Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.695
Filter
1.
Curr Opin Nephrol Hypertens ; 32(6): 551-558, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37584348

ABSTRACT

PURPOSE OF REVIEW: Matrikines are cell-signalling extracellular matrix fragments and they have attracted recent attention from basic and translational scientists, due to their diverse roles in age-related disease and their potential as therapeutic agents. In kidney, the matrix undergoes remodelling by proteolytic fragmentation, so matrikines are likely to play a substantial, yet understudied, role in ageing and pathogenesis of age-related diseases. RECENT FINDINGS: This review presents an up-to-date description of known matrikines with either a confirmed or highly anticipated role in kidney ageing and disease, including their point of origin, mechanism of cleavage, a summary of known biological actions and the current knowledge which links them to kidney health. We also highlight areas of interest, such as the prospect of matrikine cross-tissue communication, and gaps in knowledge, such as the unexplored signalling potential of many kidney disease-specific matrix fragments. SUMMARY: We anticipate that knowledge of specific matrikines, and their roles in controlling processes of kidney pathology, could be leveraged for the development of exciting new future therapies through inhibition or even with their supplementation.


Subject(s)
Extracellular Matrix Proteins , Extracellular Matrix , Humans , Extracellular Matrix Proteins/physiology , Kidney
2.
Oral Dis ; 29(1): 195-205, 2023 Jan.
Article in English | MEDLINE | ID: mdl-34110666

ABSTRACT

OBJECTIVES: Pleiotrophin (PTN), a secreted extracellular matrix-associated protein, plays an important role in regulating the osteo/dentinogenic differentiation potential of dental pulp stem cells (DPSCs). Our previous study has demonstrated that PTN expression in young DPSCs was is 10-fold higher than that in aged DPSCs. However, the role of PTN on the in maintaining the stemness of senescent DPSCs remains unclear. The present study aimed to investigate the effect of PTN on senescent DPSCs in vitro. MATERIALS AND METHODS: Dental pulp stem cells were isolated from human third molars. PTN was knocked down using short hairpin RNAs to study the role of PTN on the senescence of DPSCs. DPSCs with aging performance were obtained by a replicative senescence cell model was obtained by the long-term culture of DPSCs to the 15th passage in vitro (P15). We then investigated the effect of PTN on senescent DPSCs (P15 DPSCs). Real-time RT-PCR, western blotting, alizarin red staining, quantitative calcium analysis, SA-ß-Gal staining, CFSE, and cell-counting kit-8 (CCK8) assays were used to study cellular senescence and function. RESULTS: The depletion of PTN increased the ratio of SA-ß-gal-positive cells, upregulated the expression of p16, and down-regulated the expression of TERT and p-p38. Furthermore, 50 pg/ml of PTN recombinant protein rescued these changes the altered ratio of SA-ß-gal-positive cells, decreased the expression of p16, enhanced TERT and p-p38 expression, as well as telomere activity, caused by PTN depletion and long-term culture. The15th passage cells displayed typical aging characteristic, including high ratio of SA-ß-gal-positive cells, increased aging-related gene expression, decreased proliferation rate, high level of Cyclin D expression, and impaired osteo/dentinogenic differentiation potential. However, 50 pg/ml of PTN recombinant protein could partially reverse these alteration rescue these changes. CONCLUSIONS: The present study demonstrated that PTN could protect DPSCs from senescence by improving the proliferation and osteo/dentinogenic differentiation ability, probably through the p38 MAPK pathway.


Subject(s)
Carrier Proteins , Cytokines , Dental Pulp , Stem Cells , Humans , Cell Differentiation , Cell Proliferation , Cells, Cultured , Extracellular Matrix Proteins/physiology , Osteogenesis , Recombinant Proteins/pharmacology , Stem Cells/physiology , Carrier Proteins/physiology , Cytokines/physiology
3.
Sci Rep ; 12(1): 3080, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197532

ABSTRACT

Fibrosis is a leading cause of morbidity and mortality worldwide. Although fibrosis may involve different organ systems, transforming growth factor-ß (TGFß) has been established as a master regulator of fibrosis across organs. Pirfenidone and Nintedanib are the only currently-approved drugs to treat fibrosis, specifically idiopathic pulmonary fibrosis, but their mechanisms of action remain poorly understood. To identify novel drug targets and uncover potential mechanisms by which these drugs attenuate fibrosis, we performed an integrative 'omics analysis of transcriptomic and proteomic responses to TGFß1-stimulated lung fibroblasts. Significant findings were annotated as associated with pirfenidone and nintedanib treatment in silico via Coremine. Integrative 'omics identified a co-expressed transcriptomic and proteomic module significantly correlated with TGFß1 treatment that was enriched (FDR-p = 0.04) with genes associated with pirfenidone and nintedanib treatment. While a subset of genes in this module have been implicated in fibrogenesis, several novel TGFß1 signaling targets were identified. Specifically, four genes (BASP1, HSD17B6, CDH11, and TNS1) have been associated with pirfenidone, while five genes (CLINT1, CADM1, MTDH, SYDE1, and MCTS1) have been associated with nintedanib, and MYDGF has been implicated with treatment using both drugs. Using the Clue Drug Repurposing Hub, succinic acid was highlighted as a metabolite regulated by the protein encoded by HSD17B6. This study provides new insights into the anti-fibrotic actions of pirfenidone and nintedanib and identifies novel targets for future mechanistic studies.


Subject(s)
Antifibrotic Agents/pharmacology , Computational Biology/methods , Extracellular Matrix Proteins/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Indoles/pharmacology , Pyridones/pharmacology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Antifibrotic Agents/therapeutic use , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Indoles/therapeutic use , Interleukins/genetics , Interleukins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pyridones/therapeutic use , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tensins/genetics , Tensins/metabolism
4.
Nutrients ; 14(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35057443

ABSTRACT

Vitamin K (VK) plays many important functions in the body. The most important of them include the contribution in calcium homeostasis and anticoagulation. Vascular calcification (VC) is one of the most important mechanisms of renal pathology. The most potent inhibitor of this process-matrix Gla protein (MGP) is VK-dependent. Chronic kidney disease (CKD) patients, both non-dialysed and hemodialysed, often have VK deficiency. Elevated uncarboxylated matrix Gla protein (ucMGP) levels indirectly reflected VK deficiency and are associated with a higher risk of cardiovascular events in these patients. It has been suggested that VK intake may reduce the VC and related cardiovascular risk. Vitamin K intake has been suggested to reduce VC and the associated cardiovascular risk. The role and possibility of VK supplementation as well as the impact of anticoagulation therapy on VK deficiency in CKD patients is discussed.


Subject(s)
Renal Insufficiency, Chronic , Vascular Calcification/prevention & control , Vitamin K Deficiency/complications , Vitamin K/administration & dosage , Anticoagulants/therapeutic use , Blood Coagulation/physiology , Bone and Bones/metabolism , Calcium/metabolism , Calcium-Binding Proteins/blood , Calcium-Binding Proteins/physiology , Cardiovascular Diseases/prevention & control , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/physiology , Humans , Renal Dialysis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/etiology , Vascular Calcification/complications , Vascular Calcification/therapy , Vitamin K/physiology , Vitamin K 1/administration & dosage , Vitamin K 1/metabolism , Vitamin K 2/administration & dosage , Vitamin K 2/metabolism , Vitamin K Deficiency/therapy , Matrix Gla Protein
5.
Gene ; 815: 146137, 2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35007686

ABSTRACT

The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.


Subject(s)
Biomarkers, Tumor , Extracellular Matrix Proteins/physiology , Neoplasms/etiology , Neoplasms/therapy , Cell Adhesion Molecules/physiology , Extracellular Matrix Proteins/analysis , Humans , Matrix Metalloproteinases/physiology , Osteonectin/analysis , Osteonectin/physiology , Osteopontin/physiology , Tenascin/physiology , Thrombospondin 1/physiology , Treatment Outcome
6.
Adv Wound Care (New Rochelle) ; 11(5): 234-254, 2022 05.
Article in English | MEDLINE | ID: mdl-33913776

ABSTRACT

Significance: Hypertrophic scars (HTS) are a fibroproliferative disorder that occur following deep dermal injury and affect up to 72% of burn patients. These scars result in discomfort, impaired mobility, disruption of normal function and cosmesis, and significant psychological distress. Currently, there are no satisfactory methods to treat or prevent HTS, as the cellular and molecular mechanisms are complex and incompletely understood. This review summarizes the biology of proteins in the dermal extracellular matrix (ECM), which are involved in wound healing and hypertrophic scarring. Recent Advances: New basic research continues toward understanding the diversity of cellular and molecular mechanisms of normal wound healing and hypertrophic scarring. Broadening the understanding of these mechanisms creates insight into novel methods for preventing and treating HTS. Critical Issues: Although there is an abundance of research conducted on collagen in the ECM and its relationship to HTS, there is a significant gap in understanding the role of proteoglycans and their specific isoforms in dermal fibrosis. Future Directions: Exploring the biological roles of ECM proteins and their unique isoforms in HTS, mature scars, and normal skin will further the understanding of abnormal wound healing and create a more robust understanding of what constitutes dermal fibrosis. Research into the biological roles of ECM protein isoforms and their regulation during wound healing warrants a more extensive investigation to identify their distinct biological functions in cellular processes and outcomes.


Subject(s)
Cicatrix, Hypertrophic , Extracellular Matrix Proteins , Wound Healing , Cicatrix, Hypertrophic/therapy , Dermis , Extracellular Matrix , Extracellular Matrix Proteins/physiology , Humans
7.
J Clin Invest ; 131(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34651580

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) represents a spectrum of chronic liver disease ranging from simple steatosis (NAFL) to nonalcoholic steatohepatitis (NASH). However, the molecular mechanisms of NASH progression remain incompletely understood. White adipose tissue (WAT) has emerged as an important endocrine organ and contributes not only to the initial stage of NAFLD, but also to its severity. In the current study, through transcriptomic analysis we identified increased expression of Sparcl1, a secreted glycoprotein, in the WAT from NASH mice. Plasma Sparcl1 levels were similarly elevated and positively correlated with hepatic pathological features in NASH patients. Functional studies showed that both chronic injection of recombinant Sparcl1 protein and overexpression of Sparcl1 exaggerated hepatic inflammation and liver injury in mice. In contrast, genetic ablation of Sparcl1, knockdown of Sparcl1 in WAT, and treatment with a Sparcl1-neutralizing antibody dramatically alleviated diet-induced NASH pathogenesis. Mechanistically, Sparcl1 promoted the expression of C-C motif chemokine ligand 2 (CCL2) in hepatocytes through binding to Toll-like receptor 4 (TLR4) and activation of the NF-κB/p65 signaling pathway. Genetically or pharmacologically blocking the CCL2/CCR2 pathway attenuated the hepatic inflammatory response evoked by Sparcl1. Thus, our results demonstrated an important role for Sparcl1 in NASH progression, suggesting a potential target for therapeutic intervention.


Subject(s)
Calcium-Binding Proteins/physiology , Chemokine CCL2/physiology , Extracellular Matrix Proteins/physiology , Non-alcoholic Fatty Liver Disease/etiology , Adult , Animals , Calcium-Binding Proteins/blood , Calcium-Binding Proteins/genetics , Disease Progression , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/genetics , Humans , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/physiology , Up-Regulation
8.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34638624

ABSTRACT

Bone fragility is a pathological condition caused by altered homeostasis of the mineralized bone mass with deterioration of the microarchitecture of the bone tissue, which results in a reduction of bone strength and an increased risk of fracture, even in the absence of high-impact trauma. The most common cause of bone fragility is primary osteoporosis in the elderly. However, bone fragility can manifest at any age, within the context of a wide spectrum of congenital rare bone metabolic diseases in which the inherited genetic defect alters correct bone modeling and remodeling at different points and aspects of bone synthesis and/or bone resorption, leading to defective bone tissue highly prone to long bone bowing, stress fractures and pseudofractures, and/or fragility fractures. To date, over 100 different Mendelian-inherited metabolic bone disorders have been identified and included in the OMIM database, associated with germinal heterozygote, compound heterozygote, or homozygote mutations, affecting over 80 different genes involved in the regulation of bone and mineral metabolism. This manuscript reviews clinical bone phenotypes, and the associated bone fragility in rare congenital metabolic bone disorders, following a disease taxonomic classification based on deranged bone metabolic activity.


Subject(s)
Bone Diseases, Metabolic/congenital , Bone Density/genetics , Bone Density/physiology , Bone Development/genetics , Bone Development/physiology , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/physiopathology , Bone Remodeling/genetics , Bone Remodeling/physiology , Bone Resorption/genetics , Bone Resorption/physiopathology , Calcification, Physiologic/genetics , Calcification, Physiologic/physiology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/physiology , Fractures, Bone/genetics , Fractures, Bone/physiopathology , Humans , Metabolic Networks and Pathways/genetics , Mutation , Signal Transduction/genetics
9.
J Am Soc Nephrol ; 32(11): 2777-2794, 2021 11.
Article in English | MEDLINE | ID: mdl-34716242

ABSTRACT

BACKGROUND: Autoantibodies binding to podocyte antigens cause idiopathic membranous glomerulonephritis (iMGN). However, it remains elusive how autoantibodies reach the subepithelial space because the glomerular filtration barrier (GFB) is size selective and almost impermeable for antibodies. METHODS: Kidney biopsies from patients with iMGN, cell culture, zebrafish, and mouse models were used to investigate the role of nephronectin (NPNT) regulating microRNAs (miRs) for the GFB. RESULTS: Glomerular endothelial cell (GEC)-derived miR-192-5p and podocyte-derived miR-378a-3p are upregulated in urine and glomeruli of patients with iMGN, whereas glomerular NPNT is reduced. Overexpression of miR-192-5p and morpholino-mediated npnt knockdown induced edema, proteinuria, and podocyte effacement similar to podocyte-derived miR-378a-3p in zebrafish. Structural changes of the glomerular basement membrane (GBM) with increased lucidity, splitting, and lamellation, especially of the lamina rara interna, similar to ultrastructural findings seen in advanced stages of iMGN, were found. IgG-size nanoparticles accumulated in lucidity areas of the lamina rara interna and lamina densa of the GBM in npnt-knockdown zebrafish models. Loss of slit diaphragm proteins and severe structural impairment of the GBM were further confirmed in podocyte-specific Npnt knockout mice. GECs downregulate podocyte NPNT by transfer of miR-192-5p-containing exosomes in a paracrine manner. CONCLUSIONS: Podocyte NPNT is important for proper glomerular filter function and GBM structure and is regulated by GEC-derived miR-192-5p and podocyte-derived miR-378a-3p. We hypothesize that loss of NPNT in the GBM is an important part of the initial pathophysiology of iMGN and enables autoantigenicity of podocyte antigens and subepithelial immune complex deposition in iMGN.


Subject(s)
Endothelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Glomerular Basement Membrane/metabolism , Glomerular Basement Membrane/physiopathology , Glomerulonephritis, Membranous/genetics , Kidney Glomerulus/metabolism , MicroRNAs/physiology , Animals , Antigen-Antibody Complex/analysis , Autoantigens/genetics , Autoantigens/immunology , Cells, Cultured , Coculture Techniques , Exosomes/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/physiology , Gene Expression Regulation , Gene Targeting , Glomerular Basement Membrane/immunology , Glomerular Basement Membrane/ultrastructure , Glomerulonephritis, Membranous/immunology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/physiopathology , Gold Sodium Thiosulfate , Humans , Metal Nanoparticles , Mice , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/urine , Paracrine Communication , Permeability , Podocytes/immunology , Podocytes/metabolism , Proteinuria/etiology , Transfection , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
10.
Med Sci Monit ; 27: e931238, 2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34210950

ABSTRACT

BACKGROUND Oral squamous cell carcinoma (OSCC) is a common tumor of the head and neck. Its treatment usually requires multiple modalities. Currently, there are no molecular biomarkers to guide these treatment strategies. Studies have shown that microfibril-associated protein 4 (MFAP4) is potentially useful for non-invasive assessment of various diseases; however, its biological function in tumors is still unknown. In this study, we propose that MFAP4 is a new prognostic target for OSCC. MATERIAL AND METHODS First, we collected OSCC data (GSE25099 and GSE30784 datasets) from the Gene Expression Omnibus (GEO) database and compared the differential expression of MFAP4 gene between the patients (tumor) and normal (control) groups. The comparison was done with University of California Santa Cruz Xena (https://xenabrowser.net/Datapages/), and we calculated the difference in MFAP4 gene expression between normal and tumor tissues in a pan-cancer analysis. Then, we compared the 2 groups with high and low expression of MFAP4 gene in terms of tumor mutation burden (TMB), miRNA regulation, and immune cell infiltration. RESULTS We found that the expression of MFAP4 gene was significantly decreased in tumors. Our research also showed that high expression of MFAP4 was related to better prognosis of patients and may be related to tumor gene mutation, miRNA regulation, and infiltration of different immune cells. CONCLUSIONS Our work provides evidence that expression of MFAP4 can be used as a prognostic biomarker for risk stratification of OSCC patients and elaborates on its relation with the regulation of TMB, miRNAs, and immune cell infiltration.


Subject(s)
Carrier Proteins/physiology , Extracellular Matrix Proteins/physiology , Glycoproteins/physiology , Mouth Neoplasms/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Humans
11.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205668

ABSTRACT

Matricellular proteins (MCPs) are defined as extracellular matrix (ECM) associated proteins that are important regulators and integrators of microenvironmental signals, contributing to the dynamic nature of ECM signalling. There is a growing understanding of the role of matricellular proteins in cellular processes governing tissue development as well as in disease pathogenesis. In this review, the expression and functions of different MP family members (periostin, CCNs, TSPs, SIBLINGs and others) are presented, specifically in relation to craniofacial development and the maintenance of orofacial tissues, including bone, gingiva, oral mucosa, palate and the dental pulp. As will be discussed, each MP family member has been shown to have non-redundant roles in development, tissue homeostasis, wound healing, pathology and tumorigenesis of orofacial and dental tissues.


Subject(s)
Cell Adhesion Molecules/physiology , Extracellular Matrix Proteins/physiology , Mouth/growth & development , Osteonectin/physiology , Thrombospondins/physiology , Animals , CCN Intercellular Signaling Proteins/physiology , Head and Neck Neoplasms/etiology , Humans , Mouth/embryology , Tenascin/physiology , Wound Healing
12.
Curr Osteoporos Rep ; 19(5): 500-509, 2021 10.
Article in English | MEDLINE | ID: mdl-34331667

ABSTRACT

PURPOSE OF REVIEW: Chronic kidney disease-mineral and bone disorder (CKD-MBD) has become a global health crisis with very limited therapeutic options. Dentin matrix protein 1 (DMP1) is a matrix extracellular protein secreted by osteocytes that has generated recent interest for its possible involvement in CKD-MBD pathogenesis. This is a review of DMP1 established regulation and function, and early studies implicating DMP1 in CKD-MBD. RECENT FINDINGS: Patients and mice with CKD show perturbations of DMP1 expression in bone, associated with impaired osteocyte maturation, mineralization, and increased fibroblast growth factor 23 (FGF23) production. In humans with CKD, low circulating DMP1 levels are independently associated with increased cardiovascular events. We recently showed that DMP1 supplementation lowers circulating FGF23 levels and improves bone mineralization and cardiac outcomes in mice with CKD. Mortality rates are extremely high among patients with CKD and have only marginally improved over decades. Bone disease and FGF23 excess contribute to mortality in CKD by increasing the risk of bone fractures and cardiovascular disease, respectively. Previous studies focused on DMP1 loss-of-function mutations have established its role in the regulation of FGF23 and bone mineralization. Recent studies show that DMP1 supplementation may fill a crucial therapeutic gap by improving bone and cardiac health in CKD.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder/etiology , Extracellular Matrix Proteins/physiology , Phosphoproteins/physiology , Animals , Chronic Kidney Disease-Mineral and Bone Disorder/metabolism , Chronic Kidney Disease-Mineral and Bone Disorder/pathology , Humans , Mice , Rats
13.
Mol Cell Neurosci ; 112: 103614, 2021 04.
Article in English | MEDLINE | ID: mdl-33845123

ABSTRACT

Homozygous Dab1 yotari mutant mice, Dab1yot (yot/yot) mice, have an autosomal recessive mutation of Dab1 and show reeler-like phenotype including histological abnormality of the cerebellum, hippocampus, and cerebral cortex. We here show abnormal hippocampal development of yot/yot mice where granule cells and pyramidal cells fail to form orderly rows but are dispersed diffusely in vague multiplicative layers. Possibly due to the positioning failure of granule cells and pyramidal cells and insufficient synaptogenesis, axons of the granule cells did not extend purposefully to connect with neighboring regions in yot/yot mice. We found that both hippocampal granule cells and pyramidal cells of yot/yot mice expressed proteins reactive with the anti-Dab1 antibody. We found that Y198- phosphorylated Dab1 of yot/yot mice was greatly decreased. Accordingly the downstream molecule, Akt was hardly phosphorylated. Especially, synapse formation was defective and the distribution of neurons was scattered in hippocampus of yot/yot mice. Some of neural cell adhesion molecules and hippocampus associated transcription factors of the neurons were expressed aberrantly, suggesting that the Reelin-Dab1 signaling pathway seemed to be importantly involved in not only neural migration as having been shown previously but also neural maturation and/or synaptogenesis of the mice. It is interesting to clarify whether the defective neural maturation is a direct consequence of the dysfunctional Dab1, or alternatively secondarily due to the Reelin-Dab1 intracellular signaling pathways.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hippocampus/abnormalities , Mice, Mutant Strains/abnormalities , Nerve Tissue Proteins/physiology , Serine Endopeptidases/physiology , Signal Transduction/physiology , Animals , Cell Adhesion Molecules, Neuronal/deficiency , Cell Movement , Enzyme Activation , Extracellular Matrix Proteins/deficiency , Genes, Recessive , Hippocampus/embryology , Hippocampus/metabolism , Hippocampus/pathology , Homozygote , Mice , Mice, Mutant Strains/genetics , Mice, Mutant Strains/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Reelin Protein , Serine Endopeptidases/deficiency , Synapses/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33670905

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease causing inflammation of joints, cartilage destruction and bone erosion. Biomarkers and new drug targets are actively sought and progressed to improve available options for patient treatment. The Collagen Triple Helix Repeat Containing 1 protein (CTHRC1) may have an important role as a biomarker for rheumatoid arthritis, as CTHRC1 protein concentration is significantly elevated in the peripheral blood of rheumatoid arthritis patients compared to osteoarthritis (OA) patients and healthy individuals. CTHRC1 is a secreted glycoprotein that promotes cell migration and has been implicated in arterial tissue-repair processes. Furthermore, high CTHRC1 expression is observed in many types of cancer and is associated with cancer metastasis to the bone and poor patient prognosis. However, the function of CTHRC1 in RA is still largely undefined. The aim of this review is to summarize recent findings on the role of CTHRC1 as a potential biomarker and pathogenic driver of RA progression. We will discuss emerging evidence linking CTHRC1 to the pathogenic behavior of fibroblast-like synoviocytes and to cartilage and bone erosion through modulation of the balance between bone resorption and repair.


Subject(s)
Arthritis, Rheumatoid/metabolism , Extracellular Matrix Proteins/metabolism , Animals , Arthritis, Rheumatoid/physiopathology , Biomarkers , Extracellular Matrix Proteins/physiology , Humans , Synoviocytes/metabolism , Synoviocytes/physiology , Wnt Signaling Pathway
15.
J Gastroenterol Hepatol ; 36(1): 196-203, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32537806

ABSTRACT

BACKGROUND AND AIM: Tubulointerstitial nephritis antigen-like 1 (TINAGL1), as a novel matricellular protein, has been demonstrated to participate in cancer progression, whereas the potential function of TINAGL1 in gastric cancer (GC) remains unknown. METHODS: The expression pattern of TINAGL1 in GC was examined by immunohistochemistry, ELISA, real-time polymerase chain reaction, and Western blot. Correlation between TINAGL1 and matrix metalloproteinases (MMPs) was analyzed by the GEPIA website and Kaplan-Meier plots database. The lentivirus-based TINAGL1 knockdown, CCK-8, and transwell assays were used to test the function of TINAGL1 in vitro. The role of TINAGL1 was confirmed by subcutaneous xenograft, abdominal dissemination, and lung metastasis model. Microarray experiments, ELISA, real-time polymerase chain reaction, and Western blot were used to identify molecular mechanism. RESULTS: TINAGL1 was increased in GC tumor tissues and associated with poor patient survival. Moreover, TINAGL1 significantly promoted GC cell proliferation and migration in vitro as well as facilitated GC tumor growth and metastasis in vivo. TINAGL1 expression in GC cells was accompanied with increasing MMPs including MMP2, MMP9, MMP11, MMP14, and MMP16. GEPIA database revealed that these MMPs were correlated with TINAGL1 in GC tumors and that the most highly expressed MMP was MMP2. Mechanically, TINAGL1 regulated MMP2 through the JNK signaling pathway activation. CONCLUSIONS: Our data highlight that TINAGL1 promotes GC growth and metastasis and regulates MMP2 expression, indicating that TINAGL1 may serve as a therapeutic target for GC.


Subject(s)
Cell Proliferation/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression/genetics , Lipocalins/genetics , Lipocalins/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Metastasis/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Up-Regulation/genetics , Up-Regulation/physiology , Animals , Cell Line , Cell Movement/genetics , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/physiology , Female , Humans , Lipocalins/physiology , Mice, Nude , Molecular Targeted Therapy , Stomach Neoplasms/therapy
16.
Dev Dyn ; 250(2): 274-294, 2021 02.
Article in English | MEDLINE | ID: mdl-33012048

ABSTRACT

BACKGROUND: The extracellular matrix (ECM) is a fundamental component of multicellular organisms that orchestrates developmental processes and controls cell and tissue organization. We previously identified the novel ECM protein SNED1 as a promoter of breast cancer metastasis and showed that its level of expression negatively correlated with breast cancer patient survival. Here, we sought to identify the roles of SNED1 during murine development. RESULTS: We generated two novel Sned1 knockout mouse strains and showed that Sned1 is essential since homozygous ablation of the gene led to early neonatal lethality. Phenotypic analysis of the surviving knockout mice revealed a role for SNED1 in the development of craniofacial and skeletal structures since Sned1 knockout resulted in growth defects, nasal cavity occlusion, and craniofacial malformations. Sned1 is widely expressed in embryos, notably by cell populations undergoing epithelial-to-mesenchymal transition, such as the neural crest cells. We further show that mice with a neural-crest-cell-specific deletion of Sned1 survive, but display facial anomalies partly phenocopying the global knockout mice. CONCLUSIONS: Our results demonstrate requisite roles for SNED1 during development and neonatal survival. Importantly, the deletion of 2q37.3 in humans, a region that includes the SNED1 locus, has been associated with facial dysmorphism and short stature.


Subject(s)
Extracellular Matrix Proteins/physiology , Amino Acid Sequence , Animals , Conserved Sequence , Craniofacial Abnormalities/genetics , Genes, Lethal , Growth Disorders/genetics , Mandible/abnormalities , Mice , Mice, Knockout , Nasal Cavity/abnormalities
17.
Biosci Rep ; 41(1)2021 01 29.
Article in English | MEDLINE | ID: mdl-33306121

ABSTRACT

BACKGROUND: The family with sequence similarity 20-member C (Fam20C) kinase plays important roles in physiopathological process and is responsible for majority of the secreted phosphoproteome, including substrates associated with tumor cell migration. However, it remains unclear whether Fam20C plays a role in cancers. Here, we aimed to analyze the expression and prognostic value of Fam20C in pan-cancer and to gain insights into the association between Fam20C and immune infiltration. METHODS: We analyzed Fam20C expression patterns and the associations between Fam20C expression levels and prognosis in pan-cancer via the ONCOMINE, TIMER (Tumor Immune Estimation Resource), PrognoScan, GEPIA (Gene Expression Profiling Interactive Analysis), and Kaplan-Meier Plotter databases. After that, GEPIA and TIMER databases were applied to investigate the relations between Fam20C expression and immune infiltration across different cancer types, especially BLCA (bladder urothelial carcinoma), LGG (brain lower grade glioma), and STAD (stomach adenocarcinoma). RESULTS: Compared with adjacent normal tissues, Fam20C was widely expressed across many cancers. In general, Fam20C showed a detrimental role in pan-cancer, it was positively associated with poor survival of BLCA, LGG, and STAD patients. Specifically, based on TCGA (The Cancer Genome Atlas) database, a high expression level of Fam20C was associated with worse prognostic value in stages T2-T4 and stages N0-N2 in the cohort of STAD patients. Moreover, Fam20C expression had positive associations with immune infiltration, including CD4+ T cells, macrophages, neutrophils, and dendritic cells, and other diverse immune cells in BLCA, LGG, and STAD. CONCLUSION: Fam20C may serve as a promising prognostic biomarker in pan-cancer and has positive associations with immune infiltrates.


Subject(s)
Casein Kinase I/physiology , Extracellular Matrix Proteins/physiology , Neoplasms/immunology , Casein Kinase I/genetics , Extracellular Matrix Proteins/genetics , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Neoplasms/genetics , Neoplasms/pathology , Oncogenes , Prognosis , RNA, Messenger/genetics
18.
Sci Rep ; 10(1): 18583, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33122788

ABSTRACT

The ability to ablate a gene in a given tissue by generating a conditional knockout (cKO) is crucial for determining its function in the targeted tissue. Such tissue-specific ablation is even more critical when the gene's conventional knockout (KO) is lethal, which precludes studying the consequences of its deletion in other tissues. Therefore, here we describe a successful strategy that generated a Matrix Gla floxed mouse (Mgp.floxed) by the CRISPR/Cas9 system, that subsequently allowed the generation of cKOs by local viral delivery of the Cre-recombinase enzyme. MGP is a well-established inhibitor of calcification gene, highly expressed in arteries' smooth muscle cells and chondrocytes. MGP is also one of the most abundant genes in the trabecular meshwork, the eye tissue responsible for maintenance of intraocular pressure (IOP) and development of Glaucoma. Our strategy entailed one-step injection of two gRNAs, Cas9 protein and a long-single-stranded-circular DNA donor vector (lsscDNA, 6.7 kb) containing two loxP sites in cis and 900-700 bp 5'/3' homology arms. Ocular intracameral injection of Mgp.floxed mice with a Cre-adenovirus, led to an Mgp.TMcKO mouse which developed elevated IOP. Our study discovered a new role for the Mgp gene as a keeper of physiological IOP in the eye.


Subject(s)
Calcium-Binding Proteins/physiology , Extracellular Matrix Proteins/physiology , Eye/physiopathology , Intraocular Pressure , Trabecular Meshwork/physiopathology , Animals , Base Sequence , Female , Glaucoma/physiopathology , Integrases/metabolism , Mice , Mice, Knockout , RNA, Guide, Kinetoplastida/administration & dosage , Matrix Gla Protein
19.
J Neurosci ; 40(42): 8088-8102, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32973045

ABSTRACT

Emerging evidence supports roles for secreted extracellular matrix proteins in boosting synaptogenesis, synaptic transmission, and synaptic plasticity. SPARCL1 (also known as Hevin), a secreted non-neuronal protein, was reported to increase synaptogenesis by simultaneously binding to presynaptic neurexin-1α and to postsynaptic neuroligin-1B, thereby catalyzing formation of trans-synaptic neurexin/neuroligin complexes. However, neurexins and neuroligins do not themselves mediate synaptogenesis, raising the question of how SPARCL1 enhances synapse formation by binding to these molecules. Moreover, it remained unclear whether SPARCL1 acts on all synapses containing neurexins and neuroligins or only on a subset of synapses, and whether it enhances synaptic transmission in addition to boosting synaptogenesis or induces silent synapses. To explore these questions, we examined the synaptic effects of SPARCL1 and their dependence on neurexins and neuroligins. Using mixed neuronal and glial cultures from neonatal mouse cortex of both sexes, we show that SPARCL1 selectively increases excitatory but not inhibitory synapse numbers, enhances excitatory but not inhibitory synaptic transmission, and augments NMDAR-mediated synaptic responses more than AMPAR-mediated synaptic responses. None of these effects were mediated by SPARCL1-binding to neurexins or neuroligins. Neurons from triple neurexin-1/2/3 or from quadruple neuroligin-1/2/3/4 conditional KO mice that lacked all neurexins or all neuroligins were fully responsive to SPARCL1. Together, our results reveal that SPARCL1 selectively boosts excitatory but not inhibitory synaptogenesis and synaptic transmission by a novel mechanism that is independent of neurexins and neuroligins.SIGNIFICANCE STATEMENT Emerging evidence supports roles for extracellular matrix proteins in boosting synapse formation and function. Previous studies demonstrated that SPARCL1, a secreted non-neuronal protein, promotes synapse formation in rodent and human neurons. However, it remained unclear whether SPARCL1 acts on all or on only a subset of synapses, induces functional or largely inactive synapses, and generates synapses by bridging presynaptic neurexins and postsynaptic neuroligins. Here, we report that SPARCL1 selectively induces excitatory synapses, increases their efficacy, and enhances their NMDAR content. Moreover, using rigorous genetic manipulations, we show that SPARCL1 does not require neurexins and neuroligins for its activity. Thus, SPARCL1 selectively boosts excitatory synaptogenesis and synaptic transmission by a novel mechanism that is independent of neurexins and neuroligins.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Neural Cell Adhesion Molecules/physiology , Synapses/physiology , Animals , Cerebral Cortex/cytology , Female , Male , Mice , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Primary Cell Culture , Receptors, Cell Surface , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology
20.
Biomolecules ; 10(9)2020 09 08.
Article in English | MEDLINE | ID: mdl-32911658

ABSTRACT

Fibulin-3 (also known as EGF-containing fibulin extracellular matrix protein 1 (EFEMP1)) is a secreted extracellular matrix glycoprotein, encoded by the EFEMP1 gene that belongs to the eight-membered fibulin protein family. It has emerged as a functionally unique member of this family, with a diverse array of pathophysiological associations predominantly centered on its role as a modulator of extracellular matrix (ECM) biology. Fibulin-3 is widely expressed in the human body, especially in elastic-fibre-rich tissues and ocular structures, and interacts with enzymatic ECM regulators, including tissue inhibitor of metalloproteinase-3 (TIMP-3). A point mutation in EFEMP1 causes an inherited early-onset form of macular degeneration called Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD). EFEMP1 genetic variants have also been associated in genome-wide association studies with numerous complex inherited phenotypes, both physiological (namely, developmental anthropometric traits) and pathological (many of which involve abnormalities of connective tissue function). Furthermore, EFEMP1 expression changes are implicated in the progression of numerous types of cancer, an area in which fibulin-3 has putative significance as a therapeutic target. Here we discuss the potential mechanistic roles of fibulin-3 in these pathologies and highlight how it may contribute to the development, structural integrity, and emergent functionality of the ECM and connective tissues across a range of anatomical locations. Its myriad of aetiological roles positions fibulin-3 as a molecule of interest across numerous research fields and may inform our future understanding and therapeutic approach to many human diseases in clinical settings.


Subject(s)
Extracellular Matrix Proteins/physiology , Animals , Disease Models, Animal , Extracellular Matrix/enzymology , Extracellular Matrix Proteins/chemistry , Genome-Wide Association Study , Humans , Optic Disk Drusen/congenital , Optic Disk Drusen/genetics , Optic Disk Drusen/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...