Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.103
Filtrer
1.
Arch Biochem Biophys ; 759: 110109, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39117070

RÉSUMÉ

Chronic inflammation is an important pathogenetic factor that leads to the progression of Alzheimer's disease (AD), and specialized pro-resolving lipid mediators (SPMs) play critical role in regulating inflammatory responses during AD pathogenesis. Maresin1 (MaR1) is the latest discovered SPMs, and it is found that MaR1 improves AD cognitive impairment by regulating neurotrophic pathways to protect AD synapses and reduce Aß production, which made MaR1 as candidate agent for AD treatment. Unfortunately, the underlying mechanisms are still largely known. In this study, the AD mice and cellular models were subjected to MaR1 treatment, and we found that MaR1 reduced Aß production to ameliorate AD-related symptoms and increased the expression levels of ADAM10/17, sAPPα and sAPPß to exert its anti-inflammatory role. In addition, as it was determined by Western Blot analysis, we observed that MaR1 could affected the neuroprotective signal pathways. Specifically, MaR1 downregulated p57NTR and upregulated TrkA to activate the p75NTR/TrkA signal pathway, and it could increase the expression levels of p-PI3K and p-Akt, and downregulated p-mTOR to activate the PI3K/AKT/ERK/mTOR pathway. Finally, we verified the role of ADAM10/17 in regulating AD progression, and we found that silencing of ADAM10/17 inactivated the above neuroprotective signal pathways to aggravate AD pathogenesis. In conclusion, MaR1 is verified as potential therapeutic agent for AD by eliminating Aß production, upregulating ADAM10/17, sAPPα and sAPPß, and activating the neuroprotective p75NTR/TrkA pathway and the PI3K/AKT/ERK/mTOR pathway.


Sujet(s)
Protéine ADAM10 , Maladie d'Alzheimer , Amyloid precursor protein secretases , Peptides bêta-amyloïdes , Acide docosahexaénoïque , Transduction du signal , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Animaux , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Amyloid precursor protein secretases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Acide docosahexaénoïque/pharmacologie , Acide docosahexaénoïque/métabolisme , Peptides bêta-amyloïdes/métabolisme , Souris , Inflammation/métabolisme , Projets pilotes , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Humains , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mâle
2.
Cell Death Dis ; 15(8): 599, 2024 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-39155279

RÉSUMÉ

Oral squamous cell carcinoma (OSCC) is a prevalent cancer worldwide, exhibiting unique regional prevalence. Despite advancements in diagnostics and therapy, the 5-year survival rate for patients has seen limited improvement. A deeper understanding of OSCC pathogenesis, especially its molecular underpinnings, is essential for improving detection, prevention, and treatment. In this context, noncoding RNAs, such as circular RNAs (circRNAs), have gained recognition as crucial regulators and potential biomarkers in OSCC progression. Our study highlights the discovery of previously uncharacterized circRNAs, including a SNX5 gene-derived circRNA, circSNX5, through deep sequencing of OSCC patient tissue transcriptomes. We established circSNX5's tumor-specific expression and its strong correlation with patient survival using structure-specific and quantitative PCR analyses. In vitro and in vivo experiments underscored circSNX5 RNA's regulatory role in cancer growth and metastasis. Further, our omics profiling and functional assays revealed that ADAM10 is a critical effector in circSNX5-mediated cancer progression, with circSNX5 maintaining ADAM10 expression by sponging miR-323. This novel circRNA-miRNA-mRNA regulatory axis significantly contributes to oral cancer progression and malignancy. Moreover, we discovered that circSNX5 RNA is produced via noncanonical sequential back-splicing of pre-mRNA, a process negatively regulated by the RNA-binding protein STAU1. This finding adds a new dimension to our understanding of exonic circRNA biogenesis in the eukaryotic transcriptome. Collectively, our findings offer a detailed mechanistic dissection and functional interpretation of a novel circRNA, shedding light on the role of the noncoding transcriptome in cancer biology and potentially paving the way for innovative therapeutic strategies.


Sujet(s)
Tumeurs de la bouche , ARN circulaire , Nexines de tri , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Nexines de tri/métabolisme , Nexines de tri/génétique , Tumeurs de la bouche/génétique , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Animaux , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire/génétique , Souris , Souris nude , microARN/métabolisme , microARN/génétique , Mâle , Femelle , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme
3.
Cell Mol Life Sci ; 81(1): 333, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39112663

RÉSUMÉ

Synaptic dysfunction is an early pathogenic event leading to cognitive decline in Huntington's disease (HD). We previously reported that the active ADAM10 level is increased in the HD cortex and striatum, causing excessive proteolysis of the synaptic cell adhesion protein N-Cadherin. Conversely, ADAM10 inhibition is neuroprotective and prevents cognitive decline in HD mice. Although the breakdown of cortico-striatal connection has been historically linked to cognitive deterioration in HD, dendritic spine loss and long-term potentiation (LTP) defects identified in the HD hippocampus are also thought to contribute to the cognitive symptoms of the disease. The aim of this study is to investigate the contribution of ADAM10 to spine pathology and LTP defects of the HD hippocampus. We provide evidence that active ADAM10 is increased in the hippocampus of two mouse models of HD, leading to extensive proteolysis of N-Cadherin, which has a widely recognized role in spine morphology and synaptic plasticity. Importantly, the conditional heterozygous deletion of ADAM10 in the forebrain of HD mice resulted in the recovery of spine loss and ultrastructural synaptic defects in CA1 pyramidal neurons. Meanwhile, normalization of the active ADAM10 level increased the pool of synaptic BDNF protein and activated ERK neuroprotective signaling in the HD hippocampus. We also show that the ADAM10 inhibitor GI254023X restored LTP defects and increased the density of mushroom spines enriched with GluA1-AMPA receptors in HD hippocampal neurons. Notably, we report that administration of the TrkB antagonist ANA12 to HD hippocampal neurons reduced the beneficial effect of GI254023X, indicating that the BDNF receptor TrkB contributes to mediate the neuroprotective activity exerted by ADAM10 inhibition in HD. Collectively, these findings indicate that ADAM10 inhibition coupled with TrkB signaling represents an efficacious strategy to prevent hippocampal synaptic plasticity defects and cognitive dysfunction in HD.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Hippocampe , Maladie de Huntington , Potentialisation à long terme , Protéines membranaires , Récepteur trkB , Transduction du signal , Animaux , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Maladie de Huntington/métabolisme , Maladie de Huntington/anatomopathologie , Souris , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Récepteur trkB/métabolisme , Récepteur trkB/antagonistes et inhibiteurs , Potentialisation à long terme/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Facteur neurotrophique dérivé du cerveau/métabolisme , Modèles animaux de maladie humaine , Cadhérines/métabolisme , Épines dendritiques/métabolisme , Épines dendritiques/anatomopathologie , Neuroprotection , Mâle , Souris de lignée C57BL , Plasticité neuronale , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , Souris knockout
4.
Cell Mol Life Sci ; 81(1): 342, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39123091

RÉSUMÉ

A Disintegrin And Metalloproteinase 10 (ADAM10) plays a pivotal role in shaping neuronal networks by orchestrating the activity of numerous membrane proteins through the shedding of their extracellular domains. Despite its significance in the brain, the specific cellular localization of ADAM10 remains not well understood due to a lack of appropriate tools. Here, using a specific ADAM10 antibody suitable for immunostainings, we observed that ADAM10 is localized to presynapses and especially enriched at presynaptic vesicles of mossy fiber (MF)-CA3 synapses in the hippocampus. These synapses undergo pronounced frequency facilitation of neurotransmitter release, a process that play critical roles in information transfer and neural computation. We demonstrate, that in conditional ADAM10 knockout mice the ability of MF synapses to undergo this type of synaptic plasticity is greatly reduced. The loss of facilitation depends on the cytosolic domain of ADAM10 and association with the calcium sensor synaptotagmin 7 rather than ADAM10's proteolytic activity. Our findings unveil a new role of ADAM10 in the regulation of synaptic vesicle exocytosis.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Protéines membranaires , Souris knockout , Plasticité neuronale , Vésicules synaptiques , Animaux , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Plasticité neuronale/physiologie , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris , Vésicules synaptiques/métabolisme , Souris de lignée C57BL , Synapses/métabolisme , Fibres moussues de l'hippocampe/métabolisme , Hippocampe/métabolisme , Exocytose/physiologie , Terminaisons présynaptiques/métabolisme , Transmission synaptique , Synaptotagmines/métabolisme , Synaptotagmines/génétique
5.
Cell Rep Med ; 5(8): 101686, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39168104

RÉSUMÉ

Resistance to PD-1 blockade in onco-immunotherapy greatly limits its clinical application. T cell immunoglobulin and mucin domain containing-3 (Tim-3), a promising immune checkpoint target, is cleaved by ADAM10/17 to produce its soluble form (sTim-3) in humans, potentially becoming involved in anti-PD-1 resistance. Herein, serum sTim-3 upregulation was observed in non-small cell lung cancer (NSCLC) and various digestive tumors. Notably, serum sTim-3 is further upregulated in non-responding patients undergoing anti-PD-1 therapy for NSCLC and anti-PD-1-resistant cholangiocarcinoma patients. Furthermore, sTim-3 overexpression facilitates tumor progression and confers anti-PD-1 resistance in multiple tumor mouse models. Mechanistically, sTim-3 induces terminal T cell exhaustion and attenuates CD8+ T cell response to PD-1 blockade through carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1). Moreover, the ADAM10 inhibitor GI254023X, which blocks sTim-3 production, reduces tumor progression in Tim-3 humanized mice and reverses anti-PD-1 resistance in human tumor-infiltrating lymphocytes (TILs). Overall, human sTim-3 holds great predictive and therapeutic potential in onco-immunotherapy.


Sujet(s)
Marqueurs biologiques tumoraux , Lymphocytes T CD8+ , Résistance aux médicaments antinéoplasiques , Récepteur cellulaire-2 du virus de l'hépatite A , Récepteur-1 de mort cellulaire programmée , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/antagonistes et inhibiteurs , Humains , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Souris , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/immunologie , Pronostic , Marqueurs biologiques tumoraux/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lignée cellulaire tumorale , Femelle , Mâle , Tumeurs du poumon/immunologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Protéine ADAM10/métabolisme , Souris de lignée C57BL , Épuisement des cellules T
6.
Alzheimers Res Ther ; 16(1): 160, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030577

RÉSUMÉ

BACKGROUND: Alpha-lipoic acid (ALA) has a neuroprotective effect on neurodegenerative diseases. In the clinic, ALA can improve cognitive impairments in patients with Alzheimer's disease (AD) and other dementias. Animal studies have confirmed the anti-amyloidosis effect of ALA, but its underlying mechanism remains unclear. In particular, the role of ALA in amyloid-ß precursor protein (APP) metabolism has not been fully elucidated. OBJECTIVE: To investigate whether ALA can reduce the amyloidogenic effect of APP in a transgenic mouse model of AD, and to study the mechanism underlying this effect. METHODS: ALA was infused into 2-month-old APP23/PS45 transgenic mice for 4 consecutive months and their cognitive function and AD-like pathology were then evaluated. An ALA drug concentration gradient was applied to 20E2 cells in vitro to evaluate its effect on the expression of APP proteolytic enzymes and metabolites. The mechanism by which ALA affects APP processing was studied using GI254023X, an inhibitor of A Disintegrin and Metalloproteinase 10 (ADAM10), as well as the mitochondrial toxic drug carbonyl cyanide m-chlorophenylhydrazone (CCCP). RESULTS: Administration of ALA ameliorated amyloid plaque neuropathology in the brain tissue of APP23/PS45 mice and reduced learning and memory impairment. ALA also increased the expression of ADAM10 in 20E2 cells and the non-amyloidogenic processing of APP to produce the 83 amino acid C-terminal fragment (C83). In addition to activating autophagy, ALA also significantly promoted mitophagy. BNIP3L-knockdown reduced the mat/pro ratio of ADAM10. By using CCCP, ALA was found to regulate BNIP3L-mediated mitophagy, thereby promoting the α-cleavage of APP. CONCLUSIONS: The enhanced α-secretase cleavage of APP by ADAM10 is the primary mechanism through which ALA ameliorates the cognitive deficits in APP23/PS45 transgenic mice. BNIP3L-mediated mitophagy contributes to the anti-amyloid properties of ALA by facilitating the maturation of ADAM10. This study provides novel experimental evidence for the treatment of AD with ALA.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Précurseur de la protéine bêta-amyloïde , Dysfonctionnement cognitif , Souris transgéniques , Mitophagie , Acide lipoïque , Animaux , Acide lipoïque/pharmacologie , Mitophagie/effets des médicaments et des substances chimiques , Protéine ADAM10/métabolisme , Souris , Précurseur de la protéine bêta-amyloïde/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme , Amyloid precursor protein secretases/métabolisme , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Modèles animaux de maladie humaine , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Neuroprotecteurs/pharmacologie , Souris de lignée C57BL , Mâle
7.
J Neuroinflammation ; 21(1): 167, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956605

RÉSUMÉ

BACKGROUND: Deposition of amyloid ß, which is produced by amyloidogenic cleavage of APP by ß- and γ-secretase, is one of the primary hallmarks of AD pathology. APP can also be processed by α- and γ-secretase sequentially, to generate sAPPα, which has been shown to be neuroprotective by promoting neurite outgrowth and neuronal survival, etc. METHODS: The global expression profiles of miRNA in blood plasma samples taken from 11 AD patients as well as from 14 age and sex matched cognitively normal volunteers were analyzed using miRNA-seq. Then, overexpressed miR-140 and miR-122 both in vivo and in vitro, and knock-down of the endogenous expression of miR-140 and miR-122 in vitro. Used a combination of techniques, including molecular biology, immunohistochemistry, to detect the impact of miRNAs on AD pathology. RESULTS: In this study, we identified that two miRNAs, miR-140-3p and miR-122-5p, both targeting ADAM10, the main α-secretase in CNS, were upregulated in the blood plasma of AD patients. Overexpression of these two miRNAs in mouse brains induced cognitive decline in wild type C57BL/6J mice as well as exacerbated dyscognition in APP/PS1 mice. Although significant changes in APP and total Aß were not detected, significantly downregulated ADAM10 and its non-amyloidogenic product, sAPPα, were observed in the mouse brains overexpressing miR-140/miR-122. Immunohistology analysis revealed increased neurite dystrophy that correlated with the reduced microglial chemotaxis in the hippocampi of these mice, independent of the other two ADAM10 substrates (neuronal CX3CL1 and microglial TREM2) that were involved in regulating the microglial immunoactivity. Further in vitro analysis demonstrated that both the reduced neuritic outgrowth of mouse embryonic neuronal cells overexpressing miR-140/miR-122 and the reduced Aß phagocytosis in microglia cells co-cultured with HT22 cells overexpressing miR-140/miR-122 could be rescued by overexpressing the specific inhibitory sequence of miR-140/miR-122 TuD as well as by addition of sAPPα, rendering these miRNAs as potential therapeutic targets. CONCLUSIONS: Our results suggested that neuroprotective sAPPα was a key player in the neuropathological progression induced by dysregulated expression of miR-140 and miR-122. Targeting these miRNAs might serve as a promising therapeutic strategy in AD treatment.


Sujet(s)
Maladie d'Alzheimer , Chimiotaxie , Souris de lignée C57BL , microARN , Microglie , microARN/métabolisme , microARN/génétique , Animaux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/génétique , Souris , Humains , Microglie/métabolisme , Microglie/anatomopathologie , Mâle , Chimiotaxie/physiologie , Femelle , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Souris transgéniques , Sujet âgé , Régulation de l'expression des gènes
8.
Acta Neuropathol ; 148(1): 2, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38980441

RÉSUMÉ

Proteolytic cell surface release ('shedding') of the prion protein (PrP), a broadly expressed GPI-anchored glycoprotein, by the metalloprotease ADAM10 impacts on neurodegenerative and other diseases in animal and in vitro models. Recent studies employing the latter also suggest shed PrP (sPrP) to be a ligand in intercellular communication and critically involved in PrP-associated physiological tasks. Although expectedly an evolutionary conserved event, and while soluble forms of PrP are present in human tissues and body fluids, for the human body neither proteolytic PrP shedding and its cleavage site nor involvement of ADAM10 or the biological relevance of this process have been demonstrated thus far. In this study, cleavage site prediction and generation (plus detailed characterization) of sPrP-specific antibodies enabled us to identify PrP cleaved at tyrosin 226 as the physiological and apparently strictly ADAM10-dependent shed form in humans. Using cell lines, neural stem cells and brain organoids, we show that shedding of human PrP can be stimulated by PrP-binding ligands without targeting the protease, which may open novel therapeutic perspectives. Site-specific antibodies directed against human sPrP also detect the shed form in brains of cattle, sheep and deer, hence in all most relevant species naturally affected by fatal and transmissible prion diseases. In human and animal prion diseases, but also in patients with Alzheimer`s disease, sPrP relocalizes from a physiological diffuse tissue pattern to intimately associate with extracellular aggregated deposits of misfolded proteins characteristic for the respective pathological condition. Findings and research tools presented here will accelerate novel insight into the roles of PrP shedding (as a process) and sPrP (as a released factor) in neurodegeneration and beyond.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Maladies neurodégénératives , Humains , Protéine ADAM10/métabolisme , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Amyloid precursor protein secretases/métabolisme , Animaux , Protéines prion/métabolisme , Protéines membranaires/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Anticorps
9.
Infect Immun ; 92(8): e0013324, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38953668

RÉSUMÉ

Staphylococcus aureus α-hemolysin (Hla) is a pore-forming toxin critical for the pathogenesis of skin and soft tissue infections, which causes the pathognomonic lesion of cutaneous necrosis (dermonecrosis) in mouse models. To determine the mechanism by which dermonecrosis develops during S. aureus skin infection, mice were given control serum, Hla-neutralizing antiserum, or an inhibitor of Hla receptor [A-disintegrin and metalloprotease 10 (ADAM10) inhibitor] followed by subcutaneous infection by S. aureus, and the lesions were evaluated using immunohistochemistry and immunofluorescence. Hla induced apoptosis in the vascular endothelium at 6 hours post-infection (hpi), followed by apoptosis in keratinocytes at 24 hpi. The loss of vascular endothelial (VE)-cadherin expression preceded the loss of epithelial-cadherin expression. Hla also induced hypoxia in the keratinocytes at 24 hpi following vascular injury. Treatment with Hla-neutralizing antibody or ADAM10 inhibitor attenuated early cleavage of VE-cadherin, cutaneous hypoxia, and dermonecrosis. These findings suggest that Hla-mediated vascular injury with cutaneous hypoxia underlies the pathogenesis of S. aureus-induced dermonecrosis.


Sujet(s)
Protéine ADAM10 , Toxines bactériennes , Cadhérines , Hémolysines , Kératinocytes , Nécrose , Staphylococcus aureus , Animaux , Hémolysines/métabolisme , Hémolysines/toxicité , Souris , Toxines bactériennes/toxicité , Toxines bactériennes/métabolisme , Staphylococcus aureus/pathogénicité , Kératinocytes/microbiologie , Kératinocytes/métabolisme , Protéine ADAM10/métabolisme , Cadhérines/métabolisme , Apoptose , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Amyloid precursor protein secretases/métabolisme , Antigènes CD/métabolisme , Protéines membranaires/métabolisme , Infections cutanées à staphylocoques/microbiologie , Infections cutanées à staphylocoques/anatomopathologie , Infections cutanées à staphylocoques/immunologie , Peau/anatomopathologie , Peau/microbiologie , Femelle , Endothélium vasculaire/anatomopathologie , Endothélium vasculaire/microbiologie , Endothélium vasculaire/métabolisme , Infections à staphylocoques/microbiologie , Infections à staphylocoques/immunologie , Infections à staphylocoques/anatomopathologie , Modèles animaux de maladie humaine
10.
Nutrients ; 16(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38931216

RÉSUMÉ

Atherosclerosis is one of the most important causes of cardiovascular diseases. A disintegrin and metalloprotease (ADAM)10 and ADAM17 have been identified as important regulators of inflammation in recent years. Our study investigated the effect of inhibiting these enzymes with selective inhibitor and propolis on atherosclerosis. In our study, C57BL/6J mice (n = 16) were used in the control and sham groups. In contrast, ApoE-/- mice (n = 48) were used in the case, water extract of propolis (WEP), ethanolic extract of propolis (EEP), GW280264X (GW-synthetic inhibitor), and solvent (DMSO and ethanol) groups. The control group was fed a control diet, and all other groups were fed a high-cholesterol diet for 16 weeks. WEP (400 mg/kg/day), EEP (200 mg/kg/day), and GW (100 µg/kg/day) were administered intraperitoneally for the last four weeks. Animals were sacrificed, and blood, liver, aortic arch, and aortic root tissues were collected. In serum, total cholesterol (TC), triglycerides (TGs), and glucose (Glu) were measured by enzymatic colorimetric method, while interleukin-1ß (IL-1ß), paraoxonase-1 (PON-1), and lipoprotein-associated phospholipase-A2 (Lp-PLA2) were measured by ELISA. Tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), myeloperoxidase (MPO), interleukin-6 (IL-6), interleukin-10 (IL-10), and interleukin-12 (IL-12) levels were measured in aortic arch by ELISA and ADAM10/17 activities were measured fluorometrically. In addition, aortic root and liver tissues were examined histopathologically and immunohistochemically (ADAM10 and sortilin primary antibody). In the WEP, EEP, and GW groups compared to the case group, TC, TG, TNF-α, IL-1ß, IL-6, IL-12, PLA2, MPO, ADAM10/17 activities, plaque burden, lipid accumulation, ADAM10, and sortilin levels decreased, while IL-10 and PON-1 levels increased (p < 0.003). Our study results show that propolis can effectively reduce atherosclerosis-related inflammation and dyslipidemia through ADAM10/17 inhibition.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Dyslipidémies , Inflammation , Souris de lignée C57BL , Propolis , Animaux , Protéine ADAM10/métabolisme , Propolis/pharmacologie , Inflammation/prévention et contrôle , Dyslipidémies/traitement médicamenteux , Dyslipidémies/étiologie , Souris , Mâle , Amyloid precursor protein secretases/métabolisme , Athérosclérose/prévention et contrôle , Athérosclérose/étiologie , Cholestérol alimentaire/effets indésirables , Alimentation riche en graisse/effets indésirables , Protéines membranaires/métabolisme , Modèles animaux de maladie humaine
11.
Epigenetics ; 19(1): 2367385, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38899429

RÉSUMÉ

This research investigates the intricate dynamics of DNA methylation in the hours following CD8+ T cell activation, during a critical yet understudied temporal window. DNA methylation is an epigenetic modification central to regulation of gene expression and directing immune responses. Our investigation spanned 96-h post-activation and unveils a nuanced tapestry of global and site-specific methylation changes. We identified 15,626 significant differentially methylated CpGs spread across the genome, with the most significant changes occurring within the genes ADAM10, ICA1, and LAPTM5. While many changes had modest effect sizes, approximately 120 CpGs exhibited a log2FC above 1.5, with cell activation and proliferation pathways the most affected. Relatively few of the differentially methylated CpGs occurred along adjacent gene regions. The exceptions were seven differentially methylated gene regions, with the Human T cell Receptor Alpha Joining Genes demonstrating consistent methylation change over a 3kb window. We also investigated whether an inflammatory environment could alter DNA methylation during activation, with proliferating cells exposed to the oxidant glycine chloramine. No substantial differential methylation was observed in this context. The temporal perspective of early activation adds depth to the evolving field of epigenetic immunology, offering insights with implications for therapeutic innovation and expanding our understanding of epigenetic modulation in immune function.


Sujet(s)
Lymphocytes T CD8+ , Prolifération cellulaire , Méthylation de l'ADN , Activation des lymphocytes , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/immunologie , Humains , Ilots CpG , Épigenèse génétique , Protéine ADAM10/génétique , Protéine ADAM10/métabolisme , Protéines membranaires/génétique
12.
Cell Commun Signal ; 22(1): 322, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38863060

RÉSUMÉ

Bone resorption is driven through osteoclast differentiation by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-Β ligand (RANKL). We noted that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 are downregulated at the expression level during osteoclast differentiation of the murine monocytic cell line RAW264.7 in response to RANKL. Both proteinases are well known to shed a variety of single-pass transmembrane molecules from the cell surface. We further showed that inhibitors of ADAM10 or ADAM17 promote osteoclastic differentiation and furthermore enhance the surface expression of receptors for RANKL and M-CSF on RAW264.7 cells. Using murine bone marrow-derived monocytic cells (BMDMCs), we demonstrated that a genetic deficiency of ADAM17 or its required regulator iRhom2 leads to increased osteoclast development in response to M-CSF and RANKL stimulation. Moreover, ADAM17-deficient osteoclast precursor cells express increased levels of the receptors for RANKL and M-CSF. Thus, ADAM17 negatively regulates osteoclast differentiation, most likely through shedding of these receptors. To assess the time-dependent contribution of ADAM10, we blocked this proteinase by adding a specific inhibitor on day 0 of BMDMC stimulation with M-CSF or on day 7 of subsequent stimulation with RANKL. Only ADAM10 inhibition beginning on day 7 increased the size of developing osteoclasts indicating that ADAM10 suppresses osteoclast differentiation at a later stage. Finally, we could confirm our findings in human peripheral blood mononuclear cells (PBMCs). Thus, downregulation of either ADAM10 or ADAM17 during osteoclast differentiation may represent a novel regulatory mechanism to enhance their differentiation process. Enhanced bone resorption is a critical issue in osteoporosis and is driven through osteoclast differentiation by specific osteogenic mediators. The present study demonstrated that the metalloproteinases ADAM17 and ADAM10 critically suppress osteoclast development. This was observed for a murine cell line, for isolated murine bone marrow cells and for human blood cells by either preferential inhibition of the proteinases or by gene knockout. As a possible mechanism, we studied the surface expression of critical receptors for osteogenic mediators on developing osteoclasts. Our findings revealed that the suppressive effects of ADAM17 and ADAM10 on osteoclastogenesis can be explained in part by the proteolytic cleavage of surface receptors by ADAM10 and ADAM17, which reduces the sensitivity of these cells to osteogenic mediators. We also observed that osteoclast differentiation was associated with the downregulation of ADAM10 and ADAM17, which reduced their suppressive effects. We therefore propose that this downregulation serves as a feedback loop for enhancing osteoclast development.


Sujet(s)
Protéine ADAM10 , Protéine ADAM17 , Amyloid precursor protein secretases , Différenciation cellulaire , Régulation négative , Protéines membranaires , Ostéoclastes , Ligand de RANK , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Ostéoclastes/métabolisme , Ostéoclastes/cytologie , Animaux , Différenciation cellulaire/génétique , Souris , Régulation négative/génétique , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Humains , Ligand de RANK/métabolisme , Cellules RAW 264.7 , Facteur de stimulation des colonies de macrophages/pharmacologie , Facteur de stimulation des colonies de macrophages/métabolisme , Souris de lignée C57BL
13.
JCI Insight ; 9(11)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38855867

RÉSUMÉ

In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.


Sujet(s)
Protéine ADAM10 , Antigènes CD , Arthrite expérimentale , Polyarthrite rhumatoïde , Cadhérines , Cellules endothéliales , Récepteurs de la sphingosine-1-phosphate , Animaux , Cadhérines/métabolisme , Récepteurs de la sphingosine-1-phosphate/métabolisme , Récepteurs de la sphingosine-1-phosphate/génétique , Souris , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Antigènes CD/métabolisme , Antigènes CD/génétique , Cellules endothéliales/métabolisme , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Amyloid precursor protein secretases/métabolisme , Transduction du signal , Souris knockout , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Mâle , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Lysophospholipides/métabolisme , Perméabilité capillaire , Femelle
14.
J Biol Chem ; 300(7): 107480, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38897568

RÉSUMÉ

Phospholipase A2 receptor 1 (PLA2R1) is a 180-kDa transmembrane protein that plays a role in inflammation and cancer and is the major autoantigen in membranous nephropathy, a rare but severe autoimmune kidney disease. A soluble form of PLA2R1 has been detected in mouse and human serum. It is likely produced by proteolytic shedding of membrane-bound PLA2R1 but the mechanism is unknown. Here, we show that human PLA2R1 is cleaved by A Disintegrin And Metalloprotease 10 (ADAM10) and ADAM17 in HEK293 cells, mouse embryonic fibroblasts, and human podocytes. By combining site-directed mutagenesis and sequencing, we determined the exact cleavage site within the extracellular juxtamembrane stalk of human PLA2R1. Orthologs and paralogs of PLA2R1 are also shed. By using pharmacological inhibitors and genetic approaches with RNA interference and knock-out cellular models, we identified a major role of ADAM10 in the constitutive shedding of PLA2R1 and a dual role of ADAM10 and ADAM17 in the stimulated shedding. We did not observe evidence for cleavage by ß- or γ-secretase, suggesting that PLA2R1 may not be a substrate for regulated intramembrane proteolysis. PLA2R1 shedding occurs constitutively and can be triggered by the calcium ionophore ionomycin, the protein kinase C activator PMA, cytokines, and lipopolysaccharides, in vitro and in vivo. Altogether, our results show that PLA2R1 is a novel substrate for ADAM10 and ADAM17, producing a soluble form that is increased in inflammatory conditions and likely exerts various functions in physiological and pathophysiological conditions including inflammation, cancer, and membranous nephropathy.


Sujet(s)
Protéine ADAM10 , Protéine ADAM17 , Amyloid precursor protein secretases , Protéines membranaires , Récepteurs à la phospholipase A2 , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Humains , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Animaux , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris , Cellules HEK293 , Récepteurs à la phospholipase A2/métabolisme , Récepteurs à la phospholipase A2/génétique , Podocytes/métabolisme , Protéolyse , Domaines protéiques , Ionomycine/pharmacologie
15.
Microbiologyopen ; 13(3): e23, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38867416

RÉSUMÉ

The G protein-coupled estrogen receptor, also known as GPER1 or originally GPR30, is found in various tissues, indicating its diverse functions. It is typically present in immune cells, suggesting its role in regulating immune responses to infectious diseases. Our previous studies have shown that G-1, a selective GPER agonist, can limit the pathogenesis mediated by Staphylococcus aureus alpha-hemolysin (Hla). It aids in clearing bacteria in a mouse skin infection model and restricts the surface display of the Hla receptor, ADAM10 (a disintegrin and metalloprotease 10) in HaCaT keratinocytes. In this report, we delve into the modulation of GPER in human immune cells in relation to the NLRP3 inflammasome. We used macrophage-like differentiated THP-1 cells for our study. We found that treating these cells with G-1 reduces ATP release, decreases the activity of the caspase-1 enzyme, and lessens cell death following Hla intoxication. This is likely due to the reduced levels of ADAM10 and NLRP3 proteins, as well as the decreased display of the ADAM10 receptor in the G-1-treated THP-1 cells. Our studies, along with our previous work, suggest the potential therapeutic use of G-1 in reducing Hla susceptibility in humans. This highlights the importance of GPER in immune regulation and its potential as a therapeutic target.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Toxines bactériennes , Hémolysines , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Staphylococcus aureus , Protéine ADAM10/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Humains , Récepteurs couplés aux protéines G/agonistes , Récepteurs couplés aux protéines G/métabolisme , Hémolysines/métabolisme , Inflammasomes/métabolisme , Toxines bactériennes/métabolisme , Cellules THP-1 , Récepteurs des oestrogènes/métabolisme , Amyloid precursor protein secretases/métabolisme , Staphylococcus aureus/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/agonistes , Caspase-1/métabolisme , Adénosine triphosphate/métabolisme , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/microbiologie , Dipeptides , Acides hydroxamiques
16.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230481, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-38853546

RÉSUMÉ

Group I metabotropic glutamate receptors (Gp1-mGluRs) exert a host of effects on cellular functions, including enhancement of protein synthesis and the associated facilitation of long-term potentiation (LTP) and induction of long-term depression (LTD). However, the complete cascades of events mediating these events are not fully understood. Gp1-mGluRs trigger α-secretase cleavage of amyloid precursor protein, producing soluble amyloid precursor protein-α (sAPPα), a known regulator of LTP. However, the α-cleavage of APP has not previously been linked to Gp1-mGluR's actions. Using rat hippocampal slices, we found that the α-secretase inhibitor tumour necrosis factor-alpha protease inhibitor-1, which inhibits both disintegrin and metalloprotease 10 (ADAM10) and 17 (ADAM17) activity, blocked or reduced the ability of the Gp1-mGluR agonist (R,S)-3,5-dihydroxyphenylglycine (DHPG) to stimulate protein synthesis, metaplastically prime future LTP and elicit sub-maximal LTD. In contrast, the specific ADAM10 antagonist GI254023X did not affect the regulation of plasticity, suggesting that ADAM17 but not ADAM10 is involved in mediating these effects of DHPG. However, neither drug affected LTD that was strongly induced by either high-concentration DHPG or paired-pulse synaptic stimulation. Our data suggest that moderate Gp1-mGluR activation triggers α-secretase sheddase activity targeting APP or other membrane-bound proteins as part of a more complex signalling cascade than previously envisioned. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Sujet(s)
Amyloid precursor protein secretases , Hippocampe , Potentialisation à long terme , Dépression synaptique à long terme , Biosynthèse des protéines , Récepteurs métabotropes au glutamate , Animaux , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Rats , Récepteurs métabotropes au glutamate/métabolisme , Dépression synaptique à long terme/physiologie , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Protéine ADAM17/métabolisme , Protéine ADAM10/métabolisme , Rat Sprague-Dawley , Mâle , Méthoxyhydroxyphénylglycol/analogues et dérivés , Méthoxyhydroxyphénylglycol/pharmacologie , Protéines membranaires/métabolisme
17.
Adipocyte ; 13(1): 2339418, 2024 12.
Article de Anglais | MEDLINE | ID: mdl-38706095

RÉSUMÉ

A Disintegrin And Metalloproteinase domain-containing protein 10 (ADAM10), is involved in several metabolic and inflammatory pathways. We speculated that ADAM10 plays a modulatory role in adipose tissue inflammation and metabolism. To this end, we studied adipose tissue-specific ADAM10 knock-out mice (aKO). While young, regular chow diet-fed aKO mice showed increased insulin sensitivity, following prolonged (33 weeks) high-fat diet (HFD) exposure, aKO mice developed obesity and insulin resistance. Compared to controls, aKO mice showed less inflammatory adipokine profile despite the significant increase in adiposity. In brown adipose tissue, aKO mice on HFD had changes in CD8+ T cell populations indicating a lesser inflammatory pattern. Following HFD, both aKO and control littermates demonstrated decreased adipose tissue pro-inflammatory macrophages, and increased anti-inflammatory accumulation, without differences between the genotypes. Collectively, our observations indicate that selective deletion of ADAM10 in adipocytes results in a mitigated inflammatory response, leading to increased insulin sensitivity in young mice fed with regular diet. This state of insulin sensitivity, following prolonged HFD, facilitates energy storage resulting in increased fat accumulation which ultimately leads to the development of a phenotype of obesity and insulin resistance. In conclusion, the data indicate that ADAM10 has a modulatory effect of inflammation and whole-body energy metabolism.


Sujet(s)
Protéine ADAM10 , Tissu adipeux , Alimentation riche en graisse , Souris knockout , Animaux , Mâle , Souris , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Adipocytes/métabolisme , Tissu adipeux/métabolisme , Amyloid precursor protein secretases/métabolisme , Alimentation riche en graisse/effets indésirables , Inflammation/métabolisme , Insulinorésistance , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Obésité/métabolisme , Obésité/étiologie , Phénotype
18.
Arch Dermatol Res ; 316(6): 269, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38795191

RÉSUMÉ

Skin cutaneous melanoma (SKCM) is the skin malignancy with the highest mortality rate, and its morbidity rate is on the rise worldwide. Smoking is an independent marker of poor prognosis in melanoma. The α5-nicotinic acetylcholine receptor (α5-nAChR), one of the receptors for nicotine, is involved in the proliferation, migration and invasion of SKCM cells. Nicotine has been reported to promote the expression of a disintegrin and metalloproteinase 10 (ADAM10), which is the key gene involved in melanoma progression. Here, we explored the link between α5-nAChR and ADAM10 in nicotine-associated cutaneous melanoma. α5-nAChR expression was correlated with ADAM10 expression and lower survival in SKCM. α5-nAChR mediated nicotine-induced ADAM10 expression via STAT3. The α5-nAChR/ADAM10 signaling axis was involved in the stemness and migration of SKCM cells. Furthermore, α5-nAChR expression was associated with ADAM10 expression, EMT marker expression and stemness marker expression in nicotine-related mice homograft tissues. These results suggest the role of the α5-nAChR/ADAM10 signaling pathway in nicotine-induced melanoma progression.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Mouvement cellulaire , Évolution de la maladie , Mélanome , Protéines membranaires , Nicotine , Récepteurs nicotiniques , Facteur de transcription STAT-3 , Transduction du signal , Tumeurs cutanées , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/induit chimiquement , Tumeurs cutanées/métabolisme , Facteur de transcription STAT-3/métabolisme , Humains , Animaux , Amyloid precursor protein secretases/métabolisme , Amyloid precursor protein secretases/génétique , Nicotine/effets indésirables , Transduction du signal/effets des médicaments et des substances chimiques , Mélanome/anatomopathologie , Mélanome/métabolisme , Mélanome/induit chimiquement , Souris , Récepteurs nicotiniques/métabolisme , Récepteurs nicotiniques/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Mâle , , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques
19.
Biochem Pharmacol ; 225: 116328, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815628

RÉSUMÉ

Early stages of diabetes are characterized by elevations of insulin and glucose concentrations. Both factors stimulate reactive oxygen species (ROS) production, leading to impairments in podocyte function and disruption of the glomerular filtration barrier. Podocytes were recently shown to be an important source of αKlotho (αKL) expression. Low blood Klotho concentrations are also associated with an increase in albuminuria, especially in patients with diabetes. We investigated whether ADAM10, which is known to cleave αKL, is activated in glomeruli and podocytes under diabetic conditions and the potential mechanisms by which ADAM10 mediates ROS production and disturbances of the glomerular filtration barrier. In cultured human podocytes, high glucose increased ADAM10 expression, shedding, and activity, NADPH oxidase activity, ROS production, and albumin permeability. These effects of glucose were inhibited when cells were pretreated with an ADAM10 inhibitor or transfected with short-hairpin ADAM10 (shADAM10) or after the addition soluble Klotho. We also observed increases in ADAM10 activity, NOX4 expression, NADPH oxidase activity, and ROS production in αKL-depleted podocytes. This was accompanied by an increase in albumin permeability in shKL-expressing podocytes. The protein expression and activity of ADAM10 also increased in isolated glomeruli and urine samples from diabetic rats. Altogether, these results reveal a new mechanism by which hyperglycemia in diabetes increases albumin permeability through ADAM10 activation and an increase in oxidative stress via NOX4 enzyme activation. Moreover, αKlotho downregulates ADAM10 activity and supports redox balance, consequently protecting the slit diaphragm of podocyteσ under hyperglycemic conditions.


Sujet(s)
Protéine ADAM10 , Amyloid precursor protein secretases , Diabète expérimental , Glucuronidase , Protéines Klotho , Protéines membranaires , Podocytes , Espèces réactives de l'oxygène , Podocytes/métabolisme , Podocytes/effets des médicaments et des substances chimiques , Protéines Klotho/métabolisme , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Espèces réactives de l'oxygène/métabolisme , Humains , Animaux , Glucuronidase/métabolisme , Glucuronidase/génétique , Amyloid precursor protein secretases/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Rats , Mâle , Diabète expérimental/métabolisme , NADPH Oxidase 4/métabolisme , NADPH Oxidase 4/génétique , NADPH oxidase/métabolisme , Cellules cultivées , Glucose/métabolisme , Rat Sprague-Dawley
20.
Cell Mol Gastroenterol Hepatol ; 18(3): 101365, 2024.
Article de Anglais | MEDLINE | ID: mdl-38797477

RÉSUMÉ

BACKGROUND & AIMS: Reversion-inducing cysteine-rich protein with Kazal motifs (RECK) is an extracellular matrix regulator with anti-fibrotic effects. However, its expression and role in metabolic dysfunction-associated steatohepatitis (MASH) and hepatic fibrosis are poorly understood. METHODS: We generated a novel transgenic mouse model with RECK overexpression specifically in hepatocytes to investigate its role in Western diet (WD)-induced liver disease. Proteomic analysis and in vitro studies were performed to mechanistically link RECK to hepatic inflammation and fibrosis. RESULTS: Our results show that RECK expression is significantly decreased in liver biopsies from human patients diagnosed with MASH and correlated negatively with severity of metabolic dysfunction-associated steatotic liver disease (MASLD) and fibrosis. Similarly, RECK expression is downregulated in WD-induced MASH in wild-type mice. Hepatocyte-specific RECK overexpression significantly reduced hepatic pathology in WD-induced liver injury. Proteomic analysis highlighted changes in extracellular matrix and cell-signaling proteins. In vitro mechanistic studies linked RECK induction to reduced ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10) and ADAM17 activity, amphiregulin release, epidermal growth factor receptor activation, and stellate cell activation. CONCLUSION: Our in vivo and mechanistic in vitro studies reveal that RECK is a novel upstream regulator of inflammation and fibrosis in the diseased liver, its induction is hepatoprotective, and thus highlights its potential as a novel therapeutic in MASH.


Sujet(s)
Modèles animaux de maladie humaine , Protéines liées au GPI , Hépatocytes , Souris transgéniques , Animaux , Protéines liées au GPI/métabolisme , Protéines liées au GPI/génétique , Humains , Souris , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Protéomique , Foie/métabolisme , Foie/anatomopathologie , Protéine ADAM10/métabolisme , Protéine ADAM10/génétique , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Protéine ADAM17/métabolisme , Protéine ADAM17/génétique , Mâle , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Amyloid precursor protein secretases/métabolisme , Récepteurs ErbB/métabolisme , Transduction du signal , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Amphiréguline/métabolisme , Amphiréguline/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE