Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 124
1.
Chem Biol Drug Des ; 103(1): e14421, 2024 01.
Article En | MEDLINE | ID: mdl-38230771

Dihydromyricetin (DHM) is a bioactive flavonoid extracted from Hovenia dulcis, which has various activities. In the present study, the molecular mechanism of dihydromyricetin (DHM) in relieving liver cirrhosis was investigated through network pharmacology and experimental verification. The cell model was induced by TGF-ß1 activating the human hepatic stellate cell line (HSC; LX-2). The protein levels of α-SMA, collagen I, and collagen III and pathway-related proteins within LX-2 cells were detected using Western blot. EdU staining was conducted to detect cell proliferation. Immunofluorescence staining was performed to detect the expression levels of α-SMA and collagen I. Next, the drug targets of DHM were screened from the PubChem database. The differentially expressed genes in the liver cirrhosis dataset GSE14323 were identified. The expression of the identified drug targets in LX-2 cells was verified using qRT-PCR. The results showed that TGF-ß1 treatment notably increased LX-2 cell viability, promoted cell proliferation, and elevated α-SMA, collagen I, and collagen III protein contents. DHM treatment could partially eliminate TGF-ß1 effects, as evidenced by the inhibited cell viability and proliferation and reduced α-SMA, collagen I, and collagen III contents. After network pharmacology analysis, nine differentially expressed target genes (MMP2, PDGFRB, PARP1, BCL2L2, ABCB1, TYR, CYP2E1, SQSTM1, and IL6) in liver cirrhosis were identified. According to qRT-PCR verification, DHM could inhibit the expression of MMP2, PDGFRB, PARP1, CYP2E1, SQSTM1, and IL6, and enhance ABCB1 expression levels within LX-2 cells. Moreover, DHM inhibited mTOR and MAPK signaling pathways in TGF-ß1-induced HSCs. In conclusion, DHM could inhibit HSC activation, which may be achieved via acting on MMP2, PDGFRB, PARP1, CYP2E1, SQSTM1, IL6, and ABCB1 genes and their downstream signaling pathways, including mTOR and MAPK signaling pathway.


Flavonols , Matrix Metalloproteinase 2 , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Cytochrome P-450 CYP2E1/metabolism , Interleukin-6/metabolism , Network Pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/therapeutic use , Sequestosome-1 Protein/metabolism , Liver Cirrhosis/drug therapy , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I/therapeutic use , TOR Serine-Threonine Kinases/metabolism
2.
Curr Drug Targets ; 25(2): 135-148, 2024.
Article En | MEDLINE | ID: mdl-38213165

BACKGROUND: Astragali Radix (AR) has a long history as a traditional Chinese medicine for anti-osteoporosis (OP) treatment. The aim of the study was to explore the effect and optimal regimens of AR and its main ingredients (IAR) in OP treatment. METHODS: Eligible animal studies were searched in seven databases (PubMed, Web of Science, MEDLINE, SciELO Citation Index, Cochrane Library, China National Knowledge Infrastructure and Wanfang). The primary outcomes were bone metabolic indices. The secondary outcome measure was the anti-OP mechanism of IAR. RESULTS: 21 studies were enrolled in the study. The primary findings of the present article illustrated that IAR could significantly increase the bone mineral density (BMD), bone volume over the total volume, trabecular number, trabecular thickness, bone maximum load and serum calcium, while trabecular separation and serum C-terminal telopeptide of type 1 collagen were remarkably decreased (P < 0.05). In subgroup analysis, the BMD in the long treatment group (≥ 10 weeks) showed better effect size than the short treatment group (< 10 weeks) (P < 0.05). Modeling methods and animal sex were factors affecting serum alkaline phosphatase and osteocalcin levels. CONCLUSION: The findings suggest the possibility of developing IAR as a drug for the treatment of OP. IAR with longer treatment time may achieve better effects regardless of animal strain and age.


Osteoporosis , Animals , Osteoporosis/drug therapy , Bone Density , Collagen Type I/therapeutic use , Bone and Bones , Models, Animal
3.
Diabetes Metab J ; 48(1): 72-82, 2024 Jan.
Article En | MEDLINE | ID: mdl-38173367

BACKGRUOUND: Renal fibrosis is characterized by the accumulation of extracellular matrix proteins and interstitial fibrosis. Alantolactone is known to exert anticancer, anti-inflammatory, antimicrobial and antifungal effects; however, its effects on renal fibrosis remains unknown. Here, we investigated whether alantolactone attenuates renal fibrosis in mice unilateral ureteral obstruction (UUO) and evaluated the effect of alantolactone on transforming growth factor (TGF) signaling pathway in renal cells. METHODS: To evaluate the therapeutic effect of alantolactone, cell counting kit-8 (CCK-8) assay, histological staining, Western blot analysis, and real-time quantitative polymerase chain reaction were performed in UUO kidneys in vivo and in TGF-ß-treated renal cells in vitro. RESULTS: Alantolactone (0.25 to 4 µM) did not affect the viability of renal cells. Mice orally administered 5 mg/kg of alantolactone daily for 15 days did not show mortality or liver toxicity. Alantolactone decreased UUO-induced blood urea nitrogen and serum creatinine levels. In addition, it significantly alleviated renal tubulointerstitial damage and fibrosis and decreased collagen type I, fibronectin, and α-smooth muscle actin (α-SMA) expression in UUO kidneys. In NRK-49F cells, alantolactone inhibited TGF-ßstimulated expression of fibronectin, collagen type I, plasminogen activator inhibitor-1 (PAI-1), and α-SMA. In HK-2 cells, alantolactone inhibited TGF-ß-stimulated expression of collagen type I and PAI-1. Alantolactone inhibited UUO-induced phosphorylation of Smad3 in UUO kidneys. In addition, it not only decreased TGF-ß secretion but also Smad3 phosphorylation and translocation to nucleus in both kidney cell lines. CONCLUSION: Alantolactone improves renal fibrosis by inhibiting the TGF-ß/Smad3 signaling pathway in obstructive nephropathy. Thus, alantolactone is a potential therapeutic agent for chronic kidney disease.


Kidney Diseases , Lactones , Sesquiterpenes, Eudesmane , Ureteral Obstruction , Mice , Animals , Fibronectins/pharmacology , Fibronectins/therapeutic use , Plasminogen Activator Inhibitor 1/pharmacology , Plasminogen Activator Inhibitor 1/therapeutic use , Collagen Type I/pharmacology , Collagen Type I/therapeutic use , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction , Fibrosis
4.
Folia Histochem Cytobiol ; 61(4): 231-243, 2023.
Article En | MEDLINE | ID: mdl-38073317

INTRODUCTION: Liver fibrosis is the damage repair response following chronic liver diseases. Activated hepatic stellate cells (HSCs) are the main extracellular matrix (ECM)-producing cells and key regulators in liver fibrosis. Periplaneta americana shows prominent antifibrotic effects in liver fibrosis; however, the underlying mechanisms remain undetermined. This study aimed to elucidate the therapeutic effects of P. americana extract (PA-B) on liver fibrosis based on the regulation of the TGF-ß1/Smad signal pathway. MATERIAL AND METHODS: HSCs and Sprague Dawley rats were treated with TGF-ß1 and CCl4, respectively, to establish the hepatic fibrosis model in vitro and in vivo. The effect of PA-B on liver rat fibrosis was evaluated by biochemical (serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), hyaluronic acid (HA), laminin (LN), collagen type IV (Col-IV), pro-collagen type III (PC-III)) and histological examinations. Further, fibrogenic markers expression of alpha smooth muscle actin (α-SMA), collagen type I (Col-I), and collagen type III (Col-III), and the TGF-ß1/Smad pathway-related factors were assessed by immunofluorescence (IF), real time quantitative polymerase chain reaction (RT-qPCR), and western blotting (WB). RESULTS: Treatment of HSC-T6 cells with PA-B suppressed the expression of α-SMA, Col-I, and Col-III, downregulated the expression of TGF-ß1 receptors I and II (TßR I and TßR II, respectively), Smad2, and Smad3, and upregulated Smad7 expression. PA-B mitigates pathologic changes in the rat model of liver fibrosis, thus alleviating liver index, and improving liver function and fibrosis indices. The effects of PA-B on the expression of α-SMA, Col-I, Col-III, TßR I, TßR II, Smad2, Smad3, and Smad7 were consistent with the in vitro results, including reduced TGF-ß1 expression. CONCLUSIONS: The therapeutic effect of PA-B on liver fibrosis might involve suppression of the secretion and expression of TGF-ß1, regulation of the TGF-ß1/Smad signaling pathway, and inhibition of collagen production and secretion.


Periplaneta , Transforming Growth Factor beta1 , Rats , Animals , Transforming Growth Factor beta1/metabolism , Periplaneta/metabolism , Collagen Type III/metabolism , Rats, Sprague-Dawley , Smad Proteins/metabolism , Smad Proteins/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Signal Transduction , Collagen Type I/metabolism , Collagen Type I/pharmacology , Collagen Type I/therapeutic use
5.
Cell Mol Biol (Noisy-le-grand) ; 69(13): 102-105, 2023 Dec 10.
Article En | MEDLINE | ID: mdl-38158681

The objective of this study was to analyze the effect of curcumin (Cur) on pulmonary fibrosis (PF), so as to provide new clinical evidence for future PF treatment. To achieve these goals, the researchers set up bought human lung fibroblasts MRC-5 as a control group without treatment, a model group for PF cell modeling, and an intervention group for Cur intervention after PF modeling. Cell proliferation capacity and cellular TGF-ß1, α-SMA, Collagen I, Collagen III, Bax, N-cadherin and E-cadherin protein expression were determined. The results show that markedly enhanced cell proliferation capacity and TGF-ß1, α-SMA, Collagen I and Collagen III protein levels were observed in the model group, while the cell activity and fibrosis degree in the intervention group were significantly decreased compared with the model group (P<0.05). In addition, the intervention group exhibited lower N-cadherin and Bax with higher E-cadherin than the model group (P<0.05). In addition, the team found that the inflammatory response and oxidative stress were also more significantly improved in the intervention group (P<0.05). These experimental results tell us that Cur can ameliorate the fibrotic process of PF by inhibiting the activity of MRC-5.


Curcumin , Pulmonary Fibrosis , Humans , Pulmonary Fibrosis/drug therapy , Transforming Growth Factor beta1/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Curcumin/metabolism , bcl-2-Associated X Protein/metabolism , Fibrosis , Lung/pathology , Collagen/metabolism , Fibroblasts/metabolism , Collagen Type I/metabolism , Collagen Type I/pharmacology , Collagen Type I/therapeutic use , Cadherins/metabolism
6.
J Appl Biomed ; 21(1): 15-22, 2023 04.
Article En | MEDLINE | ID: mdl-37016776

Myocardial fibrosis is the most serious complication of viral myocarditis (VMC). This study aimed to investigate the therapeutic benefits and underlying mechanisms of lentivirus-mediated human tissue kallikrein gene transfer in myocardial fibrosis in VMC mice. We established VMC mouse model via intraperitoneal injection with Coxsackie B3 virus. The effect was then assessed after treatment with vehicle, the empty lentiviral vectors (EZ.null), and the vectors expressing hKLK1 (EZ.hKLK1) via tail vein injection for 30 days, respectively. The results showed that administering EZ.hKLK1 successfully induced hKLK1 overexpression in mouse heart. Compared with EZ.null treatment, EZ.hKLK1 administration significantly reduced the heart/weight ratio, improved cardiac function, and ameliorated myocardial inflammation in VMC mice, suggesting that hKLK1 overexpression alleviates VMC in mice. EZ.hKLK1 administration also significantly abrogated the increased myocardial collagen content, type I/III collagen ratio, TGF-ß1 mRNA and protein expression in VMC mice, suggesting that hKLK1 overexpression reduces collagen accumulation and blunts TGF-ß1 signaling in the hearts of VMC mice. In conclusion, our results suggest that hKLK1 alleviates myocardial fibrosis in VMC mice, possibly by downregulating TGF-ß1 expression.


Cardiomyopathies , Coxsackievirus Infections , Myocarditis , Mice , Humans , Animals , Myocarditis/drug therapy , Myocarditis/metabolism , Transforming Growth Factor beta1/genetics , Collagen/metabolism , Collagen/therapeutic use , Collagen Type I/genetics , Collagen Type I/therapeutic use , Coxsackievirus Infections/therapy , Coxsackievirus Infections/drug therapy , Fibrosis , Collagen Type III/therapeutic use
7.
Anal Chim Acta ; 1239: 340695, 2023 Jan 25.
Article En | MEDLINE | ID: mdl-36628763

Appropriate follow-up after treatment initiation in patients with osteoporosis is challenging. Serum biomarkers may offer more efficient monitoring of bone mineral density (BMD) than the currently used dual X-ray absorptiometry; however, significant changes in BMD often occur over at least 12 months. During teriparatide treatment for osteoporosis, monitoring with markers such as procollagen type I propeptide (PINP), which is derived from osteoblasts, can provide clinically useful information for disease management. However, rapid and cost-effective methods for detecting serum PINP are lacking, necessitating a point-of-care test (POCT) for enhanced follow-up efficiency in osteoporosis management. For the quantitative detection of PINP, we developed a high-sensitivity lateral flow immunoassay with a stacking pad (sLFIA). We established a calibration equation based on the test line/control line ratio obtained from our PINP sLFIA results of various nonspiked serum samples to calculate the PINP concentrations in 40 serum samples and compared the result with those obtained using a fully automated electrochemiluminescence immunoassay. PINP concentrations between these two methods exhibited excellent correlation (R = 0.991). In addition, we assessed the serum PINP concentrations of patients with osteoporosis treated with teriparatide. At the 3-month follow-up, their PINP levels were nearly twice as high as those at baseline, thus implying that our method can be used for osteoporosis treatment monitoring. Our findings thus indicate that the PINP sLFIA can serve as a POCT for monitoring medication response and managing osteoporosis.


Osteoporosis , Teriparatide , Humans , Teriparatide/therapeutic use , Peptide Fragments , Osteoporosis/diagnostic imaging , Osteoporosis/drug therapy , Bone Density/physiology , Collagen Type I/therapeutic use , Procollagen/therapeutic use , Biomarkers , Immunoassay
8.
RMD Open ; 8(2)2022 12.
Article En | MEDLINE | ID: mdl-36585217

OBJECTIVE: To investigate correlations between biomarkers of bone remodelling and extracellular matrix turnover with baseline disease activity and treatment response in patients with early rheumatoid arthritis (RA). METHODS: Assessing Very Early Rheumatoid arthritis Treatment-2 (AVERT-2; NCT02504268) included disease-modifying antirheumatic drug-naive, anti-citrullinated protein antibody (ACPA)-positive patients randomised to weekly subcutaneous abatacept+methotrexate (MTX) or abatacept placebo+MTX for 56 weeks. This post hoc exploratory subanalysis assessed the association between baseline disease activity and eight biomarkers (Spearman's correlation coefficient), and whether baseline biomarkers (continuous or categorical variables) could predict treatment response at weeks 24 and 52 (logistic regression). RESULTS: Patient characteristics were similar between overall (n=752) and biomarker subgroup (n=535) populations and across treatments. At baseline, neoepitopes of matrix metalloproteinase-mediated degradation products of types III and IV collagen and of C reactive protein (CRP) showed the greatest correlations with disease activity; cross-linked carboxy-terminal telopeptide of type I collagen (CTX-I) showed weak correlation. Only CTX-I predicted treatment response; baseline CTX-I levels were significantly associated with achieving Simplified Disease Activity Index remission and Disease Activity Score in 28 joints (DAS28 (CRP)) <2.6 (weeks 24 and 52), and American College of Rheumatology 70 response (week 52), in patients treated with abatacept+MTX but not abatacept placebo+MTX. CTX-I predicted significant differential response between arms for DAS28 (CRP) <2.6 (week 24). Treatment differences were greater for abatacept+MTX in patients with medium/high versus low baseline CTX-I. CONCLUSION: In MTX-naive, ACPA-positive patients with early RA, baseline CTX-I predicted treatment response to abatacept+MTX but not abatacept placebo+MTX.


Antirheumatic Agents , Arthritis, Rheumatoid , Humans , Methotrexate/therapeutic use , Abatacept/therapeutic use , Collagen Type I/therapeutic use , Anti-Citrullinated Protein Antibodies , Treatment Outcome , Drug Therapy, Combination , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/therapeutic use , C-Reactive Protein , Biomarkers
9.
Post Reprod Health ; 28(3): 149-157, 2022 Sep.
Article En | MEDLINE | ID: mdl-35938207

OBJECTIVE: Evaluate the effects of ultra-low-dose hormone therapy (Ultra-LD HT) with 17ß-estradiol 0.5 mg and norethisterone acetate 0.1 mg (E2 0.5/NETA 0.1) versus placebo on bone turnover markers (BTM) in postmenopausal women. STUDY DESIGN: A multicenter, double-blind, randomized, placebo-controlled study was performed with 107 participants who received one tablet daily of E2 0.5/NETA 0.1 or placebo for 24-weeks. Bone formation markers-N-terminal propeptide of type I procollagen (PINP) and Bone-specific alkaline phosphatase (BSAP), and bone resorption markers-C-telopeptide of type I collagen (CTX-I) and N-telopeptide crosslinked of type I collagen (NTX) were assessed before and at 12 and 24-weeks of treatment. RESULTS: Women treated with E2 0.5/NETA 0.1 had a significant reduction in the PINP marker from baseline (58.49 ± 21.12 µg/L) to week 12 (48.31 ± 20.99 µg/L) and week 24 (39.16 ± 16.50 µg/L). Placebo group, the PINP marker did not differ significantly. The analysis of the BSAP indicated a significant increase in the placebo group (13.8 ± 5.09 µg/L and 16.29 ± 4.3 µg/L, at baseline and week 24, respectively), whereas in the treatment group the values did not change. The analysis of the NTX marker showed a significant reduction only in the treatment group (43.21 ± 15.26 nM/mM and 33.89 ± 14.9 nM/mM, at baseline and week 24, respectively). CTX-I had a significant decrease in the treatment group from baseline (0.3 ± 0.16 ng/L) to week 12 (0.21 ± 0.14 ng/L) and week 24 (0.21 ± 0.12 ng/L). CONCLUSION: Women receiving E2 0.5/NETA 0.1 experienced reductions in bone resorption and formation markers, an expected effect during the anti-resorptive therapy, suggesting a protective bone effect with the Ultra-LD HT.


Bone Resorption , Osteoporosis, Postmenopausal , Alkaline Phosphatase/pharmacology , Alkaline Phosphatase/therapeutic use , Biomarkers/analysis , Bone Density , Bone Remodeling , Bone Resorption/drug therapy , Bone Resorption/prevention & control , Collagen Type I/pharmacology , Collagen Type I/therapeutic use , Double-Blind Method , Estradiol , Female , Humans , Norethindrone Acetate/pharmacology , Osteoporosis, Postmenopausal/drug therapy , Postmenopause
10.
Int Wound J ; 18(5): 598-607, 2021 Oct.
Article En | MEDLINE | ID: mdl-33666348

Hypertrophic scar (HS) is a fibrotic skin disease characterised by over-productive collagen and excessive inflammatory reaction, which can be functionally and cosmetically problematic. A scar-prone constitute will accelerate HS formation and functional disorder, which deserves systemic therapy with oral medicine. To examine the oral therapeutic effectiveness on HS with convincing evidence of gross view and histological improvement, a rabbit ear HS model was employed with oral administration of asiaticoside (AS) at the doses of 12 and 24 mg kg-1 d-1 daily for 60 consecutive days. Gross observation and histological findings showed that oral AS treatment could significantly inhibit HS formation in a dose dependent manner. Semi-quantification of scar elevation index at days 7, 15, 30, and 60, and quantitative polymerase chain reaction at days 30 and 60 also provided the evidences of reduced scar thickness and inhibited fibrotic gene expressions of collagens I, III, TGF-ß1, interleukins 1ß, 6 and 8, and enhanced gene expression of SMAD 7 and PPAR-γ with a dose-dependent manner. These results indicated that AS is likely to serve as a systemic therapeutic agent of HS treatment for those who may have scar-prone constitute via anti-inflammation, inhibiting fibrotic process, and enhancing matrix degradation.


Cicatrix, Hypertrophic , Triterpenes , Administration, Oral , Animals , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/pathology , Collagen Type I/therapeutic use , Fibroblasts/pathology , Rabbits , Triterpenes/therapeutic use
11.
J Comp Eff Res ; 9(10): 691-703, 2020 07.
Article En | MEDLINE | ID: mdl-32476449

Aim: Determine the effectiveness of purified native type I collagen matrix plus polyhexamethylene biguanide antimicrobial (PCMP) on cutaneous wounds. Materials & methods: A prospective cohort study of 307 patients (67 venous leg ulcers, 62 diabetic foot ulcers, 45 pressure ulcers, 54 post-surgical wounds and 79 other wounds) was conducted. Results: Cox wound closure for PCMP was 73% at week 32. The median time to wound closure was 17 weeks (Kaplan-Meier). The incidence of PCMP-treated wounds showing >60% reductions in areas, depths and volumes were 81, 71 and 85%, respectively. Conclusion: PCMP demonstrated clinically meaningful benefits to patients with various types of cutaneous wounds. Clinical Trial registration number: NCT03286452.


Anti-Infective Agents/therapeutic use , Biguanides/therapeutic use , Collagen Type I/therapeutic use , Wound Healing/drug effects , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Pragmatic Clinical Trials as Topic , Prospective Studies
13.
Arch Orthop Trauma Surg ; 139(1): 99-106, 2019 Jan.
Article En | MEDLINE | ID: mdl-30121776

INTRODUCTION: Cartilage defects of the knee remain a challenging problem in orthopedic surgery despite the ongoing improvements in regenerative procedures such as the autologous chondrocyte transplantation. Due to the lack of donor-site morbidity and the single-stage procedure cell-free scaffolds are an interesting alternative to cell-based procedures. But as currently mid- and long-term data are lacking, the aim of the present study was to present mid-term clinical, radiological and histological results of a cell-free collagen type I scaffolds for cartilage repair. MATERIALS AND METHODS: Twenty-eight patients were followed prospectively. Clinical evaluation using patient-reported outcome measures (KOOS, IKDC; VAS for pain, Tegner score for activity) as well as radiologic evaluation of the repair tissue (MOCART) was performed at 1 year, 2 years and 5 years. Histologic evaluation of the repair tissue was done in case of revision surgery using the ICRS II score for human cartilage repair. RESULTS: In these large cartilage defects with a mean defect size of 3.7 ± 1.9 cm2, clinical failure necessitating revision surgery was seen in 5 of 28 patients (18%). While the remaining patients showed good-to-excellent clinical results (KOOS, IKDC, VAS, Tegner), the radiologic appearance of the repair tissue showed a reduction of the MOCART score between the 2- and 5-year follow-up. Histologic evaluation of the repair tissue showed a cartilage-like appearance with no signs of inflammation or cell death but an overall medium tissue quality according to the ICRS II Score. CONCLUSION: The use of this cell-free collagen type I scaffold for large defects showed increased wear of the repair tissue and clinical failure in 18% of cases at 5-year follow-up.


Cartilage, Articular , Collagen Type I , Knee Injuries/surgery , Knee Prosthesis/adverse effects , Prosthesis Failure , Cartilage, Articular/injuries , Cartilage, Articular/surgery , Cell-Free System , Collagen Type I/adverse effects , Collagen Type I/therapeutic use , Humans , Prospective Studies
14.
Am J Physiol Endocrinol Metab ; 315(4): E650-E661, 2018 10 01.
Article En | MEDLINE | ID: mdl-29894201

Widespread use of pancreatic islet transplantation for treatment of type 1 diabetes (T1D) is currently limited by requirements for long-term immunosuppression, limited donor supply, and poor long-term engraftment and function. Upon isolation from their native microenvironment, islets undergo rapid apoptosis, which is further exacerbated by poor oxygen and nutrient supply following infusion into the portal vein. Identifying alternative strategies to restore critical microenvironmental cues, while maximizing islet health and function, is needed to advance this cellular therapy. We hypothesized that biophysical properties provided through type I oligomeric collagen macroencapsulation are important considerations when designing strategies to improve islet survival, phenotype, and function. Mouse islets were encapsulated at various Oligomer concentrations (0.5 -3.0 mg/ml) or suspended in media and cultured for 14 days, after which viability, protein expression, and function were assessed. Oligomer-encapsulated islets showed a density-dependent improvement in in vitro viability, cytoarchitecture, and insulin secretion, with 3 mg/ml yielding values comparable to freshly isolated islets. For transplantation into streptozotocin-induced diabetic mice, 500 islets were mixed in Oligomer and injected subcutaneously, where rapid in situ macroencapsulation occurred, or injected with saline. Mice treated with Oligomer-encapsulated islets exhibited rapid (within 24 h) diabetes reversal and maintenance of normoglycemia for 14 (immunocompromised), 90 (syngeneic), and 40 days (allogeneic). Histological analysis showed Oligomer-islet engraftment with maintenance of islet cytoarchitecture, revascularization, and no foreign body response. Oligomer-islet macroencapsulation may provide a useful strategy for prolonging the health and function of cultured islets and has potential as a subcutaneous injectable islet transplantation strategy for treatment of T1D.


Collagen Type I/therapeutic use , Diabetes Mellitus, Experimental/surgery , Diabetes Mellitus, Type 1/surgery , Graft Survival , Insulin Secretion , Islets of Langerhans Transplantation/methods , Islets of Langerhans/metabolism , Tissue Survival , Animals , Collagen Type I/ultrastructure , Culture Techniques , Dermis/chemistry , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Fibrillar Collagens/therapeutic use , In Vitro Techniques , Islets of Langerhans/anatomy & histology , Mice , Microscopy, Confocal , Polymerization , Swine
15.
Hum Gene Ther ; 29(8): 902-915, 2018 08.
Article En | MEDLINE | ID: mdl-29641324

A number of previous studies have indicated that the genetic variation at the collage type I alpha 1 (COLIA1) gene locus influences susceptibility to osteoporosis. However, seldom have studies reported the effect of gene delivery using an adenovirus vector carrying human recombinant COLIA1 cDNA on stimulating osteogenic activity of osteoblasts and enhancing fracture healing of ovariectomized rats. The current study was performed to demonstrate whether direct gene delivery using an adenovirus vector carrying human recombinant COLIA1 cDNA could stimulate osteogenic activity of osteoblast in vitro and enhance fracture healing of ovariectomized rats in vivo. In vitro, the tet-on system regulated COLIA1 gene adenovirus was constructed and transfected to osteoblasts. COLIA1 mRNA and collagen type I levels were assessed by reverse transcription polymerase chain reaction and enzyme-linked immunosorbent assay to determine whether adenovirus transfected successfully. Osteogenic activity of the osteoblasts was assessed by alkaline phosphatase activity, immunohistochemical staining, immunofluorescent staining, mineralized matrix formation, and extracellular calcium levels. In vivo, adenovirus-delivered COLIA1 gene was injected into the fracture site of the tibia in an ovariectomized rat model of osteoporosis, and bone callus condition was assessed to determine whether the COLIA1 gene could accelerate osteoporotic fracture healing. In vitro, the results showed that COLIA1 gene adenovirus transfection could increase osteoblast COLIA1 gene expression and collagen type I protein synthesis, increase alkaline phosphatase activity, and stimulate calcium nodules formation, which exhibited a direct osteogenic effect on the osteoblasts. In vivo, local injection of COLIA1 gene adenovirus increased collagen type I expression, restored bone mineral density, and accelerated fracture healing in ovariectomized rats, without increasing serum collagen type I and liver COLIA1 mRNA levels. This study suggests direct gene delivery using an adenovirus carrying human COLIA1 cDNA can stimulate the osteogenic activity of osteoblasts in vitro and enhance bone fracture healing in vivo. The tet-on system is an ideal gene regulatory system for effective and safe regulation of the therapeutic gene.


Collagen Type I/genetics , Genetic Therapy , Osteoporosis/therapy , Osteoporotic Fractures/therapy , Adenoviridae/genetics , Animals , Bone Density/genetics , Cell Differentiation/genetics , Collagen Type I/therapeutic use , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Humans , Osteoblasts/drug effects , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporotic Fractures/genetics , Osteoporotic Fractures/pathology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Tetracycline/therapeutic use , Transfection
16.
Wounds ; 30(3): 72-78, 2018 Mar.
Article En | MEDLINE | ID: mdl-29584602

INTRODUCTION: The management of chronic, nonhealing wounds in patients with multiple comorbidities continues to be a challenge for health care practitioners. Chronic wounds typically do not progress through the normal phases of wound healing and generally remain stagnant during the inflammatory phase, resulting in an increase in proteolytic enzymes with degradation of the extracellular matrix. Bacterial biofilm has been documented to be one of the main factors delaying wound healing, resulting in the prolongation of the inflammatory phase. OBJECTIVE: In order to control biofilm formation, sequester proteolytic enzymes, and provide a biocompatible scaffold to support healing, the investigators utilize a purified collagen matrix containing polyhexamethylene biguanide (PCMP) in a case series of 9 wounds on 8 patients with multiple comorbidities who did not respond to previous conventional or adjuvant therapy. MATERIALS AND METHODS: Wound etiologies included 3 pressure ulcers, 1 diabetic foot ulcer, 1 venous leg ulcer, 2 postsurgical wound dehiscences, 1 ulcer secondary to calciphylaxis, and 1 traumatic wound secondary to hematoma. The average wound size at the first PCMP application was 34.0 cm2, and the wounds were present for an average of 9.2 weeks prior to the first PCMP application. RESULTS: Patients received an average of 5.8 PCMP applications. Of the 6 wounds that healed, average time to closure from the first PCMP application was 10 weeks. The remaining 3 wounds demonstrated improved wound appearance with 100% granulation tissue and an average area reduction during PCMP treatment of 61.4%. CONCLUSIONS: This case series demonstrated that PCMP along with good wound care supported both wound closure and improvements in wound bed condition and area reduction on recalcitrant, nonhealing wounds of various etiologies.


Anti-Infective Agents, Local/therapeutic use , Biguanides/therapeutic use , Collagen Type I/therapeutic use , Skin Ulcer/therapy , Wound Healing/physiology , Aged , Calciphylaxis/complications , Chronic Disease , Diabetic Foot/therapy , Female , Humans , Male , Middle Aged , Pressure Ulcer/therapy , Skin Ulcer/etiology , Surgical Wound Dehiscence/therapy , Varicose Ulcer/therapy
17.
Sci Rep ; 8(1): 696, 2018 01 12.
Article En | MEDLINE | ID: mdl-29330408

Preterm premature rupture of membranes (pPROM) is a major cause of preterm birth. Recently, extracellular matrix-directed treatment is applied for wound healing. Here, we used a pregnant mouse model to test the efficacy of collagen type 1 gel for healing of the prematurely ruptured fetal membranes. Although injection of PBS into the ruptured fetal membranes resulted in 40% closure, injection of collagen type 1 improved closure rates to 90% within 72 h. Macrophages of the M2 wound healing phenotype were entrapped in the collagen layer. In primary human amnion mesenchymal cells, collagen type 1 gels activated collagen receptor discoidin domain receptor 2 (DDR2) to induce myosin light chain phosphorylation and migration of injured amnion mesenchymal cells. These findings define the mechanisms for matrix-directed therapeutics for pPROM.


Collagen Type I/therapeutic use , Fetal Membranes, Premature Rupture/drug therapy , Amnion/cytology , Animals , Cell Movement/drug effects , Collagen Type I/pharmacology , Discoidin Domain Receptor 2/antagonists & inhibitors , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Disease Models, Animal , Female , Gels/chemistry , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Pregnancy , RNA Interference , RNA, Small Interfering/metabolism
18.
Knee Surg Sports Traumatol Arthrosc ; 26(4): 1130-1136, 2018 Apr.
Article En | MEDLINE | ID: mdl-28324152

PURPOSE: Autologous matrix-induced chondrogenesis (AMIC) is a treatment for focal full-thickness cartilage defects combining microfracturing with an exogenous I/III collagen matrix (Chondro-Gide). The aim of the present study was to determine the 7 years outcomes of patients treated with the AMIC technique for knee chondral defects larger than 2 cm2. The hypothesis was that the positive short-term outcomes achieved in the previous series would not deteriorate at a 7-year follow-up. METHODS: Twenty-one patients treated with the AMIC technique were retrospectively analysed. Patients were assessed through the IKDC subjective knee evaluation questionnaire and the Lysholm scoring system. All patients underwent a complete imaging study including radiographs and magnetic resonance. The median defect size was found to be 4.3 (range 2.9-8) cm2. RESULTS: At a median follow-up of 7 (±1.4) years, the mean IKDC score improved from 31.7 (±8.9) points preoperatively, to 80.6 (±5.3) at the latest follow-up (p < 0.05). The mean Lysholm score improved from 38.8 (±12.4) points preoperatively to 72.6 (±19.5) points at the last follow-up (p < 0.05). At the last follow-up, 76.2% of patients were satisfied or extremely satisfied with their outcomes, while 66.6% of patients showed good quality repair tissue on magnetic resonance imaging. CONCLUSION: AMIC was found to be an effective method to treat full-thickness knee chondral defects larger than 2 cm2, with significant clinical and functional improvement maintained over a 7-year follow-up. LEVEL OF EVIDENCE: IV.


Cartilage, Articular/injuries , Cartilage, Articular/surgery , Chondrogenesis , Guided Tissue Regeneration/methods , Knee Injuries/surgery , Cartilage, Articular/physiology , Collagen Type I/therapeutic use , Collagen Type III/therapeutic use , Follow-Up Studies , Humans , Retrospective Studies , Treatment Outcome
19.
Acta Ortop Mex ; 31(4): 165-170, 2017.
Article En | MEDLINE | ID: mdl-29216691

INTRODUCTION: Multiple strategies have been developed looking for upgrading the consolidation rate of spine arthrodesis with autolog bone graft, but no evidence exists that adhesion with Collagen type 1 and polyvinylpyrrolidone (FibroquelMR) have application on this field. OBJECTIVE: Determine if collagen type 1 + Polyvinylpyrrolidone are effective as bone enhancer in posterolateral arthrodesis on rabbits. METHOD: Posterolateral arthrodesis in 15 New Zealand rabbits on level L5-L6 using autolog bone graft in left side (control group) and autolog bone graft + 1 ml FibroquelMR (study group) in right side of arthrodesis. Euthanasia and block resection of lumbar segment eight weeks post surgery. Radiographic analysis, manual exploration and light microscopy of fussed segments. RESULTS: Radiographic consolidation was observed in 80% in control group and 95% in study group, interleaved trabecular pattern with bone continuity and normal characteristics in 12 left sides and 14 right sides. CONCLUSION: Collagen type 1 and polyvinylpyrrolidone use is likely to have positive effect in bone consolidation process, therefore it can be recommended to use it as a bone enhancer.


INTRODUCCIÓN: Existen diversas estrategias para aumentar la tasa de consolidación de la artrodesis de columna en presencia de injerto óseo autólogo, sin aún comprobar si la adhesión de Colágena tipo I y polivinilpirrolidona (FibroquelMR) tienen aplicaciones en este campo. OBJETIVO: Determinar la efectividad de la colágena tipo I con polivinilpirrolidona como potenciador óseo en artrodesis posterolateral de conejos. MÉTODOS: Artrodesis posterolateral en 15 conejos de Nueva Zelanda L5-L6 colocando injerto autólogo del lado izquierdo (Control) e injerto autólogo + 1 ml FibroquelMR (Estudio) en el lado derecho de la artrodesis. Eutanasia con resección en bloque del segmento lumbar a las ocho semanas del postoperatorio. Análisis radiográfico, palpación manual y por microscopia de luz de los segmentos fusionados. RESULTADOS: Se observó consolidación radiográfica en 80% en grupo control y 93% en el estudio, continuidad ósea con patrón trabecular intercalado y hueso de características normales en 12 del lado izquierdo y 14 en el lado derecho. CONCLUSIONES: La utilización de Colágena tipo I y polivinilpirrolidona puede tener efectos positivos en el proceso de consolidación ósea por lo que se puede recomendar su utilización como reforzador óseo.


Bone Transplantation , Collagen Type I , Plasma Substitutes , Povidone , Spinal Fusion , Animals , Autografts , Collagen Type I/therapeutic use , Lumbar Vertebrae , Plasma Substitutes/therapeutic use , Povidone/therapeutic use , Rabbits , Spinal Fusion/methods
20.
J Dermatol Sci ; 88(3): 357-364, 2017 Dec.
Article En | MEDLINE | ID: mdl-29017796

BACKGROUND: Atopic dermatitis (AD) is a chronic, pruritic inflammatory skin disease in which type 2 allergic inflammation plays an important role. Collagen tripeptide (CTP) is a functional collagen fraction with a high content of Gly-X-Y tripeptides. OBJECTIVE: To examine the effect of CTP on inflammation in AD. METHODS: Levels of inflammatory cytokines and chemokines, such as thymus- and activation-regulated chemokine (TARC), macrophage-derived chemokine, and thymic stromal lymphopoietin (TSLP), were examined in human keratinocytes supplemented with or without CTP under AD-like inflammation. To evaluate the functional effect of CTP, a migration assay was performed using the supernatants of cultured keratinocytes treated with CTP. The signaling pathway for CTP inhibitory activity was also determined. Additionally, we conducted a clinical trial with seventeen AD patients who were assigned randomly to receive daily for 12 weeks either 3.9g of a CTP product or normal collagen peptides (CP). The eruption area, severity scoring of atopic dermatitis (SCORAD), skin hydration, transepidermal water loss (TEWL), and itching score were evaluated. The levels of TARC, serum IgE, lactate dehydrogenase, and eosinophil counts at week 12 were also compared with those at the start of administration. RESULTS: In human keratinocytes, TARC and TSLP mRNA and protein levels were inhibited significantly by CTP treatment under AD-like inflammation. Supernatants obtained from CTP-treated keratinocytes inhibited cell migration. STAT1 phosphorylation was significantly decreased by CTP in a dose-dependent manner. In the clinical trial, 13 patients (7 for CTP, 6 for CP) completed the study. The eruption area, SCORAD, and TEWL at week 12 were reduced significantly compared with the initial values in the CTP but not CP group. A significant reduction in the serum TARC level was observed only in the CTP group at week 12. Other blood parameters were not improved in either group. CONCLUSION: CTP may have therapeutic benefit for AD by inhibiting type 2-skewed allergic inflammation.


Anti-Inflammatory Agents/therapeutic use , Chemokine CCL17/metabolism , Collagen Type I/therapeutic use , Cytokines/metabolism , Dermatitis, Atopic/drug therapy , STAT1 Transcription Factor/metabolism , Administration, Oral , Adult , Anti-Inflammatory Agents/pharmacology , Cell Movement/drug effects , Cells, Cultured , Chemokine CCL17/blood , Collagen Type I/pharmacology , Dermatitis, Atopic/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Keratinocytes , Male , Middle Aged , Phosphorylation/drug effects , Severity of Illness Index , Skin/cytology , Skin/drug effects , Skin/metabolism , Thymic Stromal Lymphopoietin
...