Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 502
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Biol Lett ; 29(1): 75, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755530

RESUMO

BACKGROUND: Mechanical spinal cord injury (SCI) is a deteriorative neurological disorder, causing secondary neuroinflammation and neuropathy. ADAM8 is thought to be an extracellular metalloproteinase, which regulates proteolysis and cell adherence, but whether its intracellular region is involved in regulating neuroinflammation in microglia after SCI is unclear. METHODS: Using animal tissue RNA-Seq and clinical blood sample examinations, we found that a specific up-regulation of ADAM8 in microglia was associated with inflammation after SCI. In vitro, microglia stimulated by HMGB1, the tail region of ADAM8, promoted microglial inflammation, migration and proliferation by directly interacting with ERKs and Fra-1 to promote activation, then further activated Map3k4/JNKs/p38. Using SCI mice, we used BK-1361, a specific inhibitor of ADAM8, to treat these mice. RESULTS: The results showed that administration of BK-1361 attenuated the level of neuroinflammation and reduced microglial activation and recruitment by inhibiting the ADAM8/Fra-1 axis. Furthermore, treatment with BK-1361 alleviated glial scar formation, and also preserved myelin and axonal structures. The locomotor recovery of SCI mice treated with BK-1361 was therefore better than those without treatment. CONCLUSIONS: Taken together, the results showed that ADAM8 was a critical molecule, which positively regulated neuroinflammatory development and secondary pathogenesis by promoting microglial activation and migration. Mechanically, ADAM8 formed a complex with ERK and Fra-1 to further activate the Map3k4/JNK/p38 axis in microglia. Inhibition of ADAM8 by treatment with BK-1361 decreased the levels of neuroinflammation, glial formation, and neurohistological loss, leading to favorable improvement in locomotor functional recovery in SCI mice.


Assuntos
Proteínas ADAM , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana , Microglia , Doenças Neuroinflamatórias , Traumatismos da Medula Espinal , Animais , Camundongos , Proteínas ADAM/metabolismo , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Antígenos CD , Movimento Celular/efeitos dos fármacos , Inflamação/patologia , Inflamação/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/efeitos dos fármacos , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/tratamento farmacológico
2.
Invest New Drugs ; 40(1): 198-208, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34519970

RESUMO

BACKGROUND: Peritumoral fibroblasts are key components of the tumor microenvironment. Through remodeling of the extracellular matrix (ECM) and secretion of pro-tumorigenic cytokines, peritumoral fibroblasts foster an immunosuppressive milieu conducive to tumor cell proliferation. In this study, we investigated if peritumoral fibroblasts could be therapeutically engineered to elicit an anti-cancer response by abolishing the proteolytic activities of membrane-bound metalloproteinases involved in ECM modulation. METHODS: A high affinity, glycosylphosphatidylinositol (GPI)-anchored Tissue Inhibitor of Metalloproteinase (TIMP) named "T1PrαTACE" was created for dual inhibition of MT1-MMP and TACE. T1PrαTACE was expressed in fibroblasts and its effects on cancer cell proliferation investigated in 3D co-culture models. RESULTS: T1PrαTACE abrogated the activities of MT1-MMP and TACE in host fibroblasts. As a GPI protein, T1PrαTACE could spontaneously detach from the plasma membrane of the fibroblast to co-localize with MT1-MMP and TACE on neighboring cancer cells. In a 3D co-culture model, T1PrαTACE promoted adherence between the cancer cells and surrounding fibroblasts, which led to an attenuation in tumor development. CONCLUSION: Peritumoral fibroblasts can be modulated with the TIMP for the elimination of cancer cells. As a novel anti-tumor strategy, our approach could potentially be used in combination with conventional chemo- and immunotherapies for a more effective cancer therapy.


Assuntos
Fibroblastos/efeitos dos fármacos , Neoplasias/patologia , Inibidores Teciduais de Metaloproteinases/farmacologia , Proteínas ADAM/antagonistas & inibidores , Proteína ADAM17/antagonistas & inibidores , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos
3.
Int J Mol Sci ; 21(14)2020 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-32698506

RESUMO

Tumour growth is not solely driven by tumour cell-intrinsic mechanisms, but also depends on paracrine signals provided by the tumour micro-environment. These signals comprise cytokines and growth factors that are synthesized as trans-membrane proteins and need to be liberated by limited proteolysis also termed ectodomain shedding. Members of the family of A disintegrin and metalloproteases (ADAM) are major mediators of ectodomain shedding and therefore initiators of paracrine signal transduction. In this review, we summarize the current knowledge on how ADAM proteases on tumour cells but also on cells of the tumour micro-environment contribute to the formation of gastrointestinal tumours, and discuss how these processes can be exploited pharmacologically.


Assuntos
Proteínas ADAM/metabolismo , Neoplasias Gastrointestinais/metabolismo , Transdução de Sinais , Proteínas ADAM/antagonistas & inibidores , Animais , Citocinas/metabolismo , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/patologia , Humanos , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
4.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-33096780

RESUMO

ADAM9 (A disintegrin and a metalloprotease 9) is a membrane-anchored protein that participates in a variety of physiological functions, primarily through the disintegrin domain for adhesion and the metalloprotease domain for ectodomain shedding of a wide variety of cell surface proteins. ADAM9 influences the developmental process, inflammation, and degenerative diseases. Recently, increasing evidence has shown that ADAM9 plays an important role in tumor biology. Overexpression of ADAM9 has been found in several cancer types and is correlated with tumor aggressiveness and poor prognosis. In addition, through either proteolytic or non-proteolytic pathways, ADAM9 promotes tumor progression, therapeutic resistance, and metastasis of cancers. Therefore, comprehensively understanding the mechanism of ADAM9 is crucial for the development of therapeutic anti-cancer strategies. In this review, we summarize the current understanding of ADAM9 in biological function, pathophysiological diseases, and various cancers. Recent advances in therapeutic strategies using ADAM9-related pathways are presented as well.


Assuntos
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/patologia , Doenças Retinianas/patologia , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Doenças Neurodegenerativas/metabolismo , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Doenças Retinianas/metabolismo , Sorafenibe/farmacologia , Microambiente Tumoral
5.
Clin Sci (Lond) ; 133(1): 83-99, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30635388

RESUMO

Ectodomain shedding of extracellular and membrane proteins is of fundamental importance for cell-cell communication in neoplasias. A Disintegrin And Metalloproteinase (ADAM) proteases constitute a family of multifunctional, membrane-bound proteins with traditional sheddase functions. Their protumorigenic potential has been attributed to both, essential (ADAM10 and ADAM17) and 'dispensable' ADAM proteases (ADAM8, 9, 12, 15, and 19). Of specific interest in this review is the ADAM proteinase ADAM8 that has been identified as a significant player in aggressive malignancies including breast, pancreatic, and brain cancer. High expression levels of ADAM8 are associated with invasiveness and predict a poor patient outcome, indicating a prognostic and diagnostic potential of ADAM8. Current knowledge of substrates and interaction partners gave rise to the hypothesis that ADAM8 dysregulation affects diverse processes in tumor biology, attributable to different functional cores of the multidomain enzyme. Proteolytic degradation of extracellular matrix (ECM) components, cleavage of cell surface proteins, and subsequent release of soluble ectodomains promote cancer progression via induction of angiogenesis and metastasis. Moreover, there is increasing evidence for significance of a non-proteolytic function of ADAM8. With the disintegrin (DIS) domain ADAM8 binds integrins such as ß1 integrin, thereby activating integrin signaling pathways. The cytoplasmic domain is critical for that activation and involves focal adhesion kinase (FAK), extracellular regulated kinase (ERK1/2), and protein kinase B (AKT/PKB) signaling, further contributing to cancer progression and mediating chemoresistance against first-line therapies. This review highlights the remarkable effects of ADAM8 in tumor biology, concluding that pharmacological inhibition of ADAM8 represents a promising therapeutic approach not only for monotherapy, but also for combinatorial therapies.


Assuntos
Proteínas ADAM/metabolismo , Biomarcadores Tumorais/metabolismo , Movimento Celular , Resistencia a Medicamentos Antineoplásicos , Proteínas de Membrana/metabolismo , Neoplasias/enzimologia , Proteínas ADAM/antagonistas & inibidores , Animais , Antineoplásicos/uso terapêutico , Progressão da Doença , Humanos , Proteínas de Membrana/antagonistas & inibidores , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Inibidores de Proteases/uso terapêutico , Proteólise , Transdução de Sinais , Especificidade por Substrato
6.
Clin Exp Rheumatol ; 37 Suppl 120(5): 130-134, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31621572

RESUMO

From the time of their discovery in 1999, the aggrecanases, and ADAMTS-5 in particular, have been heavily investigated as targets for disease-modifying osteoarthritis drug (DMOAD) development. Here, we provide a brief narrative review of the discovery efforts to target these enzymes, and how this led to the current ongoing programmes that hold promise for the future. We discuss a comparison of inhibition of collagen breakdown versus inhibition of aggrecan breakdown. We then summarise existing programmes that target ADAMTS-5, including small molecule inhibitors, monoclonal neutralising antibodies and nanobodies, and gene editing technologies. We also briefly discuss the potential analgesic effects this strategy may offer in addition to its joint-protective effects.


Assuntos
Proteínas ADAM , Endopeptidases/metabolismo , Osteoartrite/enzimologia , Pró-Colágeno N-Endopeptidase , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/imunologia , Proteína ADAMTS4 , Agrecanas/metabolismo , Humanos , Osteoartrite/tratamento farmacológico , Osteoartrite/imunologia
7.
J Biol Chem ; 292(24): 10112-10122, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28455445

RESUMO

Platelets are the sole source of EGF in circulation, yet how EGF is stored or released from stimulated cells is undefined. In fact, we found platelets did not store EGF, synthesized as a single 6-kDa domain in pro-EGF, but rather expressed intact pro-EGF precursor on granular and plasma membranes. Activated platelets released high-molecular-weight (HMW)-EGF, produced by a single cleavage between the EGF and the transmembrane domains of pro-EGF. We synthesized a fluorogenic peptide encompassing residues surrounding the putative sessile arginyl residue and found stimulated platelets released soluble activity that cleaved this pro-EGF1020-1027 peptide. High throughput screening identified chymostatins, bacterial peptides with a central cyclic arginyl structure, as inhibitors of this activity. In contrast, the matrix metalloproteinase/TACE (tumor necrosis factor-α-converting enzyme) inhibitor GM6001 was ineffective. Stimulated platelets released the soluble protease ADAMDEC1, recombinant ADAMDEC1 hydrolyzed pro-EGF1020-1027, and this activity was inhibited by chymostatin and not GM6001. Biotinylating platelet surface proteins showed ADAMDEC1 hydrolyzed surface pro-EGF to HMW-EGF that stimulated HeLa EGF receptor (EGFR) reporter cells and EGFR-dependent tumor cell migration. This proteolysis was inhibited by chymostatin and not GM6001. Metabolizing pro-EGF Arg1023 to citrulline with recombinant polypeptide arginine deiminase 4 (PAD4) abolished ADAMDEC1-catalyzed pro-EGF1020-1027 peptidolysis, while pretreating intact platelets with PAD4 suppressed ADAMDEC1-, thrombin-, or collagen-induced release of HMW-EGF. We conclude that activated platelets release ADAMDEC1, which hydrolyzes pro-EGF to soluble HMW-EGF, that HMW-EGF is active, that proteolytic cleavage of pro-EGF first occurs at the C-terminal arginyl residue of the EGF domain, and that proteolysis is the regulated and rate-limiting step in generating soluble EGF bioactivity from activated platelets.


Assuntos
Proteínas ADAM/metabolismo , Plaquetas/enzimologia , Membrana Celular/enzimologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/agonistas , Ativação Plaquetária , Precursores de Proteínas/metabolismo , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/química , Proteínas ADAM/genética , Animais , Plaquetas/metabolismo , Células CHO , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cricetulus , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Hidrolases/genética , Hidrolases/metabolismo , Cinética , Peso Molecular , Oligopeptídeos/farmacologia , Inibidores de Proteases/farmacologia , Domínios e Motivos de Interação entre Proteínas , Precursores de Proteínas/química , Precursores de Proteínas/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas , Proteólise/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Solubilidade
8.
Cell Physiol Biochem ; 50(1): 136-149, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30278449

RESUMO

BACKGROUND/AIMS: Most of estrogen receptor positive breast cancer patients respond well initially to endocrine therapies, but often develop resistance during treatment with selective estrogen receptor modulators (SERMs) such as tamoxifen. Altered expression and functions of microRNAs (miRNAs) have been reportedly associated with tamoxifen resistance. Thus, it is necessary to further elucidate the function and mechanism of miRNAs in tamoxifen resistance. METHODS: Tamoxifen sensitivity was validated by using Cell Counting Kit-8 in tamoxifen-sensitive breast cancer cells (MCF-7, T47D) and tamoxifen-resistant cells (MCF-7/TAM, T47D/ TAM). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression level of miR-449a in tamoxifen-sensitive/-resistant cells and patient serums. Dual-luciferase assay was used to identify the binding of miR-449a and predicted gene ADAM22. The expression level of ADAM22 was determined by qRT-PCR and western blotting in miR-449a +/- breast cancer cells. Subsequently, rescue experiments were carried out to identify the function of ADAM22 in miR-449a-reduced tamoxifen resistance. Finally, Gene ontology (GO) and Protein-protein interaction analyses were performed to evaluate the potential mechanisms of ADAM22 in regulating tamoxifen resistance. RESULTS: MiR-449a levels were downregulated significantly in tamoxifen-resistant breast cancer cells when compared with their parental cells, as well as in clinical breast cancer serum samples. Overexpression of miR-449a re-sensitized the tamoxifen-resistant breast cancer cells, while inhibition of miR-449a conferred tamoxifen resistance in parental cells. Luciferase assay identified ADAM22 as a direct target gene of miR-449a. Additionally, silencing of ADAM22 could reverse tamoxifen resistance induced by miR-449a inhibition in ER-positive breast cancer cells. GO analysis results showed ADAM22 was mainly enriched in the biological processes of cell adhesion, cell differentiation, gliogenesis and so on. Protein-protein interaction analyses appeared that ADAM22 might regulate tamoxifen resistance through PPARG, LGI1, KRAS and LYN. CONCLUSION: Decreased miR-449a causes the upregulation of ADAM22, which induces tamoxifen resistance of breast cancer cells. These results suggest that miR-449a, functioning by targeting ADAM22, contributes to the mechanisms underlying breast cancer endocrine resistance, which may provide a potential therapeutic strategy in ER-positive breast cancers.


Assuntos
Proteínas ADAM/metabolismo , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/genética , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Tamoxifeno/farmacologia , Regiões 3' não Traduzidas , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Antagomirs/metabolismo , Antineoplásicos Hormonais/uso terapêutico , Sequência de Bases , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , PPAR gama/metabolismo , Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Tamoxifeno/uso terapêutico
9.
Biochem J ; 474(9): 1467-1479, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28264989

RESUMO

ADAM9 (A Disintegrin And Metalloprotease 9) is a membrane-anchored metalloproteinase that has been implicated in pathological retinal neovascularization and in tumor progression. ADAM9 has constitutive catalytic activity in both biochemical and cell-based assays and can cleave several membrane proteins, including epidermal growth factor and Ephrin receptor B4; yet little is currently known about the catalytic properties of ADAM9 and its post-translational regulation and inhibitor profile in cell-based assays. To address this question, we monitored processing of the membrane-anchored Ephrin receptor B4 (EphB4) by co-expressing ADAM9, with the catalytically inactive ADAM9 E > A mutant serving as a negative control. We found that ADAM9-dependent shedding of EphB4 was not stimulated by three commonly employed activators of ADAM-dependent ectodomain shedding: phorbol esters, pervanadate or calcium ionophores. With respect to the inhibitor profile, we found that ADAM9 was inhibited by the hydroxamate-based metalloprotease inhibitors marimastat, TAPI-2, BB94, GM6001 and GW280264X, and by 10 nM of the tissue inhibitor of metalloproteinases (TIMP)-3, but not by up to 20 nM of TIMP-1 or -2. Additionally, we screened a non-hydroxamate small-molecule library for novel ADAM9 inhibitors and identified four compounds that selectively inhibited ADAM9-dependent proteolysis over ADAM10- or ADAM17-dependent processing. Taken together, the present study provides new information about the molecular fingerprint of ADAM9 in cell-based assays by showing that it is not stimulated by strong activators of ectodomain shedding and by defining a characteristic inhibitor profile. The identification of novel non-hydroxamate inhibitors of ADAM9 could provide the basis for designing more selective compounds that block the contribution of ADAM9 to pathological neovascularization and cancer.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/metabolismo , Membrana Celular/enzimologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Animais , Células COS , Catálise , Membrana Celular/efeitos dos fármacos , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Camundongos
10.
Clin Exp Allergy ; 47(10): 1263-1274, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28378503

RESUMO

BACKGROUND: IL-5 causes suspended eosinophils to polarize with filamentous (F)-actin and granules at one pole and the nucleus in a specialized uropod, the "nucleopod," which is capped with P-selectin glycoprotein ligand-1 (PSGL-1). IL-5 enhances eosinophil adhesion and migration on periostin, an extracellular matrix protein upregulated in asthma by type 2 immunity mediators. OBJECTIVE: Determine how the polarized morphology evolves to foster migration of IL-5-stimulated eosinophils on a surface coated with periostin. METHODS: Blood eosinophils adhering to adsorbed periostin were imaged at different time points by fluorescent microscopy, and migration of eosinophils on periostin was assayed. RESULTS: After 10 minutes in the presence of IL-5, adherent eosinophils were polarized with PSGL-1 at the nucleopod tip and F-actin distributed diffusely at the opposite end. After 30-60 minutes, the nucleopod had dissipated such that PSGL-1 was localized in a crescent or ring away from the cell periphery, and F-actin was found in podosome-like structures. The periostin layer, detected with monoclonal antibody Stiny-1, shown here to recognize the FAS1 4 module, was cleared in wide areas around adherent eosinophils. Clearance was attenuated by metalloproteinase inhibitors or antibodies to disintegrin metalloproteinase 8 (ADAM8), a major eosinophil metalloproteinase previously implicated in asthma pathogenesis. ADAM8 was not found in podosome-like structures, which are associated with proteolytic activity in other cell types. Instead, immunoblotting demonstrated proteoforms of ADAM8 that lack the cytoplasmic tail in the supernatant. Anti-ADAM8 inhibited migration of IL-5-stimulated eosinophils on periostin. CONCLUSIONS AND CLINICAL RELEVANCE: Migrating IL-5-activated eosinophils on periostin exhibit loss of nucleopodal features and appearance of prominent podosomes along with clearance of the Stiny-1 periostin epitope. Migration and epitope clearance are both attenuated by inhibitors of ADAM8. We propose, therefore, that eosinophils remodel and migrate on periostin-rich extracellular matrix in the asthmatic airway in an ADAM8-dependent manner, making ADAM8 a possible therapeutic target.


Assuntos
Proteínas ADAM/metabolismo , Moléculas de Adesão Celular/metabolismo , Quimiotaxia de Leucócito/imunologia , Eosinófilos/imunologia , Eosinófilos/metabolismo , Interleucina-5/metabolismo , Proteínas de Membrana/metabolismo , Proteínas ADAM/antagonistas & inibidores , Adesão Celular , Células Cultivadas , Quimiotaxia de Leucócito/efeitos dos fármacos , Eosinófilos/efeitos dos fármacos , Imunofluorescência , Humanos , Interleucina-5/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Metaloproteases/antagonistas & inibidores , Metaloproteases/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Tiofenos/farmacologia
11.
Biochem J ; 473(1): e1-4, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26657033

RESUMO

The extracellular matrix of articular cartilage is structurally specialized for efficient absorption of mechanical impact. In particular, giant aggregates of the large chondroitin sulfate proteoglycan, aggrecan, with the glycosaminoglycan, hyaluronan, allow cartilage to resist compressive load. Proteolysis of aggrecan by members of the proteinase family ADAMTS (A disintegrin-like and metalloproteinase domain with thrombospondin type 1 motif), was identified as an early step in the inexorable destruction of cartilage in osteoarthritis (OA). Of the investigated proteinases, ADAMTS5 has emerged as a principal mediator of aggrecan loss in OA, convincingly so in mouse models, and with high probability in humans. ADAMTS5 has a bipartite organization, comprising a proteinase domain and an ancillary domain containing exosites for interaction with aggrecan and other substrates. In a recent issue of this journal, Santamaria et al. characterized anti-ADAMTS5 monoclonal antibodies isolated from a phage display library. By blocking the catalytic site of the ADAMTS5 immunogen with a synthetic inhibitor, the authors of the paper biased selection of antibodies to the ancillary domain. This work, together with other antibodies targeting ADAMTS5, offers diverse, high-affinity and, as far as can be determined, selective aggrecanase inhibitors. Mapping of their epitopes provided novel insights into ADAMTS5 interactions with aggrecan. These monoclonal antibodies deserve continued investigation for potential arthritis therapy, although their successful use will require a comprehensive understanding of the physiological roles of ADAMTS5, and its regulation, intrinsic properties and intermolecular interactions.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/imunologia , Anticorpos Monoclonais/imunologia , Endopeptidases/metabolismo , Osteoartrite/imunologia , Proteína ADAMTS5 , Animais , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Osteoartrite/tratamento farmacológico , Osteoartrite/enzimologia
12.
Mediators Inflamm ; 2017: 7281986, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28265178

RESUMO

Obesity is one of the most prevalent metabolic diseases in the Western world and correlates directly with insulin resistance, which may ultimately culminate in type 2 diabetes (T2D). We sought to ascertain whether the human metalloproteinase A Disintegrin and Metalloproteinase 19 (ADAM19) correlates with parameters of the metabolic syndrome in humans and mice. To determine the potential novel role of ADAM19 in the metabolic syndrome, we first conducted microarray studies on peripheral blood mononuclear cells from a well-characterised human cohort. Secondly, we examined the expression of ADAM19 in liver and gonadal white adipose tissue using an in vivo diet induced obesity mouse model. Finally, we investigated the effect of neutralising ADAM19 on diet induced weight gain, insulin resistance in vivo, and liver TNF-α levels. Significantly, we show that, in humans, ADAM19 strongly correlates with parameters of the metabolic syndrome, particularly BMI, relative fat, HOMA-IR, and triglycerides. Furthermore, we identified that ADAM19 expression was markedly increased in the liver and gonadal white adipose tissue of obese and T2D mice. Excitingly, we demonstrate in our diet induced obesity mouse model that neutralising ADAM19 therapy results in weight loss, improves insulin sensitivity, and reduces liver TNF-α levels. Our novel data suggest that ADAM19 is pro-obesogenic and enhances insulin resistance. Therefore, neutralisation of ADAM19 may be a potential therapeutic approach to treat obesity and T2D.


Assuntos
Proteínas ADAM/metabolismo , Síndrome Metabólica/metabolismo , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Animais , Dieta Hiperlipídica/efeitos adversos , Humanos , Resistência à Insulina/imunologia , Resistência à Insulina/fisiologia , Leucócitos Mononucleares/metabolismo , Fígado/metabolismo , Masculino , Síndrome Metabólica/imunologia , Síndrome Metabólica/patologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Fator de Necrose Tumoral alfa/metabolismo
13.
Int J Mol Sci ; 18(4)2017 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-28430139

RESUMO

Obesity and diabetes are major causes of morbidity and mortality globally. The current study builds upon our previous association studies highlighting that A Disintegrin And Metalloproteinase 28 (ADAM28) appears to be implicated in the pathogenesis of obesity and type 2 diabetes in humans. Our novel study characterised the expression of ADAM28 in mice with the metabolic syndrome and used molecular inhibition approaches to investigate the functional role of ADAM28 in the pathogenesis of high fat diet-induced obesity. We identified that ADAM28 mRNA and protein expression was markedly increased in the livers of mice with the metabolic syndrome. In addition, noradrenaline, the major neurotransmitter of the sympathetic nervous system, results in elevated Adam28 mRNA expression in human monocytes. Downregulation of ADAM28 with siRNA technology resulted in a lack of weight gain, promotion of insulin sensitivity/glucose tolerance and decreased liver tumour necrosis factor-α (TNF-α) levels in our diet-induced obesity mouse model as well as reduced blood urea nitrogen, alkaline phosphatase and aspartate aminotransferase. In addition, we show that ADAM28 knock-out mice also displayed reduced body weight, elevated high density lipoprotein cholesterol levels, and reductions in blood urea nitrogen, alkaline phosphatase, and aspartate aminotransferase. The results of this study provide important insights into the pathogenic role of the metalloproteinase ADAM28 in the metabolic syndrome and suggests that downregulation of ADAM28 may be a potential therapeutic strategy in the metabolic syndrome.


Assuntos
Proteínas ADAM/metabolismo , Síndrome Metabólica/etiologia , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Fosfatase Alcalina/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Nitrogênio da Ureia Sanguínea , Linhagem Celular , HDL-Colesterol/metabolismo , Dieta Hiperlipídica , Ensaio de Imunoadsorção Enzimática , Humanos , Fígado/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Norepinefrina/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
J Biol Chem ; 290(10): 6620-9, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25564618

RESUMO

ADAMDEC1 is a proteolytically active metzincin metalloprotease displaying rare active site architecture with a zinc-binding Asp residue (Asp-362). We previously demonstrated that substitution of Asp-362 for a His residue, thereby reconstituting the canonical metzincin zinc-binding environment with three His zinc ligands, increases the proteolytic activity. The protease also has an atypically short domain structure with an odd number of Cys residues in the metalloprotease domain. Here, we investigated how these rare structural features in the ADAMDEC1 metalloprotease domain impact the proteolytic activity, the substrate specificity, and the effect of inhibitors. We identified carboxymethylated transferrin (Cm-Tf) as a new ADAMDEC1 substrate and determined the primary and secondary cleavage sites, which suggests a strong preference for Leu in the P1' position. Cys(392), present in humans but only partially conserved within sequenced ADAMDEC1 orthologs, was found to be unpaired, and substitution of Cys(392) for a Ser increased the reactivity with α2-macroglobulin but not with casein or Cm-Tf. Substitution of Asp(362) for His resulted in a general increase in proteolytic activity and a change in substrate specificity was observed with Cm-Tf. ADAMDEC1 was inhibited by the small molecule inhibitor batimastat but not by tissue inhibitor of metalloproteases (TIMP)-1, TIMP-2, or the N-terminal inhibitory domain of TIMP-3 (N-TIMP-3). However, N-TIMP-3 displayed profound inhibitory activity against the D362H variants with a reconstituted consensus metzincin zinc-binding environment. We hypothesize that these unique features of ADAMDEC1 may have evolved to escape from inhibition by endogenous metalloprotease inhibitors.


Assuntos
Proteínas ADAM/química , Domínio Catalítico , Metaloproteases/química , Inibidores Teciduais de Metaloproteinases/química , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Sequência de Aminoácidos/genética , Cristalografia por Raios X , Regulação Enzimológica da Expressão Gênica , Humanos , Metaloproteases/antagonistas & inibidores , Metaloproteases/genética , Estrutura Terciária de Proteína , Proteólise , Especificidade por Substrato , Inibidores Teciduais de Metaloproteinases/metabolismo , Zinco/química
15.
Hum Mol Genet ; 23(15): 4064-76, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24643277

RESUMO

iRHOM2 is a highly conserved, catalytically inactive member of the Rhomboid family, which has recently been shown to regulate the maturation of the multi-substrate ectodomain sheddase enzyme ADAM17 (TACE) in macrophages. Dominant iRHOM2 mutations are the cause of the inherited cutaneous and oesophageal cancer-susceptibility syndrome tylosis with oesophageal cancer (TOC), suggesting a role for this protein in epithelial cells. Here, using tissues derived from TOC patients, we demonstrate that TOC-associated mutations in iRHOM2 cause an increase in the maturation and activity of ADAM17 in epidermal keratinocytes, resulting in significantly upregulated shedding of ADAM17 substrates, including EGF-family growth factors and pro-inflammatory cytokines. This activity is accompanied by increased EGFR activity, increased desmosome processing and the presence of immature epidermal desmosomes, upregulated epidermal transglutaminase activity and heightened resistance to Staphylococcal infection in TOC keratinocytes. Many of these features are consistent with the presence of a constitutive wound-healing-like phenotype in TOC epidermis, which may shed light on a novel pathway in skin repair, regeneration and inflammation.


Assuntos
Proteínas ADAM/genética , Proteínas de Transporte/genética , Epiderme/metabolismo , Neoplasias Esofágicas/genética , Queratinócitos/metabolismo , Ceratodermia Palmar e Plantar/genética , Infecções Cutâneas Estafilocócicas/genética , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/metabolismo , Proteína ADAM17 , Proteínas de Transporte/metabolismo , Citocinas/biossíntese , Desmossomos/metabolismo , Desmossomos/patologia , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Epiderme/microbiologia , Epiderme/patologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/microbiologia , Neoplasias Esofágicas/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Queratinócitos/microbiologia , Queratinócitos/patologia , Ceratodermia Palmar e Plantar/metabolismo , Ceratodermia Palmar e Plantar/microbiologia , Ceratodermia Palmar e Plantar/patologia , Masculino , Mutação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Infecções Cutâneas Estafilocócicas/metabolismo , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Staphylococcus aureus/fisiologia , Transglutaminases/genética , Transglutaminases/metabolismo
16.
Osteoarthritis Cartilage ; 24(2): 299-306, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26410555

RESUMO

OBJECTIVE: The primary goal of this study was to test the disease-modifying effect of blocking a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5 with a neutralizing monoclonal antibody (mAb) starting 4 weeks after destabilization of the medial meniscus (DMM) in the mouse. We also investigated whether ADAMTS-5 blockade reversed mechanical allodynia and decreased monocyte chemoattractant protein (MCP)-1 production by dorsal root ganglia (DRG) cells. METHODS: Ten-week old male C57BL/6 mice underwent DMM surgery and were either left untreated or treated with anti-ADAMTS-5 mAb or IgG2c isotype control mAb starting 4 weeks after surgery. Knees were collected for histopathology 4 or 12 weeks later. Mechanical allodynia was monitored biweekly in the ipsilateral hind paw through 16 weeks. DRG were collected and cultured 8 weeks after DMM for analysis of MCP-1 production. RESULTS: By 4 weeks after DMM, mild cartilage degeneration was evident in the medial compartment, small osteophytes were present, and subchondral bone sclerosis was established. By 16 weeks after surgery, significant cartilage deterioration was apparent on the medial tibial plateaux and medial femoral condyles, osteophyte size had increased, and subchondral bone sclerosis was maintained. Treatment with ADAMTS-5 mAb from week 4 to 16 after surgery slowed cartilage degeneration and osteophyte growth but did not affect subchondral bone sclerosis. Moreover, ADAMTS-5 blockade resulted in temporary reversal of mechanical allodynia, which correlated with decreased MCP-1 production by cultured DRG cells. CONCLUSIONS: This study suggests therapeutic efficacy of an ADAMTS-5 mAb in the DMM model, when therapy starts early in disease.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Anticorpos Neutralizantes/farmacologia , Quimiocina CCL2/efeitos dos fármacos , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/fisiopatologia , Nociceptividade/efeitos dos fármacos , Osteoartrite do Joelho/fisiopatologia , Joelho de Quadrúpedes/efeitos dos fármacos , Proteínas ADAM/imunologia , Proteína ADAMTS5 , Animais , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite do Joelho/metabolismo , Lesões do Menisco Tibial
17.
Anal Biochem ; 514: 42-47, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27637923

RESUMO

A disintegrin and metalloproteinase 15 (ADAM15), also known as metargidin, plays important roles in regulating inflammation, wound healing, neovascularization, and is an attractive drug target. Fluorescence resonance energy transfer (FRET)-based peptide substrates were tested to identify candidate reagents for high throughput screening and detection of ADAM15 in biological samples. ADAM15 exhibits a unique and diverse activity profile compared to other metalloproteinases. Two FRET substrates, Dabcyl-Gly-Pro-Leu-Gly-Met-Arg-Gly-Lys(FAM)-NH2 (PEPDAB011) and Dabcyl-Ala-Pro-Arg-Trp-Ile-Gln-Asp-Lys(FAM)-NH2 (PEPDAB017), which also detect activities of several matrix metalloproteinases (MMPs -2, -9, and -13), were efficiently cleaved by ADAM15 with specificity constants of 5800 M-1 s-1 and 4300 M-1 s-1, respectively. Additionally, ADAM15 efficiently processed Dabcyl-Leu-Arg-Glu-Gln-Gln-Arg-Leu-Lys-Ser-Lys(FAM)-NH2 (PEPDAB022), which is based on a physiological CD23 cleavage site, with a specificity constant (kcat/Km) of 5200 M-1 s-1. PEPDAB022 was used to screen the ability of known metalloproteinase inhibitors including TAPI-2, marimastat, GI-254023, and the Tissue Inhibitor of Metalloproteinases(TIMPs) 1 and 3 to block ADAM15 activity. Even though ADAM15 exhibits similar substrate preferences to other metalloproteinases, many broad spectrum inhibitors failed to block ADAM15 activity at concentrations as high as 50 µM. Thus, a clear need exists to develop potent and selective ADAM15 inhibitors, and the FRET substrates described herein should aid future research efforts towards this aim.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/metabolismo , Corantes Fluorescentes/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Dipeptídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Ácidos Hidroxâmicos/farmacologia , Cinética , Peptídeos/química , Peptídeos/metabolismo
18.
Alcohol Clin Exp Res ; 40(2): 273-83, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26842246

RESUMO

BACKGROUND: Farm workers in rural areas consume more alcohol than those who reside in urban areas. Occupational exposures such as agricultural work can pose hazards on the respiratory system. It is established that hog barn dust induces inflammation in the airway, including the release of cytokines such as tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and IL-8. We have shown that alcohol alters airway epithelial innate defense through changes in both nitric oxide (NO) and cAMP-dependent protein kinase A (PKA). Simultaneous exposure to hog barn dust and alcohol decreases inflammatory mediators, TNF-α, IL-6, and IL-8, in mice. Previously, mice exposed to both alcohol and hog barn dust showed a depleted amount of lymphocytes compared to mice exposed only to hog barn dust. Weakening of the innate immune response could lead to enhanced susceptibility to disease. In addition, mice that were co-exposed to hog barn dust and alcohol also experienced increased mortality. METHODS: Because we recently demonstrated that PKA activation inhibits the TNF-α sheddase, TNF-α-converting enzyme (TACE), we hypothesized that an alcohol-mediated PKA pathway blocks TACE activity and prevents the normative inflammatory response to hog barn dust exposure. To delineate these effects, we used PKA pathway inhibitors (adenylyl cyclase [AC], cAMP, and PKA) to modulate the effects of alcohol on dust-stimulated TNF-α release in the bronchial epithelial cell line, BEAS-2B. Alcohol pretreatment blocked TACE activity and TNF-α release in hog barn dust-treated cells. RESULTS: Alcohol continued to block hog barn dust-mediated TNF-α release in the presence of the particulate AC inhibitor, SQ22,536. The soluble adenylyl cyclase inhibitor, KH7, however, significantly increased the inflammatory response to hog barn dust. phosphodiesterase 4 inhibitors significantly elevated cAMP and enhanced alcohol-mediated inhibition of dust-stimulated TNF-α release. In addition, the NO synthase inhibitor, l-NMMA, also reversed the alcohol-blocking effect on dust-stimulated TNF-α. CONCLUSIONS: These data suggest that alcohol requires a soluble cyclase-generated cAMP-PKA pathway that is dependent upon the action of NO to inhibit TACE and TNF-α release. These findings support our observations that alcohol functions through a dual NO and PKA pathway in bronchial epithelial cells.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Poeira , Etanol/farmacologia , Óxido Nítrico/fisiologia , Mucosa Respiratória/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Proteínas ADAM/fisiologia , Proteína ADAM17 , Adenina/análogos & derivados , Adenina/farmacologia , Brônquios/citologia , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Humanos , Inflamação/fisiopatologia , Interleucina-6/fisiologia , Interleucina-8/fisiologia , Fator de Necrose Tumoral alfa/fisiologia
19.
Arterioscler Thromb Vasc Biol ; 35(11): 2336-42, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26338302

RESUMO

OBJECTIVE: The metalloprotease ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) regulates the size of von Willebrand factor multimers. A deficiency in ADAMTS13 activity is associated with the life-threatening disease thrombotic thrombocytopenic purpura (TTP). The vast majority of patients have acquired TTP, where circulating anti-ADAMTS13 autoantibodies are causative for the decreased ADAMTS13 activity. Current treatment consists of plasma exchange, but improved therapies are highly warranted. APPROACH AND RESULTS: We have developed a new rat model mimicking various aspects of acquired TTP to investigate the therapeutic efficacy of human recombinant ADAMTS13. A polyclonal antibody against ADAMTS13 completely blocked endogenous rat ADAMTS13 activity in Sprague-Dawley rats. When TTP was triggered using recombinant von Willebrand factor, the animals displayed severe TTP-like symptoms, such as thrombocytopenia, hemolytic anemia, and von Willebrand factor-rich thrombi in the kidneys and brain. Subsequent injection of 400, 800, or 1600 U/kg recombinant ADAMTS13 prevented full development of these symptoms. Analysis of plasma samples confirmed that recombinant ADAMTS13 was able to override circulating anti-ADAMTS13 inhibitory antibodies, resulting in restoration of ADAMTS13 activity and degradation of ultralarge von Willebrand factor multimers. CONCLUSIONS: Recombinant ADAMTS13 was shown to be effective in averting severe acquired TTP-like symptoms in rats and holds promising value for the treatment of this severe and life-threatening disease in humans.


Assuntos
Proteínas ADAM/farmacologia , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/sangue , Proteínas ADAM/imunologia , Proteína ADAMTS13 , Anemia Hemolítica/sangue , Anemia Hemolítica/induzido quimicamente , Anemia Hemolítica/prevenção & controle , Animais , Anticorpos , Modelos Animais de Doenças , Estudos de Viabilidade , Humanos , Masculino , Púrpura Trombocitopênica Trombótica/sangue , Púrpura Trombocitopênica Trombótica/induzido quimicamente , Púrpura Trombocitopênica Trombótica/imunologia , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Índice de Gravidade de Doença , Fatores de Tempo , Fator de von Willebrand
20.
Bioorg Med Chem ; 24(18): 4032-4037, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27407033

RESUMO

The metalloproteinase ADAM8 serves as a pivotal catalyst in the development of inflammatory diseases and cancer metastasis. The cyclic peptide cyclo(RLsKDK) has been shown to inhibit the enzymatic activity of ADAM8 with high specificity and potency. Herein we report a structure-activity relationship (SAR) study of cyclo(RLsKDK) that involves the synthesis and biological evaluation of the lead compound and structural analogues thereof. This study provides insight into the ligand-receptor interactions that govern the binding of cyclo(RLsKDK) to the ADAM8 disintegrin domain and represents a stepping stone for the development of new treatments for inflammatory diseases and cancer metastasis.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Proteínas de Membrana/antagonistas & inibidores , Metástase Neoplásica/tratamento farmacológico , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Proteínas ADAM/imunologia , Animais , Células COS , Chlorocebus aethiops , Humanos , Inflamação/imunologia , Proteínas de Membrana/imunologia , Metástase Neoplásica/imunologia , Peptidomiméticos/química , Peptidomiméticos/farmacologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA