Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.899
Filtrer
1.
New Phytol ; 243(3): 997-1016, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38849319

RÉSUMÉ

Jasmonic acid (JA) and gibberellin (GA) coordinately regulate plant developmental programs and environmental cue responses. However, the fine regulatory network of the cross-interaction between JA and GA remains largely elusive. In this study, we demonstrate that MdNAC72 together with MdABI5 positively regulates anthocyanin biosynthesis through an exquisite MdNAC72-MdABI5-MdbHLH3 transcriptional cascade in apple. MdNAC72 interacts with MdABI5 to promote the transcriptional activation of MdABI5 on its target gene MdbHLH3 and directly activates the transcription of MdABI5. The MdNAC72-MdABI5 module regulates the integration of JA and GA signals in anthocyanin biosynthesis by combining with JA repressor MdJAZ2 and GA repressor MdRGL2a. MdJAZ2 disrupts the MdNAC72-MdABI5 interaction and attenuates the transcriptional activation of MdABI5 by MdNAC72. MdRGL2a sequesters MdJAZ2 from the MdJAZ2-MdNAC72 protein complex, leading to the release of MdNAC72. The E3 ubiquitin ligase MdSINA2 is responsive to JA and GA signals and promotes ubiquitination-dependent degradation of MdNAC72. The MdNAC72-MdABI5 interface fine-regulates the integration of JA and GA signals at the transcriptional and posttranslational levels by combining MdJAZ2, MdRGL2a, and MdSINA2. In summary, our findings elucidate the fine regulatory network connecting JA and GA signals with MdNAC72-MdABI5 as the core in apple.


Sujet(s)
Cyclopentanes , Régulation de l'expression des gènes végétaux , Gibbérellines , Malus , Oxylipines , Protéines végétales , Transduction du signal , Ubiquitination , Oxylipines/métabolisme , Malus/génétique , Malus/métabolisme , Cyclopentanes/métabolisme , Ubiquitination/effets des médicaments et des substances chimiques , Protéines végétales/métabolisme , Protéines végétales/génétique , Gibbérellines/métabolisme , Protéolyse/effets des médicaments et des substances chimiques , Anthocyanes/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Modèles biologiques
2.
Mol Brain ; 17(1): 32, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840222

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects the motor neuron. One aspect of the neuropathology involved in ALS includes increased genomic damage and impaired DNA repair capability. The TAR-DNA binding protein 43 (TDP43) has been associated with both sporadic and familial forms of ALS, and is typically observed as cytosolic mislocalization of protein aggregates, termed TDP43 proteinopathy. TDP43 is a ubiquitous RNA/DNA binding protein with functional implications in a wide range of disease processes, including the repair of DNA double-strand breaks (DSBs). While TDP43 is widely known to regulate RNA metabolism, our lab has reported it also functions directly at the protein level to facilitate DNA repair. Here, we show that the TDP43 protein interacts with DNA mismatch repair (MMR) proteins MLH1 and MSH6 in a DNA damage-inducible manner. We utilized differentiated SH-SY5Y neuronal cultures to identify this inducible relationship using complementary approaches of proximity ligation assay (PLA) and co-immunoprecipitation (CoIP) assay. We observed that signals of TDP43 interaction with MLH1 and MSH6 increased significantly following a 2 h treatment of 10 µM methylmethanesulfonate (MMS), a DNA alkylating agent used to induce MMR repair. Likewise, we observed this effect was abolished in cell lines treated with siRNA directed against TDP43. Finally, we demonstrated these protein interactions were significantly increased in lumbar spinal cord samples of ALS-affected patients compared to age-matched controls. These results will inform our future studies to understand the mechanisms and consequences of this TDP43-MMR interaction in the context of ALS-affected neurons.


Sujet(s)
Altération de l'ADN , Protéines de liaison à l'ADN , Protéine-1 homologue de MutL , Liaison aux protéines , Humains , Protéines de liaison à l'ADN/métabolisme , Protéine-1 homologue de MutL/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Neurones/métabolisme , Adulte d'âge moyen , Mâle
3.
Tissue Cell ; 88: 102404, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38759521

RÉSUMÉ

Follicular maturation arrest is a prevalent endocrine disorder characterized by hormonal imbalance, ovarian dysfunction, and metabolic disturbances leading to Polycystic ovarian syndrome (PCOS). Tanshinone IIA (TIIA), a bioactive compound derived from Salvia miltiorrhiza, has shown promising therapeutic potential in various diseases, including cardiovascular diseases and cancer. However, its effects on reproductive health and gynecological disorders, particularly PCOS, remain poorly understood. In this study, we investigated the potential therapeutic effects of TIIA on ovarian function. Using a combination of experimental and computational approaches, we elucidated the molecular mechanisms underlying TIIA's pharmacological impact on ovarian function, follicular development, and androgen receptor signaling. Molecular docking and dynamics simulations revealed that TIIA interacts with the human androgen receptor (HAR), modulating its activity and downstream signaling pathways. Our results demonstrate that TIIA treatment alleviates PCOS-like symptoms in a zebrafish model, including improved follicular development, lowered GSI index, improved antioxidant status (SOD, CAT), decreased LDH levels, and enhanced AChE levels by regulating Tox3 and Dennd1a pathway. Our findings suggest that TIIA may hold promise as a novel therapeutic agent for the management of PCOS or ovulation induction.


Sujet(s)
Abiétanes , Follicule ovarique , Syndrome des ovaires polykystiques , Récepteurs aux androgènes , Salvia miltiorrhiza , Danio zébré , Animaux , Humains , Abiétanes/pharmacologie , Récepteurs aux androgènes/métabolisme , Salvia miltiorrhiza/composition chimique , Syndrome des ovaires polykystiques/traitement médicamenteux , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/anatomopathologie , Follicule ovarique/effets des médicaments et des substances chimiques , Follicule ovarique/métabolisme , Follicule ovarique/anatomopathologie , Femelle , Simulation de docking moléculaire , Protéines de poisson-zèbre/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques
4.
Ann Clin Lab Sci ; 54(2): 137-148, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38802154

RÉSUMÉ

OBJECTIVE: We have previously shown that the anti-cancer peptide PNC-27 kills cancer cells by co-localizing with membrane-expressed HDM-2, resulting in transmembrane pore formation causing extrusion of intracellular contents. We have also observed cancer cell mitochondrial disruption in PNC-27-treated cancer cells. Our objectives are to determine: 1. if PNC-27 binds to the p53 binding site of HDM-2 (residues 1-109) in the cancer cell membrane and 2. if this peptide causes selective disruption of cancer cell mitochondria. METHODS: For aim 1, we incubated MIA-PaCa-2 human pancreatic carcinoma cells with PNC-27 in the presence of a monoclonal antibody against the amino terminal p53 binding site of HDM-2 to determine if it, but not negative control immune serum, blocks PNC-27-induced tumor cell necrosis. For the second aim, we incubated these cells with PNC-27 in the presence of two specific dyes that highlight normal organelle function: mitotracker for mitochondria and lysotracker for lysosomes. We also performed immuno-electron microscopy (IEM) with gold-labeled anti-PNC-27 antibody on the mitochondria of these cells treated with PNC-27. RESULTS: Monoclonal antibody to the p53 binding site of HDM-2 blocks PNC-27-induced cancer cell necrosis, whereas negative control immune serum does not. The mitochondria of PNC-27-treated cancer cells fail to retain mitotracker dye while their lysosomes retain lysotracker dye. IEM of the mitochondria cancer cells reveals gold particles present on the mitochondrial membranes. CONCLUSIONS: PNC-27 binds to the p53 binding site of HDM-2 (residues 1-109) inducing transmembrane pore formation and cancer cell necrosis. Furthermore, this peptide enters cancer cells and binds to the membranes of mitochondria, resulting in their disruption.


Sujet(s)
Membrane cellulaire , Membranes mitochondriales , Protéines proto-oncogènes c-mdm2 , Humains , Membrane cellulaire/métabolisme , Membrane cellulaire/effets des médicaments et des substances chimiques , Membranes mitochondriales/métabolisme , Membranes mitochondriales/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-mdm2/métabolisme , Lignée cellulaire tumorale , Protéine p53 suppresseur de tumeur/métabolisme , Antinéoplasiques/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/anatomopathologie , Fragments peptidiques/métabolisme , Fragments peptidiques/pharmacologie , Liaison aux protéines/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Peptides/métabolisme , Nécrose
5.
PLoS One ; 19(5): e0303213, 2024.
Article de Anglais | MEDLINE | ID: mdl-38753710

RÉSUMÉ

Ischemic stroke causes a lack of oxygen and glucose supply to brain, eventually leads to severe neurological disorders. Retinoic acid is a major metabolic product of vitamin A and has various biological effects. The PI3K-Akt signaling pathway is an important survival pathway in brain. Phosphorylated Akt is important in regulating survival and apoptosis. We examined whether retinoic acid has neuroprotective effects in stroke model by regulating Akt and its downstream protein, Bad. Moreover, we investigated the relationship between retinoic acid and Bcl-2 family protein interactions. Animals were intraperitoneally administered vehicle or retinoic acid (5 mg/kg) for four days before surgery and ischemic stroke was induced by middle cerebral artery occlusion (MCAO) surgery. Neurobehavioral tests were performed 24 h after MCAO and cerebral cortical tissues were collected. Cresyl violet staining and TUNEL histochemistry were performed, Western blot and immunoprecipitation analysis were performed to elucidate the expression of various proteins. Retinoic acid reduced neurological deficits and histopathological changes, decreased the number of TUNEL-positive cells, and alleviated reduction of phospho-PDK1, phospho-Akt, and phospho-Bad expression caused by MCAO damage. Immunoprecipitation analysis showed that MCAO damage reduced the interaction between phospho-Bad and 14-3-3, which was attenuated by retinoic acid. Furthermore, retinoic acid mitigated the increase in Bcl-2/Bad and Bcl-xL/Bad binding levels and the reduction in Bcl-2/Bax and Bcl-xL/Bax binding levels caused by MCAO damage. Retinoic acid alleviated MCAO-induced increase of caspase-3 and cleaved caspase-3 expression. We demonstrate that retinoic acid prevented apoptosis against cerebral ischemia through phosphorylation of Akt and Bad, maintenance of phospho-Bad and 14-3-3 binding, and regulation of Bcl-2 family protein interactions. .


Sujet(s)
Protéines proto-oncogènes c-akt , Protéines proto-oncogènes c-bcl-2 , Trétinoïne , Protéine Bad , Animaux , Mâle , Rats , Apoptose/effets des médicaments et des substances chimiques , Protéine Bad/métabolisme , Modèles animaux de maladie humaine , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/anatomopathologie , Neuroprotecteurs/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-bcl-2/métabolisme , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Trétinoïne/pharmacologie
6.
New Phytol ; 243(1): 195-212, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38708439

RÉSUMÉ

Water plays crucial roles in expeditious growth and osmotic stress of bamboo. Nevertheless, the molecular mechanism of water transport remains unclear. In this study, an aquaporin gene, PeTIP4-3, was identified through a joint analysis of root pressure and transcriptomic data in moso bamboo (Phyllostachys edulis). PeTIP4-3 was highly expressed in shoots, especially in the vascular bundle sheath cells. Overexpression of PeTIP4-3 could increase drought and salt tolerance in transgenic yeast and rice. A co-expression pattern of PeSAPK4, PeMYB99 and PeTIP4-3 was revealed by WGCNA. PeMYB99 exhibited an ability to independently bind to and activate PeTIP4-3, which augmented tolerance to drought and salt stress. PeSAPK4 could interact with and phosphorylate PeMYB99 in vivo and in vitro, wherein they synergistically accelerated PeTIP4-3 transcription. Overexpression of PeMYB99 and PeSAPK4 also conferred drought and salt tolerance in transgenic rice. Further ABA treatment analysis indicated that PeSAPK4 enhanced water transport in response to stress via ABA signaling. Collectively, an ABA-mediated cascade of PeSAPK4-PeMYB99-PeTIP4-3 is proposed, which governs water transport in moso bamboo.


Sujet(s)
Aquaporines , Sécheresses , Régulation de l'expression des gènes végétaux , Oryza , Protéines végétales , Végétaux génétiquement modifiés , Eau , Protéines végétales/métabolisme , Protéines végétales/génétique , Eau/métabolisme , Oryza/génétique , Oryza/métabolisme , Oryza/physiologie , Aquaporines/métabolisme , Aquaporines/génétique , Transport biologique , Poaceae/génétique , Poaceae/physiologie , Modèles biologiques , Acide abscissique/métabolisme , Acide abscissique/pharmacologie , Tolérance au sel/génétique , Phosphorylation , Liaison aux protéines/effets des médicaments et des substances chimiques , Stress physiologique
7.
New Phytol ; 243(1): 362-380, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38730437

RÉSUMÉ

Plants typically activate distinct defense pathways against various pathogens. Heightened resistance to one pathogen often coincides with increased susceptibility to another pathogen. However, the underlying molecular basis of this antagonistic response remains unclear. Here, we demonstrate that mutants defective in the transcription factor ETHYLENE-INSENSITIVE 3-LIKE 2 (OsEIL2) exhibited enhanced resistance to the biotrophic bacterial pathogen Xanthomonas oryzae pv oryzae and to the hemibiotrophic fungal pathogen Magnaporthe oryzae, but enhanced susceptibility to the necrotrophic fungal pathogen Rhizoctonia solani. Furthermore, necrotroph-induced OsEIL2 binds to the promoter of OsWRKY67 with high affinity, leading to the upregulation of salicylic acid (SA)/jasmonic acid (JA) pathway genes and increased SA/JA levels, ultimately resulting in enhanced resistance. However, biotroph- and hemibiotroph-induced OsEIL2 targets OsERF083, resulting in the inhibition of SA/JA pathway genes and decreased SA/JA levels, ultimately leading to reduced resistance. Our findings unveil a previously uncharacterized defense mechanism wherein two distinct transcriptional regulatory modules differentially mediate immunity against pathogens with different lifestyles through the transcriptional reprogramming of phytohormone pathway genes.


Sujet(s)
Cyclopentanes , Régulation de l'expression des gènes végétaux , Oryza , Oxylipines , Maladies des plantes , Immunité des plantes , Protéines végétales , Rhizoctonia , Acide salicylique , Xanthomonas , Oxylipines/métabolisme , Acide salicylique/métabolisme , Cyclopentanes/métabolisme , Oryza/microbiologie , Oryza/génétique , Oryza/immunologie , Maladies des plantes/microbiologie , Maladies des plantes/immunologie , Xanthomonas/physiologie , Protéines végétales/métabolisme , Protéines végétales/génétique , Rhizoctonia/physiologie , Immunité des plantes/effets des médicaments et des substances chimiques , Mutation/génétique , Résistance à la maladie/génétique , Régions promotrices (génétique)/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques
8.
J Transl Med ; 22(1): 450, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38741146

RÉSUMÉ

BACKGROUND: Estetrol (E4) is a natural estrogen produced by the fetal liver during pregnancy. Due to its favorable safety profile, E4 was recently approved as estrogenic component of a new combined oral contraceptive. E4 is a selective ligand of estrogen receptor (ER)α and ERß, but its binding to the G Protein-Coupled Estrogen Receptor (GPER) has not been described to date. Therefore, we aimed to explore E4 action in GPER-positive Triple-Negative Breast Cancer (TNBC) cells. METHODS: The potential interaction between E4 and GPER was investigated by molecular modeling and binding assays. The whole transcriptomic modulation triggered by E4 in TNBC cells via GPER was explored through high-throughput RNA sequencing analyses. Gene and protein expression evaluations as well as migration and invasion assays allowed us to explore the involvement of the GPER-mediated induction of the plasminogen activator inhibitor type 2 (SERPINB2) in the biological responses triggered by E4 in TNBC cells. Furthermore, bioinformatics analysis was aimed at recognizing the biological significance of SERPINB2 in ER-negative breast cancer patients. RESULTS: After the molecular characterization of the E4 binding capacity to GPER, RNA-seq analysis revealed that the plasminogen activator inhibitor type 2 (SERPINB2) is one of the most up-regulated genes by E4 in a GPER-dependent manner. Worthy, we demonstrated that the GPER-mediated increase of SERPINB2 is engaged in the anti-migratory and anti-invasive effects elicited by E4 in TNBC cells. In accordance with these findings, a correlation between SERPINB2 levels and a good clinical outcome was found in ER-negative breast cancer patients. CONCLUSIONS: Overall, our results provide new insights into the mechanisms through which E4 can halt migratory and invasive features of TNBC cells.


Sujet(s)
Mouvement cellulaire , Oestétrol , Régulation de l'expression des gènes tumoraux , Inhibiteur-2 d'activateur du plasminogène , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Transduction du signal , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteurs couplés aux protéines G/métabolisme , Récepteurs des oestrogènes/métabolisme , Oestétrol/pharmacologie , Oestétrol/métabolisme , Femelle , Inhibiteur-2 d'activateur du plasminogène/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Invasion tumorale
9.
Int J Neuropsychopharmacol ; 27(5)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38695786

RÉSUMÉ

BACKGROUND: Major depressive disorder (MDD) is commonly treated with selective serotonin reuptake inhibitors (SSRIs). SSRIs inhibit the serotonin transporter (5-HTT), but the downstream antidepressant mechanism of action of these drugs is poorly understood. The serotonin 1B (5-HT1B) receptor is functionally linked to 5-HTT and 5-HT1B receptor binding and 5-HT1B receptor mRNA is reduced in the raphe nuclei after SSRI administration in primates and rodents, respectively. The effect of SSRI treatment on 5-HT1B receptor binding in patients with MDD has not been examined previously. This positron emission tomography (PET) study aimed to quantify brain 5-HT1B receptor binding changes in vivo after SSRI treatment for MDD in relation to treatment effect. METHODS: Eight unmedicated patients with moderate to severe MDD underwent PET with the 5-HT1B receptor radioligand [11C]AZ10419369 before and after 3 to 4 weeks of treatment with the SSRI escitalopram 10 mg daily. Depression severity was assessed at time of PET and after 6 to 7 weeks of treatment with the Montgomery-Åsberg Depression Rating Scale. RESULTS: We observed a significant reduction in [11C]AZ10419369 binding in a dorsal brainstem (DBS) region containing the median and dorsal raphe nuclei after escitalopram treatment (P = .036). Change in DBS [11C]AZ10419369 binding correlated with Montgomery-Åsberg Depression Rating Scale reduction after 3-4 (r = 0.78, P = .021) and 6-7 (r = 0.94, P < .001) weeks' treatment. CONCLUSIONS: Our findings align with the previously reported reduction of 5-HT1B receptor binding in the raphe nuclei after SSRI administration and support future studies testing change in DBS 5-HT1B receptor binding as an SSRI treatment response marker.


Sujet(s)
Trouble dépressif majeur , Escitalopram , Tomographie par émission de positons , Récepteur de la sérotonine de type 5-HT1B , Inbiteurs sélectifs de la recapture de la sérotonine , Récepteur de la sérotonine de type 5-HT1B/métabolisme , Mâle , Humains , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/métabolisme , Trouble dépressif majeur/imagerie diagnostique , Adulte , Femelle , Adulte d'âge moyen , Inbiteurs sélectifs de la recapture de la sérotonine/pharmacologie , Escitalopram/pharmacologie , Escitalopram/métabolisme , Encéphale/métabolisme , Encéphale/imagerie diagnostique , Encéphale/effets des médicaments et des substances chimiques , Résultat thérapeutique , Pipérazines/pharmacologie , Liaison aux protéines/effets des médicaments et des substances chimiques , Jeune adulte , Citalopram/pharmacologie , Benzopyranes , Morpholines
10.
Cell Commun Signal ; 22(1): 261, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38715108

RÉSUMÉ

BACKGROUND: Interleukin-6 (IL-6) is a multifunctional cytokine that controls the immune response, and its role has been described in the development of autoimmune diseases. Signaling via its cognate IL-6 receptor (IL-6R) complex is critical in tumor progression and, therefore, IL-6R represents an important therapeutic target. METHODS: An albumin-binding domain-derived highly complex combinatorial library was used to select IL-6R alpha (IL-6Rα)-targeted small protein binders using ribosome display. Large-scale screening of bacterial lysates of individual clones was performed using ELISA, and their IL-6Rα blocking potential was verified by competition ELISA. The binding of proteins to cells was monitored by flow cytometry and confocal microscopy on HEK293T-transfected cells, and inhibition of signaling function was examined using HEK-Blue IL-6 reporter cells. Protein binding kinetics to living cells was measured by LigandTracer, cell proliferation and toxicity by iCELLigence and Incucyte, cell migration by the scratch wound healing assay, and prediction of binding poses using molecular modeling by docking. RESULTS: We demonstrated a collection of protein variants called NEF ligands, selected from an albumin-binding domain scaffold-derived combinatorial library, and showed their binding specificity to human IL-6Rα and antagonistic effect in HEK-Blue IL-6 reporter cells. The three most promising NEF108, NEF163, and NEF172 variants inhibited cell proliferation of malignant melanoma (G361 and A2058) and pancreatic (PaTu and MiaPaCa) cancer cells, and suppressed migration of malignant melanoma (A2058), pancreatic carcinoma (PaTu), and glioblastoma (GAMG) cells in vitro. The NEF binders also recognized maturation-induced IL-6Rα expression and interfered with IL-6-induced differentiation in primary human B cells. CONCLUSION: We report on the generation of small protein blockers of human IL-6Rα using directed evolution. NEF proteins represent a promising class of non-toxic anti-tumor agents with migrastatic potential.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Récepteurs à l'interleukine-6 , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Récepteurs à l'interleukine-6/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules HEK293 , Lignée cellulaire tumorale , Liaison aux protéines/effets des médicaments et des substances chimiques
11.
New Phytol ; 242(6): 2524-2540, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38641854

RÉSUMÉ

Leaf senescence is a complex process strictly regulated by various external and endogenous factors. However, the key signaling pathway mediating leaf senescence remains unknown. Here, we show that Arabidopsis SPX1/2 negatively regulate leaf senescence genetically downstream of the strigolactone (SL) pathway. We demonstrate that the SL receptor AtD14 and MAX2 mediate the age-dependent degradation of SPX1/2. Intriguingly, we uncover an age-dependent accumulation of SLs in leaves via transcriptional activation of SL biosynthetic genes by the transcription factors (TFs) SPL9/15. Furthermore, we reveal that SPX1/2 interact with the WRKY75 subclade TFs to inhibit their DNA-binding ability and thus repress transcriptional activation of salicylic acid (SA) biosynthetic gene SA Induction-Deficient 2, gating the age-dependent SA accumulation in leaves at the leaf senescence onset stage. Collectively, our new findings reveal a signaling pathway mediating sequential activation of SL and salicylate biosynthesis for the onset of leaf senescence in Arabidopsis.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Régulation de l'expression des gènes végétaux , Lactones , Feuilles de plante , Sénescence des plantes , Facteurs de transcription , Arabidopsis/génétique , Arabidopsis/métabolisme , Arabidopsis/effets des médicaments et des substances chimiques , Feuilles de plante/métabolisme , Feuilles de plante/effets des médicaments et des substances chimiques , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Régulation de l'expression des gènes végétaux/effets des médicaments et des substances chimiques , Lactones/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Acide salicylique/métabolisme , Salicylates/métabolisme , Transduction du signal , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Voies de biosynthèse/effets des médicaments et des substances chimiques , Voies de biosynthèse/génétique
12.
SLAS Discov ; 29(3): 100154, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38521503

RÉSUMÉ

Sorafenib is a multikinase inhibitor indicated for first-line treatment of unresectable hepatocellular carcinoma. Despite its widespread use in the clinic, the existing knowledge of sorafenib mode-of-action remains incomplete. To build upon the current understanding, we used the Cellular Thermal Shift Assay (CETSA) coupled to Mass Spectrometry (CETSA-MS) to monitor compound binding to its target proteins in the cellular context on a proteome-wide scale. Among the potential sorafenib targets, we identified aldehyde dehydrogenase 2 (ALDH2), an enzyme that plays a major role in alcohol metabolism. We validated the interaction of sorafenib with ALDH2 by orthogonal methods using pure recombinant protein, proving that this interaction is not mediated by other cellular components. Moreover, we showed that sorafenib inhibits ALDH2 activity, supporting a functional role for this interaction. Finally, we were able to demonstrate that both ALDH2 protein expression and activity were reduced in sorafenib-resistant cells compared to the parental cell line. Overall, our study allowed the identification of ALDH2 as a novel sorafenib target and sheds light on its potential role in both hepatocellular carcinoma and sorafenib resistance condition.


Sujet(s)
Aldehyde dehydrogenase, mitochondrial , Carcinome hépatocellulaire , Tumeurs du foie , Protéome , Sorafénib , Sorafénib/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Aldehyde dehydrogenase, mitochondrial/génétique , Aldehyde dehydrogenase, mitochondrial/métabolisme , Lignée cellulaire tumorale , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques
13.
Plant Physiol ; 195(2): 1382-1400, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38345866

RÉSUMÉ

Brassinosteroids (BRs) are phytohormones that regulate stomatal development. In this study, we report that BR represses stomatal development in etiolated Arabidopsis (Arabidopsis thaliana) cotyledons via transcription factors BRASSINAZOLE RESISTANT 1 (BZR1) and bri1-EMS SUPPRESSOR1 (BES1), which directly target MITOGEN-ACTIVATED PROTEIN KINASE KINASE 9 (MKK9) and FAMA, 2 important genes for stomatal development. BZR1/BES1 bind MKK9 and FAMA promoters in vitro and in vivo, and mutation of the BZR1/BES1 binding motif in MKK9/FAMA promoters abolishes their transcription regulation by BZR1/BES1 in plants. Expression of a constitutively active MKK9 (MKK9DD) suppressed overproduction of stomata induced by BR deficiency, while expression of a constitutively inactive MKK9 (MKK9KR) induced high-density stomata in bzr1-1D. In addition, bzr-h, a sextuple mutant of the BZR1 family of proteins, produced overabundant stomata, and the dominant bzr1-1D and bes1-D mutants effectively suppressed the stomata-overproducing phenotype of brassinosteroid insensitive 1-116 (bri1-116) and brassinosteroid insensitive 2-1 (bin2-1). In conclusion, our results revealed important roles of BZR1/BES1 in stomatal development, and their transcriptional regulation of MKK9 and FAMA expression may contribute to BR-regulated stomatal development in etiolated Arabidopsis cotyledons.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Brassinostéroïdes , Cotylédon , Protéines de liaison à l'ADN , Régulation de l'expression des gènes végétaux , Protéines nucléaires , Stomates de plante , Arabidopsis/génétique , Arabidopsis/croissance et développement , Arabidopsis/métabolisme , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Brassinostéroïdes/métabolisme , Stomates de plante/croissance et développement , Stomates de plante/génétique , Stomates de plante/effets des médicaments et des substances chimiques , Cotylédon/génétique , Cotylédon/croissance et développement , Cotylédon/métabolisme , Cotylédon/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes végétaux/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Mutation/génétique , Régions promotrices (génétique)/génétique , Étiolement , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Liaison aux protéines/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase Kinases/métabolisme , Mitogen-Activated Protein Kinase Kinases/génétique
14.
Life Sci ; 336: 122283, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37993094

RÉSUMÉ

Chronic temporomandibular joint (TMJ) pain profoundly affects patients' quality of life. Trigeminal tumor necrosis factor-α (TNFα) plays a pivotal role in mediating TMJ pain in mice, yet the underlying epigenetic mechanisms remain enigmatic. To unravel these epigenetic intricacies, we employed a multifaceted approach. Hydroxymethylated DNA immunoprecipitation (hMeDIP) and chromatin immunoprecipitation (ChIP) followed by qPCR were employed to investigate the demethylation of TNFα gene (Tnfa) and its regulation by ten-eleven translocation methylcytosine dioxygenase 1 (TET1) in a chronic TMJ pain mouse model. The global levels of 5-hydroxymethylcytosine (5hmc) and percentage of 5hmc at the Tnfa promoter region were measured in the trigeminal ganglia (TG) and spinal trigeminal nucleus caudalis (Sp5C) following complete Freund's adjuvant (CFA) or saline treatment. TET1 knockdown and pain behavioral testing were conducted to ascertain the role of TET1-mediated epigenetic regulation of TNFα in the pathogenesis of chronic TMJ pain. Our finding revealed an increase in 5hmc at the Tnfa promoter region in both TG and Sp5C of CFA-treated mice. TET1 was upregulated in the mouse TG, and the ChIP result showed TET1 direct binding to the Tnfa promoter, with higher efficiency in the CFA-treated group. Immunofluorescence revealed the predominant expression of TET1 in trigeminal neurons. TET1 knockdown in the TG significantly reversed CFA-induced TNFα upregulation and alleviated chronic TMJ pain. In conclusion, our study implicates TET1 as a vital epigenetic regulator contributing to chronic inflammatory TMJ pain via trigeminal TNFα signaling. Targeting TET1 holds promise for epigenetic interventions in TMJ pain management.


Sujet(s)
Arthralgie , Protéines de liaison à l'ADN , Articulation temporomandibulaire , Ganglion trigéminal , Facteur de nécrose tumorale alpha , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Épigenèse génétique/génétique , Protéines de liaison à l'ADN/métabolisme , Ganglion trigéminal/physiopathologie , Arthralgie/induit chimiquement , Arthralgie/physiopathologie , Articulation temporomandibulaire/physiopathologie , Mâle , Animaux , Souris , Souris de lignée C57BL , Adjuvant Freund/pharmacologie , Régulation positive/effets des médicaments et des substances chimiques , Neurones/métabolisme , Techniques de knock-down de gènes , Régions promotrices (génétique) , Liaison aux protéines/effets des médicaments et des substances chimiques
15.
J Biol Chem ; 300(2): 105608, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38159849

RÉSUMÉ

Phototransduction in retinal rods occurs when the G protein-coupled photoreceptor rhodopsin triggers the activation of phosphodiesterase 6 (PDE6) by GTP-bound alpha subunits of the G protein transducin (GαT). Recently, we presented a cryo-EM structure for a complex between two GTP-bound recombinant GαT subunits and native PDE6, that included a bivalent antibody bound to the C-terminal ends of GαT and the inhibitor vardenafil occupying the active sites on the PDEα and PDEß subunits. We proposed GαT-activated PDE6 by inducing a striking reorientation of the PDEγ subunits away from the catalytic sites. However, questions remained including whether in the absence of the antibody GαT binds to PDE6 in a similar manner as observed when the antibody is present, does GαT activate PDE6 by enabling the substrate cGMP to access the catalytic sites, and how does the lipid membrane enhance PDE6 activation? Here, we demonstrate that 2:1 GαT-PDE6 complexes form with either recombinant or retinal GαT in the absence of the GαT antibody. We show that GαT binding is not necessary for cGMP nor competitive inhibitors to access the active sites; instead, occupancy of the substrate binding sites enables GαT to bind and reposition the PDE6γ subunits to promote catalytic activity. Moreover, we demonstrate by reconstituting GαT-stimulated PDE6 activity in lipid bilayer nanodiscs that the membrane-induced enhancement results from an increase in the apparent binding affinity of GαT for PDE6. These findings provide new insights into how the retinal G protein stimulates rapid catalytic turnover by PDE6 required for dim light vision.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 6 , Modèles moléculaires , Transducine , Cyclic Nucleotide Phosphodiesterases, Type 6/composition chimique , Cyclic Nucleotide Phosphodiesterases, Type 6/métabolisme , Guanosine triphosphate/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/enzymologie , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Transducine/composition chimique , Transducine/génétique , Transducine/métabolisme , Animaux , Bovins , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Structure quaternaire des protéines , Liaison aux protéines/effets des médicaments et des substances chimiques , Domaine catalytique , Xanthine(isobutyl-3 methyl-1)/pharmacologie , Double couche lipidique/métabolisme , Activation enzymatique
16.
Cell Oncol (Dordr) ; 47(3): 967-985, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38112979

RÉSUMÉ

PURPOSE: Nuclear accumulation of YAP/TAZ promotes tumorigenesis in several cancers, including melanoma. Although the mechanisms underlying the nuclear retention of YAP are known, those underlying the retention of TAZ remain unclear. Our study investigates a novel acetylation/deacetylation switch in TAZ, governing its subcellular localization in melanoma tumorigenesis. METHODS: Immunoprecipitation/Western blot assessed TAZ protein interactions and acetylation. SIRT5 activity was quantified with enzyme-linked immunosorbent assay. Immunofluorescence indicated TAZ nuclear localization. TEAD transcriptional activity was measured through luciferase reporter assays. ChIP detected TAZ binding to the CTGF promoter. Transwell and wound healing assays quantified melanoma cell invasiveness and migration. Metastasis was evaluated using a mouse model via tail vein injections. Clinical relevance was explored via immunohistochemical staining of patient tumors. RESULTS: CBP facilitated TAZ acetylation at K54 in response to epidermal growth factor stimulation, while SIRT5 mediated deacetylation. Acetylation correlated with phosphorylation, regulating TAZ's binding with LATS2 or TEAD. TAZ K54 acetylation enhanced its S89 phosphorylation, promoting cytosolic retention via LATS2 interaction. SIRT5-mediated deacetylation enhanced TAZ-TEAD interaction and nuclear retention. Chromatin IP showed SIRT5-deacetylated TAZ recruited to CTGF promoter, boosting transcriptional activity. In a mouse model, SIRT5 overexpression induced melanoma metastasis to lung tissue following the injection of B16F10 melanocytes via the tail vein, and this effect was prevented by verteporfin treatment. CONCLUSIONS: Our study revealed a novel mechanism of TAZ nuclear retention regulated by SIRT5-mediated K54 deacetylation and demonstrated the significance of TAZ deacetylation in CTGF expression. This study highlights the potential implications of the SIRT5/TAZ axis for treating metastatic melanoma.


Sujet(s)
Facteur de croissance du tissu conjonctif , Mélanome , Sirtuines , Acétylation/effets des médicaments et des substances chimiques , Animaux , Humains , Mélanome/anatomopathologie , Mélanome/métabolisme , Mélanome/génétique , Lignée cellulaire tumorale , Facteur de croissance du tissu conjonctif/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Sirtuines/métabolisme , Sirtuines/génétique , Souris , Facteurs de transcription/métabolisme , Transcriptional coactivator with PDZ-binding motif proteins , Régions promotrices (génétique)/génétique , Phosphorylation/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Transactivateurs/métabolisme , Noyau de la cellule/métabolisme , Carcinogenèse/anatomopathologie , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques
18.
J Biol Chem ; 299(12): 105382, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37866628

RÉSUMÉ

Proteomic studies have identified moesin (MSN), a protein containing a four-point-one, ezrin, radixin, moesin (FERM) domain, and the receptor CD44 as hub proteins found within a coexpression module strongly linked to Alzheimer's disease (AD) traits and microglia. These proteins are more abundant in Alzheimer's patient brains, and their levels are positively correlated with cognitive decline, amyloid plaque deposition, and neurofibrillary tangle burden. The MSN FERM domain interacts with the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP2) and the cytoplasmic tail of CD44. Inhibiting the MSN-CD44 interaction may help limit AD-associated neuronal damage. Here, we investigated the feasibility of developing inhibitors that target this protein-protein interaction. We have employed structural, mutational, and phage-display studies to examine how CD44 binds to the FERM domain of MSN. Interestingly, we have identified an allosteric site located close to the PIP2 binding pocket that influences CD44 binding. These findings suggest a mechanism in which PIP2 binding to the FERM domain stimulates CD44 binding through an allosteric effect, leading to the formation of a neighboring pocket capable of accommodating a receptor tail. Furthermore, high-throughput screening of a chemical library identified two compounds that disrupt the MSN-CD44 interaction. One compound series was further optimized for biochemical activity, specificity, and solubility. Our results suggest that the FERM domain holds potential as a drug development target. Small molecule preliminary leads generated from this study could serve as a foundation for additional medicinal chemistry efforts with the goal of controlling microglial activity in AD by modifying the MSN-CD44 interaction.


Sujet(s)
Maladie d'Alzheimer , Liaison aux protéines , Humains , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Domaines FERM , Antigènes CD44/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Protéomique
19.
J Virol ; 97(11): e0117123, 2023 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-37888980

RÉSUMÉ

IMPORTANCE: CD4-mimetic compounds (CD4mcs) are small-molecule inhibitors of human immunodeficiency virus (HIV-1) entry into host cells. CD4mcs target a pocket on the viral envelope glycoprotein (Env) spike that is used for binding to the receptor, CD4, and is highly conserved among HIV-1 strains. Nonetheless, naturally occurring HIV-1 strains exhibit a wide range of sensitivities to CD4mcs. Our study identifies changes distant from the binding pocket that can influence the susceptibility of natural HIV-1 strains to the antiviral effects of multiple CD4mcs. We relate the antiviral potency of the CD4mc against this panel of HIV-1 variants to the ability of the CD4mc to activate entry-related changes in Env conformation prematurely. These findings will guide efforts to improve the potency and breadth of CD4mcs against natural HIV-1 variants.


Sujet(s)
Agents antiVIH , Antigènes CD4 , Protéine d'enveloppe gp120 du VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Mimétisme moléculaire , Récepteur VIH , Humains , Agents antiVIH/composition chimique , Agents antiVIH/pharmacologie , Sites de fixation/effets des médicaments et des substances chimiques , Antigènes CD4/composition chimique , Antigènes CD4/métabolisme , Protéine d'enveloppe gp120 du VIH/composition chimique , Protéine d'enveloppe gp120 du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/classification , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Liaison aux protéines/effets des médicaments et des substances chimiques , Récepteur VIH/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques
20.
J Biol Chem ; 299(12): 105369, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37865311

RÉSUMÉ

Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.


Sujet(s)
Protéines de transport , Découverte de médicament , Défaillance cardiaque , Myofibrilles , Bibliothèques de petites molécules , Humains , Actines/métabolisme , Découverte de médicament/méthodes , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Myocarde/métabolisme , Myosines/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Liaison aux protéines/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Évaluation préclinique de médicament , Myofibrilles/effets des médicaments et des substances chimiques , Protéines de transport/métabolisme , Techniques de biocapteur , Adenosine triphosphatases/métabolisme , Muscles squelettiques/métabolisme , Protéines recombinantes/métabolisme , Activation enzymatique/effets des médicaments et des substances chimiques , Transfert d'énergie par résonance de fluorescence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...