Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Bioorg Med Chem ; 92: 117424, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37517101

ABSTRACT

Osteoarthritis is a chronic degenerative joint disease affecting millions of people worldwide, with no disease-modifying drugs currently available to treat the disease. Tissue inhibitor of metalloproteinases 3 (TIMP-3) is a potential therapeutic target in osteoarthritis because of its ability to inhibit the catabolic metalloproteinases that drive joint damage by degrading the cartilage extracellular matrix. We previously found that suramin inhibits cartilage degradation through its ability to block endocytosis and intracellular degradation of TIMP-3 by low-density lipoprotein receptor-related protein 1 (LRP1), and analysis of commercially available suramin analogues indicated the importance of the 1,3,5-trisulfonic acid substitutions on the terminal naphthalene rings for this activity. Here we describe synthesis and structure-activity relationship analysis of additional suramin analogues using ex vivo models of TIMP-3 trafficking and cartilage degradation. This showed that 1,3,6-trisulfonic acid substitution of the terminal naphthalene rings was also effective, and that the protective activity of suramin analogues depended on the presence of a rigid phenyl-containing central region, with para/para substitution of these phenyl rings being most favourable. Truncated analogues lost protective activity. The physicochemical characteristics of suramin and its analogues indicate that approaches such as intra-articular injection would be required to develop them for therapeutic use.


Subject(s)
Osteoarthritis , Tissue Inhibitor of Metalloproteinase-3 , Humans , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Tissue Inhibitor of Metalloproteinase-3/therapeutic use , Suramin/pharmacology , Suramin/metabolism , Suramin/therapeutic use , Cartilage/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Metalloproteases/metabolism , Metalloproteases/pharmacology , Metalloproteases/therapeutic use
2.
Spine (Phila Pa 1976) ; 48(7): 468-475, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36149858

ABSTRACT

STUDY DESIGN: In vitro study. OBJECTIVE: To investigate the effect of irisin on human nucleus pulposus cells (hNPCs) in vitro. SUMMARY OF BACKGROUND DATA: Physical exercise (PE) favours weight loss and ameliorates function in patients with low back pain. Although there is no biological evidence that the intervertebral disk (IVD) can respond to PE, recent studies have shown that running is associated with increased IVD hydration and hypertrophy. Irisin, a myokine released upon muscle contraction, has demonstrated anabolic effects on different cell types, including chondrocytes. MATERIALS AND METHODS: hNPCs were exposed to 5, 10, and 25 ng/mL irisin. Cell proliferation, glycosaminoglycan (GAG) content, metabolic activity, gene expression of collagen type II (COL2), matrix metalloproteinase (MMP)-13, tissue inhibitor of matrix metalloproteinase (TIMP)-1 and TIMP-3, aggrecan (ACAN), interleukin (IL)-1ß, a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5 were assessed. In addition, MTT assay and ADAMTS-5, COL2, TIMP-1, and IL-1ß gene expression were evaluated following incubation with irisin for 24 hours and subsequent culture with 10 ng/mL IL-1ß and vice versa (incubation for 24 hours with IL-1ß and subsequent culture with irisin). RESULTS: Irisin increased hNPC proliferation, metabolic activity, and GAG content, as well as COL2, ACAN, TIMP-1 and TIMP-3 gene expression, while decreasing MMP-13 and IL-1ß mRNA levels. Irisin pretreatment of hNPCs cultured in proinflammatory conditions resulted in a rescue of metabolic activity and a decrease of IL-1ß levels. Similarly, incubation of hNPCs with IL-1ß and subsequent exposure to irisin led to an increment of metabolic activity, COL2 gene expression, and a reduction of IL-1ß and ADAMTS-5 levels. CONCLUSIONS: Irisin increases hNPC proliferation, GAG content, metabolic activity, and promotes anabolic gene expression while reducing catabolic markers. Irisin may be one of the mediators by which PE and muscle tissues modulate IVD metabolism, suggesting the existence of a biological cross-talk between the muscle and IVD.


Subject(s)
Intervertebral Disc Degeneration , Intervertebral Disc , Nucleus Pulposus , Humans , Nucleus Pulposus/metabolism , Fibronectins/metabolism , Fibronectins/pharmacology , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Intervertebral Disc Degeneration/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Intervertebral Disc/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Muscles/metabolism , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/pharmacology , Cells, Cultured , Interleukin-1beta/metabolism
3.
Clin Immunol ; 238: 109023, 2022 05.
Article in English | MEDLINE | ID: mdl-35477026

ABSTRACT

BACKGROUND: Diabetic retinopathy (DR) is a frequent complication of diabetes. Recent reports have showed that circular RNAs (circRNAs) play important roles in DR progression. Herein, the aim of this study was to explore the role and molecular mechanism of circ_NNT in DR process. METHODS: Human retinal pigment epithelial cells ARPE-19 were treated with high glucose (HG) in experimental group. The expression of circ_NNT, miR-320b, and TIMP3 (TIMP Metallopeptidase Inhibitor 3) were determined using quantitative real-time polymerase chain reaction and Western blot. In vitro experiments were conducted by 5-ethynyl-2'-deoxyuridine (EdU) assay, MTT assay, flow cytometry, Western blot, and ELISA. The binding interaction was confirmed using dual-luciferase reporter and pull-down assays. RESULTS: HG stimulation led to a decrease of circ_NNT and TIMP3 expression, and an increase of miR-320b expression in ARPE-19 cells. Functionally, circ_NNT up-regulation reversed HG-evoked apoptosis and inflammation in ARPE-19 cells. Mechanistically, circ_NNT acted as a sponge for miR-320b to elevate TIMP3 expression. Further rescue experiments showed that miR-320b elevation attenuated the protective effects of circ_NNT on HG-induced ARPE-19 cells. Moreover, inhibition of miR-320b protected ARPE-19 cells against HG-evoked apoptosis and inflammation, which were abolished by TIMP3 knockdown. CONCLUSION: Circ_NNT protected ARPE-19 cells against HG-evoked apoptosis and inflammation via elevating TIMP3 through sequestering miR-320b, indicating that up-regulation of circ_NNT might contribute to the inhibition of DR process.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , MicroRNAs , Apoptosis/genetics , Diabetic Retinopathy/genetics , Glucose/pharmacology , Humans , Inflammation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Retinal Pigment Epithelium/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/pharmacology
4.
Oxid Med Cell Longev ; 2022: 9366494, 2022.
Article in English | MEDLINE | ID: mdl-35450405

ABSTRACT

Trehalose, a natural disaccharide, is synthesized by many organisms when cells are exposed to stressful stimuli. On the basis of its ability to modulate autophagy, trehalose has been considered an innovative drug for ameliorating many diseases, but its molecular mechanism is not well described. Previous findings demonstrated that trehalose plays a photoprotective role against ultraviolet (UV) B-induced damage through autophagy induction in keratinocytes. In this study, coimmunoprecipitation, label-free quantitative proteomic and parallel reaction monitoring, and western blot analysis demonstrated that trehalose promotes the interaction between tissue inhibitor of metalloproteinase (TIMP) 3 and Beclin1. Western blot and immunofluorescence staining analysis suggested that trehalose increases ATG9A localization in lysosomes and decreases its localization in the endoplasmic reticulum. Furthermore, in the presence or absence of UVB radiation, we evaluated the influence of TIMP3 and ATG9A small interfering RNA (siRNA) on the effect of trehalose on autophagy, cell death, migration, or interleukin-8 expression in keratinocytes, including HaCaT, A431, and human epidermal keratinocytes. The results revealed that in HaCaT cells, TIMP3 and ATG9A siRNA resulted in attenuation of trehalose-induced autophagy and inhibited cell death. In A431 cells, TIMP3 and ATG9A siRNA led to attenuation of trehalose-induced autophagy and cell death and inhibited migration. In human epidermal keratinocytes, trehalose-induced autophagy and inhibition of the interleukin-8 expression were blocked by ATG9A but not TIMP3 siRNA. In addition, the results of quantitative real-time PCR and immunohistochemistry analysis demonstrated the abnormal expression of TIMP3 and ATG9A in actinic keratosis and cutaneous squamous cell carcinoma skin tissues. These findings suggest the protective effects of trehalose in normal keratinocytes and its inhibitory effects on cancerous keratinocytes, possibly mediated by activation of autophagy and regulation of TIMP3 and ATG9A, providing the mechanistic basis for the potential use of trehalose in the prevention or treatment of UVB-induced skin diseases.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Autophagy , Autophagy-Related Proteins/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Interleukin-8/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Proteomics , RNA, Small Interfering/metabolism , Skin Neoplasms/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Trehalose/pharmacology , Ultraviolet Rays/adverse effects , Vesicular Transport Proteins/metabolism
5.
J Neurotrauma ; 36(9): 1416-1427, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30251917

ABSTRACT

Following spinal cord injury (SCI), inflammation amplifies damage beyond the initial insult, providing an opportunity for targeted treatments. An ideal protective therapy would reduce both edema within the lesion area and the activation/infiltration of detrimental immune cells. Previous investigations demonstrated the efficacy of intravenous injection of multipotent adult progenitor cells (MAPC®) to modulate immune response following SCI, leading to significant improvements in tissue sparing, locomotor and urological functions. Separate studies have demonstrated that tissue inhibitor of matrix metalloproteinase-3 (TIMP3) reduces blood-brain barrier permeability following traumatic brain injury in a mouse model, leading to improved functional recovery. This study examined whether TIMP3, delivered alone or in concert with MAPC cells, improves functional recovery from a contusion SCI in a rat model. The results suggest that intravenous delivery of MAPC cell therapy 1 day following acute SCI significantly improves tissue sparing and impacts functional recovery. TIMP3 treatment provided no significant benefit, and further, when co-administered with MAPC cells, it abrogated the therapeutic effects of MAPC cell therapy. Importantly, this study demonstrated for the first time that acute treatment of SCI with MAPC cells can significantly reduce the incidence of urinary tract infection (UTI) and the use of antibiotics for UTI treatment.


Subject(s)
Multipotent Stem Cells/transplantation , Recovery of Function , Spinal Cord Injuries , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Urinary Tract Infections , Adult Stem Cells/transplantation , Animals , Female , Humans , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Stem Cell Transplantation/methods , Urinary Tract Infections/epidemiology , Urinary Tract Infections/etiology
6.
Pharmacol Res Perspect ; 6(6): e00442, 2018 12.
Article in English | MEDLINE | ID: mdl-30459952

ABSTRACT

Tissue Inhibitor of Metalloproteinase 3 (TIMP3) is a secreted protein that has a great utility to inhibit elevated metalloproteinase (MMP) activity in injured tissues including infarcted cardiac tissue, inflamed vessels, and joint cartilages. An imbalance between TIMP3 and active MMP levels in the local tissue area may cause worsening of disease progression. To counter balance elevated MMP levels, exogenous administration of TIMP3 appeared to be beneficial in preclinical studies. However, the current form of WT-TIMP3 molecule has a limitation to be a therapeutic candidate due to low production yield, short plasma half-life, injection site retention, and difficulty in delivery, etc. We have engineered TIMP3 molecules by adding extra glycosylation sites or fusing with albumin, Fc, and antibody to improve pharmacokinetic properties. In general, the C-terminal fusion of TIMP3 improved expression and production in mammalian cells and extended half-lives dramatically 5-20 folds. Of note, a site-specific glycosylation at K22S/F34N resulted in a higher level of expression and better cardiac function compared to other fusion proteins in the context of left ventricle ejection fraction (LVEF) changes in a rat myocardial infarction model. It appeared that cardiac efficacy depends on a high ECM binding affinity, in which K22S/F34N and N-TIMP3 showed a higher binding to the ECM compared to other engineered molecules. In conclusion, we found that the ECM binding and sustained residence of injected TIMP3 molecules are important for cardiac tissue localization and inhibition of adverse remodeling activity.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , Matrix Metalloproteinases/metabolism , Myocardial Infarction/drug therapy , Recombinant Fusion Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Ventricular Function, Left/drug effects , ADAM17 Protein/metabolism , Animals , Cell Line , Disease Models, Animal , Disease Progression , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts , Glycosylation , Humans , Infusions, Intravenous , Injections, Intralesional , Male , Mutation , Myocardial Infarction/etiology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Tissue Inhibitor of Metalloproteinase-3/chemistry , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/therapeutic use , Treatment Outcome
7.
Mol Med Rep ; 18(2): 2061-2067, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29956789

ABSTRACT

The present study investigated the role of tissue inhibitor of matrix metalloproteinase­3 (TIMP­3) in regulating the proliferation, migration, apoptosis and activity of matrix metalloproteinase (MMP)­2 and ­9, during the development of an atherosclerotic abdominal artery aneurysm (AAA). Experiments were conducted using rabbit AAA neck (NA) smooth muscle cells (SMCs), to investigate the potential for TIMP­3 to be used as a novel stent coating in preventing aortic dilation adjacent to the AAA. The atherosclerotic AAA model was induced in New Zealand white rabbits via a 6­week high­cholesterol diet, followed by incubation of the targeted aortic region with elastase. SMCs were isolated from the aorta adjacent to the aneurysm 30 days after AAA model induction, and stimulated with 3, 10, 30 or 100 ng/ml TIMP­3. Cell proliferation was investigated using Cell Counting Kit­8 reagent, migration was examined using a Boyden chamber assay and apoptotic rate was analyzed using the Annexin V­fluorescein isothiocyanate Apoptosis Detection kit. Gelatin zymography and ELISA were used to measure the activity of MMP­2 and MMP­9, and the expression of tumor necrosis factor­α (TNF­α), respectively. Analysis of cell proliferation indicated that 10, 30 and 100 ng/ml TIMP­3 reduced cell viability. Cell migration was decreased by 10, 30 and 100 ng/ml TIMP­3. MMP­2 activity was inhibited by 10, 30 and 100 ng/ml TIMP­3, and MMP­9 activity was suppressed by 30 and 100 ng/ml TIMP­3. The protein levels of secreted TNF­α were reduced by 10, 30 and 100 ng/ml TIMP­3. The present study demonstrated the ability of 30 and 100 ng/ml TIMP­3 to attenuate migration and proliferation, and to inhibit the activity of MMP­2, MMP­9 and TNF­α secretion of NA SMCs. In conclusion, TIMP­3 may be considered a potential therapeutic drug for use in a novel drug­eluting stent, to attenuate the progressive dilation of the aortic NA.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Aorta/pathology , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Rabbits
8.
ASN Neuro ; 9(6): 1759091417745425, 2017.
Article in English | MEDLINE | ID: mdl-29198135

ABSTRACT

Tissue inhibitor of metalloproteinase-3 (TIMP-3) inhibits the activities of various metalloproteinases including matrix metalloproteinases and ADAM family proteins. In the peripheral nervous system, ADAM17, also known as TNF-α converting enzyme (TACE), cleaves the extracellular domain of Nrg1 type III, an axonal growth factor that is essential for Schwann cell myelination. The processing by ADAM17 attenuates Nrg1 signaling and inhibits Schwann cell myelination. TIMP-3 targets ADAM17, suggesting a possibility that TIMP-3 may elicit a promyelinating function in Schwann cells by relieving ADAM17-induced myelination block. To investigate this, we used a myelinating coculture system to determine the effect of TIMP-3 on Schwann cell myelination. Treatment with TIMP-3 enhanced myelin formation in cocultures, evident by an increase in the number of myelin segments and upregulated expression of Krox20 and myelin protein. The effect of TIMP-3 was accompanied by the inhibition of ADAM17 activity and an increase in Nrg1 type III signaling in cocultures. Accordingly, the N-terminus fragment of TIMP-3, which exhibits a selective inhibitory function toward ADAM17, elicited a similar myelination-promoting effect and increased Nrg1 type III activity. TIMP-3 also enhanced laminin production in cocultures, which is likely to aid Schwann cell myelination.


Subject(s)
Myelin Sheath/metabolism , Schwann Cells/drug effects , Sciatic Nerve/cytology , Tissue Inhibitor of Metalloproteinase-3/pharmacology , ADAM17 Protein/metabolism , Age Factors , Animals , Animals, Newborn , Antioxidants , Ascorbic Acid/pharmacology , Bromodeoxyuridine/metabolism , Cells, Cultured , Coculture Techniques , Fluorescence Resonance Energy Transfer , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Myelin Basic Protein/metabolism , Neurons/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Sciatic Nerve/growth & development , Sciatic Nerve/metabolism
9.
Sci Rep ; 7(1): 7961, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28801670

ABSTRACT

IgLONs are members of the immunoglobulin superfamily of cell adhesion proteins implicated in the process of neuronal outgrowth, cell adhesion and subdomain target recognition. IgLONs form homophilic and heterophilic complexes on the cell surface that repress or promote growth depending on the neuronal population, the developmental stage and surface repertoire of IgLON family members. In the present study, we identified a metalloproteinase-dependent mechanism necessary to promote growth in embryonic dorsal root ganglion cells (DRGs). Treatment of embryonic DRG neurons with pan-metalloproteinase inhibitors, tissue inhibitor of metalloproteinase-3, or an inhibitor of ADAM Metallopeptidase Domain 10 (ADAM10) reduces outgrowth from DRG neurons indicating that metalloproteinase activity is important for outgrowth. The IgLON family members Neurotrimin (NTM) and Limbic System-Associated Membrane Protein (LSAMP) were identified as ADAM10 substrates that are shed from the cell surface of DRG neurons. Overexpression of LSAMP and NTM suppresses outgrowth from DRG neurons. Furthermore, LSAMP loss of function decreases the outgrowth sensitivity to an ADAM10 inhibitor. Together our findings support a role for ADAM-dependent shedding of cell surface LSAMP in promoting outgrowth from DRG neurons.


Subject(s)
ADAM10 Protein/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Ganglia, Spinal/growth & development , Neural Cell Adhesion Molecules/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Ganglia, Spinal/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Neural Cell Adhesion Molecules/genetics , Neuronal Outgrowth , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-3/pharmacology
10.
Am J Physiol Heart Circ Physiol ; 313(4): H690-H699, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28754718

ABSTRACT

Ischemia-reperfusion (IR) and myocardial infarction (MI) cause adverse left ventricular (LV) remodeling and heart failure and are facilitated by an imbalance in matrix metalloproteinase (MMP) activation and the endogenous tissue inhibitors of metalloproteinase (TIMPs). We have identified that myocardial injections of recombinant TIMP-3 (rTIMP-3; human full length) can interrupt post-MI remodeling. However, whether and to what degree intracoronary delivery of rTIMP-3 post-IR is feasible and effective remained to be established. Pigs (25 kg) underwent coronary catheterization and balloon occlusion of the left anterior descending coronary artery (LAD) for 90 min whereby at the final 4 min, rTIMP-3 (30 mg, n = 9) or saline was infused in the distal LAD. LV echocardiography was performed at 3-28 days post-IR, and LV ejection fraction (EF) and LV end-diastolic volume were measured. LV EF fell and LV end-diastolic volume increased from baseline (pre-IR) values (66 ± 1% and 40 ± 1 ml, respectively, means ± standard deviation) in both groups; however, the extent of LV dilation was reduced in the rTIMP-3 group by 40% at 28 days post-IR (P < 0.05) and the fall in LV EF was attenuated. Despite equivalent plasma troponin levels (14 ± 3 ng/ml), computed MI size at 28 days was reduced by over 45% in the rTIMP-3 group (P < 0.05), indicating that rTIMP-3 treatment abrogated MI expansion post-IR. Plasma NH2-terminal pro-brain natriuretic peptide levels, an index of heart failure progression, were reduced by 25% in the rTIMP-3 group compared with MI saline values (P < 0.05). Although the imbalance between MMPs and TIMPs has been recognized as a contributory factor for post-MI remodeling, therapeutic strategies targeting this imbalance have not been forthcoming. This study is the first to demonstrate that a relevant delivery approach (intracoronary) using rTIMP can alter the course of post-MI remodeling.NEW & NOTEWORTHY Myocardial ischemia and reperfusion injury remain significant causes of morbidity and mortality whereby alterations in the balance between matrix metalloproteinase and tissue inhibitor of metalloproteinase have been identified as contributory biological mechanisms. This novel translational study advances the concept of targeted delivery of recombinant proteins to modify adverse myocardial remodeling in ischemia-reperfusion injury.


Subject(s)
Myocardial Infarction , Reperfusion Injury , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Coronary Vessels , Echocardiography , Infusions, Intra-Arterial , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/drug effects , Peptide Fragments/blood , Peptide Fragments/drug effects , Recombinant Proteins/pharmacology , Stroke Volume/drug effects , Swine , Troponin/blood , Troponin/drug effects
11.
Stem Cells ; 33(12): 3530-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26299440

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to have potent therapeutic effects in a number of disorders including traumatic brain injury (TBI). However, the molecular mechanism(s) underlying these protective effects are largely unknown. Herein we demonstrate that tissue inhibitor of matrix metalloproteinase-3 (TIMP3), a soluble protein released by MSCs, is neuroprotective and enhances neuronal survival and neurite outgrowth in vitro. In vivo in a murine model of TBI, intravenous recombinant TIMP3 enhances dendritic outgrowth and abrogates loss of hippocampal neural stem cells and mature neurons. Mechanistically we demonstrate in vitro and in vivo that TIMP3-mediated neuroprotection is critically dependent on activation of the Akt-mTORC1 pathway. In support of the neuroprotective effect of TIMP3, we find that intravenous delivery of recombinant TIMP3 attenuates deficits in hippocampal-dependent neurocognition. Taken together, our data strongly suggest that TIMP3 has direct neuroprotective effects that can mitigate the deleterious effects associated with TBI, an area with few if any therapeutic options.


Subject(s)
Brain Injuries/drug therapy , Cognition Disorders/drug therapy , Hippocampus/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/physiopathology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Hippocampus/pathology , Mice , Neural Stem Cells/pathology , Neurons/pathology
12.
Nat Mater ; 13(6): 653-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24681647

ABSTRACT

Inhibitors of matrix metalloproteinases (MMPs) have been extensively explored to treat pathologies where excessive MMP activity contributes to adverse tissue remodelling. Although MMP inhibition remains a relevant therapeutic target, MMP inhibitors have not translated to clinical application owing to the dose-limiting side effects following systemic administration of the drugs. Here, we describe the synthesis of a polysaccharide-based hydrogel that can be locally injected into tissues and releases a recombinant tissue inhibitor of MMPs (rTIMP-3) in response to MMP activity. Specifically, rTIMP-3 is sequestered in the hydrogels through electrostatic interactions and is released as crosslinks are degraded by active MMPs. Targeted delivery of the hydrogel/rTIMP-3 construct to regions of MMP overexpression following a myocardial infarction significantly reduced MMP activity and attenuated adverse left ventricular remodelling in a porcine model of myocardial infarction. Our findings demonstrate that local, on-demand MMP inhibition is achievable through the use of an injectable and bioresponsive hydrogel.


Subject(s)
Hydrogels/pharmacology , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Infarction/drug therapy , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Ventricular Remodeling/drug effects , Animals , Disease Models, Animal , Humans , Hydrogels/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Matrix Metalloproteinases/metabolism , Myocardial Infarction/enzymology , Myocardial Infarction/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Swine , Tissue Inhibitor of Metalloproteinase-3/chemistry
13.
Angiogenesis ; 17(1): 207-19, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24129822

ABSTRACT

The binding of vascular endothelial growth factor (VEGF) to VEGF receptor-2 (VEGFR-2) on the surface of vascular endothelial cells stimulates many steps in the angiogenic pathway. Inhibition of this interaction is proving of value in moderating the neovascularization accompanying age-related macular degeneration and in the treatment of cancer. Tissue inhibitor of metalloproteinases-3 (TIMP-3) has been shown to be a natural VEGFR-2 specific antagonist-an activity that is independent of its ability to inhibit metalloproteinases. In this investigation we localize this activity to the C-terminal domain of the TIMP-3 molecule and characterize a short peptide, corresponding to part of this domain, that not only inhibits all three VEGF-family receptors, but also fibroblast growth factor and platelet-derived growth factor receptors. This multiple-receptor inhibition may explain why the peptide was also seen to be a powerful inhibitor of tumour growth and also a partial inhibitor of arthritic joint inflammation in vivo.


Subject(s)
Arthritis/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Peptides/pharmacology , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Arthritis/metabolism , Arthritis/pathology , Female , Human Umbilical Vein Endothelial Cells , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Peptides/chemistry , Tissue Inhibitor of Metalloproteinase-3/chemistry , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Biol Reprod ; 89(5): 121, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24048576

ABSTRACT

Ovulation involves reorganization of the extracellular matrix of the follicle. This study examines the expression, localization, and potential function of the tissue inhibitor of metalloproteinase 3 (TIMP3) during ovulation in women. The dominant follicle of the menstrual cycle was collected at specified times throughout the ovulatory process: pre-, early, late, and postovulatory. For quantitative studies, the follicle was bisected; granulosa and theca cells were separated and collected. For immunohistochemistry (IHC), the intact follicle was embedded and TIMP3 was localized. Additionally, granulosa cells were collected from women undergoing in vitro fertilization and treated with increasing concentrations of recombinant TIMP3, and cell viability was assessed. Real-time PCR for TIMP3 mRNA revealed an increase in TIMP3 mRNA expression in granulosa cells from the early to the late ovulatory stage. Thecal TIMP3 mRNA expression was constitutive across the periovulatory period. TIMP3 protein was localized by IHC to the granulosa and theca cell layers in pre-, early, and late ovulatory follicles as well as to the vascular bed. The staining was most intense in the granulosa and theca cells in the late ovulatory group. Treatment of human granulosa-lutein cells with exogenous recombinant TIMP3 for 24 h decreased cell viability by 60%. Using human follicles collected throughout the periovulatory period of the menstrual cycle, we have demonstrated that TIMP3 mRNA expression increases and that TIMP3 protein is in the appropriate cellular layers to regulate proteolytic remodeling as the follicle progresses toward ovulation. In addition, we have shown that elevated levels of TIMP3 lead to decreased cell viability.


Subject(s)
Ovary/enzymology , Ovulation/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism , Adult , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Female , Gene Expression , Humans , Luteal Cells/drug effects , Luteal Cells/physiology , Menstrual Cycle/drug effects , Menstrual Cycle/genetics , Menstrual Cycle/metabolism , Oocyte Retrieval , Ovulation/genetics , Tissue Distribution , Tissue Inhibitor of Metalloproteinase-3/pharmacology
15.
J Cardiothorac Surg ; 8: 183, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23937994

ABSTRACT

BACKGROUND: Cardiovascular disorders, including coronary artery bypass graft failure and in-stent restenosis remain significant opportunities for the advancement of novel therapeutics that target neointimal hyperplasia, a characteristic of both pathologies. Gene therapy may provide a successful approach to improve the clinical outcome of these conditions, but would benefit from the development of more efficient vectors for vascular gene delivery. The aim of this study was to assess whether a novel genetically engineered Adenovirus could be utilised to produce enhanced levels of vascular gene expression. METHODS: Vascular transduction capacity was assessed in primary human saphenous vein smooth muscle and endothelial cells using vectors expressing the LacZ reporter gene. The therapeutic capacity of the vectors was compared by measuring smooth muscle cell metabolic activity and migration following infection with vectors that over-express the candidate therapeutic gene tissue inhibitor of matrix metalloproteinase-3 (TIMP-3). RESULTS: Compared to Adenovirus serotype 5 (Ad5), the novel vector Ad5T*F35++ demonstrated improved binding and transduction of human vascular cells. Ad5T*F35++ mediated expression of TIMP-3 reduced smooth muscle cell metabolic activity and migration in vitro. We also demonstrated that in human serum samples pre-existing neutralising antibodies to Ad5T*F35++ were less prevalent than Ad5 neutralising antibodies. CONCLUSIONS: We have developed a novel vector with improved vascular transduction and improved resistance to human serum neutralisation. This may provide a novel vector platform for human vascular gene transfer.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors , Capsid , Cells, Cultured , Humans , Saphenous Vein , Tissue Inhibitor of Metalloproteinase-3/analysis , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Transduction, Genetic , beta-Galactosidase/analysis
16.
Invest Ophthalmol Vis Sci ; 54(1): 864-70, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23299479

ABSTRACT

PURPOSE: Pathological neovascularization is a crucial component of proliferative retinopathies. Previous studies showed that inactivation of A disintegrin and metalloproteinase 17 (ADAM17), a membrane-anchored metalloproteinase that regulates epidermal growth factor receptor (EGFR) signaling, reduces pathological retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). Here, we tested how genetic inactivation of a physiological ADAM17 inhibitor, the tissue inhibitor of matrix metalloproteinases-3 (TIMP3), or intravitreal injection of TIMP3 or of the EGFR inhibitor erlotinib influenced the outcome of OIR. METHODS: Wild-type mice were subjected to OIR in a chamber with 75% oxygen for 5 days beginning at postnatal day 7 (P7). Upon removal from the oxygen chamber at P12, they received a single intravitreal injection of TIMP3, erlotinib, or control. The central avascular area and neovascular tufts were measured after 5 days in room air (21% oxygen) at P17. Moreover, OIR experiments were performed with Timp3-/- mice and littermate controls. RESULTS: Timp3-/- mice showed greater revascularization of the central avascular area and developed equal or fewer neovascular tufts compared to littermate controls, depending on the genetic background. Wild-type mice injected with TIMP3 or erlotinib developed fewer neovascular tufts when compared to untreated littermates. Moreover, vessel regrowth into the avascular area was reduced in TIMP3-injected mice, but not in erlotinib-injected mice. CONCLUSIONS: These studies demonstrate that TIMP3 and erlotinib inhibit pathological neovascularization in the mouse retina, most likely due to inactivation of ADAM17 and the EGFR, respectively. Thus, TIMP3 and erlotinib emerge as attractive candidate antiangiogenic compounds for prevention and treatment of proliferative retinopathies.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Quinazolines/pharmacology , Retinal Diseases/drug therapy , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Disease Models, Animal , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Intravitreal Injections , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Oxygen/toxicity , Protein Kinase Inhibitors/pharmacology , Retinal Diseases/etiology , Retinal Diseases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-3/genetics
17.
PLoS One ; 7(12): e50709, 2012.
Article in English | MEDLINE | ID: mdl-23236386

ABSTRACT

BACKGROUND: Increasing number of evidence shows that soluble factors and extracellular matrix (ECM) components provide an optimal microenvironment controlling human bone marrow mesenchymal stem cell (MSC) functions. Successful in vivo administration of stem cells lies in their ability to migrate through ECM barriers and to differentiate along tissue-specific lineages, including endothelium. Lumican, a protein of the small leucine-rich proteoglycan (SLRP) family, was shown to impede cell migration and angiogenesis. The aim of the present study was to analyze the role of lumican in the control of MSC migration and transition to functional endothelial progenitor cell (EPC). METHODOLOGY/PRINCIPAL FINDINGS: Lumican inhibited tube-like structures formation on Matrigel® by MSC, but not EPC. Since matrix metalloproteinases (MMPs), in particular MMP-14, play an important role in remodelling of ECM and enhancing cell migration, their expression and activity were investigated in the cells grown on different ECM substrata. Lumican down-regulated the MMP-14 expression and activity in MSC, but not in EPC. Lumican inhibited MSC, but not EPC migration and invasion. The inhibition of MSC migration and invasion by lumican was reversed by MMP-14 overexpression. CONCLUSION/SIGNIFICANCE: Altogether, our results suggest that lumican inhibits MSC tube-like structure formation and migration via mechanisms that involve a decrease of MMP-14 expression and activity.


Subject(s)
Cell Movement/drug effects , Chondroitin Sulfate Proteoglycans/pharmacology , Endothelial Cells/drug effects , Keratan Sulfate/pharmacology , Matrix Metalloproteinase 14/metabolism , Mesenchymal Stem Cells/drug effects , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endothelial Cells/metabolism , Humans , Lumican , Mesenchymal Stem Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-3/pharmacology
18.
Gastroenterology ; 143(5): 1277-1287.e4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22819866

ABSTRACT

BACKGROUND & AIMS: Tissue inhibitor of metalloproteinases (TIMP)-3 is an inhibitor of matrix metalloproteinases, which regulates tissue inflammation, damage, and repair. We investigated the role of TIMP-3 in intestinal inflammation in human beings and mice. METHODS: We used real-time polymerase chain reaction and flow cytometry to measure levels of TIMP-3 in intestine samples from patients with Crohn's disease (CD) and those without (controls). We also analyzed TIMP-3 levels in lamina propria mononuclear cells (LPMCs) collected from biopsy samples of individuals with or without CD (controls) and then stimulated with transforming growth factor (TGF)-ß1, as well as in biopsy samples collected from patients with CD and then incubated with a Smad7 anti-sense oligonucleotide (knock down). LPMCs and biopsy samples from patients with CD were cultured with exogenous TIMP-3 and levels of inflammatory cytokines were measured. We evaluated the susceptibility of wild-type, TIMP-3-knockout (TIMP-3-KO), and transgenic (TIMP-3-Tg) mice to induction of colitis with 2, 4, 6-trinitrobenzene-sulfonic-acid (TNBS), and the course of colitis in recombinase-activating gene-1-null mice after transfer of wild-type or TIMP-3-KO T cells. RESULTS: Levels of TIMP-3 were reduced in intestine samples from patients with CD compared with controls. Incubation of control LPMCs with TGF-ß1 up-regulated TIMP-3; knockdown of Smad7, an inhibitor of TGF-ß1, in biopsy samples from patients with CD increased levels of TIMP-3. Exogenous TIMP-3 reduced levels of inflammatory cytokines in CD LPMCs and biopsy samples. TIMP-3-KO mice developed severe colitis after administration of TNBS, whereas TIMP-3-Tg mice were resistant to TNBS-induced colitis. Reconstitution of recombinase-activating gene-1-null mice with T cells from TIMP-3-KO mice increased the severity of colitis, compared with reconstitution with wild-type T cells. CONCLUSIONS: TIMP-3 is down-regulated in inflamed intestine of patients with CD. Its expression is regulated by TGF-ß1, and knock-down of Smad7 in intestinal tissues from patient with CD up-regulates TIMP-3. Loss or reduction of TIMP-3 in mice promotes development of colitis.


Subject(s)
Colitis, Ulcerative/metabolism , Crohn Disease/metabolism , Intestinal Mucosa/metabolism , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Adult , Aged , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Cytokines/metabolism , Down-Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Intestinal Mucosa/drug effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Oligonucleotides, Antisense/pharmacology , Smad7 Protein/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/pharmacology , Transforming Growth Factor beta/pharmacology , Trinitrobenzenesulfonic Acid
19.
Matrix Biol ; 31(4): 229-33, 2012 May.
Article in English | MEDLINE | ID: mdl-22406378

ABSTRACT

Membrane microvesicle shedding is an active process and occurs in viable cells with no signs of apoptosis or necrosis. We report here that microvesicles shed by oligodendroglioma cells contain an 'aggrecanase' activity, cleaving aggrecan at sites previously identified as targets for adamalysin metalloproteinases with disintegrin and thrombospondin domains (ADAMTSs). Degradation was inhibited by EDTA, the metalloproteinase inhibitor GM6001 and by tissue inhibitor of metalloproteinases (TIMP)-3, but not by TIMP-1 or TIMP-2. This inhibitor profile indicates that the shed microvesicles contain aggrecanolytic ADAMTS(s) or related TIMP-3-sensitive metalloproteinase(s). The oligodendroglioma cells were shown to express the three most active aggrecanases, namely Adamts1, Adamts4 and Adamts5, suggesting that one or more of these enzymes may be responsible for the microvesicle activity. Microvesicles shed by rheumatoid synovial fibroblasts similarly degraded aggrecan in a TIMP-3-sensitive manner. Our findings raise the novel possibility that microvesicles may assist oligodendroglioma and rheumatoid synovial fibroblasts to invade through aggrecan-rich extracellular matrices.


Subject(s)
Cell Physiological Phenomena , Cytoplasmic Vesicles/enzymology , Endopeptidases/metabolism , Fibroblasts/enzymology , Oligodendroglioma/enzymology , ADAM Proteins/metabolism , ADAMTS5 Protein , Aggrecans/metabolism , Cytoplasmic Vesicles/physiology , Dipeptides/pharmacology , Enzyme Activation , Fibroblasts/drug effects , Humans , Proteolysis , Recombinant Proteins/metabolism , Rheumatic Fever/pathology , Tissue Inhibitor of Metalloproteinase-3/pharmacology
20.
Beijing Da Xue Xue Bao Yi Xue Ban ; 41(5): 611-2, 2009 Oct 18.
Article in Chinese | MEDLINE | ID: mdl-19829688

ABSTRACT

Osteoarthritis is mainly caused by the degenerative changes of cartilage and cartilage extracellular matrix, while Aggrecanases degradate Proteoglycans which are the major components of cartilage. This review includes three aspects: (1) We have concluded the major enzymes(ADAMTS-4 and ADAMTS-5) which regulate the metabolism of cartilage extracellular matrix. Meanwhile, we have summarized the structure of aggrecanases(ADAMTS-4 and ADAMTS-5) and introduced the function of each regional structure; (2) We have concluded the way cytokines and glycosaminoglycans regulate the metabolism of aggrecanases, and discussed the regulation and control principle of cytokines and glycosaminoglycan; (3) We have summarized the majority of inhibitors to the aggrecanases, introduced the endogenic inhibitors, and put our emphasis on the extrinsic inhibitors (chelating agents, polypeptides and so on). Through deeper research on the enzymes, it will help us further understand the pathogenesis of osteoarthritis, and open up new avenues to clinical treatment.


Subject(s)
ADAM Proteins/metabolism , Endopeptidases , Osteoarthritis/enzymology , Procollagen N-Endopeptidase/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/chemistry , ADAMTS4 Protein , ADAMTS5 Protein , Endopeptidases/chemistry , Endopeptidases/metabolism , Extracellular Matrix/enzymology , Humans , Osteoarthritis/therapy , Procollagen N-Endopeptidase/antagonists & inhibitors , Procollagen N-Endopeptidase/chemistry , Tissue Inhibitor of Metalloproteinase-3/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...