2B S6. Accordingly, dysregulation of Mklp2 levels, as commonly observed in tumour cells, leads to the lack of H2B S6 dephosphorylation during early anaphase, which might contribute to chromosomal instability.


Sujet(s)
Aurora kinase B , Protéines du cycle cellulaire , Histone , Mitose , Protein Phosphatase 1 , Aurora kinase B/métabolisme , Phosphorylation , Humains , Histone/métabolisme , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Cellules HeLa , Appareil du fuseau/métabolisme , Centromère/métabolisme , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique
10.
Carcinogenesis ; 45(9): 673-684, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-38715543

RÉSUMÉ

Esophageal cancer is one of the most common malignant tumors, and the 5-year overall survival rate is only 20%. Esophageal squamous cell carcinoma (ESCC) is the primary histological type of esophageal carcinoma in China. Protein phosphatase 1 regulatory subunit 18 (PPP1r18) is one of the actin-regulatory proteins and is able to bind to protein phosphatase 1 catalytic subunit alpha (PPP1CA). Yet, little is known about the role of PPP1r18 in ESCC. This study aimed to elucidate the biological functions of PPP1r18 in the ESCC progression. Clinical samples first confirmed that PPP1r18 expression was upregulated in ESCC, and PPP1r18 was correlated with tumor invasion depth, lymph node metastasis, distant metastasis and reduced overall survival. We then observed that PPP1r18 overexpression enhanced cell proliferation in vitro and in vivo. Mechanistically, PPP1r18 regulated tumor progression of ESCC through activating the calcineurin-mediated ERK pathway, rather than binding to PPP1CA. Collectively, our results suggest that PPP1r18 promotes ESCC progression by regulating the calcineurin-mediated ERK pathway. PPP1r18 might be a potential target for the diagnosis and treatment of ESCC.


Sujet(s)
Calcineurine , Prolifération cellulaire , Évolution de la maladie , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Régulation de l'expression des gènes tumoraux , Système de signalisation des MAP kinases , Humains , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/métabolisme , Calcineurine/métabolisme , Calcineurine/génétique , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/mortalité , Prolifération cellulaire/génétique , Animaux , Souris , Mâle , Femelle , Système de signalisation des MAP kinases/génétique , Adulte d'âge moyen , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Pronostic , Lignée cellulaire tumorale , Souris nude , Mouvement cellulaire/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Métastase lymphatique
11.
Nucleic Acids Res ; 52(12): 6866-6885, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38783162

RÉSUMÉ

The genomes of Leishmania and trypanosomes are organized into polycistronic transcription units flanked by a modified DNA base J involved in promoting RNA polymerase II (Pol II) termination. We recently characterized a Leishmania complex containing a J-binding protein, PP1 protein phosphatase 1, and PP1 regulatory protein (PNUTS) that controls transcription termination potentially via dephosphorylation of Pol II by PP1. While T. brucei contains eight PP1 isoforms, none purified with the PNUTS complex, complicating the analysis of PP1 function in termination. We now demonstrate that the PP1-binding motif of TbPNUTS is required for function in termination in vivo and that TbPP1-1 modulates Pol II termination in T. brucei and dephosphorylation of the large subunit of Pol II. PP1-1 knock-down results in increased cellular levels of phosphorylated RPB1 accompanied by readthrough transcription and aberrant transcription of the chromosome by Pol II, including Pol I transcribed loci that are typically silent, such as telomeric VSG expression sites involved in antigenic variation. These results provide important insights into the mechanism underlying Pol II transcription termination in primitive eukaryotes that rely on polycistronic transcription and maintain allelic exclusion of VSG genes.


Sujet(s)
Allèles , Protein Phosphatase 1 , Protéines de protozoaire , RNA polymerase II , Terminaison de la transcription , Trypanosoma brucei brucei , Glycoprotéines de surface variables du trypanosome , RNA polymerase II/métabolisme , RNA polymerase II/génétique , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Trypanosoma brucei brucei/génétique , Trypanosoma brucei brucei/enzymologie , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Glycoprotéines de surface variables du trypanosome/génétique , Glycoprotéines de surface variables du trypanosome/métabolisme , Phosphorylation , Transcription génétique
12.
Life Sci ; 345: 122608, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38574885

RÉSUMÉ

BACKGROUND AND AIMS: The protein phosphatase 1 regulatory inhibitor subunit 1A (PPP1R1A) has been linked with insulin secretion and diabetes mellitus. Yet, its full significance in pancreatic ß-cell function remains unclear. This study aims to elucidate the role of the PPP1R1A gene in ß-cell biology using human pancreatic islets and rat INS-1 (832/13) cells. RESULTS: Disruption of Ppp1r1a in INS-1 cells was associated with reduced insulin secretion and impaired glucose uptake; however, cell viability, ROS, apoptosis or proliferation were intact. A significant downregulation of crucial ß-cell function genes such as Ins1, Ins2, Pcsk1, Cpe, Pdx1, Mafa, Isl1, Glut2, Snap25, Vamp2, Syt5, Cacna1a, Cacna1d and Cacnb3, was observed upon Ppp1r1a disruption. Furthermore, silencing Pdx1 in INS-1 cells altered PPP1R1A expression, indicating that PPP1R1A is a target gene for PDX1. Treatment with rosiglitazone increased Ppp1r1a expression, while metformin and insulin showed no effect. RNA-seq analysis of human islets revealed high PPP1R1A expression, with α-cells showing the highest levels compared to other endocrine cells. Muscle tissues exhibited greater PPP1R1A expression than pancreatic islets, liver, or adipose tissues. Co-expression analysis revealed significant correlations between PPP1R1A and genes associated with insulin biosynthesis, exocytosis machinery, and intracellular calcium transport. Overexpression of PPP1R1A in human islets augmented insulin secretion and upregulated protein expression of Insulin, MAFA, PDX1, and GLUT1, while silencing of PPP1R1A reduced Insulin, MAFA, and GLUT1 protein levels. CONCLUSION: This study provides valuable insights into the role of PPP1R1A in regulating ß-cell function and glucose homeostasis. PPP1R1A presents a promising opportunity for future therapeutic interventions.


Sujet(s)
Cellules à insuline , Ilots pancréatiques , Protein Phosphatase 1 , Animaux , Humains , Rats , Canaux calciques/métabolisme , Lignée cellulaire , Glucose/métabolisme , Insuline/métabolisme , Sécrétion d'insuline/génétique , Cellules à insuline/métabolisme , Ilots pancréatiques/métabolisme , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme
13.
Antiviral Res ; 226: 105895, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38679165

RÉSUMÉ

Rift Valley fever virus (RVFV) is an arbovirus in the Phenuiviridae family identified initially by the large 'abortion storms' observed among ruminants; RVFV can also infect humans. In humans, there is a wide variation of clinical symptoms ranging from subclinical to mild febrile illness to hepatitis, retinitis, delayed-onset encephalitis, or even hemorrhagic fever. The RVFV is a tri-segmented negative-sense RNA virus consisting of S, M, and L segments. The L segment encodes the RNA-dependent RNA polymerase (RdRp), termed the L protein, which is responsible for both viral mRNA synthesis and genome replication. Phosphorylation of viral RdRps is known to regulate viral replication. This study shows that RVFV L protein is serine phosphorylated and identified Casein Kinase 1 alpha (CK1α) and protein phosphatase 1 alpha (PP1α) as L protein binding partners. Inhibition of CK1 and PP1 through small molecule inhibitor treatment, D4476 and 1E7-03, respectively, caused a change in the phosphorylated status of the L protein. Inhibition of PP1α resulted in increased L protein phosphorylation whereas inhibition of CK1α decreased L protein phosphorylation. It was also found that in RVFV infected cells, PP1α localized to the cytoplasmic compartment. Treatment of RVFV infected cells with CK1 inhibitors reduced virus production in both mammalian and mosquito cells. Lastly, inhibition of either CK1 or PP1 reduced viral genomic RNA levels. These data indicate that L protein is phosphorylated and that CK1 and PP1 play a crucial role in regulating the L protein phosphorylation cycle, which is critical to viral RNA production and viral replication.


Sujet(s)
Protein Phosphatase 1 , Virus de la fièvre de la vallée du Rift , Réplication virale , Virus de la fièvre de la vallée du Rift/physiologie , Virus de la fièvre de la vallée du Rift/génétique , Phosphorylation , Humains , Animaux , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Génome viral , Protéines virales/métabolisme , Protéines virales/génétique , Casein Kinase Ialpha/métabolisme , Casein Kinase Ialpha/génétique , Chlorocebus aethiops , Lignée cellulaire , RNA replicase/métabolisme , RNA replicase/génétique , Cellules Vero , ARN viral/génétique , ARN viral/métabolisme , Fièvre de la Vallée du Rift/virologie
14.
Cell Rep ; 43(4): 114069, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38602876

RÉSUMÉ

The integrated stress response (ISR) is a key cellular signaling pathway activated by environmental alterations that represses protein synthesis to restore homeostasis. To prevent sustained damage, the ISR is counteracted by the upregulation of growth arrest and DNA damage-inducible 34 (GADD34), a stress-induced regulatory subunit of protein phosphatase 1 that mediates translation reactivation and stress recovery. Here, we uncover a novel ISR regulatory mechanism that post-transcriptionally controls the stability of PPP1R15A mRNA encoding GADD34. We establish that the 3' untranslated region of PPP1R15A mRNA contains an active AU-rich element (ARE) recognized by proteins of the ZFP36 family, promoting its rapid decay under normal conditions and stabilization for efficient expression of GADD34 in response to stress. We identify the tight temporal control of PPP1R15A mRNA turnover as a component of the transient ISR memory, which sets the threshold for cellular responsiveness and mediates adaptation to repeated stress conditions.


Sujet(s)
Régions 3' non traduites , Protein Phosphatase 1 , Animaux , Humains , Souris , Régions 3' non traduites/génétique , Adaptation physiologique/génétique , Éléments riches en AU/génétique , Cellules HEK293 , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Stabilité de l'ARN/génétique , ARN messager/métabolisme , ARN messager/génétique , Stress physiologique/génétique , Tristétraproline/métabolisme , Tristétraproline/génétique
15.
Cell Death Differ ; 31(5): 683-696, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38589496

RÉSUMÉ

Protein phosphatase 1 catalytic subunit gamma (PPP1CC) promotes DNA repair and tumor development and progression, however, its underlying mechanisms remain unclear. This study investigated the molecular mechanism of PPP1CC's involvement in DNA repair and the potential clinical implications. High expression of PPP1CC was significantly correlated with radioresistance and poor prognosis in human nasopharyngeal carcinoma (NPC) patients. The mechanistic study revealed that PPP1CC bound to Ku70/Ku80 heterodimers and activated DNA-PKcs by promoting DNA-PK holoenzyme formation, which enhanced nonhomologous end junction (NHEJ) -mediated DNA repair and led to radioresistance. Importantly, BRCA1-BRCA2-containing complex subunit 3 (BRCC3) interacted with PPP1CC to enhance its stability by removing the K48-linked polyubiquitin chain at Lys234 to prevent PPP1CC degradation. Therefore, BRCC3 helped the overexpressed PPP1CC to maintain its high protein level, thereby sustaining the elevation of DNA repair capacity and radioresistance. Our study identified the molecular mechanism by which PPP1CC promotes NHEJ-mediated DNA repair and radioresistance, suggesting that the BRCC3-PPP1CC-Ku70 axis is a potential therapeutic target to improve the efficacy of radiotherapy.


Sujet(s)
Réparation de l'ADN par jonction d'extrémités , Cancer du nasopharynx , Tumeurs du rhinopharynx , Protein Phosphatase 1 , Radiotolérance , Animaux , Femelle , Humains , Mâle , Souris , Lignée cellulaire tumorale , Réparation de l'ADN , DNA-activated protein kinase/métabolisme , DNA-activated protein kinase/génétique , Autoantigène Ku/métabolisme , Autoantigène Ku/génétique , Souris nude , Cancer du nasopharynx/radiothérapie , Cancer du nasopharynx/anatomopathologie , Cancer du nasopharynx/génétique , Cancer du nasopharynx/métabolisme , Tumeurs du rhinopharynx/radiothérapie , Tumeurs du rhinopharynx/génétique , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs du rhinopharynx/métabolisme , Pronostic , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Radiotolérance/génétique
16.
Cells ; 13(8)2024 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-38667309

RÉSUMÉ

Variants of mitochondrial DNA (mtDNA) have been identified as risk factors for the development of Parkinson's disease (PD). However, the underlying pathogenetic mechanisms remain unclear. Cybrid models carrying various genotypes of mtDNA variants were tested for resistance to PD-simulating MPP+ treatment. The most resistant line was selected for transcriptome profiling, revealing specific genes potentially influencing the resistant characteristic. We then conducted protein validation and molecular biological studies to validate the related pathways as the influential factor. Cybrids carrying the W3 mtDNA haplogroup demonstrated the most resistance to the MPP+ treatment. In the transcriptome study, PPP1R15A was identified, while further study noted elevated expressions of the coding protein GADD34 across all cybrids. In the study of GADD34-related mitochondrial unfolding protein response (mtUPR), we found that canonical mtUPR, launched by the phosphate eIF2a, is involved in the resistant characteristic of specific mtDNA to MPP+ treatment. Our study suggests that a lower expression of GADD34 in the late phase of mtUPR may prolong the mtUPR process, thereby benefitting protein homeostasis and facilitating cellular resistance to PD development. We herein demonstrate that GADD34 plays an important role in PD development and should be further investigated as a target for the development of therapies for PD.


Sujet(s)
ADN mitochondrial , Haplotypes , Maladie de Parkinson , Maladie de Parkinson/génétique , Humains , ADN mitochondrial/génétique , Haplotypes/génétique , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Mitochondries/métabolisme , Mitochondries/génétique , Réponse aux protéines mal repliées/génétique
17.
FEBS J ; 291(12): 2615-2635, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38303113

RÉSUMÉ

Protein phosphatase-1 (PP1) complexed to nuclear inhibitor of PP1 (NIPP1) limits DNA repair through dephosphorylation of NIPP1-recruited substrates. However, the PP1:NIPP1 holoenzyme is completely inactive under basal conditions, hinting at a DNA damage-regulated activation mechanism. Here, we report that DNA damage caused the activation of PP1:NIPP1 after a time delay of several hours through phosphorylation of NIPP1 at the C-terminal tyrosine 335 (Y335) by a Src-family kinase. PP1:NIPP1 activation partially resulted from the dissociation of the C terminus of NIPP1 from the active site of PP1. In addition, the released Y335-phosphorylated C terminus interacted with the N terminus of NIPP1 to enhance substrate recruitment by the flanking forkhead-associated (FHA) domain. Constitutive activation of PP1:NIPP1 by knock-in of a phospho-mimicking (Y335E) NIPP1 mutant led to the hypo-phosphorylation of FHA ligands and an accumulation of DNA double-strand breaks. Our data indicate that PP1:NIPP1 activation through circularization of NIPP1 is a late response to DNA damage that contributes to the timely recovery from damage repair.


Sujet(s)
Altération de l'ADN , Protein Phosphatase 1 , src-Family kinases , Phosphorylation , Humains , Protein Phosphatase 1/métabolisme , Protein Phosphatase 1/génétique , Protein Phosphatase 1/composition chimique , src-Family kinases/métabolisme , src-Family kinases/génétique , src-Family kinases/composition chimique , Réparation de l'ADN , Régulation allostérique , Cassures double-brin de l'ADN , Cellules HEK293 , Liaison aux protéines , Protéines et peptides de signalisation intracellulaire
18.
Cell Death Dis ; 15(2): 115, 2024 02 07.
Article de Anglais | MEDLINE | ID: mdl-38326336

RÉSUMÉ

Gasdermin D (GSDMD) functions as a pivotal executor of pyroptosis, eliciting cytokine secretion following cleavage by inflammatory caspases. However, the role of posttranslational modifications (PTMs) in GSDMD-mediated pyroptosis remains largely unexplored. In this study, we demonstrate that GSDMD can undergo acetylation at the Lysine 248 residue, and this acetylation enhances pyroptosis. We identify histone deacetylase 4 (HDAC4) as the specific deacetylase responsible for mediating GSDMD deacetylation, leading to the inhibition of pyroptosis both in vitro and in vivo. Deacetylation of GSDMD impairs its ubiquitination, resulting in the inhibition of pyroptosis. Intriguingly, phosphorylation of HDAC4 emerges as a critical regulatory mechanism promoting its ability to deacetylate GSDMD and suppress GSDMD-mediated pyroptosis. Additionally, we implicate Protein phosphatase 1 (PP1) catalytic subunits (PP1α and PP1γ) in the dephosphorylation of HDAC4, thereby nullifying its deacetylase activity on GSDMD. This study reveals a complex regulatory network involving HDAC4, PP1, and GSDMD. These findings provide valuable insights into the interplay among acetylation, ubiquitination, and phosphorylation in the regulation of pyroptosis, offering potential targets for further investigation in the field of inflammatory cell death.


Sujet(s)
Gasdermines , Histone deacetylases , Protein Phosphatase 1 , Pyroptose , Histone deacetylases/génétique , Histone deacetylases/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines tumorales/métabolisme , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Maturation post-traductionnelle des protéines , Humains , Animaux , Souris , Gasdermines/métabolisme
19.
Methods Mol Biol ; 2740: 37-61, 2024.
Article de Anglais | MEDLINE | ID: mdl-38393468

RÉSUMÉ

The identification of protein phosphatase 1 (PP1) holoenzyme substrates has proven to be a challenging task. PP1 can form different holoenzyme complexes with a variety of regulatory subunits, and many of those are cell cycle regulated. Although several methods have been used to identify PP1 substrates, their cell cycle specificity is still an unmet need. Here, we present a new strategy to investigate PP1 substrates throughout the cell cycle using clustered regularly interspersed short palindromic repeats (CRISPR)-Cas9 genome editing and generate cell lines with endogenously tagged PP1 regulatory subunit (regulatory interactor of protein phosphatase one, RIPPO). RIPPOs are tagged with the auxin-inducible degron (AID) or ascorbate peroxidase 2 (APEX2) modules, and PP1 substrate identification is conducted by SILAC proteomic-based approaches. Proteins in close proximity to RIPPOs are first identified through mass spectrometry (MS) analyses using the APEX2 system; then a list of differentially phosphorylated proteins upon RIPPOs rapid degradation (achieved via the AID system) is compiled via SILAC phospho-mass spectrometry. The "in silico" overlap between the two proteomes will be enriched for PP1 putative substrates. Several methods including fluorescence resonance energy transfer (FRET), proximity ligation assays (PLA), and in vitro assays can be used as substrate validations approaches.


Sujet(s)
Protéomique , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme , Phosphorylation , Cycle cellulaire , Lignée cellulaire , Holoenzymes/composition chimique , Holoenzymes/métabolisme
20.
Mol Cancer ; 23(1): 34, 2024 02 15.
Article de Anglais | MEDLINE | ID: mdl-38360682

RÉSUMÉ

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney cancer with high aggressive phenotype and poor prognosis. Accumulating evidence suggests that circRNAs have been identified as pivotal mediators in cancers. However, the role of circRNAs in ccRCC progression remains elusive. METHODS: The differentially expressed circRNAs in 4 paired human ccRCC and adjacent noncancerous tissues ccRCC were screened using circRNA microarrays and the candidate target was selected based on circRNA expression level using weighted gene correlation network analysis (WGCNA) and the gene expression omnibus (GEO) database. CircPDHK1 expression in ccRCC and adjacent noncancerous tissues (n = 148) were evaluated along with clinically relevant information. RT-qPCR, RNase R digestion, and actinomycin D (ActD) stability test were conducted to identify the characteristics of circPDHK1. The subcellular distribution of circPDHK1 was analyzed by subcellular fractionation assay and fluorescence in situ hybridization (FISH). Immunoprecipitation-mass spectrometry (IP-MS) and immunofluorescence (IF) were employed to evaluate the protein-coding ability of circPDHK1. ccRCC cells were transfected with siRNAs, plasmids or lentivirus approach, and cell proliferation, migration and invasion, as well as tumorigenesis and metastasis in nude mice were assessed to clarify the functional roles of circPDHK1 and its encoded peptide PDHK1-241aa. RNA-sequencing, western blot analysis, immunoprecipitation (IP) and chromatin immunoprecipitation (ChIP) assays were further employed to identify the underlying mechanisms regulated by PDHK1-241aa. RESULTS: CircPDHK1 was upregulated in ccRCC tissues and closely related to WHO/ISUP stage, T stage, distant metastasis, VHL mutation and Ki-67 levels. CircPDHK1 had a functional internal ribosome entry site (IRES) and encoded a novel peptide PDHK1-241aa. Functionally, we confirmed that PDHK1-241aa and not the circPDHK1 promoted the proliferation, migration and invasion of ccRCC. Mechanistically, circPDHK1 was activated by HIF-2A at the transcriptional level. PDHK1-241aa was upregulated and interacted with PPP1CA, causing the relocation of PPP1CA to the nucleus. This thereby inhibited AKT dephosphorylation and activated the AKT-mTOR signaling pathway. CONCLUSIONS: Our data indicated that circPDHK1-encoded PDHK1-241aa promotes ccRCC progression by interacting with PPP1CA to inhibit AKT dephosphorylation. This study provides novel insights into the multiplicity of circRNAs and highlights the potential use of circPDHK1 or PDHK1-241aa as a therapeutic target for ccRCC.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Animaux , Souris , Humains , Néphrocarcinome/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , ARN circulaire/génétique , Souris nude , Hybridation fluorescente in situ , Lignée cellulaire tumorale , Transduction du signal/génétique , Tumeurs du rein/génétique , Sérine-thréonine kinases TOR/métabolisme , Prolifération cellulaire/génétique , Peptides/génétique , Régulation de l'expression des gènes tumoraux , Protein Phosphatase 1/génétique , Protein Phosphatase 1/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE