Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 932
Filtrer
1.
FASEB J ; 38(17): e70013, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39225365

RÉSUMÉ

Articular cartilage phenotypic homeostasis is crucial for life-long joint function, but the underlying cellular and molecular mechanisms governing chondrocyte stability remain poorly understood. Here, we show that the protein tyrosine phosphatase SHP2 is differentially expressed in articular cartilage (AC) and growth plate cartilage (GPC) and that it negatively regulates cell proliferation and cartilage phenotypic program. Postnatal SHP2 deletion in Prg4+ AC chondrocytes increased articular cellularity and thickness, whereas SHP2 deletion in Acan+ pan-chondrocytes caused excessive GPC chondrocyte proliferation and led to joint malformation post-puberty. These observations were verified in mice and in cultured chondrocytes following treatment with the SHP2 PROTAC inhibitor SHP2D26. Further mechanistic studies indicated that SHP2 negatively regulates SOX9 stability and transcriptional activity by influencing SOX9 phosphorylation and promoting its proteasome degradation. In contrast to published work, SHP2 ablation in chondrocytes did not impact IL-1-evoked inflammation responses, and SHP2's negative regulation of SOX9 could be curtailed by genetic or chemical SHP2 inhibition, suggesting that manipulating SHP2 signaling has translational potential for diseases of cartilage dyshomeostasis.


Sujet(s)
Cartilage articulaire , Chondrocytes , Arthrose , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Facteur de transcription SOX-9 , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Souris , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Arthrose/métabolisme , Arthrose/anatomopathologie , Prolifération cellulaire , Cellules cultivées , Souris de lignée C57BL , Souris knockout , Mâle
2.
Commun Biol ; 7(1): 1179, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39300128

RÉSUMÉ

Proteins can be targeted for degradation by engineering biomolecules that direct them to the eukaryotic ubiquitination machinery. For instance, the fusion of an E3 ubiquitin ligase to a suitable target binding domain creates a 'biological Proteolysis-Targeting Chimera' (bioPROTAC). Here we employ an analogous approach where the target protein is recruited directly to a human E2 ubiquitin-conjugating enzyme via an attached target binding domain. Through rational design and screening we develop E2 bioPROTACs that induce the degradation of the human intracellular proteins SHP2 and KRAS. Using global proteomics, we characterise the target-specific and wider effects of E2 vs. VHL-based fusions. Taking SHP2 as a model target, we also employ a route to bioPROTAC discovery based on protein display libraries, yielding a degrader with comparatively weak affinity capable of suppressing SHP2-mediated signalling.


Sujet(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protéolyse , Ubiquitin-conjugating enzymes , Humains , Ubiquitin-conjugating enzymes/métabolisme , Ubiquitin-conjugating enzymes/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Ubiquitination , Protéines proto-oncogènes p21(ras)/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/génétique , Cellules HEK293 , Protéomique/méthodes , Liaison aux protéines
3.
Nat Commun ; 15(1): 8146, 2024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-39322643

RÉSUMÉ

Mutant selective drugs targeting the inactive, GDP-bound form of KRASG12C have been approved for use in lung cancer, but resistance develops rapidly. Here we use an inhibitor, (RMC-4998) that targets RASG12C in its active, GTP-bound form, to treat KRAS mutant lung cancer in various immune competent mouse models. RAS pathway reactivation after RMC-4998 treatment could be delayed using combined treatment with a SHP2 inhibitor, which not only impacts tumour cell RAS signalling but also remodels the tumour microenvironment to be less immunosuppressive. In an immune inflamed model, RAS and SHP2 inhibitors in combination drive durable responses by suppressing tumour relapse and inducing development of immune memory. In an immune excluded model, combined RAS and SHP2 inhibition sensitises tumours to immune checkpoint blockade, leading to efficient tumour immune rejection. These preclinical results demonstrate the potential of the combination of RAS(ON) G12C-selective inhibitors with SHP2 inhibitors to sensitize tumours to immune checkpoint blockade.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protéines proto-oncogènes p21(ras) , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Animaux , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Humains , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Souris de lignée C57BL , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Mutation
4.
Int J Biochem Cell Biol ; 174: 106621, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39181599

RÉSUMÉ

BACKGROUND: Immune escape is a major obstacle to T-cell-based immunotherapy for cancers such as gastric cancer (GC). Mesoderm-specific transcript (MEST) is a tumor-promoting factor that regulates multiple oncogenic signaling pathways. However, the role of MEST-mediated immune escape is unclear. METHODS: Bioinformatics analysis of MEST expression and enrichment pathways were performed Quantitative reverse transcription PCR (qPCR) or western blot was used to detect the expression of MEST, Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2), Major histocompatibility class I (MHCI)-related genes. Cell function was assessed by Cell Counting Kit (CCK)-8, Transwell, Lactate dehydrogenase (LDH) kit, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and immunohistochemistry (IHC). Xenograft nude mice and immune-reconstructed mice were used to test the effects of different treatments on tumor growth and immune escape in vivo. RESULTS: MEST was upregulated in GC and promoted tumor proliferation, migration, and invasion. Rescue experiments revealed that TNO155 treatment or knockdown of SHP2 promoted the killing ability of CD8+ T cells and the expression of granzyme B (GZMB) and interferon-gamma (IFN-γ), and MEST overexpression reversed the effect. In vivo experiments confirmed that MEST promoted tumor growth, knockdown of MEST inhibited immune escape in GC, and that combination treatment with anti-PD-1 improved anti-tumor activity. CONCLUSION: In this study, we demonstrated that MEST inhibited IFN-γ secretion from CD8+ T cells by up-regulating SHP2, thereby downregulating MHCI expression in GC cells to promote immune escape and providing a new T cell-based therapeutic potential for GC.


Sujet(s)
Régulation négative , Souris nude , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Tumeurs de l'estomac , Échappement de la tumeur à la surveillance immunitaire , Animaux , Femelle , Humains , Mâle , Souris , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/métabolisme , Souris de lignée BALB C , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunologie , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Échappement de la tumeur à la surveillance immunitaire/génétique
5.
Cell Death Dis ; 15(8): 577, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117618

RÉSUMÉ

Shp2, a critical SH2-domain-containing tyrosine phosphatase, is essential for cellular regulation and implicated in metabolic disruptions, obesity, diabetes, Noonan syndrome, LEOPARD syndrome, and cancers. This study focuses on Shp2 in rod photoreceptor cells, revealing its enrichment, particularly in rods. Deletion of Shp2 in rods leads to age-dependent photoreceptor degeneration. Shp2 targets occludin (OCLN), a tight junction protein, and its deletion reduces OCLN expression in the retina and retinal pigment epithelium (RPE). The isolation of actively translating mRNAs from rods lacking Shp2, followed by RNA sequencing, reveals alterations in cell cycle regulation. Additionally, altered retinal metabolism is observed in retinal cells lacking Shp2. Our studies indicate that Shp2 is crucial for maintaining the structure and function of photoreceptors.


Sujet(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Dégénérescence de la rétine , Animaux , Dégénérescence de la rétine/anatomopathologie , Dégénérescence de la rétine/métabolisme , Dégénérescence de la rétine/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Souris , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/anatomopathologie , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie , Souris knockout , Vieillissement/métabolisme , Vieillissement/génétique , Occludine/métabolisme , Occludine/génétique , Souris de lignée C57BL , Délétion de gène , Rétine/métabolisme , Rétine/anatomopathologie
6.
Oncogene ; 43(36): 2737-2749, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39112517

RÉSUMÉ

The proneural-mesenchymal (PN-MES) transformation of glioma stem cells (GSCs) can significantly increase proliferation, invasion, chemotherapy tolerance, and recurrence. M2-like polarization of tumor-associated macrophages (TAMs) has a strong immunosuppressive effect, promoting tumor malignancy and angiogenesis. There is limited understanding on the interactions between GSCs and TAMs as well as their associated molecular mechanisms. In the present study, bioinformatics analysis, GSC and TAM co-culture, determination of TAM polarization phenotypes, and other in vitro experiments confirmed that CCL2 secreted by MES-GSCs promotes TAM-M2 polarization via the IKZF1-CD84-SHP2 pathway and PN-MES transformation of GSCs via the IKZF1-LRG1 pathway in TAMs. IKZF1 inhibitors could significantly reduce tumor volumes in animal glioma models and improve survival, as well as suppress TAM-M2 polarization and the GSC malignant phenotype. The results of this study indicate the important interaction between TAMs and GSCs in the glioma microenvironment as well as its role in tumor progression. The findings also suggest a novel target for follow-up clinical transformation research on the regulation of TAM function and GSCs malignant phenotype.


Sujet(s)
Chimiokine CCL2 , Gliome , Facteur de transcription Ikaros , Cellules souches tumorales , Microenvironnement tumoral , Macrophages associés aux tumeurs , Gliome/anatomopathologie , Gliome/génétique , Gliome/métabolisme , Chimiokine CCL2/métabolisme , Chimiokine CCL2/génétique , Animaux , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/anatomopathologie , Humains , Souris , Facteur de transcription Ikaros/génétique , Facteur de transcription Ikaros/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Microenvironnement tumoral/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Transduction du signal/génétique , Lignée cellulaire tumorale , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Antigènes CD/métabolisme , Antigènes CD/génétique , Régulation de l'expression des gènes tumoraux , Macrophages/métabolisme , Macrophages/anatomopathologie
7.
J Med Chem ; 67(16): 13534-13549, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39110625

RÉSUMÉ

As an oncogenic phosphatase, SHP2 acts as a converging node in the RTK-RAS-MAPK signaling pathway in cancer cells and suppresses antitumor immunity by passing signals downstream of PD-1. Here, we utilized the extra druggable pocket outside the previously identified SHP2 allosteric tunnel site by the (6,5 fused), 6 spirocyclic system. The optimized compound, JAB-3312, exhibited a SHP2 binding Kd of 0.37 nM, SHP2 enzymatic IC50 of 1.9 nM, KYSE-520 antiproliferative IC50 of 7.4 nM and p-ERK inhibitory IC50 of 0.23 nM. For JAB-3312, an oral dose of 1.0 mg/kg QD was sufficient to achieve 95% TGI in KYSE-520 xenograft model of mouse. JAB-3312 was well-tolerated in animal models, and a close correlation was observed between the plasma concentration of JAB-3312 and the p-ERK inhibition in tumors. Currently, JAB-3312 is undergoing clinical trials as a potential anticancer agent.


Sujet(s)
Antinéoplasiques , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Humains , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/synthèse chimique , Souris , Régulation allostérique/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Relation structure-activité , Tests d'activité antitumorale sur modèle de xénogreffe , Prolifération cellulaire/effets des médicaments et des substances chimiques , Découverte de médicament , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/usage thérapeutique , Antienzymes/synthèse chimique , Antienzymes/pharmacocinétique , Souris nude , Femelle , Tumeurs/traitement médicamenteux
8.
Aging (Albany NY) ; 16(17): 12263-12276, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39197167

RÉSUMÉ

The primary objective of this study was to explore the extensive implications and complex molecular interactions arising from the confluence of excessive glucocorticoids and RANKL on the differentiation process of BMM into osteoclasts, profoundly impacting osteoporosis development. The methodology encompassed X-ray analysis and HE staining for evaluating bone loss in mice, while immunohistochemical staining was utilized to observe phosphorylated SHP2 (p-SHP2) expression. The assessment of several phosphorylated and total protein expression levels, including NF-κB, SHP2, SYK, JAK2, TAK1, NFATC1, c-fos, and Cathepsin K, was conducted via Western blotting. Additional experiments, involving CCK8 and monoclonal proliferation assays, were undertaken to determine BMM proliferation capacity. Immunofluorescence staining facilitated the quantification of TRAP fluorescence intensity. In vivo analysis revealed that glucocorticoid surplus triggers SHP2 signaling pathway activation, accelerating osteoporosis progression. Western blot results demonstrated that SHP2 inhibition could decrease the expression of specific proteins such as p-NF-κB and p-SHP2, with minimal effects on p-SYK levels. In vitro findings indicated that glucocorticoid and RANKL interaction activates the SHP2 pathway through NF-κB and SYK pathways, enhancing expressions of p-JAK2, p-TAK1, NFATC1, c-fos, and Cathepsin K, thereby promoting BMM to osteoclast transformation. Conclusion: Excessive glucocorticoids and RANKL interaction advance osteoclast differentiation from BMM by activating the SYK/SHP2/NF-κB signaling pathway, expediting osteoporosis progression.


Sujet(s)
Différenciation cellulaire , Glucocorticoïdes , Macrophages , Facteur de transcription NF-kappa B , Ostéoclastes , Ostéoporose , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Ligand de RANK , Transduction du signal , Syk kinase , Animaux , Ligand de RANK/métabolisme , Ostéoclastes/métabolisme , Ostéoclastes/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Syk kinase/métabolisme , Souris , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Glucocorticoïdes/pharmacologie , Ostéoporose/métabolisme , Ostéoporose/anatomopathologie , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Femelle , Souris de lignée C57BL
9.
Aging (Albany NY) ; 16(17): 12335-12345, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39207449

RÉSUMÉ

OBJECTIVE: To investigate the effect of PD-1 monoclonal antibodies in tumor-associated macrophages on angiogenesis in cervical cancer and its mechanism of action. METHODS: The effect of PD-1 monoclonal antibodies on the progression of cervical cancer was assessed using the nude mouse xenograft model and HE staining; the impact of PD-1 monoclonal antibodies on cervical cancer cell migration was evaluated using wound healing assay and Transwell assay; the effect on vascular formation in cervical cancer cells was examined using an angiogenesis assay; the impact on the expression of related proteins was tested using Western blotting. RESULTS: PD-1 monoclonal antibodies in tumor-associated macrophages can regulate and thus inhibit the progression of cervical cancer while promoting the expression of SHP2. Additionally, Sindilizumab inhibited the expression of tissue-type fibrinogen activator K and HIF1α through the PD-1/IRE1α/SHP2 signaling pathway, which inhibited the migration and neovascularization of cervical cancer cells. CONCLUSIONS: This study discovered that PD-1 monoclonal antibodies in tumor-associated macrophages inhibit vascular generation inside cervical cancer by affecting the PD-1/IRE1α/SHP2/HIF1α signaling pathway, providing a new therapeutic target for the treatment of cervical cancer.


Sujet(s)
Sous-unité alpha du facteur-1 induit par l'hypoxie , Souris nude , Néovascularisation pathologique , Récepteur-1 de mort cellulaire programmée , Transduction du signal , Macrophages associés aux tumeurs , Tumeurs du col de l'utérus , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/immunologie , Animaux , Femelle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur-1 de mort cellulaire programmée/métabolisme , Souris , Humains , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Biol Chem ; 300(9): 107616, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39089586

RÉSUMÉ

Targeted protein degradation is an emergent and rapidly evolving therapeutic strategy. In particular, biologics-based targeted degradation modalities (bioPROTACs) are relatively under explored compared to small molecules. Here, we investigate how target affinity, cellular localization, and valency of bioPROTACs impact efficacy of targeted degradation of the oncogenic phosphatase src-homology 2 containing protein tyrosine phosphatase-2 (SHP2). We identify bivalent recruitment of SHP2 by bioPROTACs as a broadly applicable strategy to improve potency. Moreover, we demonstrate that SHP2-targeted bioPROTACs can effectively counteract gain-of-function SHP2 mutants present in cancer, which are otherwise challenging to selectively target with small molecule constructs. Overall, this study demonstrates the utility of bioPROTACs for challenging targets, and further explicates design principles for therapeutic bioPROTACs.


Sujet(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protéolyse , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Humains , Protéolyse/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tumeurs/métabolisme , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie
11.
Sci Rep ; 14(1): 20251, 2024 08 31.
Article de Anglais | MEDLINE | ID: mdl-39215017

RÉSUMÉ

Monosodium urate (MSU) crystallisation deposited in local tissues and organs induce inflammatory reactions, resulting in diseases such as gout. MSU has been recognized as a common and prevalent pathology in various clinical conditions. In this study, we investigated the role of MSU in the pathogenesis of diabetic kidney disease (DKD). We induced renal injury in diabetic kidney disease mice using streptozotocin (STZ) and assessed renal histopathological damage using Masson's trichrome staining and Collagen III immunofluorescence staining. We measured the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and uric acid (UA) using ELISA. Protein expression levels of NLRP3, p-NF-κB, SHP2, p-STAT3, and p-ERK1/2 were analyzed by Western blot. To further investigate the role of MSU in diabetic kidney disease, we conducted in vitro experiments. In our in vivo experiments, we found that compared to the Model group, there was a significant increase in interstitial fibrosis in the kidneys of mice after treatment with MSU, accompanied by elevated levels of MDA, SOD, and UA. Furthermore, the protein expression of NLRP3, p-NF-NB, SHP2, p-STAT3, and p-ERK1/2 was upregulated. In our subsequent studies on mouse fibroblasts (L929 cells), we discovered that high glucose, MSU, and TGF-ß could promote the expression of P22, GP91, NLRP3, NF-κB, p-NF-κB, p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins. Additionally, we found that SHP2 could counteract the upregulation trend induced by MSU on the expression of p-SHP2, p-EGFR, p-STAT3, and Collagen-III proteins, and inhibitors YQ128, NAC, and Cetuximab exhibited similar effects. Furthermore, immunofluorescence results indicated that SHP2 could inhibit the expression of the fibrosis marker α-SMA in L929 cells. These findings suggest that MSU can promote renal fibroblast SHP2 expression, induce oxidative stress, activate the NLRP3/NF-κB pathway, and enhance diabetic kidney disease fibroblast proliferation through the TGFß/STAT3/ERK1/2 signaling pathway, leading to renal fibrosis.


Sujet(s)
Prolifération cellulaire , Néphropathies diabétiques , Fibroblastes , Fibrose , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Mâle , Souris , Cristallisation , Diabète expérimental/métabolisme , Diabète expérimental/complications , Diabète expérimental/anatomopathologie , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibrose/métabolisme , Rein/métabolisme , Rein/anatomopathologie , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Espèces réactives de l'oxygène/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de croissance transformant bêta/métabolisme , Acide urique/métabolisme
12.
Bioorg Chem ; 151: 107661, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39067422

RÉSUMÉ

SHP2 (Src homology-2-containing protein tyrosine phosphatase 2) plays an important role in cell proliferation, survival, migration by affecting RAS-ERK, PI3K-AKT, JAK-STAT signaling pathways and so on. Overexpression or gene mutation of SHP2 is closely linked with a variety of cancers, making it a potential therapeutic target for cancer disease. In this paper, 30 target compounds bearing pyrido[1,2-a]pyrimidin-4-one core were synthesized via two-round design strategy by means of scaffold hopping protocol. It was evaluated the in vitro enzymatic inhibition and cell antiproliferation assay of these targets. 13a, designed in the first round, presented relatively good inhibitory activity, but its molecular rigidity might limit further improvement by hindering the formation of the desired "bidentate ligand", as revealed by molecular docking studies. In our second-round design, S atom as a linker was inserted into the core and the 7-aryl group to enhance the flexibility of the structure. The screening result revealed that 14i could exhibit high enzymatic activity against full-length SHP2 (IC50 = 0.104 µM), while showing low inhibitory effect on SHP2-PTP (IC50 > 50 µM). 14i also demonstrated high antiproliferative activity against the Kyse-520 cells (IC50 = 1.06 µM) with low toxicity against the human brain microvascular endothelial cells HBMEC (IC50 = 30.75 µM). 14i also displayed stronger inhibitory activities on NCI-H358 and MIA-PaCa2 cells compared to that of SHP099. Mechanistic studies revealed that 14i could induce cell apoptosis, arrest the cell cycle at the G0/G1 phase and downregulate the phosphorylation levels of Akt and Erk1/2 in Kyse-520 cells. Molecular docking and molecular dynamics studies displayed more detailed information on the binding mode and binding mechanism of 14i and SHP2. These data suggest that 14i has the potential to be a promising lead compound for our further investigation of SHP2 inhibitors.


Sujet(s)
Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Antienzymes , Simulation de docking moléculaire , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Structure moléculaire , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Tests de criblage d'agents antitumoraux , Régulation allostérique/effets des médicaments et des substances chimiques , Pyrimidinones/pharmacologie , Pyrimidinones/synthèse chimique , Pyrimidinones/composition chimique , Pyrimidines/pharmacologie , Pyrimidines/synthèse chimique , Pyrimidines/composition chimique , Pyridines/pharmacologie , Pyridines/composition chimique , Pyridines/synthèse chimique
13.
Cell Mol Life Sci ; 81(1): 294, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38977495

RÉSUMÉ

The obligate intracellular parasite Toxoplasma gondii causes life-threatening toxoplasmosis to immunocompromised individuals. The pathogenesis of Toxoplasma relies on its swift dissemination to the central nervous system through a 'Trojan Horse' mechanism using infected leukocytes as carriers. Previous work found TgWIP, a protein secreted from Toxoplasma, played a role in altering the actin cytoskeleton and promoting cell migration in infected dendritic cells (DCs). However, the mechanism behind these changes was unknown. Here, we report that TgWIP harbors two SH2-binding motifs that interact with tyrosine phosphatases Shp1 and Shp2, leading to phosphatase activation. DCs infected with Toxoplasma exhibited hypermigration, accompanying enhanced F-actin stress fibers and increased membrane protrusions such as filopodia and pseudopodia. By contrast, these phenotypes were abrogated in DCs infected with Toxoplasma expressing a mutant TgWIP lacking the SH2-binding motifs. We further demonstrated that the Rho-associated kinase (Rock) is involved in the induction of these phenotypes, in a TgWIP-Shp1/2 dependent manner. Collectively, the data uncover a molecular mechanism by which TgWIP modulates the migration dynamics of infected DCs in vitro.


Sujet(s)
Mouvement cellulaire , Cellules dendritiques , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protéines de protozoaire , Toxoplasma , Toxoplasma/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Cellules dendritiques/métabolisme , Cellules dendritiques/parasitologie , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 6/métabolisme , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Humains , Souris , rho-Associated Kinases/métabolisme , Toxoplasmose/métabolisme , Toxoplasmose/parasitologie , Toxoplasmose/anatomopathologie , Souris de lignée C57BL
14.
Nat Commun ; 15(1): 5629, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965223

RÉSUMÉ

Mutations that decrease or increase the activity of the tyrosine phosphatase, SHP2 (encoded by PTPN11), promotes developmental disorders and several malignancies by varying phosphatase activity. We uncovered that SHP2 is a distinct class of an epigenetic enzyme; upon phosphorylation by the kinase ACK1/TNK2, pSHP2 was escorted by androgen receptor (AR) to chromatin, erasing hitherto unidentified pY54-H3 (phosphorylation of histones H3 at Tyr54) epigenetic marks to trigger a transcriptional program of AR. Noonan Syndrome with Multiple Lentigines (NSML) patients, SHP2 knock-in mice, and ACK1 knockout mice presented dramatic increase in pY54-H3, leading to loss of AR transcriptome. In contrast, prostate tumors with high pSHP2 and pACK1 activity exhibited progressive downregulation of pY54-H3 levels and higher AR expression that correlated with disease severity. Overall, pSHP2/pY54-H3 signaling acts as a sentinel of AR homeostasis, explaining not only growth retardation, genital abnormalities and infertility among NSML patients, but also significant AR upregulation in prostate cancer patients.


Sujet(s)
Épigenèse génétique , Histone , Homéostasie , Souris knockout , Tumeurs de la prostate , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Récepteurs aux androgènes , Animaux , Humains , Mâle , Souris , Chromatine/métabolisme , Histone/métabolisme , Syndrome de Noonan/génétique , Syndrome de Noonan/métabolisme , Phosphorylation , Tumeurs de la prostate/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Récepteurs aux androgènes/métabolisme , Récepteurs aux androgènes/génétique , Transduction du signal
15.
Cancer Cell ; 42(7): 1217-1238.e19, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38981438

RÉSUMÉ

Although genomic anomalies in glioblastoma (GBM) have been well studied for over a decade, its 5-year survival rate remains lower than 5%. We seek to expand the molecular landscape of high-grade glioma, composed of IDH-wildtype GBM and IDH-mutant grade 4 astrocytoma, by integrating proteomic, metabolomic, lipidomic, and post-translational modifications (PTMs) with genomic and transcriptomic measurements to uncover multi-scale regulatory interactions governing tumor development and evolution. Applying 14 proteogenomic and metabolomic platforms to 228 tumors (212 GBM and 16 grade 4 IDH-mutant astrocytoma), including 28 at recurrence, plus 18 normal brain samples and 14 brain metastases as comparators, reveals heterogeneous upstream alterations converging on common downstream events at the proteomic and metabolomic levels and changes in protein-protein interactions and glycosylation site occupancy at recurrence. Recurrent genetic alterations and phosphorylation events on PTPN11 map to important regulatory domains in three dimensions, suggesting a central role for PTPN11 signaling across high-grade gliomas.


Sujet(s)
Tumeurs du cerveau , Gliome , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Transduction du signal , Humains , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Mutation , Protéomique/méthodes , Maturation post-traductionnelle des protéines , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Phosphorylation , Grading des tumeurs , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme
16.
J Med Chem ; 67(15): 13305-13323, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39066713

RÉSUMÉ

SHP2 plays a critical role in modulating tumor growth and PD-1-related signaling pathway, thereby serving as an attractive antitumor target. To date, no antitumor drugs targeting SHP2 have been approved, and hence, the search of SHP2 inhibitors with new chemical scaffolds is urgently needed. Herein, we developed a novel SHP2 allosteric inhibitor SDUY038 with a furanyl amide scaffold, demonstrating potent binding affinity (KD = 0.29 µM), enzymatic activity (IC50 = 1.2 µM) and similar binding interactions to SHP099. At the cellular level, SDUY038 exhibited pan-antitumor activity (IC50 = 7-24 µM) by suppressing pERK expression. Furthermore, SDUY038 significantly inhibited tumor growth in both xenograft and organoid models. Additionally, SDUY038 displayed acceptable bioavailability (F = 14%) and half-life time (t1/2 = 3.95 h). Conclusively, this study introduces the furanyl amide scaffold as a novel class of SHP2 allosteric inhibitors, offering promising lead compounds for further development of new antitumor therapies targeting SHP2.


Sujet(s)
Amides , Antinéoplasiques , Conception de médicament , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Animaux , Régulation allostérique/effets des médicaments et des substances chimiques , Amides/pharmacologie , Amides/composition chimique , Amides/synthèse chimique , Souris , Lignée cellulaire tumorale , Relation structure-activité , Furanes/pharmacologie , Furanes/composition chimique , Furanes/synthèse chimique , Tests d'activité antitumorale sur modèle de xénogreffe , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude
17.
Proc Natl Acad Sci U S A ; 121(30): e2407159121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39012820

RÉSUMÉ

Mutations in the tyrosine phosphatase Src homology-2 domain-containing protein tyrosine phosphatase-2 (SHP2) are associated with a variety of human diseases. Most mutations in SHP2 increase its basal catalytic activity by disrupting autoinhibitory interactions between its phosphatase domain and N-terminal SH2 (phosphotyrosine recognition) domain. By contrast, some disease-associated mutations located in the ligand-binding pockets of the N- or C-terminal SH2 domains do not increase basal activity and likely exert their pathogenicity through alternative mechanisms. We lack a molecular understanding of how these SH2 mutations impact SHP2 structure, activity, and signaling. Here, we characterize five SHP2 SH2 domain ligand-binding pocket mutants through a combination of high-throughput biochemical screens, biophysical and biochemical measurements, and molecular dynamics simulations. We show that while some of these mutations alter binding affinity to phosphorylation sites, the T42A mutation in the N-SH2 domain is unique in that it also substantially alters ligand-binding specificity, despite being 8 to 10 Å from the specificity-determining region of the SH2 domain. This mutation exerts its effect on sequence specificity by remodeling the phosphotyrosine-binding pocket, altering the mode of engagement of both the phosphotyrosine and surrounding residues on the ligand. The functional consequence of this altered specificity is that the T42A mutant has biased sensitivity toward a subset of activating ligands and enhances downstream signaling. Our study highlights an example of a nuanced mechanism of action for a disease-associated mutation, characterized by a change in protein-protein interaction specificity that alters enzyme activation.


Sujet(s)
Simulation de dynamique moléculaire , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Domaine d'homologie SRC , Protein Tyrosine Phosphatase, Non-Receptor Type 11/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/composition chimique , Humains , Domaine d'homologie SRC/génétique , Liaison aux protéines , Mutation , Phosphorylation , Sites de fixation/génétique , Phosphotyrosine/métabolisme , Ligands
18.
Sci Transl Med ; 16(753): eadj1597, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38924432

RÉSUMÉ

Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-ß and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.


Sujet(s)
Souris knockout , Neurofibromine-1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Pseudarthrose , Cellules de Schwann , Animaux , Femelle , Humains , Mâle , Souris , Différenciation cellulaire/effets des médicaments et des substances chimiques , Fibrose , Mitogen-Activated Protein Kinase Kinases/métabolisme , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Neurofibromatose de type 1/anatomopathologie , Neurofibromatose de type 1/métabolisme , Neurofibromatose de type 1/complications , Neurofibromine-1/métabolisme , Neurofibromine-1/génétique , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonistes et inhibiteurs , Pseudarthrose/anatomopathologie , Pseudarthrose/métabolisme , Pseudarthrose/congénital , Cellules de Schwann/métabolisme , Cellules de Schwann/effets des médicaments et des substances chimiques , Cellules de Schwann/anatomopathologie , Cellules souches/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Tibia/anatomopathologie
19.
Biochem Pharmacol ; 226: 116370, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38880359

RÉSUMÉ

Tendon injuries typically display limited reparative capacity, often resulting in suboptimal outcomes and an elevated risk of recurrence or rupture. While cytokines of the IL-6 family are primarily recognised for their inflammatory properties, they also have multifaceted roles in tissue regeneration and repair. Despite this, studies examining the association between IL-6 family cytokines and tendon repair remained scarce. gp130, a type of glycoprotein, functions as a co-receptor for all cytokines in the IL-6 family. Its role is to assist in the transmission of signals following the binding of ligands to receptors. RCGD423 is a gp130 modulator. Phosphorylation of residue Y759 of gp130 recruits SHP2 and SOCS3 and inhibits activation of the STAT3 pathway. In our study, RCGD423 stimulated the formation of homologous dimers of gp130 and the phosphorylation of Y759 residues without the involvement of IL-6 and IL-6R. Subsequently, the phosphorylated residues recruited SHP2, activating the downstream ERK and AKT pathways. These mechanisms ultimately promoted the migration ability of tenocytes and matrix synthesis, especially collagen I. Moreover, RCGD423 also demonstrated significant improvements in collagen content, alignment of collagen fibres, and biological and biomechanical function in a rat Achilles tendon injury model. In summary, we demonstrated a promising gp130 modulator (RCGD423) that could potentially enhance tendon injury repair by redirecting downstream signalling of IL-6, suggesting its potential therapeutic application for tendon injuries.


Sujet(s)
Tendon calcanéen , Mouvement cellulaire , Récepteur gp130 de cytokines , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Ténocytes , Animaux , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Récepteur gp130 de cytokines/métabolisme , Tendon calcanéen/métabolisme , Tendon calcanéen/traumatismes , Tendon calcanéen/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/physiologie , Rats , Protéines proto-oncogènes c-akt/métabolisme , Ténocytes/métabolisme , Ténocytes/effets des médicaments et des substances chimiques , Ténocytes/physiologie , Collagène/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Mâle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/physiologie , Traumatismes des tendons/métabolisme , Traumatismes des tendons/traitement médicamenteux
20.
Phytomedicine ; 132: 155832, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38924928

RÉSUMÉ

BACKGROUND: Lung cancer has been considered as a serious problem for the public health system. NSCLC is the main type of lung cancer, and finding improved treatments for NSCLC is a pressing concern. In this study, we have explored the efficacy of isotoosendanin (ITSN) for the treatment of NSCLC, and also explored the potential underlying mechanisms. METHODS: NSCLC cells were cultured, and colony formation, cell cycle as well as apoptosis assays have been conducted for investigating the biological functions of ITSN on NSCLC cells. Furthermore, target genes of ITSN have been predicted via PharmMapper and SuperPred database, subsequently validated using the drug affinity responsive target stability (DARTS) approach, a cellular thermal shift assay (CETSA) as well as surface plasmon resonance (SPR) analysis. Additionally, ubiquitination experiments have been conducted for the level of ubiquitination of the NSCLC cells. Finally, a nude mouse xenograft model has been established for evaluating the anti-tumor effects of ITSN in vivo. RESULTS: ITSN has shown anti-NSCLC activities both in vitro and in vivo. Mechanistically, ITSN interacts with SHP-2 through enhancing its stability and decreases the level of ubiquitination. Notably, ITSN may regulate the behaviors of NSCLC cells via affecting the JAK/STAT3 signaling, and finally, the anti-tumor effects of ITSN was partially reversed by the application of SHP-2 inhibitor or siRNA of SHP-2. CONCLUSIONS: ITSN may exert its anti-tumor effects by directly targeting SHP-2, increasing its stability and minimizing its ubiquitination. These results imply that ITSN could be a revolutionary component for treating NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Souris nude , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Facteur de transcription STAT-3 , Transduction du signal , Facteur de transcription STAT-3/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Humains , Animaux , Tumeurs du poumon/traitement médicamenteux , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques d'origine végétale/pharmacologie , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Souris de lignée BALB C , Janus kinases/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Ubiquitination/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE