Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Int J Chron Obstruct Pulmon Dis ; 17: 2589-2602, 2022.
Article in English | MEDLINE | ID: mdl-36267325

ABSTRACT

Background: It has been reported that a disintegrin and metalloproteinase 9 (ADAM9) is involved in the pathogenesis of cigarette smoke (CS)-associated chronic obstructive pulmonary disease (COPD). But how CS exposure leads to upregulation of ADAM9 remains unknown. Methods: Patients who underwent lobectomy for a solitary pulmonary nodule were enrolled and divided into three groups: non-smokers with normal lung function, smokers without COPD and smoker patients with COPD. Immunoreactivity of interleukin (IL)-17A and ADAM9 in small airways and alveolar walls was measured by immunohistochemistry. Wild-type and Il17a -/- C57BL/6 mice were exposed to CS for six months, and ADAM9 expression in the airway epithelia was measured by immunoreactivity. In addition, the protein and mRNA expression levels of IL-17A and ADAM9 were assessed in CS extract (CSE) and/or IL-17A-treated human bronchial epithelial (HBE) cells. Results: The immunoreactivity of ADAM9 was increased in the airway epithelia and alveolar walls of patients with COPD compared to that of the controls. The expression of IL-17A was also upregulated in airway epithelial cells of patients with COPD and correlated positively with the level of ADAM9. The results from the animal model showed that Il17a -/- mice were protected from emphysema induced by CS exposure, together with a reduced level of ADAM9 expression in the airway epithelia, suggesting a possible link between ADAM9 and IL-17A. Consistently, our in vitro cell model showed that CSE stimulated the expression of ADAM9 and IL-17A in HBE cells in a dose- and time-dependent manner. Recombinant IL-17A induced ADAM9 upregulation in HBE cells and had a synergistic effect with CSE, whereas blocking IL-17A inhibited CSE-induced ADAM9 expression. Further analysis revealed that IL-17A induced c-Jun N-terminal kinase (JNK) phosphorylation, thereby increasing ADAM9 expression. Conclusion: Our results revealed a novel role of IL-17A in CS-related COPD, where IL-17A contributes to ADAM9 expression by activating JNK signaling.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Humans , Mice , Animals , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Interleukin-17/genetics , Disintegrins/metabolism , Cigarette Smoking/adverse effects , Mice, Inbred C57BL , Nicotiana , Epithelial Cells/metabolism , RNA, Messenger/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Plant Extracts , Membrane Proteins/genetics , Membrane Proteins/metabolism , ADAM Proteins/genetics , ADAM Proteins/metabolism
2.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163294

ABSTRACT

Understanding the mechanisms of modulators' action on enzymes is crucial for optimizing and designing pharmaceutical substances. The acute inflammatory response, in particular, is regulated mainly by a disintegrin and metalloproteinase (ADAM) 17. ADAM17 processes several disease mediators such as TNFα and APP, releasing their soluble ectodomains (shedding). A malfunction of this process leads to a disturbed inflammatory response. Chemical protease inhibitors such as TAPI-1 were used in the past to inhibit ADAM17 proteolytic activity. However, due to ADAM17's broad expression and activity profile, the development of active-site-directed ADAM17 inhibitor was discontinued. New 'exosite' (secondary substrate binding site) inhibitors with substrate selectivity raised the hope of a substrate-selective modulation as a promising approach for inflammatory disease therapy. This work aimed to develop a high-throughput screen for potential ADAM17 modulators as therapeutic drugs. By combining experimental and in silico methods (structural modeling and docking), we modeled the kinetics of ADAM17 inhibitor. The results explain ADAM17 inhibition mechanisms and give a methodology for studying selective inhibition towards the design of pharmaceutical substances with higher selectivity.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/drug effects , ADAM17 Protein/metabolism , ADAM Proteins/metabolism , Binding Sites/drug effects , Catalytic Domain/drug effects , Computer Simulation , Drug Evaluation, Preclinical/methods , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Kinetics , Protease Inhibitors/pharmacology , Substrate Specificity/drug effects
3.
Cell Adh Migr ; 14(1): 165-181, 2020 12.
Article in English | MEDLINE | ID: mdl-32875951

ABSTRACT

The ADAMs family belongs to the transmembrane protein superfamily of zinc-dependent metalloproteases, which consists of multiple domains. These domains have independent but complementary functions that enable them to participate in multiple biological processes. Among them, ADAM9 can not only participate in the degradation of extracellular matrix as a metalloprotease, but also mediate tumor cell adhesion through its deintegrin domain, which is closely related to tumor invasion and metastasis. It is widely expressed in a variety of tumor cells and can affect the proliferation, invasion and metastasis of related cancer cells. We provide our views on current progress, its increasing importance as a strategic treatment goal, and our vision for the future of ADAM9.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/metabolism , Neoplasms/metabolism , ADAM Proteins/genetics , Evolution, Molecular , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics
4.
Med Sci Monit ; 26: e920211, 2020 Jan 12.
Article in English | MEDLINE | ID: mdl-31927559

ABSTRACT

BACKGROUND The aim of this study was to investigate the mechanisms underlying the potential effects of hydrogen-rich water (HW) on articular cartilage in a rat osteoarthritis (OA) model. MATERIAL AND METHODS A rat model of OA was established using the modified Hulth method, and rats were forced to exercise for 30 min every day 1 week after surgery for 7 weeks. Mankin's method was used to score the severity of OA. The animals were assigned into the OA group, OA+HW group, and sham operation group. After 8 weeks, the animals in the OA group had a Mankin score >8 points, and HW was administered into the knee joint. After 2 weeks of treatment, articular cartilage was obtained for pathological examination, consisting of hematoxylin and eosin, toluidine blue, and Hoechst staining, as well as quantitative real-time PCR and Western blot analyses. This combination of pharmacological and molecular biological analyses was performed to examine the mechanism underlying the protective effect of HW on articular cartilage. RESULTS The antioxidant effects of HW suppressed oxidative damage, which may have aided the inhibition of ECM-degrading enzymes (MMP3, MMP13, ADAMT4, and ADAMT5), the upregulation of Col II and aggrecan expression, and the downregulation of COX-2, iNOS, and NO expression. The results of HE staining indicated intra-articular treatment of HW attenuated cartilage degradation. However, Hoechst staining in the OA group indicated the nuclei of the fragmented chondrocytes were condensed compared to the sham operation group, and this effect was inhibited by HW. CONCLUSIONS HW showed a protective effect against the progression of OA in an animal model, which may have been mediated by its anti-oxidant and anti-apoptotic activities.


Subject(s)
Apoptosis/drug effects , Cartilage, Articular/pathology , Extracellular Matrix/metabolism , Hydrogen/therapeutic use , Osteoarthritis/drug therapy , Oxidative Stress/drug effects , Protective Agents/therapeutic use , Water/pharmacology , ADAM Proteins/metabolism , Aggrecans/metabolism , Animals , Cartilage, Articular/drug effects , Caspase 3/metabolism , Collagen Type II/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Male , Matrix Metalloproteinases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/pathology , Protective Agents/pharmacology , Rats, Sprague-Dawley , bcl-2-Associated X Protein/metabolism
5.
J Cell Sci ; 132(2)2019 01 16.
Article in English | MEDLINE | ID: mdl-30598502

ABSTRACT

The distribution of the voltage-gated Kv1 K+ channels at the axon initial segment (AIS) influences neuronal intrinsic excitability. The Kv1.1 and Kv1.2 (also known as KCNA1 and KCNA2, respectively) subunits are associated with cell adhesion molecules (CAMs), including Caspr2 (also known as CNTNAP2) and LGI1, which are implicated in autoimmune and genetic neurological diseases with seizures. In particular, mutations in the LGI1 gene cause autosomal dominant lateral temporal lobe epilepsy (ADLTE). Here, by using rat hippocampal neurons in culture, we showed that LGI1 is recruited to the AIS where it colocalizes with ADAM22 and Kv1 channels. Strikingly, the missense mutations S473L and R474Q of LGI1 identified in ADLTE prevent its association with ADAM22 and enrichment at the AIS. Moreover, we observed that ADAM22 and ADAM23 modulate the trafficking of LGI1, and promote its ER export and expression at the overall neuronal cell surface. Live-cell imaging indicated that LGI1 is co-transported in axonal vesicles with ADAM22 and ADAM23. Finally, we showed that ADAM22 and ADAM23 also associate with Caspr2 and TAG-1 (also known as CNTN2) to be selectively targeted to different axonal sub-regions. Hence, the combinatorial expression of Kv1-associated CAMs may be critical to tune intrinsic excitability in physiological and epileptogenic contexts.


Subject(s)
ADAM Proteins/metabolism , Axons/metabolism , Epilepsy, Frontal Lobe/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mutation, Missense , Sleep Wake Disorders/metabolism , ADAM Proteins/genetics , Amino Acid Substitution , Animals , Axons/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Epilepsy, Frontal Lobe/genetics , Epilepsy, Frontal Lobe/pathology , HEK293 Cells , Hippocampus , Humans , Protein Transport/genetics , Rats , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism , Sleep Wake Disorders/genetics , Sleep Wake Disorders/pathology
6.
Curr Med Chem ; 26(15): 2661-2689, 2019.
Article in English | MEDLINE | ID: mdl-29589526

ABSTRACT

The ADAMs, together with ADAMTSs and snake venom metalloproteases (SVMPs), are members of the Adamalysin family. Differences in structural organization, functions and localization are known and their domains, catalytic or non-catalytic, show key roles in the substrate recognition and protease activity. Some ADAMs, as membrane-bound enzymes, show sheddase activity. Sheddases are key to modulation of functional proteins such as the tumor necrosis factor, growth factors, cytokines and their receptors, adhesion proteins, signaling molecules and stress molecules involved in immunity. These activities take part in the regulation of several physiological and pathological processes including inflammation, tumor growth, metastatic progression and infectious diseases. On these bases, some ADAMs are currently investigated as drug targets to develop new alternative therapies in many fields of medicine. This review will be focused on these aspects.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Protease Inhibitors/therapeutic use , ADAM Proteins/chemistry , ADAM Proteins/metabolism , ADAM Proteins/physiology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Catalytic Domain , Cell Line, Tumor , Humans , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology
7.
Biochim Biophys Acta Mol Cell Res ; 1865(11 Pt A): 1598-1610, 2018 11.
Article in English | MEDLINE | ID: mdl-30327201

ABSTRACT

ADAM17, a prominent member of the "Disintegrin and Metalloproteinase" (ADAM) family, controls vital cellular functions through cleavage of transmembrane substrates including TGF-alpha, Amphiregulin (AREG) and TNF-Receptor 1 (TNFR1). We recently presented evidence that surface exposure of phosphatidylserine (PS) is pivotal for ADAM17 to exert sheddase activity. Anoctamin-6 (ANO6) has Ca2+-dependent phospholipid scramblase activity and it followed that the functions of ANO6 and ADAM17 might be linked. We report that overexpression of ANO6 in HEK293T cells led to increased Ca2+-mediated PS-exposure that was indeed accompanied by enhanced release of AREG and TGF-alpha. The effect was not observed when cells were treated with the PKC-dependent ADAM17 activator PMA. Transformation of cells with a constitutively active ANO6 mutant led to spontaneous PS-exposure and to the release of ADAM17-substrates in the absence of any stimuli. Inhibitor experiments indicated that ANO6-mediated enhancement of substrate cleavage simultaneously broadened the spectrum of participating metalloproteinases. In complementary experiments, siRNA-mediated downregulation of ANO6 was shown to decrease ionophore-mediated release of TNFR1 in human umbilical vein endothelial cells (HUVECs). We conclude that ANO6, by virtue of its scramblase activity, may play a role as an important regulator of the ADAM-network in the plasma membrane.


Subject(s)
ADAM Proteins/metabolism , Anoctamins/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , ADAM17 Protein/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Ionomycin/pharmacology , Models, Biological , Mutation , Transforming Growth Factor alpha/metabolism
8.
Fitoterapia ; 110: 181-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27012760

ABSTRACT

Diketopiperazine is a natural products found from bacteria, fungi, marine sponges, gorgonian and red algae. They are cyclic dipeptides possessing relatively simple and rigid structures with chiral nature and various side chains. The compounds in this structure class have been known to possess diverse bioactivities including antibiotic activity, anti-cancer activity, neuroprotective activity, and anti-inflammatory activity. The endothelial cell protein C receptor (EPCR) plays an important role in the cytoprotective pathway and in the activation of protein C. Endothelial cell protein C receptor (EPCR) can be shed from the cell surface, which is mediated by tumor necrosis factor-α converting enzyme (TACE). However, little is known about the effects of diketopiperazine on EPCR shedding. We investigated this issue by monitoring the effects of diketopiperazine on phorbol-12-myristate 13-acetate (PMA)-, tumor necrosis factor (TNF)-α, and interleukin (IL)-1ß-induced EPCR shedding in human umbilical vein endothelial cells (HUVECs), and cecal ligation and puncture (CLP)-mediated EPCR shedding in mice and underlying mechanism. Here, three (1-3) of diketopiperazines were isolated from two strains of marine-derived bacteria and 1-3 induced potent inhibition of PMA-, TNF-α-, IL-1ß (in HUVECs), and CLP-induced EPCR shedding (in mice) via inhibition of phosphorylation of mitogen-activated protein kinases (MAPKs) such as p38, janus kinase (JNK), and extracellular signal-regulated kinase (ERK) 1/2. 1-3 also inhibited the expression and activity of PMA-induced TACE in HUVECs suggesting that p38, ERK1/2, and JNK could be molecular targets of 1-3. These results demonstrate the potential of 1-3 as an anti-EPCR shedding reagent against PMA-mediated and CLP-mediated EPCR shedding.


Subject(s)
Antigens, CD/metabolism , Bacillus/chemistry , Diketopiperazines/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Receptors, Cell Surface/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Endothelial Protein C Receptor , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-1beta , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Seawater/microbiology , Sepsis/metabolism , Tetradecanoylphorbol Acetate , Tumor Necrosis Factor-alpha
9.
Int Immunopharmacol ; 30: 36-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26637956

ABSTRACT

Sanmiao formula (SM) is a compound prescription, which has been used in traditional Chinese medicine since the Ming Dynasty for gouty and rheumatoid arthritis treatments. However, no evidence has been unfolded to show the relationship between SM and gouty arthritis (GA), particularly inhibiting cartilage matrix degradation. In the present study, we undertook a characterization of anti-GA activity of SM using an in vivo rat model induced by potassium oxonate and cold bath together with in vitro studies with chondrocytes for further molecular characterization. Potassium oxonate and cold bath rats were treated with SM at doses of 7.2g/kg per day for 5days. SM treatments significantly suppressed the swelling rate and the severe pathologic changes in the joints of the animals in gout model. Inflammatory factors count by ELISA analysis, SM exhibited inhibition on IL-1ß and TNF-α. Moreover, histological analysis of the joints and SM-serum substantially interfered with the MSU-induced expression of glycosaminoglycans (GAG), up-regulated the content of proteoglycan. Importantly, SM interfered with GA-augmented expression of matrix metalloproteinases (MMPs) -3 and aggrecanases (ADAMTS)-4, which are considered to be key enzymes in cartilage matrix degradation, and simultaneously augmented GA-reduced tissue inhibitors of metalloproteinases (TIMPs) -1 and -3 expression in the joints and chondrocytes. Therefore, SM is looking forward to be a potential novel agent that could prevent cartilage matrix degradation effectively in gouty arthritis, and this provides a new target for development of new medicines.


Subject(s)
Arthritis, Gouty/drug therapy , Chondrocytes/drug effects , Complex Mixtures/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Joints/drug effects , Matrilin Proteins/metabolism , Medicine, Chinese Traditional , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS4 Protein , Animals , Arthritis, Gouty/chemically induced , Cells, Cultured , Chondrocytes/physiology , Down-Regulation , Humans , Hydrolysis/drug effects , Hydrolysis/radiation effects , Interleukin-1beta/metabolism , Joints/pathology , Male , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Models, Animal , Oxonic Acid/administration & dosage , Procollagen N-Endopeptidase/genetics , Procollagen N-Endopeptidase/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
10.
Am J Physiol Renal Physiol ; 310(6): F534-46, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26697977

ABSTRACT

Circulating and renal activity of angiotensin-converting enzyme 2 (ACE2) is increased in non-obese diabetic (NOD) mice. Because paricalcitol has been reported to protect against diabetic nephropathy, we investigated the role of paricalcitol in modulating ACE2 in these mice. In addition, renal ADAM17, a metalloprotease implied in ACE2 shedding, was assessed. NOD female and non-diabetic control mice were studied for 21 days after diabetes onset and divided into various treatment groups. Diabetic animals received either vehicle; 0.4 or 0.8 µg/kg paricalcitol, aliskiren, or a combination of paricalcitol and aliskiren. We then studied the effect of paricalcitol on ACE2 expression in proximal tubular epithelial cells. Paricalcitol alone or in combination with aliskiren resulted in significantly reduced circulating ACE2 activity in NOD mice but there were no changes in urinary albumin excretion. Serum renin activity was significantly decreased in mice that received aliskiren but no effect was found when paricalcitol was used alone. Renal content of ADAM17 was significantly decreased in animals that received a high dose of paricalcitol. Renal and circulating oxidative stress (quantified by plasma H2O2 levels and immunolocalization of nitrotyrosine) were reduced in high-dose paricalcitol-treated mice compared with non-treated diabetic mice. In culture, paricalcitol incubation resulted in a significant increase in ACE2 expression compared with nontreated cells. In NOD mice with type 1 diabetes, paricalcitol modulates ACE2 activity, ADAM17, and oxidative stress renal content independently from the glycemic profile and urinary albumin excretion. In tubular cells, paricalcitol may modulate ACE2 by blocking its shedding. In the early stage of diabetic nephropathy, paricalcitol treatment counterbalances the effect of diabetes on circulating ACE2 activity. Our results suggest that additional use of paricalcitol may be beneficial in treating patients with diabetes under standard therapeutic strategies.


Subject(s)
ADAM Proteins/metabolism , Diabetic Nephropathies/prevention & control , Ergocalciferols/therapeutic use , Kidney/drug effects , Peptidyl-Dipeptidase A/blood , ADAM17 Protein , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure , Diabetes Mellitus, Experimental , Drug Evaluation, Preclinical , Ergocalciferols/pharmacology , Female , Kidney/metabolism , Mice, Inbred NOD , Oxidative Stress/drug effects , Proteinuria/prevention & control , Random Allocation , Renin/metabolism
11.
PLoS One ; 10(8): e0134967, 2015.
Article in English | MEDLINE | ID: mdl-26252478

ABSTRACT

Immunotherapy is emerging as a supplement to conventional cancer treatment, and identifying antigen targets for specific types of cancer is critical to optimizing therapeutic efficacy. Cancer/testis antigens are highly promising targets for immunotherapy due to their cancer-specific expression and antigenic properties, but the expression patterns of most of the more than 200 identified cancer/testis antigens in various cancers remain largely uncharacterized. In this study, we investigated the expression of the cancer/testis antigens ADAM2, CALR3 and SAGE1 in lung and breast cancer, the two most frequent human cancers, with the purpose of providing novel therapeutic targets for these diseases. We used a set of previously uncharacterized antibodies against the cancer/testis antigens ADAM2, CALR3 and SAGE1 to investigate their expression in a large panel of normal tissues as well as breast and lung cancers. Staining for the well-characterized MAGE-A proteins was included for comparison. Immunohistochemical staining confirmed previous mRNA analysis demonstrating that ADAM2, CALR3 and SAGE1 proteins are confined to testis in normal individuals. Negative tissues included plancenta, which express many other CT antigens, such as MAGE-A proteins. Surprisingly, we detected no ADAM2, CALR3 and SAGE1 in the 67 lung cancers (mainly non-small lung cancer) and 189 breast cancers, while MAGE-A proteins were present in 15% and 7-16% of these tumor types, respectively. Treatment with DNA methyltransferase inhibitors has been proposed as an attractive strategy to increase the expression of cancer/testis antigens in tumors before immunotargeting; however, neither ADAM2, CALR3 nor SAGE1 could be significantly induced in lung and breast cancer cell lines using this strategy. Our results suggest that ADAM2, CALR3 and SAGE1 cancer/testis antigens are not promising targets for immunotherapy of breast and lung cancer.


Subject(s)
ADAM Proteins/metabolism , Antigens, Neoplasm/metabolism , Breast Neoplasms/metabolism , Calreticulin/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Azacitidine/pharmacology , Azacitidine/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cohort Studies , Female , Fertilins , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma-Specific Antigens/metabolism
12.
Am J Physiol Heart Circ Physiol ; 309(5): H926-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26254330

ABSTRACT

We previously reported that type 2 angiotensin-converting enzyme (ACE2) compensatory activity is impaired by the disintegrin and metalloprotease 17 (ADAM17), and lack of ACE2 is associated with oxidative stress in neurogenic hypertension. To investigate the relationship between ADAM17 and oxidative stress, Neuro2A cells were treated with ANG II (100 nM) 24 h after vehicle or α-lipoic acid (LA, 500 µM). ADAM17 expression was increased by ANG II (120.5 ± 9.1 vs. 100.2 ± 0.8%, P < 0.05) and decreased after LA (69.0 ± 0.3 vs. 120.5 ± 9.1%, P < 0.05). In another set of experiments, LA reduced ADAM17 (92.9 ± 5.3 vs. 100.0 ± 11.2%, P < 0.05) following its overexpression. Moreover, ADAM17 activity was reduced by LA in ADAM17-overexpressing cells [109.5 ± 19.8 vs. 158.0 ± 20.0 fluorescence units (FU)·min(-1)·µg protein(-1), P < 0.05], in which ADAM17 overexpression increased oxidative stress (114.1 ± 2.5 vs. 101.0 ± 1.0%, P < 0.05). Conversely, LA-treated cells attenuated ADAM17 overexpression-induced oxidative stress (76.0 ± 9.1 vs. 114.1 ± 2.5%, P < 0.05). In deoxycorticosterone acetate (DOCA)-salt hypertensive mice, a model in which ADAM17 expression and activity are increased, hypertension was blunted by pretreatment with LA (119.0 ± 2.4 vs. 131.4 ± 2.2 mmHg, P < 0.05). In addition, LA improved dysautonomia and baroreflex sensitivity. Furthermore, LA blunted the increase in NADPH oxidase subunit expression, as well as the increase in ADAM17 and decrease in ACE2 activity in the hypothalamus of DOCA-salt hypertensive mice. Taken together, these data suggest that LA might preserve ACE2 compensatory activity by breaking the feedforward cycle between ADAM17 and oxidative stress, resulting in a reduction of neurogenic hypertension.


Subject(s)
ADAM Proteins/metabolism , Antioxidants/pharmacology , Hypertension/metabolism , Oxidative Stress , Thioctic Acid/pharmacology , ADAM Proteins/genetics , ADAM17 Protein , Angiotensin II/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Antioxidants/therapeutic use , Baroreflex , Cell Line, Tumor , Hypertension/drug therapy , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/metabolism , Peptidyl-Dipeptidase A/metabolism , Thioctic Acid/therapeutic use
13.
J Nutr ; 145(6): 1218-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25926412

ABSTRACT

BACKGROUND: Growing evidence suggests that disintegrin and metalloprotease (ADAM) 17 (ADAM17) and ADAM10 contribute to the pathogenesis of vascular diseases. ADAM17 promotes inflammatory processes by liberating tumor necrosis factor α, interleukin 6 receptor (IL-6R), and tumor necrosis factor receptor 1 (TNFR1). ADAM17 and ADAM10 modulate vascular permeability by cleaving endothelial adhesion molecules such as junctional adhesion molecule A (JAM-A) and vascular endothelial cadherin (VE-cadherin), respectively. OBJECTIVE: This study was designed to investigate whether a link might exist between the protective effects of fish oil (FO) supplementation against atherosclerosis and ADAM function. METHODS: Male LDL receptor knockout (LDLR(-/-)) mice and male wild-type (WT) mice were fed a Western diet (200 g/kg fat, 1.5 g/kg cholesterol) containing either 20% lard (LDLR(-/-)-lard and WT-lard groups) or 10% lard combined with 10% FO (LDLR(-/-)-FO and WT-FO groups) for 12 wk. Atherosclerotic lesion development and fatty acid composition of liver microsomes were evaluated. ADAM10 and ADAM17 expression was determined by quantitative real-time polymerase chain reaction and immunoblot analyses. Concentrations of soluble ADAM substrates in plasma and liver extracts were measured by ELISA. RESULTS: Diets supplemented with FO markedly reduced development of early atherosclerotic lesions in LDLR(-/-) mice (LDLR(-/-)-lard group vs. LDLR(-/-)-FO group mean ± SD: 29.6 ± 6.1% vs. 22.5 ± 4.2%, P < 0.05). This was not accompanied by changes in expression of ADAM17 or ADAM10 in the aorta or liver. No dietary effects on circulating TNFR1 (LDLR(-/-)-lard group vs. LDLR(-/-)-FO group mean ± SD: 1.22 ± 0.23 vs. 1.39 ± 0.28, P > 0.2) or IL-6R (1.06 ± 0.12 vs. 0.98 ± 0.09 fold of WT-lard group, P > 0.1), classical substrates of ADAM17 on macrophages, and neutrophil granulocytes were observed. However, a reduction in atherosclerotic lesions in the LDLR(-/-)-FO group was accompanied by a significant reduction in the circulating endothelial cell adhesion molecules JAM-A (LDLR(-/-)-lard group vs. LDLR(-/-)-FO group mean ± SD: 1.42 ± 0.20 vs. 0.95 ± 0.56 fold of WT-lard group, P < 0.05), intercellular adhesion molecule 1 (1.15 ± 0.14 vs. 0.88 ± 0.17 fold of WT-lard group, P < 0.05), and VE-cadherin (0.88 ± 0.12 vs. 0.72 ± 0.15 fold of WT-lard group, P < 0.05), reflecting reduced ADAM activity in endothelial cells. CONCLUSION: FO exerted an antiatherogenic effect on male LDLR(-/-) mice that was accompanied by a reduced release of ADAM17 and ADAM10 substrates from endothelial cells. It is suggested that FO-decreased ADAM activity contributes to improved endothelial barrier function and thus counteracts intimal lipoprotein insudation and macrophage accumulation.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Dietary Supplements , Fish Oils/pharmacology , Membrane Proteins/metabolism , ADAM Proteins/genetics , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Diet, Western/adverse effects , Dietary Fats/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Intercellular Adhesion Molecule-1/metabolism , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, LDL/genetics , Receptors, LDL/metabolism
14.
APMIS ; 123(8): 697-705, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26011228

ABSTRACT

This study aimed to observe the effects of Se deficiency on epiphyseal plates of two generation DA rats fed with artificial total synthetic low Se diet. All F0 and F1 DA rats were fed with synthetic low Se diet (SeD group) and low Se diet supplied with Se (SeS group). The levels of selenium and enzyme activities of GPx were detected in plasma of the rats. General growth of bone and articular cartilage was measured macroscopically and microscopically. The epiphyseal plate of femur heads or tibia were obtained to histological and immunohistochemical examinations. The cartilage from left knee joints and femur heads was used to detect the gene expression of collagens, ADAMTSs and several selenoproteins by RT-qPCR. Two generation SeD rats showed Se insufficiency status. The thicknesses of the femur and tibial epiphyseal plates in both F0 and F1 SeD rats were significantly less than that of SeS rats. In F1 generation, SeD rats showed much fewer proliferative chondrocyte layers than SeS ones. Importantly, two generation SeD rats both showed significantly more serious pathological changes of epiphyseal plates. In two generation rats, gene expressions of COL II, GPx1 and GPx4 were significantly down-regulated in SeD rats than SeS ones; meanwhile ADAMTS-4 showed an up-regulated expression in cartilage. Dietary Se deficiency can apparently cause epiphyseal plate lesion and decrease cartilage type II collagen production and GPx1 activity in two generation DA rats fed with the artificial total synthesis low Se diet.


Subject(s)
Growth Plate/abnormalities , Selenium/blood , Selenium/deficiency , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS4 Protein , Animals , Chondrocytes , Collagen Type II/genetics , Collagen Type II/metabolism , Diet , Dietary Supplements , Down-Regulation , Female , Femur/abnormalities , Femur/drug effects , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Growth Plate/drug effects , Male , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats , Up-Regulation , Glutathione Peroxidase GPX1
15.
BMC Evol Biol ; 15: 22, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25879701

ABSTRACT

BACKGROUND: The A Disintegrin-like and Metalloproteinase domain with Thrombospondin-1 motifs (ADAMTS) enzymes comprise 19 mammalian zinc-dependent metalloproteinases (metzincins) with homologues in a wide range of invertebrates. ADAMTS enzymes have a broad range of functions in development and diseases due to their extracellular matrix remodelling activity. Here, we report a detailed characterisation of their evolutionary conservation across vertebrates. RESULTS: Using bioinformatics complemented with de novo sequencing, gene sequences for ADAMTS enzymes were obtained from a variety of organisms. Detailed evolutionary analyses revealed a high level of conservation across vertebrates with evidence of ADAMTS gene expansion during two rounds of whole genome duplication (WGD) in vertebrates, while tandem duplication events and gene loss were also apparent. However, the additional round of teleost-specific WGD did not have a significant effect on ADAMTS gene family members suggesting their conserved roles have remained constant in teleost fish. Quantitative reverse-transcriptase polymerase chain reaction analysis revealed dynamic expression of adamts genes throughout zebrafish embryonic development reflecting the key conserved roles they play in vertebrate embryogenesis. Notably, several adamts mRNAs were maternally expressed with a dramatic increase in mRNA levels coinciding with zygotic expression and organogenesis. Broad adamts mRNA expression was also demonstrated in several adult organs indicating potential roles in adult homeostasis. CONCLUSIONS: Our data highlight the evolution of the ADAMTS gene family through duplication processes across metazoans supplemented by a burst of amplification through vertebrate WGD events. It also strongly posits the zebrafish as a potential model species to further elucidate the function of ADAMTS enzymes during vertebrate development.


Subject(s)
Evolution, Molecular , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish/genetics , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM Proteins/metabolism , Animals , Gene Duplication , Gene Expression Regulation, Developmental , Genome , Metalloendopeptidases/metabolism , Phylogeny , Protein Structure, Tertiary , Vertebrates/genetics , Zebrafish/embryology , Zebrafish Proteins/metabolism
16.
Fitoterapia ; 100: 179-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25510322

ABSTRACT

Aspalathin (Asp) and nothofagin (Not) are two major active dihydrochalcones found in green rooibos, which have been reported for their anti-oxidant activity. Increasing evidence has demonstrated that beyond its role in the activation of protein C, endothelial cell protein C receptor (EPCR) is also involved in vascular inflammation. EPCR activity is markedly changed by ectodomain cleavage and its release as the soluble EPCR. EPCR can be shed from the cell surface, which is mediated by tumor necrosis factor-α converting enzyme (TACE). However, little is known about the effects of Asp and Not on EPCR shedding. Our results demonstrated that Asp and Not induced potent inhibition of phorbol-12-myristate 13-acetate (PMA)-, tumor necrosis factor (TNF)-α-, interleukin (IL)-1ß, and cecal ligation and puncture (CLP)-induced EPCR shedding. Asp and Not also inhibited the expression and activity of PMA-induced TACE in endothelial cells. Asp and Not also suppressed CLP-induced protein C decrease in mice and thrombin generation in HUVECs. In addition, treatment with Asp and Not resulted in reduced PMA-stimulated phosphorylation of p38, extracellular regulated kinase (ERK) 1/2, and c-Jun N-terminal kinase (JNK). These results demonstrate the potential of Asp and Not as an anti-sEPCR shedding reagent against PMA and CLP-mediated EPCR shedding.


Subject(s)
Blood Coagulation Factors/metabolism , Chalcones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Receptors, Cell Surface/metabolism , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Aspalathus , Humans , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Phosphorylation , Tetradecanoylphorbol Acetate/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Chin J Integr Med ; 21(3): 196-203, 2015 Mar.
Article in English | MEDLINE | ID: mdl-23860800

ABSTRACT

OBJECTIVE: Although chondroprotective activities have been documented for polysaccharides, the potential target of different polysaccharide may differ. The study was aimed to explore the effect of glucan HBP-A in chondrocyte monolayer culture and chondrocytes-alginate hydrogel constructs in vivo, especially on the expression of type II collagen. METHODS: Chondrocytes isolated from rabbit articular cartilage were cultured and verified by immunocytochemical staining of type II collagen. Chondrocyte viability was assessed after being treated with HBP-A in different concentrations. Morphological status of chondrocytes-alginate hydrogel constructs in vitro was observed by scanning electron microscope (SEM). The constructs were treated with HBP-A and then injected to nude mice subcutaneously. Six weeks after transplantation, the specimens were observed through transmission electron microscopy (TEM). The mRNA expressions of disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTs-5), aggrecan and type II collagen in both monolayer culture and constructs were determined by real time polymerase chain reaction (PCR). The expression of type II collagen and matrix metalloproteinases-3 (MMP-3) in chondrocyte monolayer culture was also tested through Western blot and enzyme linked immunosorbent assay (ELISA), respectively. RESULTS: MMP-3 secretion and ADAMTs-5 mRNA expression in vitro were inhibited by HBP-A at 0.3 mg/mL concentration. In morphological study, there were significant appearance of collagen in those constructs treated by HBP-A. Accordingly, in both chondrocyte monolayer culture and chondrocytes-alginate hydrogel constructs, the expression of type II collagen was increased significantly in HBP-A group when compared with control group (P<0.001). CONCLUSIONS: The study documented that the potential pharmacological target of glucan HBP-A in chondrocytes monolayer culture and tissue engineered cartilage in vivo may be concerned with the inhibition of catabolic enzymes MMP-3, ADAMTs-5, and increasing of type II collagen expression.


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/metabolism , Collagen Type II/metabolism , Glucans/pharmacology , Tissue Engineering/methods , ADAM Proteins/genetics , ADAM Proteins/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Alginates/pharmacology , Animals , Cartilage, Articular/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Collagen Type II/genetics , Female , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Immunohistochemistry , Matrix Metalloproteinase 3/metabolism , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits
18.
Anal Biochem ; 452: 34-42, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24534253

ABSTRACT

ADAM12 belongs to the A disintegrin and metalloprotease (ADAM) family of secreted sheddases activating extracellular growth factors such as epidermal growth factor receptor (EGFR) ligands and tumor necrosis factor-alpha (TNF-α). ADAM proteases, most notably ADAM17 (TNF-α-converting enzyme), have long been investigated as pharmaceutical drug targets; however, due to lack of potency and in vivo side effects, none of the small-molecule inhibitors discovered so far has made it beyond clinical testing. Ongoing research on novel selective inhibitors of ADAMs requires reliable biochemical assays to validate molecular probes from large-scale screening efforts. Here we describe an electrophoretic mobility shift assay for ADAM12 based on the identification of an optimized peptide substrate that is characterized by excellent performance and reproducibility.


Subject(s)
ADAM Proteins/metabolism , Electrophoretic Mobility Shift Assay/methods , Membrane Proteins/metabolism , Peptides/metabolism , ADAM12 Protein , Amino Acid Sequence , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Hydroxamic Acids/pharmacology , Molecular Sequence Data , Peptides/chemistry , Protease Inhibitors/pharmacology , Reproducibility of Results
19.
Neurobiol Aging ; 35(5): 1045-54, 2014 May.
Article in English | MEDLINE | ID: mdl-24332448

ABSTRACT

Huperzine A (HupA), a natural inhibitor of acetylcholinesterase derived from a plant, is a licensed anti-Alzheimer's disease (AD) drug in China and a nutraceutical in the United States. In addition to acting as an acetylcholinesterase inhibitor, HupA possesses neuroprotective properties. However, the relevant mechanism is unknown. Here, we showed that the neuroprotective effect of HupA was derived from a novel action on brain iron regulation. HupA treatment reduced insoluble and soluble beta amyloid levels, ameliorated amyloid plaques formation, and hyperphosphorylated tau in the cortex and hippocampus of APPswe/PS1dE9 transgenic AD mice. Also, HupA decreased beta amyloid oligomers and amyloid precursor protein levels, and increased A Disintegrin And Metalloprotease Domain 10 (ADAM10) expression in these treated AD mice. However, these beneficial effects of HupA were largely abolished by feeding the animals with a high iron diet. In parallel, we found that HupA decreased iron content in the brain and demonstrated that HupA also has a role to reduce the expression of transferrin-receptor 1 as well as the transferrin-bound iron uptake in cultured neurons. The findings implied that reducing iron in the brain is a novel mechanism of HupA in the treatment of Alzheimer's disease.


Subject(s)
Alkaloids/pharmacology , Alkaloids/therapeutic use , Alzheimer Disease/drug therapy , Brain/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Iron/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phytotherapy , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , ADAM Proteins/metabolism , ADAM10 Protein , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Phosphorylation/drug effects , Plaque, Amyloid/drug therapy , Receptors, Transferrin/metabolism , tau Proteins/metabolism
20.
Ann Rheum Dis ; 73(9): 1719-27, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23852692

ABSTRACT

BACKGROUND: An exogenous supplement of n-3 polyunsaturated fatty acids (PUFAs) has been reported to prevent osteoarthritis (OA) through undefined mechanisms. OBJECTIVE: This study investigated the effect of alterations in the composition of endogenous PUFAs on OA, and associations of PUFAs with mammalian target of rapamycin complex 1 (mTORC1) signalling, a critical autophagy pathway in fat-1 transgenic (TG) mice. METHODS: fat-1 TG and wild-type mice were used to create an OA model by resecting the medial meniscus. The composition of the endogenous PUFAs in mouse tissues was analysed by gas chromatography, and the incidence of OA was evaluated by micro-computed tomography (micro-CT), scanning electron microscopy and histological methods. Additionally, primary chondrocytes were isolated and cultured. The effect of exogenous and endogenous PUFAs on mTORC1 activity and autophagy in chondrocytes was assessed. RESULTS: The composition of endogenous PUFAs of TG mice was optimised both by increased n-3 PUFAs and decreased n-6 PUFAs, which significantly alleviated the articular cartilage destruction and osteophytosis in the OA model (p<0.01), decreased protein expression of matrix metalloproteinase-13 (MMP-13) and ADAMTS-5 (a disintegrin and metalloproteinase with thrombospondin motifs) in the articular cartilage (p<0.01) and reduced chondrocyte number and loss of cartilage extracellular matrix. Both exogenous and endogenous n-3 PUFAs downregulated mTORC1 activity and promoted autophagy in articular chondrocytes. Conversely, mTORC1 pathway activation suppressed autophagy in articular chondrocytes. CONCLUSIONS: Enhancement of the synthesis of endogenous n-3 PUFAs from n-6 PUFAs can delay the incidence of OA, probably through inhibition of mTORC1, promotion of autophagy and cell survival in cartilage chondrocytes. Future investigation into the role of the endogenous n-6/n-3 PUFAs composition in OA prevention and treatment is warranted.


Subject(s)
Arthritis, Experimental/prevention & control , Fatty Acids, Omega-3/biosynthesis , Multiprotein Complexes/physiology , Osteoarthritis/prevention & control , TOR Serine-Threonine Kinases/physiology , ADAM Proteins/metabolism , ADAMTS5 Protein , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Autophagy/physiology , Cadherins/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/ultrastructure , Chondrocytes/pathology , Disease Progression , Fatty Acids, Omega-3/physiology , Fatty Acids, Omega-6/biosynthesis , Female , Matrix Metalloproteinase 13/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Osteoarthritis/etiology , Osteoarthritis/pathology , Proteoglycans/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL