Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Sci Rep ; 11(1): 12437, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34127702

ABSTRACT

Calcineurin inhibitors, such as Cyclosporin (CsA), are the mainstay of anti-rejection therapy in solid organ transplants but can paradoxically induce progressive nephropathy characterised by renal dysfunction and interstitial fibrosis. Lysyl oxidases (LOXs), a group of enzymes that catalyse extracellular matrix (ECM) crosslinking, were shown to implicate in tissue scarring. It is hypothesized that inhibition of these enzymes may render therapeutic effects against CsA-induced nephropathy. In this study, 6-to-8 weeks old C57BL/6 J mice were administered saline or CsA (30 mg/kg/day s.c) for 16 weeks. At 8 weeks, CsA-treated animals were divided into 5 groups respectively treated with: (1) vehicle, (2) PXS-5505 (Pan-LOX inhibitor), (3) PXS-5382 (LOX-like 2 inhibitor), (4) PXS-5505 for 4 weeks then PXS-5382 for 4 weeks (sequential therapy), and (5) Telmisartan (standard therapy). Our results indicate that CsA administration significantly increased the levels of blood urea nitrogen, glomerular and tubular injury, tubulointerstitial fibrosis, inflammation and oxidative stress in mouse kidney. These changes were associated with upregulated mRNA expression of LOX and LOXL2. Administration of Pan-LOX or LOXL2 inhibitors or the sequential therapy suppressed the expression of ECM proteins (α-SMA, FN and COL1A), matrix metalloproteases (MMP)2 and 9, inflammatory markers (TNFα and MCP-1) and TGF-ß1-Smad3 signalling. Among all regimens including telmisartan, only Pan-LOX inhibitor PXS-5505 was able to attenuate uraemia. Collectively, our study suggests that Pan-LOX and LOXL2 inhibition can attenuate progressive nephropathy due to CsA administration.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Cyclosporine/adverse effects , Extracellular Matrix Proteins/antagonists & inhibitors , Kidney Diseases/drug therapy , Protective Agents/therapeutic use , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Amino Acid Oxidoreductases/metabolism , Animals , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Extracellular Matrix Proteins/metabolism , Fibrosis , Humans , Kidney Diseases/chemically induced , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Protective Agents/pharmacology , Protein-Lysine 6-Oxidase/metabolism
2.
Oncogene ; 40(23): 3959-3973, 2021 06.
Article in English | MEDLINE | ID: mdl-33986509

ABSTRACT

Metastasis is a major cause of cancer-related deaths. Tumor-intrinsic properties can determine whether tumor metastasis occurs or not. Here, by comparing the gene expression patterns in primary colorectal cancer (CRC) patients with or without metastasis, we found that Collagen Triple Helix Repeat Containing 1 (CTHRC1) in primary CRC served as a metastasis-associated gene. Animal experiments verified that CTHRC1 secreted by CRC cells promoted hepatic metastasis, which was closely correlated with macrophage infiltration. Depletion of macrophages by liposomal clodronate largely abolished the promoting effect of CTHRC1 on CRC liver metastasis. Furthermore, we demonstrated that CTHRC1 modulated macrophage polarization to M2 phenotypes through TGF-ß signaling. A mechanistic study revealed that CTHRC1 bound directly to TGF-ß receptor II and TGF-ß receptor III, stabilized the TGF-ß receptor complex, and activated TGF-ß signaling. The combination treatment of CTHRC1 monoclonal antibody and anti-PD-1 blocking antibody effectively suppressed CRC hepatic metastasis. Taken together, our data demonstrated that CTHRC1 is an intrinsic marker of CRC metastasis and further revealed that CTHRC1 promoted CRC liver metastasis by reshaping infiltrated macrophages through TGF-ß signaling, suggesting that CTHRC1 could be a potential biomarker for the early prediction of and a therapeutic target of CRC hepatic metastasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colorectal Neoplasms/metabolism , Extracellular Matrix Proteins/metabolism , Liver Neoplasms/secondary , Macrophages/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Databases, Genetic , Disease Models, Animal , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Survival Rate , Treatment Outcome
3.
Neurosci Lett ; 755: 135902, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33865939

ABSTRACT

Glaucoma is a progressive optic neuropathy in more than 25 % of cases in patients with permanent blindness. The microRNA is implicated in modulating the cellular function of the trabecular meshwork (TM). The aim of this study is to investigate the role of miR-137 in glaucoma and illustrate the potential molecular mechanisms. We show that miR-137 was down-regulated in H2O2-induced human trabecular meshwork cells (HTMCs), and overexpression of miR-137 attenuated H2O2-induced cell growth inhibition, apoptosis and elevated extracellular matrix (ECM) protein expression. In addition, miR-137 blocked the activation of YAP/TAZ by directly targeting src. Overexpression of src or activation of the YAP/TAZ pathway partly abrogated the effects of miR-137 on H2O2-induced cell viability and apoptosis and dampened the inhibition effect on ECM protein expression. In conclusion, miR-137 promotes cell growth and inhibits extracellular matrix protein expression in H2O2-induced human trabecular meshwork cells via the YAP/TAZ pathway by targeting src. Hence, miR-137 might be used as a novel therapeutic target to treat glaucoma.


Subject(s)
Cell Proliferation/physiology , Extracellular Matrix Proteins/biosynthesis , Hydrogen Peroxide/toxicity , MicroRNAs/biosynthesis , Proto-Oncogene Proteins pp60(c-src)/metabolism , Trabecular Meshwork/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Gene Expression , Humans , MicroRNAs/genetics , Oxidative Stress/drug effects , Oxidative Stress/physiology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/genetics , Trabecular Meshwork/drug effects
4.
Inflammation ; 44(5): 1831-1842, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33846931

ABSTRACT

Collagen triple helix repeat containing 1 (CTHRC1), a secreted glycoprotein, is widely expressed in many tissues. It has been recently defined as a novel marker for rheumatoid arthritis (RA), a systemic inflammatory disorder. However, the precise role of CTHRC1 in other chronic inflammatory diseases, like periodontal disease, remains unclear. This research aimed to explore the presence of CTHRC1 in periodontal inflammation, determine the precise role in inflammatory response modulation in periodontal ligament cells (PDLCs), and explore its underlying mechanisms. In vivo gingival crevicular fluid (GCF) and gingivae were obtained from healthy people and chronic periodontitis patients. Maxillary tissues of mice with or without ligature-induced periodontitis were immunostained for CTHRC1. In vitro human PDLCs were treated with tumor necrosis factor alpha (TNF-α) to mimic the inflammatory environment. Small interfering RNA (siRNA) was used to silence CTHRC1. SB203580 was used to inhibit the p38 mitogen-activated protein kinase (MAPK) pathway. CTHRC1 was highly expressed in GCF and gingival tissues of periodontitis patients. Animal models also revealed the same tendency. CTHRC1 knockdown promoted inflammatory cytokine production and activated the p38 MAPK signaling pathway in PDLCs. Inhibiting the p38 MAPK signaling pathway partially attenuated the inflammatory responses. This study revealed that CTHRC1 was highly expressed in periodontitis and suggested that CTHRC1 might play an important role in modulating periodontal inflammation.


Subject(s)
Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/biosynthesis , Gene Knockdown Techniques/methods , Inflammation Mediators/metabolism , MAP Kinase Signaling System/physiology , Periodontal Ligament/metabolism , Adolescent , Animals , Cells, Cultured , Child , Enzyme Activation/physiology , Extracellular Matrix Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Periodontal Ligament/pathology , Periodontitis/genetics , Periodontitis/metabolism , Periodontitis/pathology , Young Adult
5.
Cell Rep ; 34(10): 108829, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33691102

ABSTRACT

Neuronal synapse formation is critical for brain development and depends on secreted factors from astrocytes. Here, we report that small extracellular vesicles (EVs) secreted from primary astrocytes, but not from neurons or C6 glioma cells, greatly enhance spine and synapse formation by primary cortical neurons. A comparative proteomics analysis of small EVs from astrocytes, neurons, and C6 glioma cells identified fibulin-2 as a promising EV cargo to regulate synaptogenesis. Treatment of cortical neurons with recombinant fibulin-2 increased the formation of spines and synapses, similar to the effect of small EVs. In addition, treatment of neurons with fibulin-2 or astrocyte-derived small EVs led to increased phosphorylation of Smad2, an indicator of TGF-ß signaling. Finally, the effects of fibulin-2 and astrocyte-derived small EVs on synapse formation were reversed by inhibiting transforming growth factor ß (TGF-ß) signaling. These data suggest a model in which astrocyte EVs promote synapse formation via fibulin-2-mediated activation of TGF-ß signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Vesicles/metabolism , Signal Transduction , Synapses/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cells, Cultured , Dendritic Spines/physiology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
6.
Arterioscler Thromb Vasc Biol ; 41(4): 1309-1318, 2021 04.
Article in English | MEDLINE | ID: mdl-33626909
7.
Eur J Med Chem ; 210: 113088, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33316691

ABSTRACT

The family with sequence similarity 20, member C (Fam20C), a Golgi casein kinase, has been recently regarded as a potential therapeutic target for the treatment of triple negative breast cancer (TNBC). Lacking enzyme activity center has been becoming an obstacle to the development of small-molecule inhibitors of Fam20C. Herein, we combined in silico high-throughput screening with chemical synthesis methods to obtain a new small-molecule Fam20C inhibitor 3r, which exhibited desired anti-proliferative activities against MDA-MB-231 cells and also inhibited migration. Subsequently, the enzymatic assay, molecular docking, and molecular dynamics (MD) simulations were carried out for validating that 3r could bind to Fam20C. In addition, 3r was found to induce apoptosis via the mitochondrial pathway in MDA-MB-231 cells as well as to inhibit cell migration. Moreover, we demonstrated that 3r inhibited tumor growth in vivo and thereby having a good therapeutic potential on TNBC. Taken together, these results suggest that 3r may be a novel Fam20C inhibitor with anti-proliferative and apoptosis-inducing activities, which would shed light on discovering more small-molecule drugs for the future TNBC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Casein Kinase I/antagonists & inhibitors , Drug Discovery , Extracellular Matrix Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Casein Kinase I/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Extracellular Matrix Proteins/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
8.
Biochem Pharmacol ; 189: 114390, 2021 07.
Article in English | MEDLINE | ID: mdl-33359068

ABSTRACT

Matrix Gla protein (MGP), an extracellular matrix protein, has been widely reported to participate in the tumorigenic process and is abnormally expressed in several tumors. However, the role of MGP in colorectal cancer (CRC) remains unknown. Chemotherapy resistance represents a significant limitation in the treatment of CRC. Here, a comprehensive bioinformatics analysis revealed that MGP, which is overexpressed in CRC, might act as one of the critical genes conferring resistance to oxaliplatin (OXA). Furthermore, we found that MGP overexpression in tumor tissue might be correlated with cancer stage and patient prognosis, consistent with the bioinformatics analysis. The upregulation of MGP may act as an independent risk factor for CRC. The knockdown of MGP or inhibition of MGP expression significantly increased the sensitivity of the CRC cell lines to OXA. Suppression of MGP may reverse OXA resistance by upregulating copper transporter 1 (CTR1) and downregulating ATP7A and ATP7B. When used in combination with OXA, the inhibition of MGP reduced cancer cell proliferation, invasion, and migration and increased cell apoptosis in vitro. Suppression of MGP or OXA treatment alone significantly inhibited tumor growth in the CRC mouse model. Additionally, we found that OXA might promote the antitumor immune response in vivo. In summary, our study is the first to provide evidence that MGP expression confers OXA chemotherapy resistance in CRC and provides novel strategies to overcome chemotherapy resistance in CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Extracellular Matrix Proteins/antagonists & inhibitors , Oxaliplatin/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Databases, Genetic , Drug Resistance, Neoplasm/physiology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Oxaliplatin/therapeutic use , Matrix Gla Protein
9.
J Invest Dermatol ; 141(6): 1482-1492.e4, 2021 06.
Article in English | MEDLINE | ID: mdl-33242499

ABSTRACT

Systemic sclerosis a chronic, fibrotic disorder associated with high disease-specific mortality and morbidity. Cutaneous manifestations include dermal thickening and obliteration of dermal adipose tissue. Accumulation of low-molecular-weight hyaluronan, which signals through the receptor for hyaluronan-mediated motility, RHAMM, leads to progressive fibrosis and is correlated with increased severity of systemic sclerosis. The purpose of this study is to test the efficacy of two function-blocking RHAMM peptides, NPI-110 and NPI-106, in reducing skin fibrosis in a bleomycin-induced mouse model of systemic sclerosis. NPI-110 reduced visible measures of fibrosis (dermal thickness and collagen production, deposition, and organization) and profibrotic gene expression (Tgfb1, c-Myc, Col1a1, Col3a1). NPI-110 treatment also increased the expression of the antifibrotic adipokines perilipin and adiponectin. Both RHAMM peptides strongly reduced dermal RHAMM expression, predicting that dermal fibroblasts are peptide targets. Transcriptome and cell culture analyses using Rhamm-/- and Rhamm-rescued dermal fibroblasts reveal a TGFß1/RHAMM/MYC signaling axis that promotes fibrogenic gene expression and myofibroblast differentiation. RHAMM function‒blocking peptides suppress this signaling and prevent TGFß1-induced myofibroblast differentiation. These results suggest that inhibiting RHAMM signaling will offer a treatment method for cutaneous fibrosis in systemic sclerosis.


Subject(s)
Adipokines/metabolism , Extracellular Matrix Proteins/antagonists & inhibitors , Hyaluronan Receptors/antagonists & inhibitors , Peptides/pharmacology , Scleroderma, Systemic/drug therapy , Skin/pathology , Animals , Bleomycin/administration & dosage , Bleomycin/toxicity , Cell Differentiation/drug effects , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Fibrosis , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Knockout , Peptides/therapeutic use , Proto-Oncogene Proteins c-myc/metabolism , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/pathology , Signal Transduction/drug effects , Skin/drug effects , Transforming Growth Factor beta1/metabolism
10.
J Ethnopharmacol ; 269: 113761, 2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33383114

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Peel of Citrus reticulata, a Chinese herbal drug with functions of regulating Qi and expelling phlegm, has been used for the treatment of lung related diseases in Chinese medicine for a long time. Its detailed effects on collagen in anti-idiopathic pulmonary fibrosis (IPF) is still unclear. AIM OF THE STUDY: To explore the effects of citrus alkaline extract (CAE) on collagen synthesis, crosslinking and deposition in pulmonary fibrosis and understand the possible signal pathways involved in the activity. MATERIALS AND METHODS: CAE was prepared from C. reticulata. Bleomycin-induced pulmonary fibrosis mouse model was applied. Pulmonary fibrosis of lung was estimated with histopathology analysis, and collagen deposition was evaluated with immunohistochemistry. Collagen crosslinking related biomarkers and enzymes were analyzed with chemical methods, immunohistochemical and western blot analyses. RESULTS: CAE oral administration lowered hydroxyproline content, inhibited the collagen deposition including expressions of collagen I and III, and relieved bleomycin-induced pulmonary fibrosis in mice model. The productions of a collagen crosslink pyridinoline and crosslinking related enzymes including lysyl oxidase (LOX), lysyl oxidase-like protein 1 (LOXL1) in lung were suppressed by CAE treatment. Furthermore, the protein expressions of TGF-ß1 and Smad3 levels in lungs were also downregulated by CAE. CONCLUSIONS: This study demonstrated that CAE inhibited collagen synthesis, crosslinking and deposition, and ameliorated bleomycin-induced pulmonary fibrosis. Preliminary mechanism study revealed that CAE exerted its bioactivity at least via downregulation of TGF-ß1/Smad3 pathway. Our findings provided a great potential in fighting IPF based on CAE.


Subject(s)
Citrus/chemistry , Collagen Type III/metabolism , Collagen Type I/metabolism , Plant Extracts/pharmacology , Pulmonary Fibrosis/drug therapy , Administration, Oral , Alkalies/chemistry , Amino Acid Oxidoreductases/antagonists & inhibitors , Amino Acid Oxidoreductases/metabolism , Amino Acids/metabolism , Animals , Bleomycin/toxicity , Collagen Type III/genetics , Disease Models, Animal , Down-Regulation/drug effects , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/metabolism , Hydroxyproline/metabolism , Mice, Inbred C57BL , Plant Extracts/administration & dosage , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics
12.
J Gene Med ; 22(12): e3288, 2020 12.
Article in English | MEDLINE | ID: mdl-33047833

ABSTRACT

BACKGROUND: Dentin matrix protein 1 (DMP1) is highly expressed in mineralized tooth and bone, playing a critical role in mineralization and phosphate metabolism. One important role for the expression of DMP1 in the nucleus of preosteoblasts is the up-regulation of osteoblast-specific genes such as osteocalcin and alkaline phosphatase1 . The present study aimed to investigate the potential application of human DMP1 promoter as an indicator marker of osteoblastic differentiation. METHODS: In the present study, we developed DMP1 promoter-DsRed-GFP knock-in mesenchymal stem cell (MSCs) via the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system that enabled automatic detection of osteoblast differentiation. With the application of a homology-directed knock-in strategy, a 2-kb fragment of DMP1 promoter, which was inserted upstream of the GFP and DsRed reporter cassette, was integrated into the human ROSA locus to generate double fluorescent cells. We further differentiated MSCs under osteogenic media to monitor the fate of MSCs. First, cells were transfected using CRISPR/Cas9 plasmids, which culminated in MSCs with a green fluorescence intensity, then GFP-positive cells were selected using puromycin. Second, the GFP-positive MSCs were differentiated toward osteoblasts, which demonstrated an increased red fluorescence intensity. The osteoblast differentiation of MSCs was also verified by performing alkaline phosphatase and Alizarin Red assays. RESULTS: We have exploited the DMP1 promoter as a predictive marker of MSC differentiation toward osteoblasts. Using the CRISPR/Cas9 technology, we have identified a distinctive change in the fluorescence intensities of GFP knock-in (green) and osteoblast differentiated MSCs 2 . CONCLUSIONS: The data show that DMP1-DsRed-GFP knock-in MSCs through CRISPR/Cas9 technology provide a valuable indicator for osteoblast differentiation. Moreover, The DMP1 promoter might be used as a predictive marker of MSCs differentiated toward osteoblasts.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation , Extracellular Matrix Proteins/antagonists & inhibitors , Gene Knock-In Techniques/methods , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis , Phosphoproteins/antagonists & inhibitors , Cell Proliferation , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic
13.
Sci Adv ; 6(31)2020 07.
Article in English | MEDLINE | ID: mdl-32937590

ABSTRACT

The outbreak of the highly contagious and deadly severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as coronavirus disease 2019 (COVID-19), has posed a serious threat to public health across the globe, calling for the development of effective diagnostic markers and therapeutics. Here, we report a highly reliable severity diagnostic biomarker, acetylated 676th lysine transforming growth factor-beta-induced protein (TGFBIp K676Ac). TGFBIp K676Ac was consistently elevated in the blood of patients with SARS-CoV-2 pneumonia (n = 113), especially in patients in the intensive care unit (ICU) compared to non-ICU patients. Patients' blood samples showed increased cytokines and lymphopenia, which are exemplary indicators of SARS-CoV-2 pneumonia. Treatment with TGFBIp neutralizing antibodies suppressed the cytokine storm. The increased level of TGFBIp K676Ac in ICU patients suggests the promise of this protein as a reliable severity diagnostic biomarker for severe SARS-CoV-2 disease.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/diagnosis , Cytokine Release Syndrome/diagnosis , Extracellular Matrix Proteins/immunology , Leukocytes, Mononuclear/immunology , Pneumonia, Viral/diagnosis , Protein Processing, Post-Translational , Respiratory Insufficiency/diagnosis , Transforming Growth Factor beta/immunology , Acetylation , Antibodies, Neutralizing/pharmacology , Betacoronavirus/immunology , Biomarkers/blood , COVID-19 , Case-Control Studies , Coronavirus Infections/blood , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Gene Expression , Humans , Intensive Care Units , Leukocyte Count , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/virology , Lung/blood supply , Lung/drug effects , Lung/pathology , Lung/virology , Lysine/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Primary Cell Culture , Prognosis , Respiratory Insufficiency/blood , Respiratory Insufficiency/immunology , Respiratory Insufficiency/pathology , SARS-CoV-2 , Severity of Illness Index , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics
14.
Drug Des Devel Ther ; 14: 3577-3587, 2020.
Article in English | MEDLINE | ID: mdl-32943845

ABSTRACT

BACKGROUND: As a substrate of apoER2, Reelin has been verified to exert neuroprotection by preventing memory impairment. Pinocembrin is the most abundant natural flavonoid found in propolis, and it has been used to exert neuroprotection, blood-brain barrier protection, anti-oxidation, and inflammation diminishing, both in vitro and in vivo. However, the roles and molecular mechanisms of pinocembrin in neurobehavioral outcomes and neuronal repair after vascular dementia are still under investigation. PURPOSE: To explore the role of pinocembrin in the involvement of the Reelin-dab1 signaling pathway in improving memory impairment, both in cell culture and animals experiments. MATERIAL AND METHODS: Behavioral tests were conducted on day 48 to confirm the protection of pinocembrin against cognitive impairment. Cell and molecular biology experiments demonstrated that the Reelin-dab1 pathway mediates the underlying mechanism of cognitive improvement by pinocembrin. RESULTS: It was showed that pinocembrin alleviated learning and memory deficits induced by vascular dementia, by inducing the expression of Reelin, apoER2, and p-dab1 in the hippocampus. The expression of Reelin and p-dab1 was both inhibited following Reelin RNA interference in SH-SY5Y prior to oxygen glucose deprivation (OGD) injury, suggesting that Reelin played a core role in pinocembrin's effect on OGD in vitro. CONCLUSION: Pinocembrin improves the cognition via the Reelin-dab1 signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cognitive Dysfunction/drug therapy , Dementia, Vascular/drug therapy , Extracellular Matrix Proteins/metabolism , Flavanones/pharmacology , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Behavior, Animal/drug effects , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Dementia, Vascular/metabolism , Dementia, Vascular/pathology , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/genetics , Humans , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction/drug effects
15.
Exp Cell Res ; 397(1): 112301, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32979364

ABSTRACT

Vascular calcification (VC) is a major risk factor for increasing cardiovascular morbidity and mortality in patients with chronic kidney disease (CKD). Indoxyl sulfate (IS), a representative uremic toxin, is closely associated with VC in CKD patients. Matrix Gla protein (MGP) plays pivotal role in VC as a calcification inhibitor. The aim of this work was to explore whether MGP was involved in IS-induced VC. Here, we demonstrated the role of MGP in the IS-induced osteogenic differentiation of human aortic smooth muscle cells (HASMCs). The methods included Von Kossa staining, immunohistochemistry, Alizarin Red staining, quantitative real-time PCR and western blotting. MGP was decreased in calcified arteries both in CKD patients and rats. In vitro, IS suppressed MGP expression in HASMCs by activating ROS/NF-κB signaling in parallel with osteogenic differentiation, which was mitigated by inhibiting ROS and NF-κB with diphenyleneiodonium and Bay11-7082. Further investigation showed that IS induced NF-κB-responsive microRNA (miR)-155-5p mediating MGP downregulation. Overexpression of miR-155-5p with mimics aggravated IS-induced MGP reduction and osteogenic differentiation. In contrast, these conditions were diminished by silencing miR-155-5p. We demonstrate that IS promotes the HASMCs phenotype switch by suppressing MGP expression via ROS/NF-κB/miR-155-5p signaling and provide a new insight for the pathogenesis of IS-induced VC.


Subject(s)
Calcium-Binding Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/antagonists & inhibitors , Gene Expression Regulation/drug effects , Indican/pharmacology , Muscle, Smooth, Vascular/pathology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/complications , Vascular Calcification/pathology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , MicroRNAs/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , NF-kappa B/genetics , Osteogenesis , Rats , Rats, Sprague-Dawley , Vascular Calcification/etiology , Vascular Calcification/metabolism , Matrix Gla Protein
16.
Cells ; 9(9)2020 09 22.
Article in English | MEDLINE | ID: mdl-32972039

ABSTRACT

Tumor growth and invasion occurs through a dynamic interaction between cancer and stromal cells, which support an aggressive niche. MicroRNAs are thought to act as tumor messengers to "corrupt" stromal cells. We previously demonstrated that miR-9, a known metastamiR, is released by triple negative breast cancer (TNBC) cells to enhance the transition of normal fibroblasts (NFs) into cancer-associated fibroblast (CAF)-like cells. EGF containing fibulin extracellular matrix protein 1 (EFEMP1), which encodes for the ECM glycoprotein fibulin-3, emerged as a miR-9 putative target upon miRNA's exogenous upmodulation in NFs. Here we explored the impact of EFEMP1 downmodulation on fibroblast's acquisition of CAF-like features, and how this phenotype influences neoplastic cells to gain chemoresistance. Indeed, upon miR-9 overexpression in NFs, EFEMP1 resulted downmodulated, both at RNA and protein levels. The luciferase reporter assay showed that miR-9 directly targets EFEMP1 and its silencing recapitulates miR-9-induced pro-tumoral phenotype in fibroblasts. In particular, EFEMP1 siRNA-transfected (si-EFEMP1) fibroblasts have an increased ability to migrate and invade. Moreover, TNBC cells conditioned with the supernatant of NFs transfected with miR-9 or si-EFEMP1 became more resistant to cisplatin. Overall, our results demonstrate that miR-9/EFEMP1 axis is crucial for the conversion of NFs to CAF-like cells under TNBC signaling.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cell Transformation, Neoplastic/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Transformed , Cell Movement/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cisplatin/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Mice, SCID , MicroRNAs/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Xenograft Model Antitumor Assays
17.
Oncol Rep ; 44(5): 2143-2151, 2020 11.
Article in English | MEDLINE | ID: mdl-32901854

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is one of the most lethal gynecological malignancies; however, the precise molecular mechanisms have not been fully characterized. Fibulin­5 (FBLN­5) is an extracellular matrix (ECM) glycoprotein, and plays a crucial role in maintaining the stability of ECM structures, regulating cell proliferation and tumorigenesis. In the present study, the expression of FBLN­5, as determined by western blot analysis and immunohistochemistry, was significantly increased in normal fallopian tube (FT) samples compared with that in HGSOC samples, and decreased FBLN5 expression was associated with unfavorable prognosis of HGSOC. Functional characterization revealed that FBLN5 overexpression significantly inhibited migration, invasion and proliferation abilities of ovarian cancer cells in vitro. Furthermore, micro (mi)RNA­27a­3p (miR­27a­3p) was revealed to be increased in HGSOC, and dual­luciferase reporter assay indicated that miR­27a­3p was functioned as a negative regulator of FBLN5 by directly binding with its 3'­untranslated region. Collectively, FBLN5 expression was associated with prognosis, proliferation, and metastasis in HGSOC. We hypothesized that FBLN5 was targeted by miR­27a­3p and may serve as a biomarker and provide a new therapeutic approach for the treatment of HGSOC.


Subject(s)
Cystadenocarcinoma, Serous/metabolism , Extracellular Matrix Proteins/metabolism , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Adult , Aged , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease Progression , Extracellular Matrix Proteins/antagonists & inhibitors , Female , Humans , MicroRNAs/genetics , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , Survival Rate
18.
Biochem Biophys Res Commun ; 527(1): 283-288, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446381

ABSTRACT

Lysyl oxidase (LOX) is involved in fibrosis by catalyzing collagen cross-linking. Previous work observed that Triptolide (TPL) alleviated radiation-induced pulmonary fibrosis (RIPF), but it is unknown whether the anti-RIPF effect of TPL is related to LOX. In a mouse model of RIPF, we found that LOX persistently increased in RIPF which was significantly lowered by TPL. Excessive LOX aggravated fibrotic lesions in RIPF, while LOX inhibition mitigated RIPF. Irradiation enhanced the transcription and synthesis of LOX by lung fibroblasts through IKKß/NFκB activation, and siRNA knockdown IKKß largely abolished LOX production. By interfering radiation induced IKKß activation, TPL prevented NFκB nuclear translocation and DNA binding, and potently decreased LOX synthesis. Our results demonstrate that the anti-RIPF effect of TPL is associated with reduction of LOX production which mediated by inhibition of IKKß/NFκB pathway.


Subject(s)
Diterpenes/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , I-kappa B Kinase/antagonists & inhibitors , Phenanthrenes/pharmacology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Pulmonary Fibrosis/drug therapy , Radiation Injuries/drug therapy , Animals , Diterpenes/administration & dosage , Dose-Response Relationship, Drug , Epoxy Compounds/administration & dosage , Epoxy Compounds/pharmacology , Extracellular Matrix Proteins/biosynthesis , Female , I-kappa B Kinase/metabolism , Injections, Intravenous , Mice , Mice, Inbred C57BL , Molecular Structure , Phenanthrenes/administration & dosage , Protein-Lysine 6-Oxidase/biosynthesis , Pulmonary Fibrosis/metabolism , Radiation Injuries/metabolism , Structure-Activity Relationship
19.
Int J Oncol ; 57(1): 183-196, 2020 07.
Article in English | MEDLINE | ID: mdl-32319595

ABSTRACT

Cyclovirobuxine D (CVB­D) is an alkaloid, which is mainly derived from Buxus microphylla. It has been reported that CVB­D has positive effects on breast cancer, gastric cancer and other malignant tumors. However, to the best of our knowledge, there are no reports regarding the effects of CVB­D on colorectal cancer (CRC). The purpose of the present study was to determine the anticancer effects of CVB­D and further elucidate its molecular mechanism(s). DLD­1 and LoVo cell lines were selected to evaluate the antitumor effect of CVB­D. Cytotoxicity, viability and proliferation were evaluated by the MTT and colony formation assays. Flow cytometry was used to detect the effects on apoptosis and the cell cycle in CVB­D­treated CRC cells. The migration and invasion abilities of CRC cells were examined by wound healing and Transwell assays. In addition, RNA sequencing, bioinformatics analysis and western blotting were performed to investigate the target of drug action and clarify the molecular mechanisms. A xenograft model was established using nude mice, and ultrasound was employed to assess the preclinical therapeutic effects of CVB­D in vivo. It was identified that CVB­D inhibited the proliferation, migration, stemness, angiogenesis and epithelial­mesenchymal transition of CRC cells, and induced apoptosis and S­phase arrest. In addition, CVB­D significantly inhibited the growth of xenografts. It is notable that CVB­D exerted anticancer effects in CRC cells partly by targeting collagen triple helix repeat containing 1 (CTHRC1), which may be upstream of the AKT and ERK pathways. CVB­D exerted anticancer effects through the CTHRC1­AKT/ERK­Snail signaling pathway. Targeted therapy combining CTHRC1 with CVB­D may offer a promising novel therapeutic approach for CRC treatment.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colonic Neoplasms/drug therapy , Drugs, Chinese Herbal/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Drugs, Chinese Herbal/therapeutic use , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/genetics , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , RNA-Seq , Snail Family Transcription Factors/metabolism , Xenograft Model Antitumor Assays
20.
Sci Rep ; 10(1): 6358, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286390

ABSTRACT

Vaginal delivery with obstetrical trauma is a risk factor for pelvic organ prolapse later in life. Loss of fibulin-5 (FBLN5), an elastogenesis-promoting cellular matrix protein, results in prolapse in mice. Here, we evaluated effects of pregnancy, parturition, and obstetrical injury on FBLN5 content, elastic fibers, biomechanics, and histomorphology of the vaginal wall in rats. Further, we analyzed the effects of actinonin, a protease inhibitor, on obstetrical injury of the vaginal wall. Vaginal FBLN5 decreased significantly in pregnancy, and injury resulted in further downregulation. Stiffness of the vaginal wall decreased 82% in pregnant rats and 74% (p = 0.019) with injury relative to uninjured vaginal delivery controls at 3d. Actinonin ameliorated loss of FBLN5, rescued injury-induced loss of elastic fibers and biomechanical properties after parturition, and reduced the area of injury 10-fold. We conclude that pregnancy and parturition have a profound impact on vaginal FBLN5 and biomechanics of the vaginal wall. Further, obstetrical injury has significant deleterious impact on recovery of the vaginal wall from pregnancy. Actinonin, a non-specific matrix metalloprotease inhibitor, improved recovery of the parturient vaginal wall after obstetrical injury.


Subject(s)
Extracellular Matrix Proteins/genetics , Pelvic Organ Prolapse/drug therapy , Recombinant Proteins/genetics , Vagina/drug effects , Wound Healing/genetics , Animals , Delivery, Obstetric/adverse effects , Extracellular Matrix Proteins/antagonists & inhibitors , Female , Humans , Hydroxamic Acids/pharmacology , Obstetric Surgical Procedures/adverse effects , Pelvic Organ Prolapse/etiology , Pelvic Organ Prolapse/genetics , Pregnancy , Pregnancy Complications/drug therapy , Pregnancy Complications/physiopathology , Pregnancy Complications/prevention & control , Protease Inhibitors/pharmacology , Rats , Risk Factors , Uterine Prolapse/drug therapy , Uterine Prolapse/physiopathology , Uterine Prolapse/prevention & control , Vagina/physiopathology , Vagina/surgery , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL