Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 210
1.
Cell Commun Signal ; 22(1): 322, 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38863060

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.


ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases , Cell Differentiation , Down-Regulation , Membrane Proteins , Osteoclasts , RANK Ligand , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Osteoclasts/metabolism , Osteoclasts/cytology , Animals , Cell Differentiation/genetics , Mice , Down-Regulation/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Humans , RANK Ligand/metabolism , RAW 264.7 Cells , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/metabolism , Mice, Inbred C57BL
2.
JCI Insight ; 9(11)2024 Jun 10.
Article En | MEDLINE | ID: mdl-38855867

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.


ADAM10 Protein , Antigens, CD , Arthritis, Experimental , Arthritis, Rheumatoid , Cadherins , Endothelial Cells , Sphingosine-1-Phosphate Receptors , Animals , Cadherins/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Mice , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Antigens, CD/metabolism , Antigens, CD/genetics , Endothelial Cells/metabolism , Humans , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/genetics , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Signal Transduction , Mice, Knockout , Membrane Proteins/metabolism , Membrane Proteins/genetics , Male , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Capillary Permeability , Female
3.
Adipocyte ; 13(1): 2339418, 2024 Dec.
Article En | MEDLINE | ID: mdl-38706095

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.


ADAM10 Protein , Adipose Tissue , Diet, High-Fat , Mice, Knockout , Animals , Male , Mice , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Adipocytes/metabolism , Adipose Tissue/metabolism , Amyloid Precursor Protein Secretases/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Membrane Proteins/genetics , Obesity/metabolism , Obesity/etiology , Phenotype
4.
Arch Dermatol Res ; 316(6): 269, 2024 May 25.
Article En | MEDLINE | ID: mdl-38795191

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.


ADAM10 Protein , Amyloid Precursor Protein Secretases , Cell Movement , Disease Progression , Melanoma , Membrane Proteins , Nicotine , Receptors, Nicotinic , STAT3 Transcription Factor , Signal Transduction , Skin Neoplasms , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Skin Neoplasms/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Humans , Animals , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Nicotine/adverse effects , Signal Transduction/drug effects , Melanoma/pathology , Melanoma/metabolism , Melanoma/chemically induced , Mice , Receptors, Nicotinic/metabolism , Receptors, Nicotinic/genetics , Cell Line, Tumor , Cell Movement/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Male , Melanoma, Cutaneous Malignant , Female , Cell Proliferation/drug effects
5.
Biochem Pharmacol ; 225: 116328, 2024 Jul.
Article En | MEDLINE | ID: mdl-38815628

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.


ADAM10 Protein , Amyloid Precursor Protein Secretases , Diabetes Mellitus, Experimental , Glucuronidase , Klotho Proteins , Membrane Proteins , Podocytes , Reactive Oxygen Species , Podocytes/metabolism , Podocytes/drug effects , Klotho Proteins/metabolism , ADAM10 Protein/metabolism , ADAM10 Protein/genetics , Reactive Oxygen Species/metabolism , Humans , Animals , Glucuronidase/metabolism , Glucuronidase/genetics , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Rats , Male , Diabetes Mellitus, Experimental/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidases/metabolism , Cells, Cultured , Glucose/metabolism , Rats, Sprague-Dawley
6.
Nat Commun ; 15(1): 541, 2024 Jan 15.
Article En | MEDLINE | ID: mdl-38225245

Efferocytic clearance of apoptotic cells in general, and T cells in particular, is required for tissue and immune homeostasis. Transmembrane mucins are extended glycoproteins highly expressed in the cell glycocalyx that function as a barrier to phagocytosis. Whether and how mucins may be regulated during cell death to facilitate efferocytic corpse clearance is not well understood. Here we show that normal and transformed human T cells express a subset of mucins which are rapidly and selectively removed from the cell surface during apoptosis. This process is mediated by the ADAM10 sheddase, the activity of which is associated with XKR8-catalyzed flipping of phosphatidylserine to the outer leaflet of the plasma membrane. Mucin clearance enhances uptake of apoptotic T cells by macrophages, confirming mucins as an enzymatically-modulatable barrier to efferocytosis. Together these findings demonstrate a glycocalyx regulatory pathway with implications for therapeutic intervention in the clearance of normal and transformed apoptotic T cells.


Efferocytosis , Mucins , Humans , T-Lymphocytes/metabolism , Apoptosis , Phagocytosis , ADAM10 Protein/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases
7.
Cell Mol Biol (Noisy-le-grand) ; 69(13): 167-173, 2023 Dec 10.
Article En | MEDLINE | ID: mdl-38158671

APP, well-studied in the development of Alzheimer's disease, has been recently identified as the key gene correlated with TSCC. Here, we investigate the function of APP and its proteolytic cleavage by ADAM10 in the pathogenesis of TSCC. A total of 63 TSCC patients and 30 healthy controls were included and the results of IHC assay showed high expressions of ADAM10 and APP in TSCC tissues compared to paired para-carcinoma tissues. Interestingly, APP expression in TSCC patients was correlated with ADAM10 expression and their combined expression was related to the poor patients' survival. We found that APP was ɑ-cleaved in TSCC cells to form sAPPα, and the serum level of sAPPα but not sAPPß in TSCC patients was higher than healthy controls. Both overexpression with full-length APP and sAPPα promoted TSCC cell proliferation, migration and invasion. Downregulation of APP or ADAM10 by siRNA decreased the generation of sAPPα and inhibited the activity of ERK1/2 and p38 pathways, thereby reducing TSCC cell proliferation, migration and invasion. Treatment with ERK1/2 or p38 agonist or sAPPα overexpression reversed the effects of APP or ADAM10 knockdown. In conclusion, our data demonstrated the pathogenic roles of APP cleaved by ADAM10 to activate ERK1/2 and p38 pathways in TSCC cells. Both high expressions of ADAM10 and APP were related to poor prognosis. Targeting APP cleaved by ADAM10 might be a potential strategy in TSCC treatment.


Carcinoma, Squamous Cell , Tongue Neoplasms , Humans , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Tongue Neoplasms/genetics , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , ADAM10 Protein/genetics , Cell Proliferation , Tongue/metabolism , Tongue/pathology , Cell Line, Tumor , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism
8.
PLoS Pathog ; 19(11): e1011759, 2023 Nov.
Article En | MEDLINE | ID: mdl-37967063

Hepatitis C virus (HCV) exploits the four entry factors CD81, scavenger receptor class B type I (SR-BI, also known as SCARB1), occludin, and claudin-1 as well as the co-factor epidermal growth factor receptor (EGFR) to infect human hepatocytes. Here, we report that the disintegrin and matrix metalloproteinase 10 (ADAM10) associates with CD81, SR-BI, and EGFR and acts as HCV host factor. Pharmacological inhibition, siRNA-mediated silencing and genetic ablation of ADAM10 reduced HCV infection. ADAM10 was dispensable for HCV replication but supported HCV entry and cell-to-cell spread. Substrates of the ADAM10 sheddase including epidermal growth factor (EGF) and E-cadherin, which activate EGFR family members, rescued HCV infection of ADAM10 knockout cells. ADAM10 did not influence infection with other enveloped RNA viruses such as alphaviruses and a common cold coronavirus. Collectively, our study reveals a critical role for the sheddase ADAM10 as a HCV host factor, contributing to EGFR family member transactivation and as a consequence to HCV uptake.


Hepacivirus , Hepatitis C , Humans , Hepacivirus/physiology , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Virus Internalization , Carrier Proteins , ErbB Receptors/metabolism , Tetraspanin 28/genetics , Tetraspanin 28/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism
9.
Platelets ; 34(1): 2288213, 2023 Dec.
Article En | MEDLINE | ID: mdl-38031964

Platelet-specific collagen receptor glycoprotein (GP)VI is stable on the surface of circulating platelets but undergoes ectodomain cleavage on activated platelets. Activation-dependent GPVI metalloproteolysis is primarily mediated by A Disintegrin And Metalloproteinase (ADAM) 10. Regulation of platelet ADAMs activity is not well-defined however Tissue Inhibitors of Metalloproteinases (TIMPs) may play a role. As levels of TIMPs on platelets and the control of ADAMs-mediated shedding by TIMPs has not been evaluated, we quantified the levels of TIMPs on the surface of resting and activated platelets from healthy donors by flow cytometry and multiplex ELISA. Variable levels of all TIMPs could be detected on platelets. Plasma contained significant quantities of TIMP1 and TIMP2, but only trace amounts of TIMP3 and TIMP4. Recombinant TIMP3 strongly ablated resting and activated platelet ADAM10 activity, when monitored using a quenched fluorogenic peptide substrate with ADAM10 specificity. Whilst ADAM10-specific inhibitor GI254023X or ethylenediamine tetraacetic acid (EDTA) could modulate ligand-initiated shedding of GPVI, only recombinant TIMP2 achieved a modest (~20%) inhibition. We conclude that some platelet TIMPs are able to modulate platelet ADAM10 activity but none strongly regulate ligand-dependent shedding of GPVI. Our findings provide new insights into the regulation of platelet receptor sheddase activity.


What do we know? Platelet receptor GPVI initiates platelet adhesion and aggregation and is proteolytically cleaved from the activated platelet surfaceThe metalloproteinases responsible belong to the ADAMs family of enzymes which are inhibited by TIMPsWhat did we discover? Plasma contains significant amounts of TIMP1 and TIMP2Circulating platelets bear significant amounts of TIMPs 1, 2, and 3Recombinant TIMP3 strongly inhibits resting and activated platelet ADAM10 activityExogenous addition of TIMP2 mildly blocked ligand-initiated shedding of GPVIWhat is the impact? TIMPs may modulate ADAM10 activity under resting conditions and stabilize GPVI levels in response to platelet activationAnti-GPVI agents are being evaluated as anti-thrombotic agents, however, acute loss of GPVI in trauma or settings of thrombocytopenia is linked with clinical bleedingUnderstanding how GPVI levels are regulated is important as agents that modulate GPVI function are emerging as important therapeutics for clinical applications in Thrombosis and Hemostasis fields.


Blood Platelets , Platelet Membrane Glycoproteins , Humans , Ligands , ADAM10 Protein/genetics , Peptides/pharmacology , Metalloproteases , Platelet Activation , Membrane Proteins , Amyloid Precursor Protein Secretases
10.
J Clin Invest ; 133(23)2023 Dec 01.
Article En | MEDLINE | ID: mdl-37788087

The endothelium plays a critical role in the host response to infection and has been a focus of investigation in sepsis. While it is appreciated that intravascular thrombus formation, severe inflammation, and loss of endothelial integrity impair tissue oxygenation during sepsis, the precise molecular mechanisms that lead to endothelial injury remain poorly understood. We demonstrate here that endothelial ADAM10 was essential for the pathogenesis of Staphylococcus aureus sepsis, contributing to α-toxin-mediated (Hla-mediated) microvascular thrombus formation and lethality. As ADAM10 is essential for endothelial development and homeostasis, we examined whether other major human sepsis pathogens also rely on ADAM10-dependent pathways in pathogenesis. Mice harboring an endothelium-specific knockout of ADAM10 were protected against lethal Pseudomonas aeruginosa and Streptococcus pneumoniae sepsis, yet remained fully susceptible to group B streptococci and Candida albicans sepsis. These studies illustrate a previously unknown role for ADAM10 in sepsis-associated endothelial injury and suggest that understanding pathogen-specific divergent host pathways in sepsis may enable more precise targeting of disease.


Bacteremia , Candidiasis , Sepsis , Thrombosis , Vascular System Injuries , Animals , Humans , Mice , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Endothelium/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Sepsis/genetics , Sepsis/metabolism
11.
J Alzheimers Dis ; 95(2): 399-405, 2023.
Article En | MEDLINE | ID: mdl-37545238

The prevalence of Alzheimer's disease is greater in women, but the underlying mechanisms remain to be elucidated. We herein demonstrated that α-secretase ADAM10 was downregulated and ADAM10 inhibitor sFRP1 was upregulated in 5xFAD mice. While there were no sex effects on ADAM10 protein and sFRP1 mRNA levels, female 5xFAD and age-matched non-transgenic mice exhibited higher levels of sFRP1 protein than corresponding male mice. Importantly, female 5xFAD mice accumulated more Aß than males, and sFRP1 protein levels were positively associated with Aß42 levels in 5xFAD mice. Our study suggests that sFRP1 is associated with amyloid pathology in a sex-dependent manner.


Alzheimer Disease , Amyloid beta-Protein Precursor , Animals , Female , Male , Mice , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain/pathology , Disease Models, Animal , Mice, Transgenic , Up-Regulation
12.
Liver Int ; 43(10): 2275-2291, 2023 10.
Article En | MEDLINE | ID: mdl-37545390

BACKGROUND & AIMS: Notch1 activation promotes ICC progression and is associated with chemoresistance; however, therapies directly targeting Notch1 showed severe adverse effects. Notch1 activation is mediated by ADAM10, a molecular scissor that separates the target protein from its substrates in the cell membrane. Tspan15 regulates ADAM10 function, but the role of Tspan15 in ICC progression is unclear. METHODS: Tspan15, ADAM10, and Notch1 expression and activation in fresh surgical specimens from 80 ICC patients and ICC cells were evaluated by immunohistochemistry, RT-PCR, western blotting, and flow cytometry. RESULTS: Tspan15 expression was increased in ICC compared with adjacent liver tissue, and high Tspan15 expression was an independent factor for poor prognosis. In ICC with high Tspan15 expression, vascular invasion, lymph node metastasis, and haematogenous recurrence were increased. Tspan15 was co-expressed with ADAM10 in ICC, and associated with the expression of stemness and EMT markers. In ICC cells, Tspan15 induced ADAM10 activation by mediating the translocation of activated m-ADAM10 from the cytoplasm to the surface of the cell membrane, which further activated Notch1 by separating the intracellular domain of Notch1 from its extracellular domain, leading to enhancement of CSC-like properties and EMT. This signalling was associated with enhanced chemoresistance against gemcitabine and cisplatin. Inhibition of Tspan15 or ADAM10 is a promising therapeutic strategy in ICC, as Tspan15 or ADAM10 knockdown or treatment with ADAM10 inhibitor reduced chemoresistance and invasiveness by suppressing Notch1-mediated CSC-like properties and EMT. CONCLUSIONS: Tspan15-ADAM10-Notch1 signalling is associated with aggressive tumour progression and poor prognosis in ICC.


Drug Resistance, Neoplasm , Neoplasms , Humans , Drug Resistance, Neoplasm/genetics , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Signal Transduction , Neoplastic Stem Cells/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Cell Line, Tumor , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism
13.
Mol Cancer ; 22(1): 107, 2023 07 08.
Article En | MEDLINE | ID: mdl-37422628

BACKGROUND: Acute leukemias represent deadly malignancies that require better treatment. As a challenge, treatment is counteracted by a microenvironment protecting dormant leukemia stem cells. METHODS: To identify responsible surface proteins, we performed deep proteome profiling on minute numbers of dormant patient-derived xenograft (PDX) leukemia stem cells isolated from mice. Candidates were functionally screened by establishing a comprehensive CRISPR‒Cas9 pipeline in PDX models in vivo. RESULTS: A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) was identified as an essential vulnerability required for the survival and growth of different types of acute leukemias in vivo, and reconstitution assays in PDX models confirmed the relevance of its sheddase activity. Of translational importance, molecular or pharmacological targeting of ADAM10 reduced PDX leukemia burden, cell homing to the murine bone marrow and stem cell frequency, and increased leukemia response to conventional chemotherapy in vivo. CONCLUSIONS: These findings identify ADAM10 as an attractive therapeutic target for the future treatment of acute leukemias.


Leukemia , Proteomics , Humans , Mice , Animals , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , CRISPR-Cas Systems , Membrane Proteins/genetics , Membrane Proteins/metabolism , Leukemia/genetics , Disease Models, Animal , Tumor Microenvironment , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism
14.
Biomater Adv ; 152: 213516, 2023 Sep.
Article En | MEDLINE | ID: mdl-37348330

In the lung, pulmonary epithelial cells undergo mechanical stretching during ventilation. The associated cellular mechanoresponse is still poorly understood at the molecular level. Here, we demonstrate that activation of the mechanosensitive cation channel Piezo1 in a human epithelial cell line (H441) and in primary human lung epithelial cells induces the proteolytic activity of the metalloproteinases ADAM10 and ADAM17 at the plasma membrane. These ADAMs are known to convert cell surface expressed proteins into soluble and thereby play major roles in proliferation, barrier regulation and inflammation. We observed that chemical activation of Piezo1 promotes cleavage of substrates that are specific for either ADAM10 or ADAM17. Activation of Piezo1 also induced the synthesis and ADAM10/17-dependent release of the growth factor amphiregulin (AREG). In addition, junctional adhesion molecule A (JAM-A) was shed in an ADAM10/17-dependent manner resulting in a reduction of cell contacts. Stretching experiments combined with Piezo1 knockdown further demonstrated that mechanical activation promotes shedding via Piezo1. Most importantly, high pressure ventilation of murine lungs increased AREG and JAM-A release into the alveolar space, which was reduced by a Piezo1 inhibitor. Our study provides a novel link between stretch-induced Piezo1 activation and the activation of ADAM10 and ADAM17 in lung epithelium. This may help to understand acute respiratory distress syndrome (ARDS) which is induced by ventilation stress and goes along with perturbed epithelial permeability and release of growth factors.


Amyloid Precursor Protein Secretases , Lung , Humans , Mice , Animals , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Lung/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Epithelial Cells/metabolism , Ion Channels/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Metalloproteases/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism
15.
Genet Test Mol Biomarkers ; 27(5): 165-171, 2023 May.
Article En | MEDLINE | ID: mdl-37257180

Objective: To explore the abnormal expression of ADAM10, its cause, and its clinical value in the prognosis of cervical lesions. Methods: The abnormal expression of ADAM10 was explored using the Gene Expression Profiling Interactive Analysis database, and the abnormal expression in cervical lesions was verified using immunohistochemistry (IHC). The transfection effect of shRNA was evaluated using real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The expression of ADAM10 in cells was analyzed using western blotting. Results: ADAM10 was highly expressed in multiple cancers. As the disease progressed, the expression of ADAM10 gradually increased (p < 0.05). Patients with higher expression of ADAM10 had poorer survival outcomes than those with lower expression levels (p < 0.05). The expression levels of ADAM10 decreased after expression levels of E6 was inhibited. Conclusion: ADAM10 is highly expressed in cervical cancer; the higher the expression levels, the worse the survival outcome. HPV E6 is the critical driver of the elevated expression of ADAM10 in cervical cancer.


Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/genetics , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Uterine Cervical Neoplasms/genetics
16.
Int J Mol Sci ; 24(7)2023 Mar 24.
Article En | MEDLINE | ID: mdl-37047093

ADAM10 is the main α-secretase acting in the non-amyloidogenic processing of APP. We hypothesized that certain rare ADAM10 variants could increase the risk for AD by conferring the age-related downregulation of α-secretase. The ADAM10 gene was sequenced in 103 AD cases (82% familial) and 96 cognitively preserved nonagenarians. We examined rare variants (MAF < 0.01) and determined their potential association in the AD group with lower CSF protein levels, as analyzed by means of ELISA, and Western blot (species of 50 kDa, 55 kDa, and 80 kDa). Rare variants were found in 15.5% of AD cases (23% early-onset, 8% late-onset) and in 12.5% of nonagenarians, and some were group-specific. All were intronic variants except Q170H, found in three AD cases and one nonagenarian. The 3'UTR rs74016945 (MAF = 0.01) was found in 6% of the nonagenarians (OR 0.146, p = 0.057). Altogether, ADAM10 total levels or specific species were not significantly different when comparing AD with controls or carriers of rare variants versus non-carriers (except a Q170H carrier exhibiting low levels of all species), and did not differ according to the age at onset or APOE genotype. We conclude that ADAM10 exonic variants are uncommon in AD cases, and the presence of rare intronic variants (more frequent in early-onset cases) is not associated with decreased protein levels in CSF.


Alzheimer Disease , Aged, 80 and over , Humans , ADAM Proteins/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Cerebrospinal Fluid Proteins/analysis , Cerebrospinal Fluid Proteins/metabolism
17.
Histol Histopathol ; 38(12): 1453-1464, 2023 Dec.
Article En | MEDLINE | ID: mdl-36912070

Lung cancer, one of the most frequently diagnosed cancers, causes a huge number of mortalities globally. Among lung cancers, non-small cell lung cancer (NSCLC) is the most recorded. Despite accumulating research, the molecular basis of NSCLC progression remains poorly known. Therefore, we aim to assess the function of NCK1-AS1 in NSCLC and elucidate the molecular mechanism. Firstly, we quantified the NCK1-AS1 level in tumors and adjacent healthy tissues. NCK1-AS1 was significantly upregulated in NSCLC tumors, which was associated with poor prognosis in patients. Silencing NCK1-AS1 significantly inhibited the proliferation, migration, and invasion, as well as the EMT of NSCLC cell lines. Starbase bioinformatic prediction revealed that NCK1-AS1 targets miR-361-5p which acts to regulate ADAM10 gene expression. Our result showed that NCK1-AS1 upregulation markedly reduced miR-361-5p mRNA expression, while increasing ADAM10 expression. For the first time, we demonstrated that NCK1-AS1 regulates the miR-361-5p/ADAM10 axis, thereby promoting NSCLC progression. NCK1-AS1 might be developed as a therapeutic target for treating NSCLC.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , RNA, Long Noncoding/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism
18.
J Cell Mol Med ; 27(8): 1144-1152, 2023 04.
Article En | MEDLINE | ID: mdl-36946281

A disintegrin and metalloproteinase domain-containing protein 10 (ADAM-10) involves in the tumour progression, but the impacts of single-nucleotide polymorphism (SNP) of ADAM-10 on oral squamous cell carcinoma (OSCC) remain unclear. The aim of this study was to investigate the influence of SNP of ADAM-10 on the clinical features of OSCC in male Taiwanese. Five loci of ADAM-10 SNPs including rs653765 (C/T), rs2305421 (A/G), rs514049 (A/C), rs383902 (T/C) and rs2054096 (A/T) were genotyped by TaqMan allelic discrimination in 1138 OSCC patients and 1199 non-OSCC individuals. The ADAM-10 SNP rs2305421 GG (AOR: 1.399, 95% CI: 1.045-1.874, p = 0.024) and G allele (AOR: 1.170, 95% CI: 1.012-1.351, p = 0.034) illustrated a significantly higher genotypic frequencies in the OSCC group compared to the distribution of the ADAM-10 SNP rs2305421 AA wild type. In the subgroup analysis, the ADAM-10 SNP rs383902 TC+CC was significantly correlated to tumour size larger than T2 in betel quid chewer (AOR: 1.375, 95% CI: 1.010-1.872, p = 0.043), while the ADAM-10 SNP rs653765 CT+TT was significantly associated with tumour size larger than T2 in cigarette smoker (AOR: 1.346, 95% CI: 1.023-1.772, p = 0.034). The results from The Cancer Genome Atlas revealed highest ADAM-10 mRNA level in T2 stage of current smokers with head and neck squamous cell carcinoma (HNSCC). In conclusions, the ADAM-10 SNP rs2305421 G allele is associated with the presence of OSCC, and the ADAM-10 SNP rs383902 TC+CC and ADAM-10 SNP rs653765 CT+TT correlates to large tumour size in specific conditions.


ADAM10 Protein , Carcinoma, Squamous Cell , Mouth Neoplasms , Humans , Male , ADAM10 Protein/genetics , Carcinoma, Squamous Cell/genetics , Case-Control Studies , Genetic Predisposition to Disease , Head and Neck Neoplasms , Mouth Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Risk Factors , Squamous Cell Carcinoma of Head and Neck
19.
Fish Physiol Biochem ; 49(2): 321-334, 2023 Apr.
Article En | MEDLINE | ID: mdl-36964830

The ADAMs (a disintegrin and metalloproteinase) play regulatory roles in cell adhesion, migration and proteolysis. To explore the origin and evolution of ADAMs, this study identified the homologs of adam10 and adam17 in Lampetra morii and Lampetra japonica. Sequence analysis revealed that they share the same genomic structures with their counterparts in jawed vertebrates. The putative proteins possess conserved motifs, including a furin cut site (RXXR) for precursor processing, an enzyme catalytic motif (HEXGEHXXGXXH) for hydrolysis, and a Ca2+-binding motif (CGNXXXEXGEXCD) for stabilizing protein structure. In addition, a substrate recognition domain is present at the membrane-proximal region of lamprey ADAM17. The cytoplasmic region of lamprey ADAM10 contains a potential threonine phosphorylation site which has been shown to be activated by protein kinase C (PKC) in mammals. Both the adam10 and adam17 genes were constitutively expressed in the brain, kidney, and gills and were differentially regulated in the primary blood leukocytes by lipopolysaccharide (LPS) and pokeweed mitogen (PWM). Adam10 was induced by LPS but not PWM; conversely, adam17 was induced by PWM but not LPS. Taken together, our results suggest that the activation pathways and functions of ADAM10 and ADAM17 are conserved in agnathans.


ADAM Proteins , Lampreys , Animals , ADAM Proteins/genetics , ADAM Proteins/metabolism , Lampreys/genetics , Phylogeny , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , ADAM10 Protein/genetics , Mammals/metabolism
20.
Oncogene ; 42(18): 1438-1452, 2023 05.
Article En | MEDLINE | ID: mdl-36922678

Peritoneal metastasis is a key feature of advanced ovarian cancer, but the critical protein required for ovarian cancer metastasis and progression is yet to be defined. Thus, an unbiased high throughput and in-depth study is warranted to unmask the mechanism. Transcriptomic sequencing of paired primary ovarian tumors and metastases unveiled that MAP4K4, a serine/threonine kinase belongs to the Ste20 family of kinases, was highly expressed in metastatic sites. Increased MAP4K4 expression in metastasis was further validated in other independent patients, with higher MAP4K4 expression associated with poorer survival, higher level of CA125 and more advanced FIGO stage. Down regulation of MAP4K4 inhibited cancer cell adhesion, migration, and invasion. Notably, MAP4K4 was found to stabilize N-cadherin. Further results showed that MAP4K4 mediated phosphorylation of ADAM10 at Ser436 results in suppression of N-cadherin cleavage by ADAM10, leading to N-cadherin stabilization. Pharmacologic inhibition of MAP4K4 abrogated peritoneal metastases. Overall, our data reveal MAP4K4 as a significant promoter in ovarian cancer metastasis. Targeting MAP4K4 may be a potential therapeutic approach for ovarian cancer patients.


Cadherins , Ovarian Neoplasms , Humans , Female , Cadherins/genetics , Cadherins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ovarian Neoplasms/pathology , Phosphorylation , Cell Adhesion , ADAM10 Protein/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
...