1
|
Wang Z, Chen Y, Li W, Gao C, Zhang J, Zang X, Zhao Z, Fan H, Zhao Y. Identification and validation of diagnostic biomarkers and immune infiltration in dilated cardiomyopathies with heart failure and construction of diagnostic model. Gene 2025; 934:149007. [PMID: 39427832 DOI: 10.1016/j.gene.2024.149007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/14/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Dilated cardiomyopathy (DCM) is characterized by immune cell infiltration and can readily progress to heart failure (HF). In the study, differential expression analysis, enrichment analysis, and protein-protein interaction (PPI) network analysis were performed on DCM with HF-related datasets. The CytoHubba was used to identify hub genes. Diagnostic biomarkers were obtained by validating their expression and diagnostic value in another external dataset, and a diagnostic model was constructed. Finally, single-sample gene set enrichment analysis (ssGSEA) was used to predict immune cell infiltration in cardiac samples. The associations between diagnostic biomarkers and immune cells were investigated. The NetworkAnalyst and miRDB databases were used to predict transcription factors and microRNAs, followed by establishing regulatory networks. The DSigDB database was used to predict drug candidates. Subsequently, a mouse model of DCM with HF was used to validate the expression levels of these genes. The present study revealed that differentially expressed genes were enriched in the extracellular matrix organization, cardiac muscle hypertrophy, and other immune-related biological processes. OMD and THBS4 were finally identified, and the nomogram has satisfactory prediction and strong calibration ability. In addition, the two diagnostic biomarkers exhibited significant associations with multiple immune infiltrating cells. Finally, two TFs, 65 microRNAs, and 10 drug candidates were obtained. In animal experiments, two diagnostic biomarkers showed expression trends consistent with the results of bioinformatic analysis. OMD and THBS4 have been identified as hub immune-related diagnostic biomarkers for DCM with HF. Our research provides novel insights into the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Zhaodi Wang
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Yihan Chen
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Weidong Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Henan Provincial Key Lab for Control of Coronary Heart Disease, Zhengzhou University Central China Fuwai Hospital, Zhengzhou 450000, China
| | - Jing Zhang
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaobiao Zang
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Zhihan Zhao
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Hongkun Fan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Yonghui Zhao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, China; Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
2
|
Zhou Y, Shi W, Kimura R, Chai Y, Tagaya M. Self-Assembly of Cyclic-Bending Collagen Fibrils by Polyimide Films. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:22602-22613. [PMID: 39412338 DOI: 10.1021/acs.langmuir.4c02340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The cyclic-bending morphologies of the fibrils formed by the self-assembly of type I collagen (Col) are closely related to the mechanisms of various diseases. Therefore, studies that allow the self-assembly of Col molecules to form cyclic-bending fibrils in vitro are vitally important. In this study, we successfully achieved the cyclic-bending shapes (specifically, a regular hexagonal shape) of Col molecules by controlling the steric structures of polyimide (PI) molecular chains through the film formation process. Specifically, when a single layer of PI film was baked, the PI molecular chains within the film bent in the direction parallel to the substrate surface plane. Repeating the layering and baking processes resulted in 3D structures of the PI molecular chains, which were oriented in the direction perpendicular to the substrate surface plane. This three-dimensional bending would result from the PI molecular chain interactions between the upper and lower layers. When the Col molecules were reacted on these film surfaces, they recognized the structures of the PI molecular chains and self-assembled to form cyclic-bending Col fibrils. Especially, in PI films subjected to three cycles of layering and baking, hemicircular-shaped Col fibrils were observed to be regularly arrayed. Additionally, these regularly cyclic-bending fibrils were aligned in the uniaxial direction through a uniaxial rubbing treatment of the PI films. This successful research is significant both as a method for controlling the morphologies of Col fibrils and as a study that explores the biomedical implications of Col fibril cyclic-bending in the living body.
Collapse
Affiliation(s)
- Yanni Zhou
- Department of Materials Science and Technology, Nagaoka University of Technology, Kamitomioka 1603-1, Nagaoka, Niigata 940-2188, Japan
| | - Wanyu Shi
- Department of Materials Science and Technology, Nagaoka University of Technology, Kamitomioka 1603-1, Nagaoka, Niigata 940-2188, Japan
- Research Fellow of the Japan Society for the Promotion of Science (DC), 5-3-1 Koji-machi, Chiyoda-ku, Tokyo102-0083, Japan
| | - Reo Kimura
- Department of Materials Science and Technology, Nagaoka University of Technology, Kamitomioka 1603-1, Nagaoka, Niigata 940-2188, Japan
| | - Yadong Chai
- Department of Materials Science and Technology, Nagaoka University of Technology, Kamitomioka 1603-1, Nagaoka, Niigata 940-2188, Japan
| | - Motohiro Tagaya
- Department of Materials Science and Technology, Nagaoka University of Technology, Kamitomioka 1603-1, Nagaoka, Niigata 940-2188, Japan
| |
Collapse
|
3
|
Zhu Z, Huang F, Gao M, Liu M, Zhang Y, Tang L, Wu J, Yu H, He C, Chen J, Yang Z, Chen Z, Li Y, Chen H, Lei T, Zeng F, Cui Y. Osteogenic-Like Microenvironment of Renal Interstitium Induced by Osteomodulin Contributes to Randall's Plaque Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405875. [PMID: 39225583 PMCID: PMC11516157 DOI: 10.1002/advs.202405875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Calcium oxalate (CaOx) kidney stones are common and recurrent, lacking pharmacological prevention. Randall's plaques (RPs), calcium deposits in renal papillae, serve as niduses for some CaOx stones. This study explores the role of osteogenic-like cells in RP formation resembling ossification. CaP crystals deposit around renal tubules, interstitium, and blood vessels in RP tissues. Human renal interstitial fibroblasts (hRIFs) exhibit the highest osteogenic-like differentiation potential compared to chloride voltage-gated channel Ka positive tubular epithelial cells, aquaporin 2 positive collecting duct cells, and vascular endothelial cells, echoing the upregulated osteogenic markers primarily in hRIFs within RP tissues. Utilizing RNA-seq, osteomodulin (OMD) is found to be upregulated in hRIFs within RP tissues and hRIFs following osteogenic induction. Furthermore, OMD colocalizes with CaP crystals and calcium vesicles within RP tissues. OMD can enhance osteogenic-like differentiation of hRIFs in vitro and in vivo. Additionally, crystal deposits are attenuated in mice with Omd deletion in renal interstitial fibroblasts following CaOx nephrocalcinosis induction. Mechanically, a positive feedback loop of OMD/BMP2/BMPR1A/RUNX2/OMD drives hRIFs to adopt osteogenic-like fates, by which OMD induces osteogenic-like microenvironment of renal interstitium to participate in RP formation. We identify OMD upregulation as a pathological feature of RP, paving the way for preventing CaOx stones.
Collapse
Affiliation(s)
- Zewu Zhu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Internal MedicineSection EndocrinologyYale University School of MedicineNew HavenCT06519USA
| | - Fang Huang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Meng Gao
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Minghui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Youjie Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Liang Tang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Jian Wu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hao Yu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Cheng He
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhongqing Yang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhiyong Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hequn Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ting Lei
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310006China
| | - Feng Zeng
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yu Cui
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
4
|
Zhao W, von Kroge S, Jadzic J, Milovanovic P, Sihota P, Luther J, Brylka L, von Brackel FN, Bockamp E, Busse B, Amling M, Schinke T, Yorgan TA. Osteomodulin deficiency in mice causes a specific reduction of transversal cortical bone size. J Bone Miner Res 2024; 39:1025-1041. [PMID: 38722812 PMCID: PMC11301521 DOI: 10.1093/jbmr/zjae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 08/07/2024]
Abstract
Skeletal growth, modeling, and remodeling are regulated by various molecules, one of them being the recently identified osteoanabolic factor WNT1. We have previously reported that WNT1 transcriptionally activates the expression of Omd, encoding Osteomodulin (OMD), in a murine mesenchymal cell line, which potentially explained the skeletal fragility of mice with mutational WNT1 inactivation, since OMD has been shown to regulate type I collagen fibril formation in vitro. In this study we confirmed the strong induction of Omd expression in a genome-wide expression analysis of transfected cells, and we obtained further evidence for Omd being a direct target gene of WNT1. To assess the in vivo relevance of this regulation, we crossed Omd-deficient mice with a mouse line harboring an inducible, osteoblast-specific Wnt1 transgene. After induction of Wnt1 expression for 1 or 3 weeks, the osteoanabolic potency of WNT1 was not impaired despite the Omd deficiency. Since current knowledge regarding the in vivo physiological function of OMD is limited, we next focused on skeletal phenotyping of wild-type and Omd-deficient littermates, in the absence of a Wnt1 transgene. Here we did not observe an impact of Omd deficiency on trabecular bone parameters by histomorphometry and μCT either. Importantly, however, male and female Omd-deficient mice at the ages of 12 and 24 weeks displayed a slender bone phenotype with significantly smaller long bones in the transversal dimension, while the longitudinal bone growth remained unaffected. Although mechanical testing revealed no significant changes explained by impaired bone material properties, atomic force microscopy of the femoral bone surface of Omd-deficient mice revealed moderate changes at the nanostructural level, indicating altered regulation of collagen fibril formation and aggregation. Taken together, our data demonstrate that, although OMD is dispensable for the osteoanabolic effect of WNT1, its deficiency in mice specifically modulates transversal cortical bone morphology.
Collapse
Affiliation(s)
- Wenbo Zhao
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Jelena Jadzic
- Center of Bone Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Petar Milovanovic
- Center of Bone Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Praveer Sihota
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Julia Luther
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Laura Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Felix N von Brackel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Ernesto Bockamp
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Timur A Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| |
Collapse
|
5
|
Bai Y, Wu P, Zhang Q, Lin F, Hu L, Zhang Z, Huang W, Xiao Y, Zuo Q. Decorin in the spatial control of collagen mineralization. MATERIALS HORIZONS 2024; 11:3396-3407. [PMID: 38690683 DOI: 10.1039/d3mh02216a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Understanding the molecular mechanism by which the periodontal ligament (PDL) is maintained uncalcified between two mineralized tissues (cementum and bone) may facilitate the functional repair and regeneration of the periodontium complex, disrupted in the context of periodontal diseases. However, research that explores the control of type I collagen (COL I) mineralization fails to clarify the detailed mechanism of regulating spatial collagen mineralization, especially in the periodontium complex. In the present study, decorin (DCN), which is characterized as abundant in the PDL region and rare in mineralized tissues, was hypothesized to be a key regulator in the spatial control of collagen mineralization. The circular dichroism results confirmed that DCN regulated the secondary structure of COL I, and the surface plasmon resonance results indicated that COL I possessed a higher affinity for DCN than for other mineralization promoters, such as DMP-1, OPN, BSP and DSPP. These features of DCN may contribute to blocking intrafibrillar mineralization in COL I fibrils during the polymer-induced liquid-precursor mineralization process when the fibrils are cross-linked with DCN. This effect was more remarkable when the fibrils were phosphorylated by sodium trimetaphosphate, as shown by the observation of a tube-like morphology via TEM and mineral sheath via SEM. This study enhances the understanding of the role of DCN in mineralization regulation among periodontal tissues. This provides insights for the development of biomaterials for the regeneration of interfaces between soft and hard tissues.
Collapse
Affiliation(s)
- Yuming Bai
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Peng Wu
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Qiufang Zhang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Feng Lin
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Ling Hu
- Department of Pharmacy and Pharmaceutical Sciences, Xiamen Medical College, Xiamen, PR China
| | - Zhisheng Zhang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Wenxia Huang
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| | - Yin Xiao
- School of Medicine and Dentistry, Griffith University, Gold Coast, Australia
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Qiliang Zuo
- Stomatological Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, PR China.
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, PR China
- Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen, PR China
| |
Collapse
|
6
|
Rojas MG, Pereira-Simon S, Zigmond ZM, Varona Santos J, Perla M, Santos Falcon N, Stoyell-Conti FF, Salama A, Yang X, Long X, Duque JC, Salman LH, Tabbara M, Martinez L, Vazquez-Padron RI. Single-Cell Analyses Offer Insights into the Different Remodeling Programs of Arteries and Veins. Cells 2024; 13:793. [PMID: 38786017 PMCID: PMC11119253 DOI: 10.3390/cells13100793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins at the single-cell level, with a focus on cell populations with remodeling potential. Upper arm arteries (brachial) and veins (basilic/cephalic) from 30 organ donors were compared using a combination of bulk and single-cell RNA sequencing, proteomics, flow cytometry, and histology. The cellular atlases of six arteries and veins demonstrated a 7.8× higher proportion of contractile smooth muscle cells (SMCs) in arteries and a trend toward more modulated SMCs. In contrast, veins showed a higher abundance of endothelial cells, pericytes, and macrophages, as well as an increasing trend in fibroblasts. Activated fibroblasts had similar proportions in both types of vessels but with significant differences in gene expression. Modulated SMCs and activated fibroblasts were characterized by the upregulation of MYH10, FN1, COL8A1, and ITGA10. Activated fibroblasts also expressed F2R, POSTN, and COMP and were confirmed by F2R/CD90 flow cytometry. Activated fibroblasts from veins were the top producers of collagens among all fibroblast populations from both types of vessels. Venous fibroblasts were also highly angiogenic, proinflammatory, and hyper-responders to reactive oxygen species. Differences in wall structure further explain the significant contribution of fibroblast populations to remodeling in veins. Fibroblasts are almost exclusively located outside the external elastic lamina in arteries, while widely distributed throughout the venous wall. In line with the above, ECM-targeted proteomics confirmed a higher abundance of fibrillar collagens in veins vs. more basement ECM components in arteries. The distinct cellular compositions and transcriptional programs of reparative populations in arteries and veins may explain differences in acute and chronic wall remodeling between vessels. This information may be relevant for the development of antistenotic therapies.
Collapse
Affiliation(s)
- Miguel G. Rojas
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Simone Pereira-Simon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | | | - Javier Varona Santos
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Mikael Perla
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Nieves Santos Falcon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Filipe F. Stoyell-Conti
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Alghidak Salama
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xiaochun Long
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Juan C. Duque
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Loay H. Salman
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY 12208, USA
| | - Marwan Tabbara
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Laisel Martinez
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
- Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA;
| |
Collapse
|
7
|
Davaapil H, Hopkins J, Bonnin N, Papadaki V, Leung A, Kosuge H, Tashima T, Nakakido M, Sekido R, Tsumoto K, Sagoo MS, Ohnuma SI. PRELP secreted from mural cells protects the function of blood brain barrier through regulation of endothelial cell-cell integrity. Front Cell Dev Biol 2023; 11:1147625. [PMID: 37936982 PMCID: PMC10626469 DOI: 10.3389/fcell.2023.1147625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/13/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction: Proline/arginine-rich end leucine-rich repeat protein (PRELP), is a small secreted proteoglycan expressed by pericytes and vascular smooth muscle cells surrounding the brain vasculature of adult mouse. Methods: We utilised a Prelp knockout (Prelp -/-) mouse model to interrogate vasculature integrity in the brain alongside performing in vitro assays to characterise PRELP application to endothelial cells lines. Our findings were supplemented with RNA expression profiling to elucidate the mechanism of how PRELP maintains neurovasculature function. Results: Prelp -/- mice presented with neuroinflammation and reducedneurovasculature integrity, resulting in IgG and dextran leakage in the cerebellum and cortex. Histological analysis of Prelp -/- mice revealed reducedcell-cell integrity of the blood brain barrier, capillary attachment of pericytes andastrocyte end-feet. RNA-sequencing analysis found that cell-cell adhesion andinflammation are affected in Prelp -/- mice and gene ontology analysis as well as gene set enrichment analysis demonstrated that inflammation related processes and adhesion related processes such as epithelial-mesenchymal transition and apical junctions were significantly affected, suggesting PRELP is a regulator of cell-cell adhesion. Immunofluorescence analysis showed that adhesion junction protein expression levels of cadherin, claudin-5, and ZO-1, was suppressed in Prelp -/- mice neurovasculature. Additionally, in vitro studies revealed that PRELP application to endothelial cells enhances cell-cell integrity, induces mesenchymal-endothelial transition and inhibits TGF-β mediated damage to cell-cell adhesion. Discussion: Our study indicates that PRELP is a novel endogenous secreted regulator of neurovasculature integrity and that PRELP application may be a potential treatment for diseases associated with neurovascular damage.
Collapse
Affiliation(s)
| | - Jack Hopkins
- UCL Institute of Ophthalmology, UCL, London, Untited Kingdom
| | - Nadia Bonnin
- UCL Institute of Ophthalmology, UCL, London, Untited Kingdom
| | | | - Alex Leung
- UCL Institute of Ophthalmology, UCL, London, Untited Kingdom
| | - Hirofumi Kosuge
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takumi Tashima
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakakido
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryohei Sekido
- UCL Institute of Ophthalmology, UCL, London, Untited Kingdom
| | - Kouhei Tsumoto
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mandeep S. Sagoo
- UCL Institute of Ophthalmology, UCL, London, Untited Kingdom
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, London, Untited Kingdom
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, Royal London Hospital, London, Untited Kingdom
| | | |
Collapse
|
8
|
Gesteira TF, Verma S, Coulson-Thomas VJ. Small leucine rich proteoglycans: Biology, function and their therapeutic potential in the ocular surface. Ocul Surf 2023; 29:521-536. [PMID: 37355022 PMCID: PMC11092928 DOI: 10.1016/j.jtos.2023.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Small leucine rich proteoglycans (SLRPs) are the largest family of proteoglycans, with 18 members that are subdivided into five classes. SLRPs are small in size and can be present in tissues as glycosylated and non-glycosylated proteins, and the most studied SLRPs include decorin, biglycan, lumican, keratocan and fibromodulin. SLRPs specifically bind to collagen fibrils, regulating collagen fibrillogenesis and the biomechanical properties of tissues, and are expressed at particularly high levels in fibrous tissues, such as the cornea. However, SLRPs are also very active components of the ECM, interacting with numerous growth factors, cytokines and cell surface receptors. Therefore, SLRPs regulate major cellular processes and have a central role in major fundamental biological processes, such as maintaining corneal homeostasis and transparency and regulating corneal wound healing. Over the years, mutations and/or altered expression of SLRPs have been associated with various corneal diseases, such as congenital stromal corneal dystrophy and cornea plana. Recently, there has been great interest in harnessing the various functions of SLRPs for therapeutic purposes. In this comprehensive review, we describe the structural features and the related functions of SLRPs, and how these affect the therapeutic potential of SLRPs, with special emphasis on the use of SLRPs for treating ocular surface pathologies.
Collapse
Affiliation(s)
| | - Sudhir Verma
- College of Optometry, University of Houston, USA; Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi, India
| | | |
Collapse
|
9
|
Chen J, Cao G, Li L, Cai Q, Dunne N, Li X. Modification of polyether ether ketone for the repairing of bone defects. Biomed Mater 2022; 17:042001. [PMID: 35395651 DOI: 10.1088/1748-605x/ac65cd] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/08/2022] [Indexed: 11/12/2022]
Abstract
Bone damage as a consequence of disease or trauma is a common global occurrence. For bone damage treatment-bone implant materials are necessary across three classifications of surgical intervention (i.e. fixation, repair, and replacement). Many types of bone implant materials have been developed to meet the requirements of bone repair. Among them, polyether ether ketone (PEEK) has been considered as one of the next generation of bone implant materials, owing to its advantages related to good biocompatibility, chemical stability, x-ray permeability, elastic modulus comparable to natural bone, as well as the ease of processing and modification. However, as PEEK is a naturally bioinert material, some modification is needed to improve its integration with adjacent bones after implantation. Therefore, it has become a very hot topic of biomaterials research and various strategies for the modification of PEEK including blending, 3D printing, coating, chemical modification and the introduction of bioactive and/or antibacterial substances have been proposed. In this systematic review, the recent advances in modification of PEEK and its application prospect as bone implants are summarized, and the remaining challenges are also discussed.
Collapse
Affiliation(s)
- Junfeng Chen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, People's Republic of China
| | - Guangxiu Cao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, People's Republic of China
| | - Linhao Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, People's Republic of China
| | - Qiang Cai
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, People's Republic of China
| | - Nicholas Dunne
- Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, People's Republic of China
| |
Collapse
|
10
|
Skenteris NT, Seime T, Witasp A, Karlöf E, Wasilewski GB, Heuschkel MA, Jaminon AM, Oduor L, Dzhanaev R, Kronqvist M, Lengquist M, Peeters FE, Söderberg M, Hultgren R, Roy J, Maegdefessel L, Arnardottir H, Bengtsson E, Goncalves I, Quertermous T, Goettsch C, Stenvinkel P, Schurgers LJ, Matic L. Osteomodulin attenuates smooth muscle cell osteogenic transition in vascular calcification. Clin Transl Med 2022; 12:e682. [PMID: 35184400 PMCID: PMC8858609 DOI: 10.1002/ctm2.682] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 11/28/2021] [Accepted: 12/02/2021] [Indexed: 12/29/2022] Open
Abstract
RATIONALE Vascular calcification is a prominent feature of late-stage diabetes, renal and cardiovascular disease (CVD), and has been linked to adverse events. Recent studies in patients reported that plasma levels of osteomodulin (OMD), a proteoglycan involved in bone mineralisation, associate with diabetes and CVD. We hypothesised that OMD could be implicated in these diseases via vascular calcification as a common underlying factor and aimed to investigate its role in this context. METHODS AND RESULTS In patients with chronic kidney disease, plasma OMD levels correlated with markers of inflammation and bone turnover, with the protein present in calcified arterial media. Plasma OMD also associated with cardiac calcification and the protein was detected in calcified valve leaflets by immunohistochemistry. In patients with carotid atherosclerosis, circulating OMD was increased in association with plaque calcification as assessed by computed tomography. Transcriptomic and proteomic data showed that OMD was upregulated in atherosclerotic compared to control arteries, particularly in calcified plaques, where OMD expression correlated positively with markers of smooth muscle cells (SMCs), osteoblasts and glycoproteins. Immunostaining confirmed that OMD was abundantly present in calcified plaques, localised to extracellular matrix and regions rich in α-SMA+ cells. In vivo, OMD was enriched in SMCs around calcified nodules in aortic media of nephrectomised rats and in plaques from ApoE-/- mice on warfarin. In vitro experiments revealed that OMD mRNA was upregulated in SMCs stimulated with IFNγ, BMP2, TGFβ1, phosphate and β-glycerophosphate, and by administration of recombinant human OMD protein (rhOMD). Mechanistically, addition of rhOMD repressed the calcification process of SMCs treated with phosphate by maintaining their contractile phenotype along with enriched matrix organisation, thereby attenuating SMC osteoblastic transformation. Mechanistically, the role of OMD is exerted likely through its link with SMAD3 and TGFB1 signalling, and interplay with BMP2 in vascular tissues. CONCLUSION We report a consistent association of both circulating and tissue OMD levels with cardiovascular calcification, highlighting the potential of OMD as a clinical biomarker. OMD was localised in medial and intimal α-SMA+ regions of calcified cardiovascular tissues, induced by pro-inflammatory and pro-osteogenic stimuli, while the presence of OMD in extracellular environment attenuated SMC calcification.
Collapse
Affiliation(s)
- Nikolaos T. Skenteris
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
| | - Till Seime
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Anna Witasp
- Division of Renal MedicineDepartment of Clinical SciencesIntervention and TechnologyKarolinska InstituteStockholmSweden
| | - Eva Karlöf
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Grzegorz B. Wasilewski
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Nattopharma ASA, OsloNorway
| | - Marina A. Heuschkel
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Department of Internal Medicine I‐CardiologyMedical FacultyRWTH Aachen University, Aachen, Germany
| | - Armand M.G. Jaminon
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
| | - Loureen Oduor
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Robert Dzhanaev
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Biointerface GroupHelmholtz Institute for Biomedical EngineeringRWTH Aachen UniversityAachenGermany
| | - Malin Kronqvist
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Mariette Lengquist
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Frederique E.C.M. Peeters
- Department of Cardiology and CARIMSchool for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtNetherlands
| | - Magnus Söderberg
- CardiovascularRenal and Metabolism SafetyClinical Pharmacology and Safety SciencesR&D, AstraZenecaGothenburgSweden
| | - Rebecka Hultgren
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Joy Roy
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| | - Lars Maegdefessel
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
- Klinikum rechts der IsarDepartment for Vascular and Endovascular SurgeryTechnical University MunichMunichGermany
| | - Hildur Arnardottir
- Cardiovascular Medicine UnitDepartment of MedicineKarolinska InstituteStockholmSweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö and CardiologySkåne University HospitalLund UniversityLundSweden
| | - Thomas Quertermous
- Department of Cardiovascular Medicine, University of StanfordStanfordCaliforniaUSA
| | - Claudia Goettsch
- Department of Internal Medicine I‐CardiologyMedical FacultyRWTH Aachen University, Aachen, Germany
| | - Peter Stenvinkel
- Division of Renal MedicineDepartment of Clinical SciencesIntervention and TechnologyKarolinska InstituteStockholmSweden
| | - Leon J. Schurgers
- Department of Biochemistry and CARIMSchool for Cardiovascular DiseasesMaastricht UniversityMaastrichtNetherlands
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen UniversityAachenGermany
| | - Ljubica Matic
- Division of Vascular SurgeryDepartment of Molecular Medicine and SurgeryKarolinska InstituteStockholmSweden
| |
Collapse
|
11
|
Hosiriluck N, Kashio H, Takada A, Mizuguchi I, Arakawa T. The profiling and analysis of gene expression in human periodontal ligament tissue and fibroblasts. Clin Exp Dent Res 2022; 8:658-672. [PMID: 35106969 PMCID: PMC9209801 DOI: 10.1002/cre2.533] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 09/24/2021] [Accepted: 12/28/2021] [Indexed: 01/03/2023] Open
Abstract
Objectives The periodontal ligament (PDL) is an important component of periodontium to support dental structure in the alveolar socket. Regeneration of PDL tissue is an effective treatment option for periodontal disease and the profiling of genes involved in this process will be informative. Therefore, our study aims to accurately delineate the profiling of gene expression for PDL tissue regeneration. Materials and Methods We isolated PDL tissues and PDL fibroblasts (PDLFs) from premolar teeth, which were extracted from healthy periodontal status patients undergoing orthodontic treatment. Messenger RNA (mRNA) expression in PDL tissue and PDLFs were analyzed using Cap analysis gene expression, which is a second‐generation sequencing technique to create profiling. We also determined the protein expression using Western blot. Results Collagens (type I, III, and VI), noncollagenous proteins (periostin and osteonectin), and proteoglycans (asporin, lumican, decorin, and osteomodulin) were highly expressed in PDL tissue. Integrin, β1 was also expressed in PDL tissue. On comparison of gene expression between PDL tissue and PDLFs, four PDL marker genes, osteopontin, asporin, periostin, and osteonectin, were decreased in PDLFs. The genes for gene regulation were also highly expressed. Conclusions Our study demonstrated the overall profiling of mRNA expression in PDL tissue and analyzed the important genes which may be useful for providing specific information for the reconstruction of PDL. We also identified the difference in gene expression between PDL tissue and PDLFs which might provide insights towards PDL regeneration.
Collapse
Affiliation(s)
- Nattakarn Hosiriluck
- Division of Biochemistry, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Hokkaido, Japan
| | - Haruna Kashio
- Division of Orthodontics and Dentofacial Orthopedics, Department of Oral Growth and Development, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Hokkaido, Japan
| | - Ayuko Takada
- Division of Biochemistry, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Hokkaido, Japan
| | - Itaru Mizuguchi
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Toshiya Arakawa
- Division of Biochemistry, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu-cho, Hokkaido, Japan
| |
Collapse
|
12
|
Ma X, Mo C, Huang L, Cao P, Shen L, Gui C. An Robust Rank Aggregation and Least Absolute Shrinkage and Selection Operator Analysis of Novel Gene Signatures in Dilated Cardiomyopathy. Front Cardiovasc Med 2022; 8:747803. [PMID: 34970603 PMCID: PMC8713643 DOI: 10.3389/fcvm.2021.747803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Objective: Dilated cardiomyopathy (DCM) is a heart disease with high mortality characterized by progressive cardiac dilation and myocardial contractility reduction. The molecular signature of dilated cardiomyopathy remains to be defined. Hence, seeking potential biomarkers and therapeutic of DCM is urgent and necessary. Methods: In this study, we utilized the Robust Rank Aggregation (RRA) method to integrate four eligible DCM microarray datasets from the GEO and identified a set of significant differentially expressed genes (DEGs) between dilated cardiomyopathy and non-heart failure. Moreover, LASSO analysis was carried out to clarify the diagnostic and DCM clinical features of these genes and identify dilated cardiomyopathy derived diagnostic signatures (DCMDDS). Results: A total of 117 DEGs were identified across the four microarrays. Furthermore, GO analysis demonstrated that these DEGs were mainly enriched in the regulation of inflammatory response, the humoral immune response, the regulation of blood pressure and collagen–containing extracellular matrix. In addition, KEGG analysis revealed that DEGs were mainly enriched in diverse infected signaling pathways. Moreover, Gene set enrichment analysis revealed that immune and inflammatory biological processes such as adaptive immune response, cellular response to interferon and cardiac muscle contraction, dilated cardiomyopathy are significantly enriched in DCM. Moreover, Least absolute shrinkage and selection operator (LASSO) analyses of the 18 DCM-related genes developed a 7-gene signature predictive of DCM. This signature included ANKRD1, COL1A1, MYH6, PERELP, PRKACA, CDKN1A, and OMD. Interestingly, five of these seven genes have a correlation with left ventricular ejection fraction (LVEF) in DCM patients. Conclusion: Our present study demonstrated that the signatures could be robust tools for predicting DCM in clinical practice. And may also be potential treatment targets for clinical implication in the future.
Collapse
Affiliation(s)
- Xiao Ma
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Changhua Mo
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liangzhao Huang
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Peidong Cao
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Louyi Shen
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chun Gui
- Department of Cardiology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Vollersen N, Zhao W, Rolvien T, Lange F, Schmidt FN, Sonntag S, Shmerling D, von Kroge S, Stockhausen KE, Sharaf A, Schweizer M, Karsak M, Busse B, Bockamp E, Semler O, Amling M, Oheim R, Schinke T, Yorgan TA. The WNT1 G177C mutation specifically affects skeletal integrity in a mouse model of osteogenesis imperfecta type XV. Bone Res 2021; 9:48. [PMID: 34759273 PMCID: PMC8580994 DOI: 10.1038/s41413-021-00170-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/28/2021] [Accepted: 06/27/2021] [Indexed: 12/27/2022] Open
Abstract
The recent identification of homozygous WNT1 mutations in individuals with osteogenesis imperfecta type XV (OI-XV) has suggested that WNT1 is a key ligand promoting the differentiation and function of bone-forming osteoblasts. Although such an influence was supported by subsequent studies, a mouse model of OI-XV remained to be established. Therefore, we introduced a previously identified disease-causing mutation (G177C) into the murine Wnt1 gene. Homozygous Wnt1G177C/G177C mice were viable and did not display defects in brain development, but the majority of 24-week-old Wnt1G177C/G177C mice had skeletal fractures. This increased bone fragility was not fully explained by reduced bone mass but also by impaired bone matrix quality. Importantly, the homozygous presence of the G177C mutation did not interfere with the osteoanabolic influence of either parathyroid hormone injection or activating mutation of LRP5, the latter mimicking the effect of sclerostin neutralization. Finally, transcriptomic analyses revealed that short-term administration of WNT1 to osteogenic cells induced not only the expression of canonical WNT signaling targets but also the expression of genes encoding extracellular matrix modifiers. Taken together, our data demonstrate that regulating bone matrix quality is a primary function of WNT1. They further suggest that individuals with WNT1 mutations should profit from existing osteoanabolic therapies.
Collapse
Affiliation(s)
- Nele Vollersen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Wenbo Zhao
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Fabiola Lange
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Felix Nikolai Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Stephan Sonntag
- PolyGene AG, 8153, Rümlang, Switzerland.,ETH Phenomics Center (EPIC), ETH Zürich, 8092, Zürich, Switzerland
| | | | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Kilian Elia Stockhausen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ahmed Sharaf
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center, Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ernesto Bockamp
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, D 55131, Mainz, Germany
| | - Oliver Semler
- Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, University of Cologne, 50937, Cologne, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
14
|
Matsushima N, Miyashita H, Kretsinger RH. Sequence features, structure, ligand interaction, and diseases in small leucine rich repeat proteoglycans. J Cell Commun Signal 2021; 15:519-531. [PMID: 33860400 DOI: 10.1007/s12079-021-00616-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/25/2021] [Indexed: 12/26/2022] Open
Abstract
Small leucine rich repeat proteoglycans (SLRPs) are a group of active components of the extracellular matrix in all tissues. SLRPs bind to collagens and regulate collagen fibril growth and fibril organization. SLRPs also interact with various cytokines and extracellular compounds, which lead to various biological functions such cell adhesion and signaling, proliferation, and differentiation. Mutations in SLRP genes are associated with human diseases. Now crystal structures of five SLRPs are available. We describe some features of amino acid sequence and structures of SLRPs. We also review ligand interactions and then discuss the interaction surfaces. Furthermore, we map mutations associated with human diseases and discuss possible effects on structures by the mutations.
Collapse
Affiliation(s)
- Norio Matsushima
- Division of Bioinformatics, Institute of Tandem Repeats, Noboribetsu, 059-0464, Japan.
- Center for Medical Education, Sapporo Medical University, Sapporo, 060-8556, Japan.
| | - Hiroki Miyashita
- Division of Bioinformatics, Institute of Tandem Repeats, Noboribetsu, 059-0464, Japan
- Hokubu Rinsho Co., Ltd, Sapporo, 060⎼0061, Japan
| | - Robert H Kretsinger
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA
| |
Collapse
|
15
|
Xu Y, Gu Y, Ji W, Dong Q. Activation of the extracellular-signal-regulated kinase (ERK)/c-Jun N-terminal kinase (JNK) signal pathway and osteogenic factors in subchondral bone of patients with knee osteoarthritis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:663. [PMID: 33987361 PMCID: PMC8106020 DOI: 10.21037/atm-21-1215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/22/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND The objectives of this study was to explore the activation of the extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathway and osteogenesis-related factors in the subchondral bone of patients with knee osteoarthritis (OA). METHODS Ten patients with primary OA who underwent total knee arthroplasty in the Department of Arthritis Surgery of our hospital were enrolled, and subchondral bone tissue samples were obtained during the operation. He staining and saffron staining were used to observe the arrangement of chondrocytes in the patient tissues. The protein expression levels of JNK, p-JNK, ERK, p-ERK, Runx2 and OMD in subchondral bone were detected by Western Blot. Knee osteoarthritis mice were established. He staining was used to observe the arrangement of subchondral bone cells in the knee joint of mice. Cellular mineralized nodules were determined by alizarin red staining. RESULTS Firstly, in general and staining, it was observed that the subchondral bone lesions of knee OA participants were obvious. Compared with normal knee joints, the levels of phosphorylation-c-Jun N-terminal kinase (P-JNK) and phosphorylation-extracellular-signal-regulated kinase (P-ERK) in the subchondral bone of knee arthritis participants were significantly increased (P<0.05). The level of osteomodulin (OMD) was significantly reduced (P<0.05). Secondly, compared with normal mice, the levels of JNK, P-JNK, OMD, ERK, and P-ERK in the model group were significantly different (P<0.05). At 2-8 weeks, the JNK and P-JNK levels in the mice model group increased significantly over time (P<0.05), and the OMD level decreased significantly over time (P<0.05). The levels of ERK and P-ERK fluctuated over time. Thirdly, osteoblasts were treated with different concentrations of anisomycin, and stained with alizarin red after continuous culture for 24 and 48 h, respectively. It was found that all the cells were stained with orange-red mineralized nodules. As the concentration of anisomycin was increased, the number of cell mineralization nodules was significantly larger, and the positive rate of chemical nodules increased. Different concentrations of anisomycin were given to interfere with the osteoblasts of mice. When anisomycin was administered at a dose of 25 ng, the OMD level reached the highest level. When the concentration of anisomycin was increased, the osteocalcin (OCN) level also showed an upward trend. CONCLUSIONS The process by which the JNK signaling pathway regulates OMD may be closely related to the pathological changes of subchondral bone in patients with knee OA, and is involved in the occurrence and development of knee arthritis.
Collapse
Affiliation(s)
- Yaofeng Xu
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Suzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, China
| | - Yuguo Gu
- Department of Orthopedics, Suzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, China
| | - Wanbo Ji
- Department of Orthopedics, Suzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, China
| | - Qirong Dong
- Department of Orthopedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
16
|
Osteomodulin positively regulates osteogenesis through interaction with BMP2. Cell Death Dis 2021; 12:147. [PMID: 33542209 PMCID: PMC7862363 DOI: 10.1038/s41419-021-03404-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Osteomodulin (OMD), a member of the small leucine-rich proteoglycan family, distributes in mineralized tissues and is positively regulated by bone morphogenetic protein 2 (BMP2). However, the exact function of OMD during mineralization and its association with BMP2 remain poorly understood. Herein, the expression pattern of OMD during osteogenesis was investigated in human dental pulp stem cells. Silencing OMD gene significantly suppressed the alkaline phosphatase activity, mineralized nodule formation and osteogenesis-associated gene transcription. Besides, OMD could enhance BMP2-induced expression of SP7 and RUNX2 with concentration dependence in vitro. Rat mandibular bone defect model revealed that scaffolds injected with the combination of OMD and suboptimal BMP2 exhibited more mature and abundant mineralized bone than that treated with OMD or suboptimal BMP2 alone. Mechanistically, OMD could bind to BMP2 via its terminal leucine-rich repeats and formed complexes with BMP2 and its membrane receptors, thus promoting BMP/SMAD signal transduction. In addition, OMD was a putative target gene of SMAD4, which plays a pivotal role in this pathway. Collectively, these data elucidate that OMD may act as a positive coordinator in osteogenesis through BMP2/SMADs signaling.
Collapse
|
17
|
Rajasekaran S, Soundararajan DCR, Tangavel C, Nayagam SM, K S SV, R S, Matchado MS, Muthurajan R, Shetty AP, Kanna RM, K D. Uncovering molecular targets for regenerative therapy in degenerative disc disease: do small leucine-rich proteoglycans hold the key? Spine J 2021; 21:5-19. [PMID: 32344061 DOI: 10.1016/j.spinee.2020.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Small leucine-rich proteoglycans (SLRPs) play an essential role in extracellular matrix (ECM) organization and function. Recently, dysregulation of SLRPs has been implicated in degenerative disc disease (DDD). An in-depth analysis using high-throughput proteomic sequencing might provide valuable information on their implications in health and disease. PURPOSE To utilize proteomics for analyzing the expression of SLRPs in fetal, healthy adult, and degenerated discs, to identify possible molecular targets to halt or reverse the degenerative process. STUDY DESIGN Experimental analysis. METHODS Proteomic signatures of 8 magnetic resonance imaging (MRI) normal lumbar discs (ND) [harvested from brain dead alive organ donors] were compared to 8 fetal disc samples (FD) [harvested from fetal spines devoid of congenital anomalies following spontaneous or medical termination of pregnancy] and 8 degenerate discs (DD) [collected from patients undergoing fusion surgery]. The various functional pathways along with the differential expression of SLRPs and the associated changes in collagens, large proteoglycans (LLRPs), matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) have been analyzed further using bioinformatics. This project was self-funded by the Ganga Orthopedic Research and Education Foundation. RESULTS ESI-LC-MS/MS analysis revealed a total of 1,029 proteins in FD, 1,785 proteins in ND, and 1,775 proteins in DD. Fetal disc proteins were engaged mainly in ribosomal pathways (indicating active proliferation and regenerative potential). The healthy adult discs (ND) primarily participated in ECM maintenance and basic metabolic pathways, whereas the unique proteins of DD group were involved in inflammatory (Complement and coagulation cascades, Systemic Lupus Erythematosus and Leukocyte transendothelial migration) pathways and infective (Staphylococcus aureus infection, Prion diseases, Amoebiasis, Pertussis, and Legionellosis) channels which favor the recent concepts of inflammaging and subclinical infection as causes of DDD. Analysis of SLRPs revealed the upregulation of Biglycan in FDs and downregulation of Lumican, Decorin, Prolargin, and Chondroadherin in the DD group. The universal decrease in the abundance of SLRPs in the DD group was associated with an increase in MMPs and a reduction in TIMPs, collagen and LLRP content. CONCLUSIONS Our study documents the influence of SLRPs in the maintenance of disc health and also the need for future research in using them for disc regeneration. CLINICAL SIGNIFICANCE The various SLRPs that we identified are all known to have a beneficial influence on ECM integrity and a negative effect on the degenerative process at different stages in the evolution of degeneration. Biglycan, which is abundantly present in a fetus, may be suitable for regenerative therapy, and the other SLRPs like Lumican, Prolargin, Decorin, and Chondroadherin may serve the same purpose and/or as biomarkers.
Collapse
Affiliation(s)
| | | | - Chitraa Tangavel
- Ganga Research Centre, No 91, Mettupalayam Rd, Coimbatore 641030, India
| | | | - Sri Vijayanand K S
- Department of Spine Surgery, Ganga Hospital, 313, Mettupalayam Rd, Coimbatore, India
| | - Sunmathi R
- Ganga Research Centre, No 91, Mettupalayam Rd, Coimbatore 641030, India
| | | | - Raveendran Muthurajan
- Department of Plant Biotechnology, Tamil Nadu agricultural university, Coimbatore 641003, India
| | - Ajoy Prasad Shetty
- Department of Spine Surgery, Ganga Hospital, 313, Mettupalayam Rd, Coimbatore, India
| | - Rishi Mugesh Kanna
- Department of Spine Surgery, Ganga Hospital, 313, Mettupalayam Rd, Coimbatore, India
| | - Dharmalingam K
- Aravind Medical Research Foundation, Madurai 625020, India
| |
Collapse
|
18
|
Whitmore EK, Martin D, Guvench O. Constructing 3-Dimensional Atomic-Resolution Models of Nonsulfated Glycosaminoglycans with Arbitrary Lengths Using Conformations from Molecular Dynamics. Int J Mol Sci 2020; 21:ijms21207699. [PMID: 33080973 PMCID: PMC7589010 DOI: 10.3390/ijms21207699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/24/2022] Open
Abstract
Glycosaminoglycans (GAGs) are the linear carbohydrate components of proteoglycans (PGs) and are key mediators in the bioactivity of PGs in animal tissue. GAGs are heterogeneous, conformationally complex, and polydisperse, containing up to 200 monosaccharide units. These complexities make studying GAG conformation a challenge for existing experimental and computational methods. We previously described an algorithm we developed that applies conformational parameters (i.e., all bond lengths, bond angles, and dihedral angles) from molecular dynamics (MD) simulations of nonsulfated chondroitin GAG 20-mers to construct 3-D atomic-resolution models of nonsulfated chondroitin GAGs of arbitrary length. In the current study, we applied our algorithm to other GAGs, including hyaluronan and nonsulfated forms of dermatan, keratan, and heparan and expanded our database of MD-generated GAG conformations. Here, we show that individual glycosidic linkages and monosaccharide rings in 10- and 20-mers of hyaluronan and nonsulfated dermatan, keratan, and heparan behave randomly and independently in MD simulation and, therefore, using a database of MD-generated 20-mer conformations, that our algorithm can construct conformational ensembles of 10- and 20-mers of various GAG types that accurately represent the backbone flexibility seen in MD simulations. Furthermore, our algorithm efficiently constructs conformational ensembles of GAG 200-mers that we would reasonably expect from MD simulations.
Collapse
Affiliation(s)
- Elizabeth K. Whitmore
- Department of Pharmaceutical Sciences and Administration, University of New England School of Pharmacy, 716 Stevens Avenue, Portland, ME 04103, USA; (E.K.W.); (D.M.)
- Graduate School of Biomedical Science and Engineering, University of Maine, 5775 Stodder Hall, Orono, ME 04469, USA
| | - Devon Martin
- Department of Pharmaceutical Sciences and Administration, University of New England School of Pharmacy, 716 Stevens Avenue, Portland, ME 04103, USA; (E.K.W.); (D.M.)
- Graduate School of Biomedical Science and Engineering, University of Maine, 5775 Stodder Hall, Orono, ME 04469, USA
| | - Olgun Guvench
- Department of Pharmaceutical Sciences and Administration, University of New England School of Pharmacy, 716 Stevens Avenue, Portland, ME 04103, USA; (E.K.W.); (D.M.)
- Graduate School of Biomedical Science and Engineering, University of Maine, 5775 Stodder Hall, Orono, ME 04469, USA
- Correspondence: ; Tel.: +1-207-221-4171
| |
Collapse
|
19
|
Reiss RA, Lowe TC, Sena JA, Makhnin O, Connick MC, Illescas PE, Davis CF. Bio-activating ultrafine grain titanium: RNA sequencing reveals enhanced mechano-activation of osteoconduction on nanostructured substrates. PLoS One 2020; 15:e0237463. [PMID: 32970688 PMCID: PMC7514099 DOI: 10.1371/journal.pone.0237463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/27/2020] [Indexed: 02/02/2023] Open
Abstract
Titanium is essentially absent from biological systems yet reliably integrates into bone. To achieve osseointegration, titanium must activate biological processes without entering cells, defining it as a bio-activating material. Nanostructuring bulk titanium reduces grain size, increases strength, and improves other quantifiable physical properties, including cytocompatibility. The biological processes activated by increasing grain boundary availability were detected with total RNA-sequencing in mouse pre-osteoblasts grown for 72 hours on nanometrically smooth substrates of either coarse grain or nanostructured ultrafine grain titanium. The average grain boundary length under cells on the conventional coarse grain substrates is 273.0 μm, compared to 70,881.5 μm for cells adhered to the nanostructured ultrafine grain substrates; a 260-fold difference. Cells on both substrates exhibit similar expression profiles for genes whose products are critical for mechanosensation and transduction of cues that trigger osteoconduction. Biological process Gene Ontology term enrichment analysis of differentially expressed genes reveals that cell cycle, chromatin modification, telomere maintenance, and RNA metabolism processes are upregulated on ultrafine grain titanium. Processes related to immune response, including apoptosis, are downregulated. Tumor-suppressor genes are upregulated while tumor-promoting genes are downregulated. Upregulation of genes involved in chromatin remodeling and downregulation of genes under the control of the peripheral circadian clock implicate both processes in the transduction of mechanosensory information. Non-coding RNAs may also play a role in the response. Merging transcriptomics with well-established mechanobiology principles generates a unified model to explain the bio-activating properties of titanium. The modulation of processes is accomplished through chromatin remodeling in which the nucleus responds like a rheostat to grain boundary concentration. This convergence of biological and materials science reveals a pathway toward understanding the biotic-abiotic interface and will inform the development of effective bio-activating and bio-inactivating materials.
Collapse
Affiliation(s)
- Rebecca A. Reiss
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Terry C. Lowe
- George S. Ansell Department of Metallurgical and Materials Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| | - Johnny A. Sena
- National Center for Genome Resources, Santa Fe, New Mexico, United States of America
| | - Oleg Makhnin
- Mathematics Department, New Mexico Institute of Mining and Technology, Socorro, New Mexico, United States of America
| | - Melanie C. Connick
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Patrick E. Illescas
- Biology Department, New Mexico Institution of Mining and Technology, Socorro, New Mexico, United States of America
| | - Casey F. Davis
- George S. Ansell Department of Metallurgical and Materials Engineering, Colorado School of Mines, Golden, Colorado, United States of America
| |
Collapse
|
20
|
Antimicrobial Properties of Extracellular Matrix Scaffolds for Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9641456. [PMID: 31911931 PMCID: PMC6930736 DOI: 10.1155/2019/9641456] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/13/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022]
Abstract
The necessity to manufacture graft materials with superior biocompatibility capabilities and biodegradability characteristics for tissue regeneration has led to the production of extracellular matrix- (ECM-) based scaffolds. Among their advantages are better capacity to allow cell colonization, which enables its successful integration into the tissue surrounding the area to be repaired. In addition, it has been shown that some of these scaffolds have antimicrobial activity, preventing possible infections; therefore, it could be used as an alternative to control surgical infection and decrease the use of antimicrobial agents. The purpose of this review is to collect the existing information about antimicrobial activity of the ECM and their components.
Collapse
|