1
|
Miguez PA, Bash E, Musskopf ML, Tuin SA, Rivera-Concepcion A, Chapple ILC, Liu J. Control of tissue homeostasis by the extracellular matrix: Synthetic heparan sulfate as a promising therapeutic for periodontal health and bone regeneration. Periodontol 2000 2024; 94:510-531. [PMID: 37614159 PMCID: PMC10891305 DOI: 10.1111/prd.12515] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023]
Abstract
Proteoglycans are core proteins associated with carbohydrate/sugar moieties that are highly variable in disaccharide composition, which dictates their function. These carbohydrates are named glycosaminoglycans, and they can be attached to proteoglycans or found free in tissues or on cell surfaces. Glycosaminoglycans such as hyaluronan, chondroitin sulfate, dermatan sulfate, keratan sulfate, and heparin/heparan sulfate have multiple functions including involvement in inflammation, immunity and connective tissue structure, and integrity. Heparan sulfate is a highly sulfated polysaccharide that is abundant in the periodontium including alveolar bone. Recent evidence supports the contention that heparan sulfate is an important player in modulating interactions between damage associated molecular patterns and inflammatory receptors expressed by various cell types. The structure of heparan sulfate is reported to dictate its function, thus, the utilization of a homogenous and structurally defined heparan sulfate polysaccharide for modulation of cell function offers therapeutic potential. Recently, a chemoenzymatic approach was developed to allow production of many structurally defined heparan sulfate carbohydrates. These oligosaccharides have been studied in various pathological inflammatory conditions to better understand their function and their potential application in promoting tissue homeostasis. We have observed that specific size and sulfation patterns can modulate inflammation and promote tissue maintenance including an anabolic effect in alveolar bone. Thus, new evidence provides a strong impetus to explore heparan sulfate as a potential novel therapeutic agent to treat periodontitis, support alveolar bone maintenance, and promote bone formation.
Collapse
Affiliation(s)
- PA Miguez
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - E Bash
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - ML Musskopf
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - SA Tuin
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - A Rivera-Concepcion
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - ILC Chapple
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, Birmingham’s NIHR BRC in Inflammation Research, University of Birmingham and Birmingham Community Health Foundation Trust, Birmingham UK Iain Chapple
| | - J Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Drysdale A, Unsworth AJ, White SJ, Jones S. The Contribution of Vascular Proteoglycans to Atherothrombosis: Clinical Implications. Int J Mol Sci 2023; 24:11854. [PMID: 37511615 PMCID: PMC10380219 DOI: 10.3390/ijms241411854] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
The vascular extracellular matrix (ECM) produced by endothelial and smooth muscle cells is composed of collagens and glycoproteins and plays an integral role in regulating the structure and function of the vascular wall. Alteration in the expression of these proteins is associated with endothelial dysfunction and has been implicated in the development and progression of atherosclerosis. The ECM composition of atherosclerotic plaques varies depending on plaque phenotype and vulnerability, with distinct differences observed between ruptured and erodes plaques. Moreover, the thrombi on the exposed ECM are diverse in structure and composition, suggesting that the best antithrombotic approach may differ depending on plaque phenotype. This review provides a comprehensive overview of the role of proteoglycans in atherogenesis and thrombosis. It discusses the differential expression of the proteoglycans in different plaque phenotypes and the potential impact on platelet function and thrombosis. Finally, the review highlights the importance of this concept in developing a targeted approach to antithrombotic treatments to improve clinical outcomes in cardiovascular disease.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (A.J.U.)
| | - Amanda J. Unsworth
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (A.J.U.)
| | - Stephen J. White
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK;
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (A.D.); (A.J.U.)
| |
Collapse
|
3
|
Zheng XQ, Huang J, Lin JL, Song CL. Pathophysiological mechanism of acute bone loss after fracture. J Adv Res 2023; 49:63-80. [PMID: 36115662 PMCID: PMC10334135 DOI: 10.1016/j.jare.2022.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Acute bone loss after fracture is associated with various effects on the complete recovery process and a risk of secondary fractures among patients. Studies have reported similarities in pathophysiological mechanisms involved in acute bone loss after fractures and osteoporosis. However, given the silence nature of bone loss and bone metabolism complexities, the actual underlying pathophysiological mechanisms have yet to be fully elucidated. AIM OF REVIEW To elaborate the latest findings in basic research with a focus on acute bone loss after fracture. To briefly highlight potential therapeutic targets and current representative drugs. To arouse researchers' attention and discussion on acute bone loss after fracture. KEY SCIENTIFIC CONCEPTS OF REVIEW Bone loss after fracture is associated with immobilization, mechanical unloading, blood supply damage, sympathetic nerve regulation, and crosstalk between musculoskeletals among other factors. Current treatment strategies rely on regulation of osteoblasts and osteoclasts, therefore, there is a need to elucidate on the underlying mechanisms of acute bone loss after fractures to inform the development of efficacious and safe drugs. In addition, attention should be paid towards ensuring long-term skeletal health.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
| |
Collapse
|
4
|
Brown M, Zhu S, Taylor L, Tabrizian M, Li-Jessen NY. Unraveling the Relevance of Tissue-Specific Decellularized Extracellular Matrix Hydrogels for Vocal Fold Regenerative Biomaterials: A Comprehensive Proteomic and In Vitro Study. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200095. [PMID: 37547672 PMCID: PMC10398787 DOI: 10.1002/anbr.202200095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Decellularized extracellular matrix (dECM) is a promising material for tissue engineering applications. Tissue-specific dECM is often seen as a favorable material that recapitulates a native-like microenvironment for cellular remodeling. However, the minute quantity of dECM derivable from small organs like the vocal fold (VF) hampers manufacturing scalability. Small intestinal submucosa (SIS), a commercial product with proven regenerative capacity, may be a viable option for VF applications. This study aims to compare dECM hydrogels derived from SIS or VF tissue with respect to protein content and functionality using mass spectrometry-based proteomics and in vitro studies. Proteomic analysis reveals that VF and SIS dECM share 75% of core matrisome proteins. Although VF dECM proteins have greater overlap with native VF, SIS dECM shows less cross-sample variability. Following decellularization, significant reductions of soluble collagen (61%), elastin (81%), and hyaluronan (44%) are noted in VF dECM. SIS dECM contains comparable elastin and hyaluronan but 67% greater soluble collagen than VF dECM. Cells deposit more neo-collagen on SIS than VF-dECM hydrogels, whereas neo-elastin (~50 μg/scaffold) and neo-hyaluronan (~ 6 μg/scaffold) are comparable between the two hydrogels. Overall, SIS dECM possesses reasonably similar proteomic profile and regenerative capacity to VF dECM. SIS dECM is considered a promising alternative for dECM-derived biomaterials for VF regeneration.
Collapse
Affiliation(s)
- Mika Brown
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
| | - Shirley Zhu
- Department of Microbiology and Immunology 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| | - Lorne Taylor
- The Proteomics Platform, McGill University Health Center 1001 Decarie Boulevard Montreal Suite E01.5056 Montreal, Quebec, H4A 3J1, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- Department of Bioengineering, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
- Faculty of Dentistry, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
| | - Nicole Y.K. Li-Jessen
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- School of Communication Sciences and Disorders, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Department of Otolaryngology - Head and Neck Surgery, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Research Institute of McGill University Health Center, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| |
Collapse
|
5
|
Shainer R, Kram V, Kilts TM, Li L, Doyle AD, Shainer I, Martin D, Simon CG, Zeng-Brouwers J, Schaefer L, Young MF. Biglycan regulates bone development and regeneration. Front Physiol 2023; 14:1119368. [PMID: 36875017 PMCID: PMC9979216 DOI: 10.3389/fphys.2023.1119368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Endochondral bone development and regeneration relies on activation and proliferation of periosteum derived-cells (PDCs). Biglycan (Bgn), a small proteoglycan found in extracellular matrix, is known to be expressed in bone and cartilage, however little is known about its influence during bone development. Here we link biglycan with osteoblast maturation starting during embryonic development that later affects bone integrity and strength. Biglycan gene deletion reduced the inflammatory response after fracture, leading to impaired periosteal expansion and callus formation. Using a novel 3D scaffold with PDCs, we found that biglycan could be important for the cartilage phase preceding bone formation. The absence of biglycan led to accelerated bone development with high levels of osteopontin, which appeared to be detrimental to the structural integrity of the bone. Collectively, our study identifies biglycan as an influencing factor in PDCs activation during bone development and bone regeneration after fracture.
Collapse
Affiliation(s)
- Reut Shainer
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Vardit Kram
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Tina M. Kilts
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Li Li
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Andrew D. Doyle
- NIDCR Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Inbal Shainer
- Department Genes-Circuits-Behavior, Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Daniel Martin
- NIDCD/NIDCR Genomics and Computational Biology Core, National Institutes of Health, Bethesda, MD, United States
| | - Carl G. Simon
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| | - Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt, Germany
| | - Marian F. Young
- Molecular Biology of Bones and Teeth Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | | |
Collapse
|
6
|
Liu Y, Qin ZQ, Zheng Y, Wu J, Yang G, Tan Q, Zhu G, Liu K, Mei H. New insights into pathogenesis of congenital pseudarthrosis of tibia in children using periosteum proteomics analysis. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9374. [PMID: 35933588 DOI: 10.1002/rcm.9374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/28/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
RATIONALE The exact etiology and pathogenesis of congenital pseudarthrosis of tibia (CPT) are not clear. Quantitative proteomics analysis plays a vital role in disease pathology research. Tandem mass tag (TMT)-based proteomics techniques were employed to identify and analyze the differentially expressed proteins (DEP) in the tibia periosteum tissues of CPT patients. METHODS The samples were divided into three groups: CPT with NF1 group, CPT without NF1 group (non-NF1-CPT), and control group (patients with open tibial fracture). A fold change ≥1.5 or ≤0.66 and P-value <0.05 were used as the thresholds to screen DEPs. Subsequently, bioinformatics resources such as online tools DAVID and String were used to generate gene ontology (GO) annotation, KEGG pathways enrichment, and protein-protein interaction (PPI) network for these DEPs. RESULTS The results show that a total of 347 proteins were differentially expressed in NF1-CPT groups, 212 of which were upregulated and 135 were downregulated. There were more DEPs in non-NF1-CPT groups; we identified 467 DEPs, including 281 upregulated and 186 downregulated. Among them, NF1-CPT groups and non-NF1-CPT groups shared 231 DEPs, and the remaining 230 DEPs showed the same expression trend in the two disease groups, with 117 upregulated and 113 downregulated. In particular, 116 proteins were altered only in NF1-CPT groups (94 were upregulated and 22 were downregulated), whereas 236 proteins were altered only in non-NF1-CPT groups (164 were upregulated and 72 were downregulated). Finally, compared with non-NF1-CPT groups, 47 proteins changed 1.5-fold and P-value < 0.05 in NF1-CPT groups. CONCLUSIONS To sum up, we found that common DEPS in periosteum of NF1-CPT and non-NF1-CPT groups are mainly involved in cell matrix assembly, cell adhesion, AKT-PI3K signal pathway activation, and vascular agglutination, which indicate that these are the pathological characteristics of CPT. The osteogenic ability is weak, the osteoclastic ability is strong, the vascular lumen is narrow, the invasive growth and the proliferation of fibroblasts are enhanced in CPT patients.
Collapse
Affiliation(s)
- Yaoxi Liu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Zhen Qi Qin
- Medical School, Fuyang Normal University, Fuyang, China
| | - Yu Zheng
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Jiangyan Wu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Ge Yang
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Qian Tan
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Guanghui Zhu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Kun Liu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| | - Haibo Mei
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The School of Pediatrics, Heng Yang Medical School, University of South China, Changsha City, Hunan Province, 410007, China
| |
Collapse
|
7
|
Hu S, Li P, Wang C, Liu X. Expression and the Prognostic Value of Biglycan in Gastric Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2656480. [PMID: 36110576 PMCID: PMC9470332 DOI: 10.1155/2022/2656480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022]
Abstract
Background Biglycan (BGN) is a family member of small leucine-rich repeat proteoglycans. High expression of BGN might enhance the invasion and metastasis in some types of tumors. Here, the prognostic significance of BGN was evaluated in gastric cancer. Material and Methods. Two independent Gene Expression Omnibus (GEO) gastric cancer microarray datasets (n = 64 and n = 432) were collected for this study. Kaplan-Meier analysis was applied to evaluate if BGN impacts the outcomes of gastric cancer. Protein-protein interaction (PPI) analysis was performed on gastric cancer-related genes and BGN targets, and those interactions with confidence interval (CI) ≥ 0.7 were chosen to construct a PPI network. The gene set enrichment analysis (GSEA) was used to explore BGN and cancer-related gene signatures. Gene Transcription Regulation Database (GTRD) and ALGGEN-PROMO predicted the transcription factor binding sites (TFBSs) of the BGN promoter. BGN protein level in gastric cancer tissue was determined by immunohistochemistry (IHC). Bioinformatic analysis predicted the putative TFs of BGN. Results For gastric cancer, the mRNA expression level of BGN in tumor tissue was significantly higher than that in normal tissue. Kaplan-Meier analysis showed that higher expression of BGN mRNA was significantly associated with more reduced recurrence-free survival (RFS). GSEA results suggested that BGN was significantly enriched in gene signatures related to metastasis and poor prognosis, revealing that BGN might be associated with cell proliferation, poor differentiation, and high invasiveness of gastric cancer. Meanwhile, the putative TFs, including AR, E2F1, and TCF4, were predicted by bioinformatic analysis and also significantly correlated with expression of BGN in mRNA levels. Conclusion High expression of BGN mRNA was significantly related to poor prognosis, which suggested that BGN was a potential prognostic biomarker and therapeutic target of gastric cancer.
Collapse
Affiliation(s)
- Sizhe Hu
- Department of Gastrointestinal Surgery, Affiliated Dongyang People's Hospital of Wenzhou Medical University, Dongyang, Zhejiang 322100, China
| | - Peipei Li
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China
| | - Chenying Wang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, China
| | - Xiyong Liu
- Sino-American Cancer Foundation, Covina, CA 91722, USA
| |
Collapse
|
8
|
Beach ZM, Dekhne MS, Rodriguez AB, Weiss SN, Adams TH, Adams SM, Sun M, Birk DE, Soslowsky LJ. Decorin knockdown is beneficial for aged tendons in the presence of biglycan expression. Matrix Biol Plus 2022; 15:100114. [PMID: 35818471 PMCID: PMC9270257 DOI: 10.1016/j.mbplus.2022.100114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/19/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
Decorin and biglycan are two major small leucine-rich proteoglycans (SLRPs) present in the tendon extracellular matrix that facilitate collagen fibrillogenesis, tissue turnover, and cell signal transduction. Previously, we demonstrated that knockout of decorin prevented the decline of tendon mechanical properties that are associated with aging. The objective of this study was to determine the effects of decorin and biglycan knockdown on tendon structure and mechanics in aged tendons using tamoxifen-inducible knockdown models. We hypothesized that the knockdown of decorin and compound knockdown of decorin and biglycan would prevent age-related declines in tendon mechanics and structure compared to biglycan knockdown and wild-type controls, and that these changes would be exacerbated as the tendons progress towards geriatric ages. To achieve this objective, we created tamoxifen-inducible mouse knockdown models to target decorin and biglycan gene inactivation without the abnormal tendon development associated with traditional knockout models. Knockdown of decorin led to increased midsubstance modulus and decreased stress relaxation in aged tendons. However, these changes were not sustained in the geriatric tendons. Knockdown in biglycan led to no changes in mechanics in the aged or geriatric tendons. Contrary to our hypothesis, the compound decorin/biglycan knockdown tendons did not resemble the decorin knockdown tendons, but resulted in increased viscoelastic properties in the aged and geriatric tendons. Structurally, knockdown of SLRPs, except for the 570d I-Dcn-/-/Bgn-/- group, resulted in alterations to the collagen fibril diameter relative to wild-type controls. Overall, this study identified the differential roles of decorin and biglycan throughout tendon aging in the maintenance of tendon structural and mechanical properties and revealed that the compound decorin and biglycan knockdown phenotype did not resemble the single gene decorin or biglycan models and was detrimental to tendon properties throughout aging.
Collapse
|
9
|
Enthesis Healing Is Dependent on Scaffold Interphase Morphology—Results from a Rodent Patellar Model. Cells 2022; 11:cells11111752. [PMID: 35681447 PMCID: PMC9179925 DOI: 10.3390/cells11111752] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
The use of multiphasic scaffolds to treat injured tendon-to-bone entheses has shown promising results in vitro. Here, we used two versions of a biphasic silk fibroin scaffold to treat an enthesis defect created in a rat patellar model in vivo. One version presented a mixed transition between the bony and the tendon end of the construct (S-MT) while this transition was abrupt in the second version (S-AT). At 12 weeks after surgery, the S-MT scaffold promoted better healing of the injured enthesis, with minimal undesired ossification of the insertion area. The expression of tenogenic and chondrogenic markers was sustained for longer in the S-MT-treated group and the tangent modulus of the S-MT-treated samples was similar to the native tissue at 12 weeks while that of the S-AT-treated enthesis was lower. Our study highlights the important role of the transition zone of multiphasic scaffolds in the treatment of complex interphase tissues such as the tendon-to-bone enthesis.
Collapse
|
10
|
Briffa JF, Bevens W, Gravina S, Said JM, Wlodek ME. Pregnant biglycan knockout mice have altered cardiorenal adaptations and a shorter gestational length, but do not develop a pre-eclamptic phenotype. Placenta 2022; 119:52-62. [PMID: 35150975 DOI: 10.1016/j.placenta.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Pre-eclampsia complicates 4.6% of pregnancies and is linked to impaired placentation; likely due to dysregulated vasculogenesis/angiogenesis. Proteoglycans, such as biglycan, are located on the endothelial surface of fetal capillaries. Biglycan is reduced in the placenta of pregnancies complicated by fetal growth restriction and pre-eclampsia. Importantly, biglycan stimulates angiogenesis in numerous tissues. Therefore, this study investigated whether biglycan knockdown in mice results in a pre-eclamptic phenotype. METHODS Wild-type (WT) and Bgn-/- mice underwent cardiorenal measurements prior to and during pregnancy. One cohort of mice underwent post-mortem on gestational day 18 (E18) and another cohort underwent post-mortem on postnatal day 1 (PN1), with maternal and offspring tissues of relevance collected. RESULTS Bgn-/- dams had increased heart rate (+9%, p < 0.037) and reduced systolic (-11%, p < 0.001), diastolic (-15%, p < 0.001), and mean arterial (-12%, p < 0.001) pressures at all ages investigated compared to WT. Additionally, Bgn-/- dams had reduced urine flow rate (-64%, p < 0.001) as well as reduced urinary excretions (-49%, p < 0.004) during late gestation compared to WT. Bgn-/- pups had higher body weight (+8%, p = 0.004; E18 only) and a higher liver-to-brain weight ratio (+43%, p < 0.001). Placental weight was unaltered with only minor changes in vasculogenic and angiogenic gene abundances detected, which did not correlate to changes in protein expression. DISCUSSION This study demonstrated that total knockdown of biglycan is not associated with features of pre-eclampsia.
Collapse
Affiliation(s)
- J F Briffa
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - W Bevens
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Gravina
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - J M Said
- Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, 3010, Australia; Maternal Fetal Medicine, Sunshine Hospital, Western Health, St Albans, VIC, 3021, Australia
| | - M E Wlodek
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, 3010, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
11
|
Chen CG, Iozzo RV. Extracellular matrix guidance of autophagy: a mechanism regulating cancer growth. Open Biol 2022; 12:210304. [PMID: 34982945 PMCID: PMC8727153 DOI: 10.1098/rsob.210304] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix (ECM) exists as a dynamic network of biophysical and biochemical factors that maintain tissue homeostasis. Given its sensitivity to changes in the intra- and extracellular space, the plasticity of the ECM can be pathological in driving disease through aberrant matrix remodelling. In particular, cancer uses the matrix for its proliferation, angiogenesis, cellular reprogramming and metastatic spread. An emerging field of matrix biology focuses on proteoglycans that regulate autophagy, an intracellular process that plays both critical and contextual roles in cancer. Here, we review the most prominent autophagic modulators from the matrix and the current understanding of the cellular pathways and signalling cascades that mechanistically drive their autophagic function. We then critically assess how their autophagic functions influence tumorigenesis, emphasizing the complexities and stage-dependent nature of this relationship in cancer. We highlight novel emerging data on immunoglobulin-containing and proline-rich receptor-1, heparanase and thrombospondin 1 in autophagy and cancer. Finally, we further discuss the pro- and anti-autophagic modulators originating from the ECM, as well as how these proteoglycans and other matrix constituents specifically influence cancer progression.
Collapse
Affiliation(s)
- Carolyn G. Chen
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
12
|
Abstract
Understanding the properties of bone is of both fundamental and clinical relevance. The basis of bone’s quality and mechanical resilience lies in its nanoscale building blocks (i.e., mineral, collagen, non-collagenous proteins, and water) and their complex interactions across length scales. Although the structure–mechanical property relationship in healthy bone tissue is relatively well characterized, not much is known about the molecular-level origin of impaired mechanics and higher fracture risks in skeletal disorders such as osteoporosis or Paget’s disease. Alterations in the ultrastructure, chemistry, and nano-/micromechanics of bone tissue in such a diverse group of diseased states have only been briefly explored. Recent research is uncovering the effects of several non-collagenous bone matrix proteins, whose deficiencies or mutations are, to some extent, implicated in bone diseases, on bone matrix quality and mechanics. Herein, we review existing studies on ultrastructural imaging—with a focus on electron microscopy—and chemical, mechanical analysis of pathological bone tissues. The nanometric details offered by these reports, from studying knockout mice models to characterizing exact disease phenotypes, can provide key insights into various bone pathologies and facilitate the development of new treatments.
Collapse
|
13
|
Cui D, Kong N, Ding L, Guo Y, Yang W, Yan F. Ultrathin 2D Titanium Carbide MXene (Ti 3 C 2 T x ) Nanoflakes Activate WNT/HIF-1α-Mediated Metabolism Reprogramming for Periodontal Regeneration. Adv Healthc Mater 2021; 10:e2101215. [PMID: 34586717 PMCID: PMC11468541 DOI: 10.1002/adhm.202101215] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/08/2021] [Indexed: 12/17/2022]
Abstract
Periodontal defect regeneration in severe periodontitis relies on the differentiation and proliferation of periodontal ligament cells (PDLCs). Recently, an emerging 2D nanomaterial, MXene (Ti3 C2 Tx ), has gained more and more attention due to the extensive antibacterial and anticancer activity, while its potential biomedical application on tissue regeneration remains unclear. Through a combination of experimental and multiscale simulation schemes, Ti3 C2 Tx has exhibited satisfactory biocompatibility and induced distinguish osteogenic differentiation of human PDLCs (hPDLCs), with upregulated osteogenesis-related genes. Ti3 C2 Tx manages to activate the Wnt/β-catenin signaling pathway by enhancing the Wnt-Frizzled complex binding, thus stabilizing HIF-1α and altering metabolic reprogramming into glycolysis. In vivo, hPDLCs pretreated by Ti3 C2 Tx display excellent performance in new bone formation and osteoclast inhibition with enhanced RUNX2, HIF-1α, and β-catenin in an experimental rat model of periodontal fenestration defects, indicating that this material has high efficiency of periodontal regeneration promotion. It is demonstrated in this work that Ti3 C2 Tx has highly efficient therapeutic effects in osteogenic differentiation and periodontal defect repairment.
Collapse
Affiliation(s)
- Di Cui
- Nanjing Stomatological HospitalMedical School of Nanjing UniversityNanjingJiangsu210008China
| | - Na Kong
- School of Life and Environmental ScienceDeakin UniversityWaurn PondsVictoria3216Australia
| | - Liang Ding
- Nanjing Stomatological HospitalMedical School of Nanjing UniversityNanjingJiangsu210008China
| | - Yachong Guo
- Kuang Yaming Honors SchoolNanjing UniversityNanjing210023China
- Institute Theory of PolymersLeibniz‐Institut für Polymerforschung DresdenDresden01069Germany
| | - Wenrong Yang
- School of Life and Environmental ScienceDeakin UniversityWaurn PondsVictoria3216Australia
| | - Fuhua Yan
- Nanjing Stomatological HospitalMedical School of Nanjing UniversityNanjingJiangsu210008China
| |
Collapse
|
14
|
Roles of Two Small Leucine-Rich Proteoglycans Decorin and Biglycan in Pregnancy and Pregnancy-Associated Diseases. Int J Mol Sci 2021; 22:ijms221910584. [PMID: 34638928 PMCID: PMC8509074 DOI: 10.3390/ijms221910584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/18/2022] Open
Abstract
Two small leucine-rich proteoglycans (SLRP), decorin and biglycan, play important roles in structural–functional integrity of the placenta and fetal membranes, and their alterations can result in several pregnancy-associated diseases. In this review, we briefly discuss normal placental structure and functions, define and classify SLRPs, and then focus on two SLRPs, decorin (DCN) and biglycan (BGN). We discuss the consequences of deletions/mutations of DCN and BGN. We then summarize DCN and BGN expression in the pregnant uterus, myometrium, decidua, placenta, and fetal membranes. Actions of these SLRPs as ligands are then discussed in the context of multiple binding partners in the extracellular matrix and cell surface (receptors), as well as their alterations in pathological pregnancies, such as preeclampsia, fetal growth restriction, and preterm premature rupture of membranes. Lastly, we raise some unanswered questions as food for thought.
Collapse
|
15
|
Neill T, Kapoor A, Xie C, Buraschi S, Iozzo RV. A functional outside-in signaling network of proteoglycans and matrix molecules regulating autophagy. Matrix Biol 2021; 100-101:118-149. [PMID: 33838253 PMCID: PMC8355044 DOI: 10.1016/j.matbio.2021.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Proteoglycans and selected extracellular matrix constituents are emerging as intrinsic and critical regulators of evolutionarily conversed, intracellular catabolic pathways. Often, these secreted molecules evoke sustained autophagy in a variety of cell types, tissues, and model systems. The unique properties of proteoglycans have ushered in a paradigmatic shift to broaden our understanding of matrix-mediated signaling cascades. The dynamic cellular pathway controlling autophagy is now linked to an equally dynamic and fluid signaling network embedded in a complex meshwork of matrix molecules. A rapidly emerging field of research encompasses multiple matrix-derived candidates, representing a menagerie of soluble matrix constituents including decorin, biglycan, endorepellin, endostatin, collagen VI and plasminogen kringle 5. These matrix constituents are pro-autophagic and simultaneously anti-angiogenic. In contrast, perlecan, laminin α2 chain, and lumican have anti-autophagic functions. Mechanistically, each matrix constituent linked to intracellular catabolic events engages a specific cell surface receptor that often converges on a common core of the autophagic machinery including AMPK, Peg3 and Beclin 1. We consider this matrix-evoked autophagy as non-canonical given that it occurs in an allosteric manner and is independent of nutrient availability or prevailing bioenergetics control. We propose that matrix-regulated autophagy is an important outside-in signaling mechanism for proper tissue homeostasis that could be therapeutically leveraged to combat a variety of diseases.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christopher Xie
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
16
|
Chen CG, Iozzo RV. Angiostatic cues from the matrix: Endothelial cell autophagy meets hyaluronan biology. J Biol Chem 2020; 295:16797-16812. [PMID: 33020183 PMCID: PMC7864073 DOI: 10.1074/jbc.rev120.014391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/02/2020] [Indexed: 01/21/2023] Open
Abstract
The extracellular matrix encompasses a reservoir of bioactive macromolecules that modulates a cornucopia of biological functions. A prominent body of work posits matrix constituents as master regulators of autophagy and angiogenesis and provides molecular insight into how these two processes are coordinated. Here, we review current understanding of the molecular mechanisms underlying hyaluronan and HAS2 regulation and the role of soluble proteoglycan in affecting autophagy and angiogenesis. Specifically, we assess the role of proteoglycan-evoked autophagy in regulating angiogenesis via the HAS2-hyaluronan axis and ATG9A, a novel HAS2 binding partner. We discuss extracellular hyaluronan biology and the post-transcriptional and post-translational modifications that regulate its main synthesizer, HAS2. We highlight the emerging group of proteoglycans that utilize outside-in signaling to modulate autophagy and angiogenesis in cancer microenvironments and thoroughly review the most up-to-date understanding of endorepellin signaling in vascular endothelia, providing insight into the temporal complexities involved.
Collapse
Affiliation(s)
- Carolyn G Chen
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
17
|
Johnston E, Buckley M. Relative Protein Abundances and Biological Ageing in Whole Skeletal Elements. J Proteome Res 2020; 20:538-548. [PMID: 33089684 DOI: 10.1021/acs.jproteome.0c00555] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Establishing biological age is an integral part of forensic investigations, currently achieved through morphological methods with varying degrees of accuracy. Furthermore, biological ageing is much easier in juveniles than in adults, at which point traditional ageing methods struggle. Therefore, biomolecular approaches are considered of great interest, with several protein markers already recognized for their potential forensic significance. However, previous studies have typically relied on subsampling different parts of skeletal elements. Here, we attempt to evaluate the proteome of complete elements using a rat model. In the analysis of specimens spanning beyond adulthood (1 week to 1.5 years), we observed 729 unique proteins across 33 samples (three for each sex for each of the five (female) or six (male)), five of which represent newly identified proteins in relation to age estimation: vimentin, osteopontin, matrilin-1, apolipoprotein A-I, and prothrombin. Most of these follow the trend of decreasing abundance through age, with the exception of prothrombin that increases. We consider the combined use of these relative abundances, along with those of previously noted fetuin-A, biglycan, albumin, and chromogranin-A signatures, as being of potential value to the development of an age estimation tool worthy of further evaluation in forensic contexts.
Collapse
Affiliation(s)
- Elizabeth Johnston
- Department of Earth and Environmental Sciences, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Michael Buckley
- Department of Earth and Environmental Sciences, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| |
Collapse
|
18
|
Pechanec MY, Boyd TN, Baar K, Mienaltowski MJ. Adding exogenous biglycan or decorin improves tendon formation for equine peritenon and tendon proper cells in vitro. BMC Musculoskelet Disord 2020; 21:627. [PMID: 32967653 PMCID: PMC7513506 DOI: 10.1186/s12891-020-03650-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Background Tendon injuries amount to one of the leading causes of career-ending injuries in horses due to the inability for tendon to completely repair and the high reinjury potential. As a result, novel therapeutics are necessary to improve repair with the goal of decreasing leg lameness and potential reinjury. Small leucine-rich repeat proteoglycans (SLRPs), a class of regulatory molecules responsible for collagen organization and maturation, may be one such therapeutic to improve tendon repair. Before SLRP supplementation can occur in vivo, proper evaluation of the effect of these molecules in vitro needs to be assessed. The objective of this study was to evaluate the effectiveness of purified bovine biglycan or decorin on tendon proper and peritenon cell populations in three-dimensional tendon constructs. Methods Equine tendon proper or peritenon cell seeded fibrin three-dimensional constructs were supplemented with biglycan or decorin at two concentrations (5 nM or 25 nM). The functionality and ultrastructural morphology of the constructs were assessed using biomechanics, collagen content analysis, transmission electron microscopy (TEM), and gene expression by real time – quantitative polymerase chain reaction (RT-qPCR). Results SLRP supplementation affected both tendon proper and peritenon cells-seeded constructs. With additional SLRPs, material and tensile properties of constructs strengthened, though ultrastructural analyses indicated production of similar-sized or smaller fibrils. Overall expression of tendon markers was bolstered more in peritenon cells supplemented with either SLRP, while supplementation of SLRPs to TP cell-derived constructs demonstrated fewer changes in tendon and extracellular matrix markers. Moreover, relative to non-supplemented tendon proper cell-seeded constructs, SLRP supplementation of the peritenon cells showed increases in mechanical strength, material properties, and collagen content. Conclusions The SLRP-supplemented peritenon cells produced constructs with greater mechanical and material properties than tendon proper seeded constructs, as well as increased expression of matrix assembly molecules. These findings provide evidence that SLRPs should be further investigated for their potential to improve tendon formation in engineered grafts or post-injury.
Collapse
Affiliation(s)
- Monica Y Pechanec
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Tannah N Boyd
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Keith Baar
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, 195 Briggs Hall, One Shields Ave, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California Davis School of Medicine, 195 Briggs Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Michael J Mienaltowski
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
19
|
Abstract
The skeleton is highly vascularized due to the various roles blood vessels play in the homeostasis of bone and marrow. For example, blood vessels provide nutrients, remove metabolic by-products, deliver systemic hormones, and circulate precursor cells to bone and marrow. In addition to these roles, bone blood vessels participate in a variety of other functions. This article provides an overview of the afferent, exchange and efferent vessels in bone and marrow and presents the morphological layout of these blood vessels regarding blood flow dynamics. In addition, this article discusses how bone blood vessels participate in bone development, maintenance, and repair. Further, mechanical loading-induced bone adaptation is presented regarding interstitial fluid flow and pressure, as regulated by the vascular system. The role of the sympathetic nervous system is discussed in relation to blood vessels and bone. Finally, vascular participation in bone accrual with intermittent parathyroid hormone administration, a medication prescribed to combat age-related bone loss, is described and age- and disease-related impairments in blood vessels are discussed in relation to bone and marrow dysfunction. © 2020 American Physiological Society. Compr Physiol 10:1009-1046, 2020.
Collapse
Affiliation(s)
- Rhonda D Prisby
- Bone Vascular and Microcirculation Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
20
|
Kram V, Shainer R, Jani P, Meester JAN, Loeys B, Young MF. Biglycan in the Skeleton. J Histochem Cytochem 2020; 68:747-762. [PMID: 32623936 DOI: 10.1369/0022155420937371] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Small leucine rich proteoglycans (SLRPs), including Biglycan, have key roles in many organ and tissue systems. The goal of this article is to review the function of Biglycan and other related SLRPs in mineralizing tissues of the skeleton. The review is divided into sections that include Biglycan's role in structural biology, signaling, craniofacial and long bone homeostasis, remodeled skeletal tissues, and in human genetics. While many cell types in the skeleton are now known to be affected by Biglycan, there are still unanswered questions about its mechanism of action(s).
Collapse
Affiliation(s)
- Vardit Kram
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Reut Shainer
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Priyam Jani
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| | - Josephina A N Meester
- Laboratory of Cardiogenetics, Center of Medical Genetics, University Hospital Antwerp, University of Antwerp, Antwerp, Belgium
| | - Bart Loeys
- Laboratory of Cardiogenetics, Center of Medical Genetics, University Hospital Antwerp, University of Antwerp, Antwerp, Belgium
| | - Marian F Young
- Molecular Biology of Bones and Teeth Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, U.S. Department of Health & Human Services, Bethesda, Maryland
| |
Collapse
|
21
|
Abstract
The present work focuses on the application of time-of-flight secondary ion mass spectrometry (ToF-SIMS) in osteoporotic bone research. In order to demonstrate the benefit, the authors present concrete application examples of ToF-SIMS in three different areas of bone research. ToF-SIMS as a mass spectrometric imaging technique allows simultaneous visualization of mineralized and nonmineralized bone tissue as well as implanted biomaterials and bone implant interphases. In the first example, the authors show that it is possible to study the incorporation and distribution of different components released from bone filler materials into bone with a single mass spectrometric measurement. This not only enables imaging of nonstained bone cross sections but also provides further insights beyond histologically obtained information. Furthermore, they successfully identified several mass fragments as markers for newly formed cartilage tissue and growth joint in bone. Different modes of ToF-SIMS as well as different SIMS instruments (IONTOF's TOF.SIMS 5 and M6 Hybrid SIMS, Ionoptika's J105) were used to identify these mass signals and highlight the high versatility of this method. In the third part, bone structure of cortical rat bone was investigated from bone sections embedded in technovit (polymethyl methacrylate, PMMA) and compared to cryosections. In cortical bone, they were able to image different morphological features, e.g., concentric arrangement of collagen fibers in so-called osteons as well as Haversian canals and osteocytes. In summary, the study provides examples of application and shows the strength of ToF-SIMS as a promising analytical method in the field of osteoporotic bone research.
Collapse
|
22
|
Pellicani R, Poletto E, Andreuzzi E, Paulitti A, Doliana R, Bizzotto D, Braghetta P, Colladel R, Tarticchio G, Sabatelli P, Bucciotti F, Bressan G, Iozzo RV, Colombatti A, Bonaldo P, Mongiat M. Multimerin-2 maintains vascular stability and permeability. Matrix Biol 2020; 87:11-25. [DOI: 10.1016/j.matbio.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022]
|
23
|
Xue H, Niu P, Liu Y, Sun Y. Glycosylation of DMP1 promotes bone reconstruction in long bone defects. Biochem Biophys Res Commun 2020; 526:1125-1130. [PMID: 32331833 DOI: 10.1016/j.bbrc.2020.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/08/2020] [Indexed: 02/04/2023]
Abstract
The regeneration of bone defects is necessary for the successful healing. During the process of healing, callus plays crucial roles in providing the stable bone-reconstruction environment. The callus is consisted of various large molecules including collagen proteins, non-collagen proteins and proteoglycans (PGs), which are involved in maintaining mechanical strength and interacting with cytokines and grow factors in the injury sites. Recently, our data have found that the PG form of Dentin Matrix Protein 1 (DMP1-PG), which is a newly identified PG, was richly expressed in the bone defect sites. Previous researches have demonstrated the special role of DMP1-PG in chondrogenesis and endochondral ossification, however, the knowledge about the role of DMP1-PG in bone defect repair is still limited. To further detect the potential function of DMP1-PG in the defect healing, we employed a bone defect intramembranous ossification model using the glycosylation site mutant DMP1-PG (S89-G89, S89G-DMP1) mouse. The morphologic changes of calluses and abnormal expression levels of osteogenesis genes were displayed in the injury sites in S89G-DMP1 mice. In addition, impaired BMP-Smad signaling pathway was observed due to the deficiency of DMP1-PG. Collectively, our findings indicated that the DMP1-PG is one of key proteoglycans in the process of defect healing via regulating the osteogenesis.
Collapse
Affiliation(s)
- Hui Xue
- Department of Stomatology, The First Affiliated Hospital of Qiqihaer Medical University, Qiqihaer, Heilongjiang, 161041, China; Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| | - Pingping Niu
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Yang Liu
- Department of Stomatology, The First Affiliated Hospital of Qiqihaer Medical University, Qiqihaer, Heilongjiang, 161041, China
| | - Yao Sun
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.
| |
Collapse
|
24
|
Pang X, Dong N, Zheng Z. Small Leucine-Rich Proteoglycans in Skin Wound Healing. Front Pharmacol 2020; 10:1649. [PMID: 32063855 PMCID: PMC6997777 DOI: 10.3389/fphar.2019.01649] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Healing of cutaneous wounds is a complex and well-coordinated process requiring cooperation among multiple cells from different lineages and delicately orchestrated signaling transduction of a diversity of growth factors, cytokines, and extracellular matrix (ECM) at the wound site. Most skin wound healing in adults is imperfect, characterized by scar formation which results in significant functional and psychological sequelae. Thus, the reconstruction of the damaged skin to its original state is of concern to doctors and scientists. Beyond the traditional treatments such as corticosteroid injection and radiation therapy, several growth factors or cytokines-based anti-scarring products are being or have been tested in clinical trials to optimize skin wound healing. Unfortunately, all have been unsatisfactory to date. Currently, accumulating evidence suggests that the ECM not only functions as the structural component of the tissue but also actively modulates signal transduction and regulates cellular behaviors, and thus, ECM should be considered as an alternative target for wound management pharmacotherapy. Of particular interest are small leucine-rich proteoglycans (SLRPs), a group of the ECM, which exist in a wide range of connecting tissues, including the skin. This manuscript summarizes the most current knowledge of SLRPs regarding their spatial-temporal expression in the skin, as well as lessons learned from the genetically modified animal models simulating human skin pathologies. In this review, particular focus is given on the diverse roles of SLRP in skin wound healing, such as anti-inflammation, pro-angiogenesis, pro-migration, pro-contraction, and orchestrate transforming growth factor (TGF)β signal transduction, since cumulative investigations have indicated their therapeutic potential on reducing scar formation in cutaneous wounds. By conducting this review, we intend to gain insight into the potential application of SLRPs in cutaneous wound healing management which may pave the way for the development of a new generation of pharmaceuticals to benefit the patients suffering from skin wounds and their sequelae.
Collapse
Affiliation(s)
- Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nuo Dong
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
25
|
Abstract
Purpose: Biglycan is a proteoglycan of the small leucine-rich repeat family. It is present in all connective tissues and plays key structural and signaling roles. This review aimed to compile available evidence in the characteristics and distribution of biglycan and its glycosylated and non-glycosylated forms in connective tissues with a specific focus on the contribution to homeostasis of bone and changes of biglycan structure with aging.Methods: The Pubmed database was searched and included the terms "biglycan", "proteoglycans", "glycosaminoglycans", "bone", "osteoblast", "osteocyte", "osteoclast", "aging", "inflammation", "cartilage". Abstracts were appraised and a series of original articles and reviews studied to generate this narrative review.Results: Based on the search, biglycan significantly affects bone development and homeostasis and can be significantly changed by the aging process in several connective tissues, which in turn affects the behavior of tissue and cell responses in aged networks. Further, as the understanding of the various forms of biglycan in vivo is expanded and the function of its components in vitro is dissected, this proteoglycan can potentially serve as a therapeutic or biomarker molecule to detect tissue destruction.Conclusions: Biglycan is a key player in skeletal bone homeostasis, and overall, there is more evidence on the role of biglycan in development and less in the adult physiological or diseased young and aged systems. Further understanding of its conformation, degradation peptides and post-translational modifications will be required to understand the role of biglycan in bone maintenance and to support the development of treatments for age-related bone dysfunctions.
Collapse
Affiliation(s)
- Patricia A Miguez
- Adams School of Dentistry, Division of Comprehensive Oral Health, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
26
|
Rigoglio NN, Rabelo ACS, Borghesi J, de Sá Schiavo Matias G, Fratini P, Prazeres PHDM, Pimentel CMMM, Birbrair A, Miglino MA. The Tumor Microenvironment: Focus on Extracellular Matrix. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:1-38. [PMID: 32266651 DOI: 10.1007/978-3-030-40146-7_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular matrix (ECM) regulates the development and maintains tissue homeostasis. The ECM is composed of a complex network of molecules presenting distinct biochemical properties to regulate cell growth, survival, motility, and differentiation. Among their components, proteoglycans (PGs) are considered one of the main components of ECM. Its composition, biomechanics, and anisotropy are exquisitely tuned to reflect the physiological state of the tissue. The loss of ECM's homeostasis is seen as one of the hallmarks of cancer and, typically, defines transitional events in tumor progression and metastasis. In this chapter, we discuss the types of proteoglycans and their roles in cancer. It has been observed that the amount of some ECM components is increased, while others are decreased, depending on the type of tumor. However, both conditions corroborate with tumor progression and malignancy. Therefore, ECM components have an increasingly important role in carcinogenesis and this leads us to believe that their understanding may be a key in the discovery of new anti-tumor therapies. In this book, the main ECM components will be discussed in more detail in each chapter.
Collapse
Affiliation(s)
- Nathia Nathaly Rigoglio
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Carolina Silveira Rabelo
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Jessica Borghesi
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gustavo de Sá Schiavo Matias
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Paula Fratini
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Alexander Birbrair
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
27
|
Stovall KE, Tran TDN, Suantawee T, Yao S, Gimble JM, Adisakwattana S, Cheng H. Adenosine triphosphate enhances osteoblast differentiation of rat dental pulp stem cells via the PLC-IP 3 pathway and intracellular Ca 2+ signaling. J Cell Physiol 2019; 235:1723-1732. [PMID: 31301074 DOI: 10.1002/jcp.29091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/11/2019] [Indexed: 01/01/2023]
Abstract
Intracellular Ca2+ signals are essential for stem cell function and play a significant role in the differentiation process. Dental pulp stem cells (DPSCs) are a potential source of stem cells; however, the mechanisms controlling cell differentiation remain largely unknown. Utilizing rat DPSCs, we examined the effect of adenosine triphosphate (ATP) on osteoblast differentiation and characterized its mechanism of action using real-time Ca 2+ imaging analysis. Our results revealed that ATP enhanced osteogenesis as indicated by Ca 2+ deposition in the extracellular matrix via Alizarin Red S staining. This was consistent with upregulation of osteoblast genes BMP2, Mmp13, Col3a1, Ctsk, Flt1, and Bgn. Stimulation of DPSCs with ATP (1-300 µM) increased intracellular Ca 2+ signals in a concentration-dependent manner, whereas histamine, acetylcholine, arginine vasopressin, carbachol, and stromal-cell-derived factor-1α failed to do so. Depletion of intracellular Ca 2+ stores in the endoplasmic reticulum by thapsigargin abolished the ATP responses which, nevertheless, remained detectable under extracellular Ca 2+ free condition. Furthermore, the phospholipase C (PLC) inhibitor U73122 and the inositol triphosphate (IP 3 ) receptor inhibitor 2-aminoethoxydiphenyl borate inhibited the Ca 2+ signals. Our findings provide a better understanding of how ATP controls osteogenesis in DPSCs, which involves a Ca 2+ -dependent mechanism via the PLC-IP 3 pathway. This knowledge could help improve osteogenic differentiation protocols for tissue regeneration of bone structures.
Collapse
Affiliation(s)
- Kelsie E Stovall
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Tran D N Tran
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Tanyawan Suantawee
- Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Jeffrey M Gimble
- LaCell LLC, New Orleans Bioinnovation Center, New Orleans, Louisiana.,Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, Louisiana
| | - Sirichai Adisakwattana
- Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Henrique Cheng
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
28
|
de Sousa Gomes P, Daugela P, Poskevicius L, Mariano L, Fernandes MH. Molecular and Cellular Aspects of Socket Healing in the Absence and Presence of Graft Materials and Autologous Platelet Concentrates: a Focused Review. J Oral Maxillofac Res 2019; 10:e2. [PMID: 31620264 PMCID: PMC6788423 DOI: 10.5037/jomr.2019.10302] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The present manuscript aims to critically detail the physiologic process of socket healing, in the absence or presence of grafting materials or platelet concentrates, addressing the associated molecular and cellular events that culminate in the restoration of the lost tissue architecture and functionality. MATERIAL AND METHODS An electronic search in the National Library of Medicine database MEDLINE through its online site PubMed and Web of Science from inception until May 2019 was conducted to identify articles concerning physiologic process of socket healing, in the absence or presence of grafting materials or platelet concentrates. The search was restricted to English language articles without time restriction. Additionally, a hand search was carried out in oral surgery, periodontology and dental implants related journals. RESULTS In total, 122 literature sources were obtained and reviewed. The detailed biological events, at the molecular and cellular level, that occur in the alveolus after tooth extraction and socket healing process modulated by grafting materials or autologous platelet concentrates were presented as two entities. CONCLUSIONS Tooth extraction initiates a convoluted set of orderly biological events in the alveolus, aiming wound closure and socket healing. The healing process comprises a wide range of events, regulated by the interplay of cytokines, chemokines and growth factors that determine cellular recruitment, proliferation and differentiation in the healing milieu, in a space- and time-dependent choreographic interplay. Additionally, the healing process may further be modulated by the implantation of grafting materials or autologous platelet concentrates within the tooth socket, aiming to enhance the regenerative outcome.
Collapse
Affiliation(s)
- Pedro de Sousa Gomes
- BoneLab, Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U. Porto, PortoPortugal.
- LAQV/REQUIMTE, Faculty of Dental Medicine, U. Porto, PortoPortugal.
| | - Povilas Daugela
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Lukas Poskevicius
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Lorena Mariano
- BoneLab, Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U. Porto, PortoPortugal.
| | - Maria Helena Fernandes
- BoneLab, Laboratory for Bone Metabolism and Regeneration, Faculty of Dental Medicine, U. Porto, PortoPortugal.
- LAQV/REQUIMTE, Faculty of Dental Medicine, U. Porto, PortoPortugal.
| |
Collapse
|
29
|
Xue H, Tao D, Weng Y, Fan Q, Zhou S, Zhang R, Zhang H, Yue R, Wang X, Wang Z, Sun Y. Glycosylation of dentin matrix protein 1 is critical for fracture healing via promoting chondrogenesis. Front Med 2019; 13:575-589. [DOI: 10.1007/s11684-019-0693-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/25/2019] [Indexed: 10/26/2022]
|
30
|
Veselá B, Švandová E, Bobek J, Lesot H, Matalová E. Osteogenic and Angiogenic Profiles of Mandibular Bone-Forming Cells. Front Physiol 2019; 10:124. [PMID: 30837894 PMCID: PMC6389724 DOI: 10.3389/fphys.2019.00124] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/31/2019] [Indexed: 11/24/2022] Open
Abstract
The mandible is a tooth-bearing structure involving one of the most prominent bones of the facial region. Mesenchymal cell condensation is the first morphological sign of osteogenesis, and several studies have focused on this stage also in the mandible. Little information is available about the early post-condensation period, during which avascular soft condensation turns into vascularized bone, and all three major bone cell types, osteoblasts, osteocytes, and osteoclasts, differentiate. In the mouse first lower molar region, the post-condensation period corresponds to the prenatal days 13–15. If during this critical period, when osteogenesis reaches the point of major bone cell differentiation, vascularization already has to play a critical role, one should be able to show molecular changes which support both types of cellular events. The aim of the present report was to follow in organ context the expression of major osteogenic and angiogenic markers and identify those that are up- or downregulated during this period. To this end, PCR Array was applied covering molecules involved in osteoblastic cell proliferation, commitment or differentiation, extracellular matrix (ECM) deposition, mineralisation, osteocyte maturation, angiogenesis, osteoclastic differentiation, and initial bone remodeling. From 161 analyzed osteogenic and angiogenic factors, the expression of 37 was altered when comparing the condensation stage with the bone stage. The results presented here provide a molecular survey of the early post-condensation stage of mandibular/alveolar bone development which has not yet been investigated in vivo.
Collapse
Affiliation(s)
- Barbora Veselá
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia.,Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Eva Švandová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia.,Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Jan Bobek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Hervé Lesot
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Eva Matalová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia.,Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| |
Collapse
|
31
|
Small leucine-rich proteoglycans and matrix metalloproteinase-14: Key partners? Matrix Biol 2019; 75-76:271-285. [DOI: 10.1016/j.matbio.2017.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 11/19/2022]
|
32
|
Abdul Kadir L, Stacey M, Barrett-Jolley R. Emerging Roles of the Membrane Potential: Action Beyond the Action Potential. Front Physiol 2018; 9:1661. [PMID: 30519193 PMCID: PMC6258788 DOI: 10.3389/fphys.2018.01661] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/02/2018] [Indexed: 01/03/2023] Open
Abstract
Whilst the phenomenon of an electrical resting membrane potential (RMP) is a central tenet of biology, it is nearly always discussed as a phenomenon that facilitates the propagation of action potentials in excitable tissue, muscle, and nerve. However, as ion channel research shifts beyond these tissues, it became clear that the RMP is a feature of virtually all cells studied. The RMP is maintained by the cell’s compliment of ion channels. Transcriptome sequencing is increasingly revealing that equally rich compliments of ion channels exist in both excitable and non-excitable tissue. In this review, we discuss a range of critical roles that the RMP has in a variety of cell types beyond the action potential. Whereas most biologists would perceive that the RMP is primarily about excitability, the data show that in fact excitability is only a small part of it. Emerging evidence show that a dynamic membrane potential is critical for many other processes including cell cycle, cell-volume control, proliferation, muscle contraction (even in the absence of an action potential), and wound healing. Modulation of the RMP is therefore a potential target for many new drugs targeting a range of diseases and biological functions from cancer through to wound healing and is likely to be key to the development of successful stem cell therapies.
Collapse
Affiliation(s)
- Lina Abdul Kadir
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Michael Stacey
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States
| | - Richard Barrett-Jolley
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
33
|
de Melo Pereira D, Habibovic P. Biomineralization-Inspired Material Design for Bone Regeneration. Adv Healthc Mater 2018; 7:e1800700. [PMID: 30240157 DOI: 10.1002/adhm.201800700] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/23/2018] [Indexed: 12/22/2022]
Abstract
Synthetic substitutes of bone grafts, such as calcium phosphate-based ceramics, have shown some good clinical successes in the regeneration of large bone defects and are currently extensively used. In the past decade, the field of biomineralization has delivered important new fundamental knowledge and techniques to better understand this fascinating phenomenon. This knowledge is also applied in the field of biomaterials, with the aim of bringing the composition and structure, and hence the performance, of synthetic bone graft substitutes even closer to those of the extracellular matrix of bone. The purpose of this progress report is to critically review advances in mimicking the extracellular matrix of bone as a strategy for development of new materials for bone regeneration. Lab-made biomimicking or bioinspired materials are discussed against the background of the natural extracellular matrix, starting from basic organic and inorganic components, and progressing into the building block of bone, the mineralized collagen fibril, and finally larger, 2D and 3D constructs. Moreover, bioactivity studies on state-of-the-art biomimicking materials are discussed. By addressing these different topics, an overview is given of how far the field has advanced toward a true bone-mimicking material, and some suggestions are offered for bridging current knowledge and technical gaps.
Collapse
Affiliation(s)
- Daniel de Melo Pereira
- MERLN Institute for Technology-Inspired Regenerative Medicine; Maastricht University; P.O. Box 616 6200 MD Maastricht The Netherlands
| | - Pamela Habibovic
- MERLN Institute for Technology-Inspired Regenerative Medicine; Maastricht University; P.O. Box 616 6200 MD Maastricht The Netherlands
| |
Collapse
|
34
|
Safflower yellow promotes angiogenesis through p-VHL/ HIF-1α/VEGF signaling pathway in the process of osteogenic differentiation. Biomed Pharmacother 2018; 107:1736-1743. [PMID: 30257392 DOI: 10.1016/j.biopha.2018.06.119] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/21/2018] [Accepted: 06/21/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Safflower yellow (SY) is an active component ofCarthamus tinctorius L. that is widely used in orthopedics. This study aimed to evaluate the role of SY in angiogenesis and osteogenic differentiation. METHODS The migration and in vitro angiogenesis of SY (4.5, 9.0, 18 μg/ml)-treated human umbilical vein endothelial cells (HUVEC-12) were assessed by transwell and tube formation assay, respectively. Osteogenic differentiation ability was detected by alkaline phosphatase (ALP) and Alizarin Red S staining. The mRNA and protein expressions of related markers were determined by RT-qPCR and Western blot. RESULTS The migration and tube formation ability of HUVEC-12 were promoted by SY. Furthermore, SY facilitated the angiogenesis and osteogenic differentiation in the co-culture of HUVEC-12 and BMSCs by increasing hypoxia inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), Angiopoietin-2 (Ang-2), ALP, runt-related transcription factor 2 (Runx2) and osteopontin-1 (OPN-1) levels. Inhibition of HIF-1α expression by 3-(5-hydroxymethl-2-furyl)-1-benzylindazole (YC-1), restrained SY-induced proliferation, migration and angiogenesis of HUVEC-12 and the increased protein levels of VEGF, Ang-2, ALP, Runx2 and OPN-1. Finally, WD repeat and SOCS box-containing protein-1 (WSB-1)/Von Hippel-Lindau protein (p-VHL) pathway was involved in the beneficial effect of SY. CONCLUSION SY promotes osteogenic differentiation via enhancing angiogenesis by regulating pVHL/HIF-1α/VEGF signaling pathway.
Collapse
|
35
|
Zhang Y, Wang D, Li M, Wei X, Liu S, Zhao M, Liu C, Wang X, Jiang X, Li X, Zhang S, Bergquist J, Wang B, Yang C, Mi J, Tian G. Quantitative Proteomics of TRAMP Mice Combined with Bioinformatics Analysis Reveals That PDGF-B Regulatory Network Plays a Key Role in Prostate Cancer Progression. J Proteome Res 2018; 17:2401-2411. [PMID: 29863873 DOI: 10.1021/acs.jproteome.8b00158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transgenic adenocarcinoma of the mouse prostate (TRAMP) mice is a widely used transgenic animal model of prostate cancer (PCa). We performed a label-free quantitative proteomics analysis combined with a bioinformatics analysis on the entire prostate protein extraction from TRAMP mice and compared it with WT littermates. From 2379 total identified proteins, we presented a modest mice prostate reference proteome containing 919 proteins. 61 proteins presented a significant expression difference between two groups. The integrative bioinformatics analysis predicted the overexpression of platelet-derived growth factor B (PDGF-B) in tumor tissues and supports the hypothesis of the PDGF-B signaling network as a key upstream regulator in PCa progression. Furthermore, we demonstrated that Crenolanib, a novel PDGF receptor inhibitor, inhibited PCa cell proliferation in a dose-dependent manner. Finally, we revealed the importance of PDGF-B regulatory network in PCa progression, which will assist us in understanding the role and mechanisms of PDGF-B in promoting cancer growth and provide valuable knowledge for future research on anti-PDGF therapy.
Collapse
Affiliation(s)
- Yuan Zhang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Dan Wang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Radiology , Affiliated Hospital of Binzhou Medical University , 661 Second Huanghe Road , Binzhou , Shandong Province 256603 , China
| | - Min Li
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Xiaodan Wei
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Shuang Liu
- College of Enology , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Miaoqing Zhao
- Department of Pathology , Provincial Hospital Affiliated to Shandong University , No. 324 Jingwu Weiqi Road , Jinan , Shandong Province 250021 , China
| | - Chu Liu
- Department of Urology , Yantai Yuhuangding Hospital , Zhifu District, No. 20, Yuhuangding East Road , Yantai , Shandong Province 264000 , China
| | - Xizhen Wang
- Imaging Center , Affiliated Hospital of Weifang Medical University , Kuiwen District, No. 465, Yuhe Road , Weifang , Shandong Province 256603 , China
| | - Xingyue Jiang
- Department of Radiology , Affiliated Hospital of Binzhou Medical University , 661 Second Huanghe Road , Binzhou , Shandong Province 256603 , China
| | - Xuri Li
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Shuping Zhang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Jonas Bergquist
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Chemistry - BMC , Uppsala University , P.O. Box 599, Husargatan 3 , Uppsala 75124 , Sweden
| | - Bin Wang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Chunhua Yang
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| | - Jia Mi
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China.,Department of Chemistry - BMC , Uppsala University , P.O. Box 599, Husargatan 3 , Uppsala 75124 , Sweden
| | - Geng Tian
- Medicine and Pharmacy Research Center , Binzhou Medical University , Laishan District, No. 346, Guanhai Road , Yantai , Shandong Province 264003 , China
| |
Collapse
|
36
|
Identification of the effector domain of biglycan that facilitates BMP-2 osteogenic function. Sci Rep 2018; 8:7022. [PMID: 29728612 PMCID: PMC5935668 DOI: 10.1038/s41598-018-25279-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
Abstract
We have reported that recombinant biglycan (BGN) core protein accelerates bone formation in vivo by enhancing bone morphogenetic protein (BMP)-2 function. The purpose of the present study was to identify the specific domain (“effector”) within the BGN core protein that facilitates BMP-2 osteogenic function. Thus, we generated various recombinant and synthetic peptides corresponding to several domains of BGN, and tested their effects on BMP-2 functions in vitro. The results demonstrated that the leucine-rich repeats 2–3 domain (LRR2-3) of BGN significantly enhanced the BMP-2 induced Smad1/5/9 phosphorylation, osteogenic gene expression, and alkaline phosphatase activity in myogenic C2C12 cells. Furthermore, addition of LRR2-3 to osteoblastic MC3T3-E1 cells accelerated in vitro mineralization without compromising the quality of the mineral and matrix. These data indicate that LRR2-3 is, at least in part, responsible for BGN’s ability to enhance BMP-2 osteogenic function, and it could be useful for bone tissue regeneration.
Collapse
|
37
|
Extracellular matrix: The driving force of mammalian diseases. Matrix Biol 2018; 71-72:1-9. [PMID: 29625183 DOI: 10.1016/j.matbio.2018.03.023] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 12/31/2022]
Abstract
Like the major theme of a Mozart concerto, the immense and pervasive extracellular matrix drives each movement and ultimately closes the symphony, embracing a unique role as the fundamental mediator for most, if not all, ensuing intracellular events. As such, it comes as no surprise that the mechanism of just about every known disease can be traced back to some part of the matrix, typically in the form of an abnormal amount or activity level of a particular matrix component. These defects considerably affect downstream signaling axes leading to overt cellular dysfunction, organ failure, and death. From skin to bone, from vessels to brain, from eyes to all the internal organs, the matrix plays an incredible role as both a cause and potential means to reverse diseases. Human malaises including connective tissue disorders, muscular dystrophy, fibrosis, and cancer are all extracellular matrix-driven diseases. The ability to understand and modulate these matrix-related mechanisms may lead to the future discovery of novel therapeutic options for these patients.
Collapse
|
38
|
Procopio N, Williams A, Chamberlain AT, Buckley M. Forensic proteomics for the evaluation of the post-mortem decay in bones. J Proteomics 2018; 177:21-30. [PMID: 29407476 DOI: 10.1016/j.jprot.2018.01.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/31/2022]
Abstract
Current methods for evaluation the of post-mortem interval (PMI) of skeletal remains suffer from poor accuracy due to the great number of variables that affect the diagenetic process and to the lack of specific guidelines to address this issue. During decomposition, proteins can undergo cumulative decay over the time, resulting in a decrease in the range and abundance of proteins present (i.e., the proteome) in different tissues as well as in an increase of post-translational modifications occurring in these proteins. In this study, we investigate the applicability of bone proteomic analyses to simulated forensic contexts, looking for specific biomarkers that may help the estimation of PMI, as well as evaluate a previously discovered marker for the estimation of biological age. We noticed a reduction of particular plasma and muscle proteins with increasing PMIs, as well as an increased deamidation of biglycan, a protein with a role in modulating bone growth and mineralization. We also corroborated our previous results regarding the use of fetuin-A as a potential biomarker for the estimation of age-at-death, demonstrating the applicability and the great potential that proteomics may have towards forensic sciences. SIGNIFICANCE The estimation of the post-mortem interval has a key role in forensic investigations, however nowadays it still suffers from poor reliability, especially when body tissues are heavily decomposed. Here we propose for the first time the application of bone proteomics to the estimation of the time elapsed since death and found several new potential biomarkers to address this, demonstrating the applicability of proteomic analyses to forensic sciences.
Collapse
Affiliation(s)
- Noemi Procopio
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - Anna Williams
- School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| | - Andrew T Chamberlain
- School of Earth and Environmental Sciences, The University of Manchester, Stopford Building, 99 Oxford Road, Manchester, M13 9PG, UK
| | - Michael Buckley
- Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK.
| |
Collapse
|
39
|
Subbarayan K, Leisz S, Wickenhauser C, Bethmann D, Massa C, Steven A, Seliger B. Biglycan-mediated upregulation of MHC class I expression in HER-2/neu-transformed cells. Oncoimmunology 2018; 7:e1373233. [PMID: 29632715 DOI: 10.1080/2162402x.2017.1373233] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/22/2017] [Accepted: 08/25/2017] [Indexed: 01/16/2023] Open
Abstract
The extracellular matrix protein biglycan (BGN) has oncogenic or tumor suppressive potential depending on the cellular origin. HER-2/neu overexpression in murine fibroblasts and human model systems is inversely correlated with BGN expression. Upon its restoration BGNhigh HER-2/neu+ fibroblasts were less tumorigenic in immune competent mice when compared to BGNlow/neg HER-2/neu+ cells, which was associated with enhanced immune cell responses and higher frequencies of immune effector cells in tumors and peripheral blood. The increased immunogenicity of BGNhigh HER-2/neu+ fibroblasts appears to be due to upregulated MHC class I surface antigens and reduced expression levels of transforming growth factor (TGF)-β isoforms and the TGF-β receptor 1 suggesting a link between BGN, TGF-β pathway and HER-2/neu-mediated downregulation of MHC class I antigens. Treatment of BGNlow/neg HER-2/neu+ cells with recombinant BGN or an inhibitor of TGF-β enhanced MHC class I surface antigens in BGNlow/neg HER-2/neu-overexpressing murine fibroblasts, which was mediated by a transcriptional upregulation of major MHC class I antigen processing components. Furthermore, BGN expression in HER-2/neu+ cells was accompanied by an increased expression of the proteoglycan decorin (DCN). Since recombinant DCN also elevated MHC class I surface expression in BGNlow/neg HER-2/neu+ cells, both proteoglycans might act synergistically. This was in accordance with in silico analyses of mRNA data obtained from The Cancer Genome Atlas (TCGA) dataset available for breast cancer (BC) patients. Thus, our data provide for the first time evidence that proteoglycan signatures are modulated by HER-2/neu and linked to MHC class I-mediated immune escape associated with an altered TGF-β pathway.
Collapse
Affiliation(s)
- Karthikeyan Subbarayan
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - Sandra Leisz
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - Daniel Bethmann
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - Chiara Massa
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - André Steven
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle/ Saale, Germany
| |
Collapse
|
40
|
Ou Z, Cheng Q, Chen Y, Chen T, Rong X, Long F, Zhang X, Liang Q, Feng Z. Chemical characterization of wound ointment (WO) and its effects on fracture repair: a rabbit model. Chin Med 2017; 12:31. [PMID: 29093746 PMCID: PMC5661936 DOI: 10.1186/s13020-017-0152-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
Background Wound ointment (WO), a kind of Chinese medicine, can significantly promote fracture healing. The study aimed at analyzing the chemical composition and the effects of WO on fracture of rabbits and tried to explore the corresponding molecular mechanism in cytokine. Methods The qualitative and quantitative analysis of WO was conducted by liquid chromatography-mass spectrometry (LC–MS). Fifty-four Zealand mature male rabbits were randomly divided into 3 groups: Control group, Yunnan Baiyao (YB) group and WO group. All the rabbits suffered a fracture of right radius and were then stabilized with an external fixator. Treated with different methods, fracture healing was observed. The bone specimens were subjected to radiograph, immunohistochemistry (IHC) analysis, hematoxylin–eosin staining (HE), western blot and enzyme linked immunosorbent assay (ELISA). Results A total of 12 active compositions were detected by LC–MS. Radiographs showed a considerably better bone healing and remodeling of the fracture in WO group. HE experiments showed that a large number of osteoclasts appeared in the early stage when treated with WO. In immunohistochemistry (IHC), western blot and ELISA test, significant increases in vascular endothelial growth factor (VEGF) expression were observed in WO group compared with other two groups. Conclusions Wound ointment contained active compositions which efficiently promoted fracture healing through increasing the expression of VEGF. Trial Registration Not applicable Electronic supplementary material The online version of this article (doi:10.1186/s13020-017-0152-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhixue Ou
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Qi Cheng
- Department of Traumatic Orthopedics, Daye Hospital of Traditional Chinese Medicine, Daye, 435100 Hubei China
| | - Yueping Chen
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Tao Chen
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Xiangbin Rong
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Feipan Long
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Xiaoyun Zhang
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Qinghua Liang
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| | - Zhe Feng
- Department of Traumatic Orthopedics and Hand Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Block B, No 10 Huadong Road, Nanning, 530011 Guangxi China
| |
Collapse
|
41
|
Tyekucheva S, Bowden M, Bango C, Giunchi F, Huang Y, Zhou C, Bondi A, Lis R, Van Hemelrijck M, Andrén O, Andersson SO, Watson RW, Pennington S, Finn SP, Martin NE, Stampfer MJ, Parmigiani G, Penney KL, Fiorentino M, Mucci LA, Loda M. Stromal and epithelial transcriptional map of initiation progression and metastatic potential of human prostate cancer. Nat Commun 2017; 8:420. [PMID: 28871082 PMCID: PMC5583238 DOI: 10.1038/s41467-017-00460-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/29/2017] [Indexed: 01/02/2023] Open
Abstract
While progression from normal prostatic epithelium to invasive cancer is driven by molecular alterations, tumor cells and cells in the cancer microenvironment are co-dependent and co-evolve. Few human studies to date have focused on stroma. Here, we performed gene expression profiling of laser capture microdissected normal non-neoplastic prostate epithelial tissue and compared it to non-transformed and neoplastic low-grade and high-grade prostate epithelial tissue from radical prostatectomies, each with its immediately surrounding stroma. Whereas benign epithelium in prostates with and without tumor were similar in gene expression space, stroma away from tumor was significantly different from that in prostates without cancer. A stromal gene signature reflecting bone remodeling and immune-related pathways was upregulated in high compared to low-Gleason grade cases. In validation data, the signature discriminated cases that developed metastasis from those that did not. These data suggest that the microenvironment may influence prostate cancer initiation, maintenance, and metastatic progression.Stromal cells contribute to tumor development but the mechanisms regulating this process are still unclear. Here the authors analyze gene expression profiles in the prostate and show that stromal gene signature changes ahead of the epithelial gene signature as prostate cancer initiates and progresses.
Collapse
Affiliation(s)
- Svitlana Tyekucheva
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Clyde Bango
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Francesca Giunchi
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Ying Huang
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Chensheng Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Arrigo Bondi
- Department of Surgical Pathology, Maggiore Hospital, Largo Nigrisoli 2, 40133, Bologna, Italy
| | - Rosina Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Mieke Van Hemelrijck
- King's College London, Division of Cancer Studies, Translational Oncology & Urology Research, Guy's Hospital, London, SE1 9RT, UK
| | - Ove Andrén
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - Sven-Olof Andersson
- Department of Urology, School of Health and Medical Sciences, Örebro University Hospital, Örebro, SE 701 85, Sweden
| | - R William Watson
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen Pennington
- School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Stephen P Finn
- Department of Histopathology and Morbid Anatomy, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Neil E Martin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Giovanni Parmigiani
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Kathryn L Penney
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Michelangelo Fiorentino
- Department of Pathology, Addarii Institute of Oncology, S.Orsola-Malpighi Teaching Hospital, University of Bologna, Viale Ercolani 4/2, 40138, Bologna, Italy
| | - Lorelei A Mucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA, 02115, USA
| | - Massimo Loda
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
- The Broad Institute, 415 Main St, Cambridge, MA, 02142, USA.
| |
Collapse
|
42
|
Westergren-Thorsson G, Bagher M, Andersson-Sjöland A, Thiman L, Löfdahl CG, Hallgren O, Bjermer L, Larsson-Callerfelt AK. VEGF synthesis is induced by prostacyclin and TGF-β in distal lung fibroblasts from COPD patients and control subjects: Implications for pulmonary vascular remodelling. Respirology 2017; 23:68-75. [PMID: 28834088 DOI: 10.1111/resp.13142] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 05/15/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Involvement of pulmonary vascular remodelling is a characteristic sign in COPD. Vascular mediators such as vascular endothelial growth factor (VEGF) and prostacyclin may regulate fibroblast activity. The objective was to study the synthesis of VEGF and interactions with prostacyclin and transforming growth factor (TGF)-β1 in lung fibroblasts from patients with COPD and healthy control subjects. To further explore the autocrine role of synthesized VEGF on fibroblast activity, studies were performed in human lung fibroblasts (HFL-1). METHODS Primary distal lung fibroblast cultures were established from healthy individuals and from COPD patients (GOLD stage IV). Lung fibroblasts were stimulated with the prostacyclin analogue iloprost and the profibrotic stimuli TGF-β1 . VEGF synthesis was measured in the cell culture medium. Changes in proliferation rate, migration and synthesis of the extracellular matrix (ECM) proteins proteoglycans were analysed after stimulations with VEGF-A isoform 165 (VEGF165 ; 1-10 000 pg/mL) in HFL-1. RESULTS Iloprost and TGF-β1 significantly increased VEGF synthesis in both fibroblasts from COPD patients and control subjects. TGF-β1 -induced VEGF synthesis was significantly reduced by the cyclooxygenase inhibitor indomethacin in fibroblasts from COPD patients. VEGF significantly increased proliferation rate and migration capacity in HFL-1. VEGF also significantly increased synthesis of the ECM proteins biglycan and perlecan. The VEGF receptors (VEGFR), VEGFR1, VEGFR2 and VEGFR3, were all expressed in primary lung fibroblasts and HFL-1. CONCLUSION VEGF is synthesized in high amounts by distal lung fibroblasts and may have a crucial role in ongoing vascular remodelling processes in the distal lung compartments.
Collapse
Affiliation(s)
| | - Mariam Bagher
- Department of Respiratory, Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Annika Andersson-Sjöland
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Lena Thiman
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Claes-Göran Löfdahl
- Department of Respiratory, Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Oskar Hallgren
- Department of Respiratory, Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory, Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | | |
Collapse
|
43
|
Schaefer L, Tredup C, Gubbiotti MA, Iozzo RV. Proteoglycan neofunctions: regulation of inflammation and autophagy in cancer biology. FEBS J 2017; 284:10-26. [PMID: 27860287 PMCID: PMC5226885 DOI: 10.1111/febs.13963] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/27/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022]
Abstract
Inflammation and autophagy have emerged as prominent issues in the context of proteoglycan signaling. In particular, two small, leucine-rich proteoglycans, biglycan and decorin, play pivotal roles in the regulation of these vital cellular pathways and, as such, are intrinsically involved in cancer initiation and progression. In this minireview, we will address novel functions of biglycan and decorin in inflammation and autophagy, and analyze new emerging signaling events triggered by these proteoglycans, which directly or indirectly modulate these processes. We will critically discuss the dual role of proteoglycan-driven inflammation and autophagy in tumor biology, and delineate the potential mechanisms through which soluble extracellular matrix constituents affect the microenvironment associated with inflammatory and neoplastic diseases.
Collapse
Affiliation(s)
- Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Claudia Tredup
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Maria A. Gubbiotti
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
44
|
Ionic liquid-based method for direct proteome characterization of velvet antler cartilage. Talanta 2016; 161:541-546. [DOI: 10.1016/j.talanta.2016.08.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 11/19/2022]
|
45
|
Extracellular Matrix, a Hard Player in Angiogenesis. Int J Mol Sci 2016; 17:ijms17111822. [PMID: 27809279 PMCID: PMC5133823 DOI: 10.3390/ijms17111822] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins, glycoproteins, proteoglycans, and polysaccharides. Through multiple interactions with each other and the cell surface receptors, not only the ECM determines the physical and mechanical properties of the tissues, but also profoundly influences cell behavior and many physiological and pathological processes. One of the functions that have been extensively explored is its impingement on angiogenesis. The strong impact of the ECM in this context is both direct and indirect by virtue of its ability to interact and/or store several growth factors and cytokines. The aim of this review is to provide some examples of the complex molecular mechanisms that are elicited by these molecules in promoting or weakening the angiogenic processes. The scenario is intricate, since matrix remodeling often generates fragments displaying opposite effects compared to those exerted by the whole molecules. Thus, the balance will tilt towards angiogenesis or angiostasis depending on the relative expression of pro- or anti-angiogenetic molecules/fragments composing the matrix of a given tissue. One of the vital aspects of this field of research is that, for its endogenous nature, the ECM can be viewed as a reservoir to draw from for the development of new more efficacious therapies to treat angiogenesis-dependent pathologies.
Collapse
|
46
|
Wu QH, Ma Y, Ruan CC, Yang Y, Liu XH, Ge Q, Kong LR, Zhang JW, Yan C, Gao PJ. Loss of osteoglycin promotes angiogenesis in limb ischaemia mouse models via modulation of vascular endothelial growth factor and vascular endothelial growth factor receptor 2 signalling pathway. Cardiovasc Res 2016; 113:70-80. [DOI: 10.1093/cvr/cvw220] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/29/2016] [Accepted: 09/21/2016] [Indexed: 01/22/2023] Open
|
47
|
Ma P, Yan W, Tian Y, Wang J, Feng JQ, Qin C, Cheng YSL, Wang X. Inactivation of Fam20B in Joint Cartilage Leads to Chondrosarcoma and Postnatal Ossification Defects. Sci Rep 2016; 6:29814. [PMID: 27405802 PMCID: PMC4942823 DOI: 10.1038/srep29814] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/24/2016] [Indexed: 12/13/2022] Open
Abstract
During endochondral ossification, chondrocytes embed themselves in a proteoglycan-rich matrix during the proliferation-maturation transition. Accumulating evidence shows that proteoglycans are essential components for chondrocyte proliferation and differentiation. When we conditionally inactivated FAM20B (Family with sequence similarity 20 member-B), which is a newly identified xylose kinase essential for glycosaminoglycan (GAG) formation on the protein core of proteoglycans, from the dental mesenchyme using Osr2-Cre, which is also strongly expressed in joint cartilage, we found chondrosarcoma in the knee joint and remarkable defects of postnatal ossification in the long bones. Mechanistic analysis revealed that the defects were associated with gain of function in multiple signaling pathways in the epiphyseal chondrocytes, such as those derived by WNT, BMP, and PTHrP/IHH molecules, suggesting that the FAM20B-catalyzed proteoglycans are critical mediators for a signaling balance in the regulatory network controlling chondrocyte differentiation and proliferation. In particular, we demonstrated that the WNT inhibitor was able to rescue part of the bone defects in Osr2-Cre;Fam20Bfl/fl mice, indicating that FAM20B-catalyzed proteoglycans regulate postnatal endochondral ossification partially through the mediation of WNT signaling.
Collapse
Affiliation(s)
- Pan Ma
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America.,Department of Oral Implantology, Beijing Stomatological Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenjuan Yan
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| | - Ye Tian
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| | - Jingya Wang
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| | - Jian Q Feng
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| | - Chunlin Qin
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| | - Yi-Shing Lisa Cheng
- Department of Diagnostic Sciences, Texas A&M University Baylor College of Dentistry, 3302 Gaston Ave, Dallas, TX, United States of America
| | - Xiaofang Wang
- Department of Biomedical Sciences and Center for Craniofacial Research and Diagnosis, Texas A&M University Baylor College of Dentistry, Dallas, Texas, United States of America
| |
Collapse
|
48
|
Abstract
The skeleton is unique from all other tissues in the body because of its ability to mineralize. The incorporation of mineral into bones and teeth is essential to give them strength and structure for body support and function. For years, researchers have wondered how mineralized tissues form and repair. A major focus in this context has been on the role of the extracellular matrix, which harbors key regulators of the mineralization process. In this introductory minireview, we will review some key concepts of matrix biology as it related to mineralized tissues. Concurrently, we will highlight the subject of this special issue covering many aspects of mineralized tissues, including bones and teeth and their associated structures cartilage and tendon. Areas of emphasis are on the generation and analysis of new animal models with permutations of matrix components as well as the development of new approaches for tissue engineering for repair of damaged hard tissue. In assembling key topics on mineralized tissues written by leaders in our field, we hope the reader will get a broad view of the topic and all of its fascinating complexities.
Collapse
Affiliation(s)
- Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States.
| |
Collapse
|
49
|
|
50
|
Myren M, Kirby DJ, Noonan ML, Maeda A, Owens RT, Ricard-Blum S, Kram V, Kilts TM, Young MF. Biglycan potentially regulates angiogenesis during fracture repair by altering expression and function of endostatin. Matrix Biol 2016; 52-54:141-150. [PMID: 27072616 DOI: 10.1016/j.matbio.2016.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/31/2022]
Abstract
The small proteoglycan biglycan (Bgn) is highly expressed in the organic matrix of bone and plays a role in bone formation. Previous work implicated Bgn in vessel growth during bone healing [1]. By infusing barium sulfate (BaSO4) into WT and Bgn-deficient mice we discovered the positive effect of Bgn in modulating angiogenesis during fracture healing. Using micro-computed tomography angiography we found significant differences in the vessel size and volume among other parameters. To further understand the mechanistic basis for this, we explored the relationship between Bgn and the anti-angiogenic protein endostatin. Immunohistochemistry (IHC) showed co-localization of Bgn and endostatin in regions of bone formation, with increased endostatin staining in Bgn-KO compared to WT at 14days post-fracture. To further elucidate the relationship between Bgn and endostatin, an endothelial cell tube formation assay was used. This study showed that endothelial cells treated with endostatin had significantly decreased vessel length and vessel branches compared to untreated cells, while cells treated with endostatin and Bgn at a 1:1M ratio had vessel length and vessel branches comparable to untreated cells. This indicated that Bgn was able to mitigate the inhibitory effect of endostatin on endothelial cell growth. In summary, these results suggest that Bgn is needed for proper blood vessel formation during fracture healing, and one mechanism by which Bgn impacts angiogenesis is through inhibition of endostatin.
Collapse
Affiliation(s)
- Maja Myren
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - David J Kirby
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Megan L Noonan
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Azusa Maeda
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Rick T Owens
- Life Cell Corporation, Branchburg, NJ 08876, United States
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246 CNRS - University Lyon 1, ICBMS, 69622 Villeurbanne, France
| | - Vardit Kram
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Tina M Kilts
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, NIDCR, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|