Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Haematologica ; 107(4): 909-920, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34109776

ABSTRACT

Shedding of ADAM10 substrates, like TNFa or CD30, can affect both anti-tumor immune response and antibody-drug-conjugate (ADC)-based immunotherapy. We have published two new ADAM10 inhibitors, LT4 and MN8 able to prevent such shedding in Hodgkin lymphoma (HL). Since tumor tissue architecture deeply influences the outcome of anti-cancer treatments, we set up a new threedimensional (3D) culture systems to verify whether ADAM10 inhibitors can contribute to, or enhance, the anti-lymphoma effects of the ADC brentuximab-vedotin (BtxVed). In order to recapitulate some aspects of lymphoma structure and architecture, we assembled two 3D culture models: mixed spheroids made of HL lymph node (LN) mesenchymal stromal cells (MSC) and Reed Sternberg/Hodgkin lymphoma cells (HL cells) or collagen scaffolds repopulated with LN-MSC and HL cells. In these 3D systems we found that: i) the ADAM10 inhibitors LT4 and MN8 reduce ATP content or glucose consumption, related to cell proliferation, increasing lactate dehydrogenase release as a cell damage hallmark; ii) these events are paralleled by mixed spheroids size reduction and inhibition of CD30 and TNFa shedding; iii) the effects observed can be reproduced in repopulated HL LN-derived matrix or collagen scaffolds; iv) ADAM10 inhibitors enhance the anti-lymphoma effect of the anti-CD30 ADC BtxVed both in conventional cultures and in repopulated scaffolds. Thus, we provide evidence for a direct and combined antilymphoma effect of ADAM10 inhibitors with BtxVed, leading to the improvement of ADC effects; this is documented in 3D models recapitulating features of the LN microenvironment, that can be proposed as a reliable tool for anti-lymphoma drug testing.


Subject(s)
ADAM10 Protein/antagonists & inhibitors , Brentuximab Vedotin/therapeutic use , Hodgkin Disease , Immunoconjugates , Lymphoma , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Humans , Immunoconjugates/therapeutic use , Ki-1 Antigen , Lymphoma/drug therapy , Membrane Proteins , Tumor Microenvironment
2.
Anticancer Res ; 41(5): 2307-2320, 2021 May.
Article in English | MEDLINE | ID: mdl-33952456

ABSTRACT

BACKGROUND/AIM: The association between MHC class I polypeptide-related sequence A (MICA) and hepatocellular carcinoma (HCC) development was identified in our previous genome-wide association study. Decreasing soluble MICA (sMICA) through MICA sheddases suppression facilitates natural killer (NK) cell-mediated cytotoxicity. The expression of ADAM9 in HCC has been correlated with poor prognosis, and our recent study showed that its suppression contributes to cancer elimination by decreasing sMICA. MATERIALS AND METHODS: Human HCC cell line PLC/PRF/5 and HepG2 cells were used. sMICA levels were measured by ELISA. Expression of retinoid X receptors (RXRs) and retinoic acid receptors (RARs) was knocked down by siRNA. RESULTS: In our screening of FDA-approved drugs in vitro, retinoids were found to be efficient ADAM9 and ADAM10 inhibitors. Treatment with retinoids reduced sMICA levels in human HCC cells. Interestingly, the effects were abrogated by depletion of the retinoid receptor RXRα. CONCLUSION: Retinoids can be potential novel agents for HCC treatment.


Subject(s)
ADAM Proteins/metabolism , ADAM10 Protein/metabolism , Histocompatibility Antigens Class I/metabolism , Membrane Proteins/metabolism , Retinoids/pharmacology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Biocatalysis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Hep G2 Cells , Histocompatibility Antigens Class I/genetics , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Molecular Structure , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , RNA Interference , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Retinoids/chemistry , Solubility
3.
Neoplasia ; 23(4): 415-428, 2021 04.
Article in English | MEDLINE | ID: mdl-33839455

ABSTRACT

Trop-2 is a transmembrane signal transducer that can induce cancer growth. Using antibody targeting and N-terminal Edman degradation, we show here that Trop-2 undergoes cleavage in the first thyroglobulin domain loop of its extracellular region, between residues R87 and T88. Molecular modeling indicated that this cleavage induces a profound rearrangement of the Trop-2 structure, which suggested a deep impact on its biological function. No Trop-2 cleavage was detected in normal human tissues, whereas most tumors showed Trop-2 cleavage, including skin, ovary, colon, and breast cancers. Coimmunoprecipitation and mass spectrometry analysis revealed that ADAM10 physically interacts with Trop-2. Immunofluorescence/confocal time-lapse microscopy revealed that the two molecules broadly colocalize at the cell membrane. We show that ADAM10 inhibitors, siRNAs and shRNAs abolish the processing of Trop-2, which indicates that ADAM10 is an effector protease. Proteolysis of Trop-2 at R87-T88 triggered cancer cell growth both in vitro and in vivo. A corresponding role was shown for metastatic spreading of colon cancer, as the R87A-T88A Trop-2 mutant abolished xenotransplant metastatic dissemination. Activatory proteolysis of Trop-2 was recapitulated in primary human breast cancers. Together with the prognostic impact of Trop-2 and ADAM10 on cancers of the skin, ovary, colon, lung, and pancreas, these data indicate a driving role of this activatory cleavage of Trop-2 on malignant progression of tumors.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation/physiology , Membrane Proteins/metabolism , Neoplasms/pathology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Amino Acid Sequence/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Epithelial Cells/metabolism , HCT116 Cells , HT29 Cells , Humans , MCF-7 Cells , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Models, Molecular , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Proteolysis , Signal Transduction , Transplantation, Heterologous
4.
Cancer Res ; 81(12): 3255-3269, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33526513

ABSTRACT

Stromal fibrosis activates prosurvival and proepithelial-to-mesenchymal transition (EMT) pathways in pancreatic ductal adenocarcinoma (PDAC). In patient tumors treated with neoadjuvant stereotactic body radiation therapy (SBRT), we found upregulation of fibrosis, extracellular matrix (ECM), and EMT gene signatures, which can drive therapeutic resistance and tumor invasion. Molecular, functional, and translational analysis identified two cell-surface proteins, a disintegrin and metalloprotease 10 (ADAM10) and ephrinB2, as drivers of fibrosis and tumor progression after radiation therapy (RT). RT resulted in increased ADAM10 expression in tumor cells, leading to cleavage of ephrinB2, which was also detected in plasma. Pharmacologic or genetic targeting of ADAM10 decreased RT-induced fibrosis and tissue tension, tumor cell migration, and invasion, sensitizing orthotopic tumors to radiation killing and prolonging mouse survival. Inhibition of ADAM10 and genetic ablation of ephrinB2 in fibroblasts reduced the metastatic potential of tumor cells after RT. Stimulation of tumor cells with ephrinB2 FC protein reversed the reduction in tumor cell invasion with ADAM10 ablation. These findings represent a model of PDAC adaptation that explains resistance and metastasis after RT and identifies a targetable pathway to enhance RT efficacy. SIGNIFICANCE: Targeting a previously unidentified adaptive resistance mechanism to radiation therapy in PDAC tumors in combination with radiation therapy could increase survival of the 40% of PDAC patients with locally advanced disease.See related commentary by Garcia Garcia et al., p. 3158 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3255/F1.large.jpg.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Carcinoma, Pancreatic Ductal/radiotherapy , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Gamma Rays/adverse effects , Membrane Proteins/metabolism , Pancreatic Neoplasms/radiotherapy , Radiation Injuries/pathology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , Antifibrotic Agents/therapeutic use , Apoptosis , Carcinoma, Pancreatic Ductal/pathology , Cell Movement , Cell Proliferation , Ephrin-B2/blood , Female , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/pathology , Prognosis , Radiation Injuries/drug therapy , Radiation Injuries/etiology , Radiation Injuries/metabolism , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Cells ; 10(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33504031

ABSTRACT

To efficiently cross the endothelial barrier during inflammation, neutrophils first firmly adhere to the endothelial surface using the endothelial adhesion molecule ICAM-1. Upon actual transmigration, the release from ICAM-1 is required. While Integrin LFA1/Mac1 de-activation is one described mechanism that leads to this, direct cleavage of ICAM-1 from the endothelium represents a second option. We found that a disintegrin and metalloprotease 10 (ADAM10) cleaves the extracellular domain of ICAM-1 from the endothelial surface. Silencing or inhibiting endothelial ADAM10 impaired the efficiency of neutrophils to cross the endothelium, suggesting that neutrophils use endothelial ADAM10 to dissociate from ICAM-1. Indeed, when measuring transmigration kinetics, neutrophils took almost twice as much time to finish the diapedesis step when ADAM10 was silenced. Importantly, we found increased levels of ICAM-1 on the transmigrating neutrophils when crossing an endothelial monolayer where such increased levels were not detected when neutrophils crossed bare filters. Using ICAM-1-GFP-expressing endothelial cells, we show that ICAM-1 presence on the neutrophils can also occur by membrane transfer from the endothelium to the neutrophil. Based on these findings, we conclude that endothelial ADAM10 contributes in part to neutrophil transendothelial migration by cleaving ICAM-1, thereby supporting the release of neutrophils from the endothelium during the final diapedesis step.


Subject(s)
ADAM10 Protein/metabolism , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Transendothelial and Transepithelial Migration , ADAM10 Protein/antagonists & inhibitors , Cell Adhesion , Endothelium/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans
6.
Sci Immunol ; 5(49)2020 07 17.
Article in English | MEDLINE | ID: mdl-32680952

ABSTRACT

Mechanisms of resistance to cancer immunotherapy remain poorly understood. Lymphocyte activation gene-3 (LAG3) signaling is regulated by a disintegrin and metalloprotease domain-containing protein-10 (ADAM10)- and ADAM17-mediated cell surface shedding. Here, we show that mice expressing a metalloprotease-resistant, noncleavable LAG3 mutant (LAG3NC) are resistant to PD1 blockade and fail to mount an effective antitumor immune response. Expression of LAG3NC intrinsically perturbs CD4+ T conventional cells (Tconvs), limiting their capacity to provide CD8+ T cell help. Furthermore, the translational relevance for these observations is highlighted with an inverse correlation between high LAG3 and low ADAM10 expression on CD4+ Tconvs in the peripheral blood of patients with head and neck squamous cell carcinoma, which corresponded with poor prognosis. This correlation was also observed in a cohort of patients with skin cancers and was associated with increased disease progression after standard-of-care immunotherapy. These data suggest that subtle changes in LAG3 inhibitory receptor signaling can act as a resistance mechanism with a substantive effect on patient responsiveness to immunotherapy.


Subject(s)
Antigens, CD/immunology , Drug Resistance, Neoplasm/immunology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antigens, CD/blood , Antigens, CD/genetics , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Immunotherapy , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Transgenic , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , Transcriptome , Lymphocyte Activation Gene 3 Protein
7.
Front Immunol ; 11: 499, 2020.
Article in English | MEDLINE | ID: mdl-32265938

ABSTRACT

Generating inhibitors for A Disintegrin And Metalloproteinase 10 (ADAM10), a zinc-dependent protease, was heavily invested in by the pharmaceutical industry starting over 20 years ago. There has been much enthusiasm in basic research for these inhibitors, with a multitude of studies generating significant data, yet the clinical trials have not replicated the same results. ADAM10 is ubiquitously expressed and cleaves many important substrates such as Notch, PD-L1, EGFR/HER ligands, ICOS-L, TACI, and the "stress related molecules" MIC-A, MIC-B and ULBPs. This review goes through the most recent pre-clinical data with inhibitors as well as clinical data supporting the use of ADAM10 inhibitor use in cancer and autoimmunity. It additionally addresses how ADAM10 inhibitor therapy can be improved and if inhibitor therapy can be paired with other drug treatments to maximize effectiveness in various disease states. Finally, it examines the ADAM10 substrates that are important to each disease state and if any of these substrates or ADAM10 itself is a potential biomarker for disease.


Subject(s)
ADAM10 Protein/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Autoimmune Diseases/drug therapy , Membrane Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protease Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Clinical Trials as Topic , Dipeptides/pharmacology , Dipeptides/therapeutic use , Drug Evaluation, Preclinical , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Multicenter Studies as Topic , Neoplasms/enzymology , Neoplasms/immunology , Protease Inhibitors/pharmacology , Receptors, Notch/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Substrate Specificity
8.
Molecules ; 25(3)2020 Feb 03.
Article in English | MEDLINE | ID: mdl-32028607

ABSTRACT

Accumulation of ß-amyloid (Aß) in the brain has been implicated in the pathology of Alzheimer's disease (AD). Aß is produced from the Aß precursor protein (APP) through the amyloidogenic pathway by ß-, and γ-secretase. Alternatively, APP can be cleaved by α-, and γ-secretase, precluding the production of Aß. Thus, stimulating α-secretase mediated APP processing is considered a therapeutic option not only for decreasing Aß production but for increasing neuroprotective sAPPα. We have previously reported that 7-deoxy-trans-dihydronarciclasine (E144), the active component of Lycoris chejuensis, decreases Aß production by attenuating APP level, and retarding APP maturation. It can also improve cognitive function in the AD model mouse. In this study, we further analyzed the activating effect of E144 on α-secretase. Treatment of E144 increased sAPPα, but decreased ß-secretase products from HeLa cells stably transfected with APP. E144 directly activated ADAM10 and ADAM17 in a substrate-specific manner both in cell-based and in cell-free assays. The Lineweaver-Burk plot analysis revealed that E144 enhanced the affinities of A Disintegrin and Metalloproteinases (ADAMs) towards the substrate. Consistent with this result, immunoprecipitation analysis showed that interactions of APP with ADAM10 and ADAM17 were increased by E144. Our results indicate that E144 might be a novel agent for AD treatment as a substrate-specific activator of α-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Isoquinolines/pharmacology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/metabolism , Enzyme Activation , Humans , Isoquinolines/chemistry , Molecular Structure , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
10.
BMC Cancer ; 19(1): 958, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31619190

ABSTRACT

BACKGROUND: We analyzed the changes in permeability of endothelial cell layers after photon irradiation, with a focus on the metalloproteases ADAM10 and ADAM17, and on VE-cadherin, components crucial for the integrity of endothelial intercellular junctions, and their roles in the transmigration of cancer cells through endothelial cell monolayers. METHODS: Primary HUVEC were irradiated with 2 or 4 Gy photons at a dose rate of 5 Gy/min. The permeability of an irradiated endothelial monolayer for macromolecules and tumor cells was analyzed in the presence or absence of the ADAM10/17 inhibitors GI254023X and GW280264X. Expression of ADAM10, ADAM17 and VE-Cadherin in endothelial cells was quantified by immunoblotting and qRT. VE-Cadherin was additionally analyzed by immunofluorescence microscopy and ELISA. RESULTS: Ionizing radiation increased the permeability of endothelial monolayers and the transendothelial migration of tumor cells. This was effectively blocked by a selective inhibition (GI254023X) of ADAM10. Irradiation increased both, the expression and activity of ADAM10, which led to increased degradation of VE-cadherin, but also led to higher rates of VE-cadherin internalization. Increased degradation of VE-cadherin was also observed when endothelial monolayers were exposed to tumor-cell conditioned medium, similar to when exposed to recombinant VEGF. CONCLUSIONS: Our results suggest a mechanism of irradiation-induced increased permeability and transendothelial migration of tumor cells based on the activation of ADAM10 and the subsequent change of endothelial permeability through the degradation and internalization of VE-cadherin.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/radiation effects , Human Umbilical Vein Endothelial Cells/radiation effects , Membrane Proteins/metabolism , Proteolysis/radiation effects , Radiation, Ionizing , Transendothelial and Transepithelial Migration/radiation effects , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Cell Line, Tumor , Dipeptides/pharmacology , Endothelial Cells/metabolism , Humans , Hydroxamic Acids/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Permeability/radiation effects , Radiotherapy/adverse effects , Signal Transduction/radiation effects , Transendothelial and Transepithelial Migration/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
11.
Sci Rep ; 9(1): 14901, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624271

ABSTRACT

CD44 fragmentation is enhanced in chondrocytes of osteoarthritis (OA) patients. We hypothesized that mechanical stress-induced enhancement of CD44-intracellular domain (CD44-ICD) production plays an important role in the de-differentiation of chondrocytes and OA. This study aimed to assess the relationship between CD44-ICD and chondrocyte gene expression. Monolayer cultured primary bovine articular chondrocytes (BACs) were subjected to cyclic tensile strain (CTS) loading. ADAM10 inhibitor (GI254023X) and γ-secretase inhibitor (DAPT) were used to inhibit CD44 cleavage. In overexpression experiments, BACs were electroporated with a plasmid encoding CD44-ICD. CTS loading increased the expression of ADAM10 and subsequent CD44 cleavage, while decreasing the expression of SOX9, aggrecan, and type 2 collagen (COL2). Overexpression of CD44-ICD also resulted in decreased expression of these chondrocyte genes. Both GI254023X and DAPT reduced the production of CD44-ICD upon CTS loading, and significantly rescued the reduction of SOX9 expression by CTS loading. Chemical inhibition of CD44-ICD production also rescued aggrecan and COL2 expression following CTS loading. Our findings suggest that CD44-ICD is closely associated with the de-differentiation of chondrocytes. Excessive mechanical stress loading promoted the de-differentiation of BACs by enhancing CD44 cleavage and CD44-ICD production. Suppression of CD44 cleavage has potential as a novel treatment strategy for OA.


Subject(s)
Cartilage, Articular/pathology , Cell Dedifferentiation/drug effects , Chondrocytes/drug effects , Hyaluronan Receptors/metabolism , Osteoarthritis/drug therapy , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cartilage, Articular/cytology , Cattle , Cells, Cultured , Chondrocytes/pathology , Diamines/pharmacology , Diamines/therapeutic use , Dipeptides/pharmacology , Dipeptides/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Male , Osteoarthritis/pathology , Primary Cell Culture , Protein Domains/drug effects , Stress, Mechanical , Thiazoles/pharmacology , Thiazoles/therapeutic use
12.
Cancer Lett ; 467: 50-57, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31593799

ABSTRACT

ADAM proteases are multi domain transmembrane metalloproteases that cleave a range of cell surface proteins and activate signaling pathways implicated in tumor progression, including those mediated by Notch, EFGR, and the Eph receptors. Consequently, they have emerged as key therapeutic targets in the efforts to inhibit tumor initiation and progression. To that end, two main approaches have been taken to develop ADAM antagonists: (i) small molecule inhibitors, and (ii) monoclonal antibodies. In this mini-review we describe the distinct features of ADAM proteases, particularly of ADAM10 and ADAM17, their domain organization, conformational rearrangements, regulation, as well as their emerging importance as therapeutic targets in cancer. Further, we highlight an anti-ADAM10 monoclonal antibody that we have recently developed, which has shown significant promise in inhibiting Notch signaling and deterring growth of solid tumors in pre-clinical settings.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/metabolism , Neoplasms/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/chemistry , ADAM10 Protein/metabolism , ADAM17 Protein/chemistry , ADAM17 Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Catalytic Domain , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , Protein Conformation , Protein Domains
13.
Nat Neurosci ; 22(7): 1075-1088, 2019 07.
Article in English | MEDLINE | ID: mdl-31209379

ABSTRACT

Microglia rapidly respond to changes in neural activity and inflammation to regulate synaptic connectivity. The extracellular signals, particularly neuron-derived molecules, that drive these microglial functions at synapses remain a key open question. Here we show that whisker lesioning, known to dampen cortical activity, induces microglia-mediated synapse elimination. This synapse elimination is dependent on signaling by CX3CR1, the receptor for microglial fractalkine (also known as CXCL1), but not complement receptor 3. Furthermore, mice deficient in CX3CL1 have profound defects in synapse elimination. Single-cell RNA sequencing revealed that Cx3cl1 is derived from cortical neurons, and ADAM10, a metalloprotease that cleaves CX3CL1 into a secreted form, is upregulated specifically in layer IV neurons and in microglia following whisker lesioning. Finally, inhibition of ADAM10 phenocopies Cx3cr1-/- and Cx3cl1-/- synapse elimination defects. Together, these results identify neuron-to-microglia signaling necessary for cortical synaptic remodeling and reveal that context-dependent immune mechanisms are utilized to remodel synapses in the mammalian brain.


Subject(s)
ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , CX3C Chemokine Receptor 1/physiology , Chemokine CX3CL1/physiology , Membrane Proteins/physiology , Microglia/physiology , Sensorimotor Cortex/physiopathology , Touch/physiology , Vibrissae/injuries , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Animals , CX3C Chemokine Receptor 1/deficiency , CX3C Chemokine Receptor 1/genetics , Cell Count , Female , Gene Expression Regulation , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfluidic Analytical Techniques , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/pathology , Signal Transduction/physiology , Single-Cell Analysis , Transcriptome , Vibrissae/physiology
14.
EMBO Mol Med ; 11(4)2019 04.
Article in English | MEDLINE | ID: mdl-30833305

ABSTRACT

The metalloprotease ADAM10 is a drug target in Alzheimer's disease, where it cleaves the amyloid precursor protein (APP) and lowers amyloid-beta. Yet, ADAM10 has additional substrates, which may cause mechanism-based side effects upon therapeutic ADAM10 activation. However, they may also serve-in addition to APP-as biomarkers to monitor ADAM10 activity in patients and to develop APP-selective ADAM10 activators. Our study demonstrates that one such substrate is the neuronal cell adhesion protein NrCAM ADAM10 controlled NrCAM surface levels and regulated neurite outgrowth in vitro in an NrCAM-dependent manner. However, ADAM10 cleavage of NrCAM, in contrast to APP, was not stimulated by the ADAM10 activator acitretin, suggesting that substrate-selective ADAM10 activation may be feasible. Indeed, a whole proteome analysis of human CSF from a phase II clinical trial showed that acitretin, which enhanced APP cleavage by ADAM10, spared most other ADAM10 substrates in brain, including NrCAM Taken together, this study demonstrates an NrCAM-dependent function for ADAM10 in neurite outgrowth and reveals that a substrate-selective, therapeutic ADAM10 activation is possible and may be monitored with NrCAM.


Subject(s)
ADAM10 Protein/metabolism , Alzheimer Disease/pathology , Cell Adhesion Molecules/metabolism , ADAM10 Protein/antagonists & inhibitors , Acitretin/pharmacology , Acitretin/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Methylaspartate/pharmacology , Neuronal Outgrowth/drug effects , Neurons/cytology , Neurons/metabolism , Proteolysis/drug effects , Proteome/analysis , Proteome/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Substrate Specificity , Tetraspanins/genetics , Tetraspanins/metabolism
15.
J Natl Cancer Inst ; 111(6): 609-619, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30265336

ABSTRACT

BACKGROUND: Resistance to HER2-targeted therapy with trastuzumab still remains a major challenge in HER2-amplified tumors. Here we investigated the potential role of MEL-18, a polycomb group gene, as a novel prognostic marker for trastuzumab resistance in HER2-positive (HER2+) breast cancer. METHODS: The genetic alteration of MEL-18 and its clinical relevance were examined in multiple breast cancer cohorts including METABRIC (n = 1,980), TCGA (n = 825), and our clinical specimens (n = 213, trastuzumab-treated HER2+ cases). MEL-18 amplification was validated by fluorescence in situ hybridization (FISH) analysis. The MEL-18 effect on trastuzumab response was confirmed by in vitro cell viability assays and an in vivo xenograft experiment (n = 7 per group). Gene expression microarray and receptor tyrosine kinase array were performed to identify the trastuzumab resistance mechanism by MEL-18 loss. All statistical tests were two-sided. RESULTS: MEL-18 was exclusively amplified in approximately 30-50% of HER2+ breast tumors and was associated with a favorable clinical outcome (disease-free survival: P = .02 in HER2+ cases, METABRIC; P = .04 in patients receiving trastuzumab). In MEL-18-amplified HER2+ breast cancer, MEL-18 depletion induced trastuzumab resistance by increasing ADAM sheddase-mediated ErbB ligand production and receptor heterodimerization. MEL-18 epigenetically silenced ADAM10/17 expression in cooperation with polycomb-repressive complex (PRC) 1 and PRC2. Combination treatment with an ADAM10/17 inhibitor and trastuzumab could overcome MEL-18 loss-mediated trastuzumab resistance in vivo (BT474/shMEL-18 xenograft: trastuzumab, mean [SD] tumor volume = 406.1 [50.1] mm3, vs trastuzumab + GW280264 30 mg/kg, mean [SD] tumor volume = 68.4 [15.6] mm3, P < .001). Consistently, trastuzumab-treated patients harboring concomitant MEL-18 amplification and low ADAM17 expression showed prolonged relapse-free survival (P = .02 in our cohort, n = 213). CONCLUSION: MEL-18 serves to prevent ligand-dependent ErbB heterodimerization and trastuzumab resistance, suggesting MEL-18 amplification as a novel biomarker for HER2+ breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Receptor, ErbB-2/antagonists & inhibitors , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Amplification , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Trastuzumab/administration & dosage , Xenograft Model Antitumor Assays
16.
Nat Microbiol ; 4(2): 216-225, 2019 02.
Article in English | MEDLINE | ID: mdl-30420785

ABSTRACT

Cell motility is essential for viral dissemination1. Vaccinia virus (VACV), a close relative of smallpox virus, is thought to exploit cell motility as a means to enhance the spread of infection1. A single viral protein, F11L, contributes to this by blocking RhoA signalling to facilitate cell retraction2. However, F11L alone is not sufficient for VACV-induced cell motility, indicating that additional viral factors must be involved. Here, we show that the VACV epidermal growth factor homologue, VGF, promotes infected cell motility and the spread of viral infection. We found that VGF secreted from early infected cells is cleaved by ADAM10, after which it acts largely in a paracrine manner to direct cell motility at the leading edge of infection. Real-time tracking of cells infected in the presence of EGFR, MAPK, FAK and ADAM10 inhibitors or with VGF-deleted and F11-deleted viruses revealed defects in radial velocity and directional migration efficiency, leading to impaired cell-to-cell spread of infection. Furthermore, intravital imaging showed that virus spread and lesion formation are attenuated in the absence of VGF. Our results demonstrate how poxviruses hijack epidermal growth factor receptor-induced cell motility to promote rapid and efficient spread of infection in vitro and in vivo.


Subject(s)
Cell Movement , Host-Pathogen Interactions , Peptides/metabolism , Signal Transduction , Vaccinia virus/physiology , Vaccinia/virology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cytopathogenic Effect, Viral/genetics , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Deletion , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Peptides/deficiency , Peptides/genetics , Signal Transduction/drug effects , Vaccinia/metabolism , Vaccinia/pathology , Vaccinia virus/genetics , Vaccinia virus/growth & development , Vaccinia virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
17.
ChemMedChem ; 13(19): 2119-2131, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30102846

ABSTRACT

A disintegrin and metalloproteinase (ADAMs) are membrane-bound metalloproteases responsible for the ectodomain shedding of various transmembrane proteins and play important roles in multiple relevant biological processes. Their altered expression is involved in several pathological conditions, and in particular ADAM10 or ADAM17 overexpression is found in various forms of cancer. To better understand how they are regulated in the cellular context, it is useful to visualize the specific ADAMs pathway by means of molecular imaging techniques. For this purpose, we synthesized bioactive fluorescent probes suitable for cell imaging and that are able to specifically target ADAM10 or ADAM17. Two previously developed ADAM17- and ADAM10-selective inhibitors were chosen for conjugation, respectively, to a Cy5.5 dye and to Cy5.5 and FITC dyes. Herein we also report the synthesis of a gold-labeled compound as an additional bioimaging probe for ADAM10. The newly synthesized ligands were found to be active in vitro on human recombinant ADAM10 and/or ADAM17, showing IC50 values in the nanomolar range and a good selectivity over matrix metalloproteinases (MMPs). Finally, these newly developed probes were successfully used for ADAMs staining on different lymphoma cell lines and lymph node mesenchymal stromal cells.


Subject(s)
ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Membrane Proteins/metabolism , ADAM10 Protein/antagonists & inhibitors , ADAM17 Protein/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antigens, CD/metabolism , Carbocyanines/chemistry , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Fetal Proteins/metabolism , Fluorescein-5-isothiocyanate/chemistry , Fluorescence , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Organogold Compounds/chemical synthesis , Organogold Compounds/chemistry , Organogold Compounds/metabolism , Organogold Compounds/pharmacology , Tumor Necrosis Factor-alpha/metabolism
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(1): 41-46, 2018 Jan.
Article in Chinese | MEDLINE | ID: mdl-29595456

ABSTRACT

Objective To study the role of a disintegrin and metalloproteinase10 (ADAM10) in shedding neural cadherin (N-cadherin) and develop an approach to interfere the process of ventricular remodeling in adriamycin-induced cardiomyopathy (ACM) rats. Methods In a rat model of ACM, the effects of intraperitoneal injection of the lentiviral RNAi vector of ADAM10 on the morphology of cardiomyocytes and contractile function were observed by HE staining and color Doppler echocardiography. The expressions of N-cadherin and C-terminal fragment 1 (CTF1) were detected by Western blotting and immunohistochemistry. Results In the in vivo experiment, a large amount of fluorescence was seen in the isolated primary cardiomyocytes, which indicated that the transfection in the rat model was successful. In the treatment group, the morphology of cardiomyocytes and function of the heart were evidently improved, N-cadherin protein expression was remarkably up-regulated and CTF1 protein was obviously down-regulated compared with the model group. Conclusion Knock-down of ADAM10 increases N-cadherin expression and decreases CTF1 expression, thus improves cardiac function in the rat model of ACM.


Subject(s)
ADAM10 Protein/physiology , Cardiomyopathies/therapy , Doxorubicin/toxicity , ADAM10 Protein/antagonists & inhibitors , Animals , Cadherins/analysis , Cardiomyopathies/chemically induced , Cardiomyopathies/physiopathology , Cardiotoxicity , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
19.
J Biol Chem ; 293(8): 2725-2734, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29317500

ABSTRACT

Theta-defensins (θ-defensins) are macrocyclic peptides expressed exclusively in granulocytes and selected epithelia of Old World monkeys. They contribute to anti-pathogen host defense responses by directly killing a diverse range of microbes. Of note, θ-defensins also modulate microbe-induced inflammation by affecting the production of soluble tumor necrosis factor (sTNF) and other proinflammatory cytokines. Here, we report that natural rhesus macaque θ-defensin (RTD) isoforms regulate sTNF cellular release by inhibiting TNF-α-converting enzyme (TACE; also known as adisintegrin and metalloprotease 17; ADAM17), the primary pro-TNF sheddase. Dose-dependent inhibition of cellular TACE activity by RTDs occurred when leukocytes were stimulated with live Escherichia coli cells as well as numerous Toll-like receptor agonists. Moreover, the relative inhibitory potencies of the RTD isoforms strongly correlated with their suppression of TNF release by stimulated blood leukocytes and THP-1 monocytes. RTD isoforms also inhibited ADAM10, a sheddase closely related to TACE. TACE inhibition was abrogated by introducing a single opening in the RTD-1 backbone, demonstrating that the intact macrocycle is required for enzyme inhibition. Enzymologic analyses showed that RTD-1 is a fast binding, reversible, non-competitive inhibitor of TACE. We conclude that θ-defensin-mediated inhibition of pro-TNF proteolysis by TACE represents a rapid mechanism for the regulation of sTNF and TNF-dependent inflammatory pathways. Molecules with structural and functional features mimicking those of θ-defensins may have clinical utility as TACE inhibitors for managing TNF-driven diseases.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Defensins/pharmacology , Leukocytes/drug effects , Monocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line , Chlorocebus aethiops , Colon/drug effects , Colon/immunology , Colon/metabolism , Defensins/chemistry , Escherichia coli/immunology , Escherichia coli/physiology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Lipopolysaccharides/toxicity , Macaca mulatta , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/pharmacology , Proteolysis/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/metabolism
20.
Anticancer Res ; 37(10): 5507-5513, 2017 10.
Article in English | MEDLINE | ID: mdl-28982863

ABSTRACT

BACKGROUND/AIM: The inhibition of a disintegrin and metalloproteinase (ADAM) has the potential to become a novel approach for natural killer (NK) cell-based cancer immunotherapy. Thus, the aim of this study was to investigate the influence of ADAM10 and ADAM17 inhibitors on expanded NK cell to enhance antibody-dependent cellular cytotoxicity (ADCC) in breast cancer cell lines. MATERIALS AND METHODS: NK cells were expanded in medium supplemented with an ADAM10 or ADAM17 inhibitor to prevent the shedding of soluble CD16/FcγRIII. The expression level of CD16 and production of interferon-gamma (IFN-γ) was detected by flow cytometry using specific antibodies. ADCC activity of expanded NK cells was estimated in trastuzumab treated breast cancer cell lines such as MCF-7, MDA-MB-231, SKBR3, and BT-474 cells. RESULTS: The ADAM17 inhibitor increased the purity of expanded NK cells to 90% after 14 days at 5 and 10 µM in vitro (p=0.043). However, the expansion rate of NK cells was decreased at 10 µM of the ADAM 17 inhibitor (p=0.043). Inhibition of ADAM10 suppressed the expansion of NK cells, although the NK purity was increased at 1 µM of the inhibitor. The expression of CD16 was significantly increased at 1 and 5 µM of the ADAM17 inhibitor (p=0.046, 0.028, respectively) during the culturing period. Inhibition of ADAM10 reduced the expression of CD16 on NK cells. The cytotoxic activity of the ADAM17 inhibitor treated NK cells against MCF-7 (p=0.039) and BT-474 (p=0.027) cells was significantly elevated. The ADCC activity of NK cells treated with 5 µM of ADAM17 inhibitor was significantly increased against SKBR-3 and BT-474 (p=0.027). Inhibition of ADAM17 increased the production of IFN-γ in expanded NK cells. CONCLUSION: The inhibition of ADAM17 enhanced the purity of expanded NK cells and the ADCC activity of these cells against trastuzumab treated breast cancer cell lines.


Subject(s)
ADAM10 Protein/antagonists & inhibitors , ADAM17 Protein/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Membrane Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Coculture Techniques , Dose-Response Relationship, Drug , Female , GPI-Linked Proteins/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/enzymology , Killer Cells, Natural/immunology , MCF-7 Cells , Membrane Proteins/metabolism , Receptors, IgG/metabolism , Time Factors , Trastuzumab/pharmacology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...