1
|
Abraham N, Pandey G, Kolipaka T, Negi M, Srinivasarao DA, Srivastava S. Exploring advancements in polysaccharide-based approaches: The cornerstone of next-generation cartilage regeneration therapeutics. Int J Biol Macromol 2025; 306:141352. [PMID: 39986526 DOI: 10.1016/j.ijbiomac.2025.141352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Cartilage regeneration poses a formidable challenge in orthopaedics due to continuous wear and tear exertion and its limited intrinsic healing capacity, which demand exploration beyond current clinical approaches. Polysaccharides emerged as promising agents for cartilage regeneration, offering biocompatibility, biodegradability, bioactivity, and ECM mimicry. This article provides an overview of the pathophysiology of cartilage diseases and current clinical approaches, followed by polysaccharide-based strategies for cartilage repair, delineating the chemical and biological properties of various polysaccharides like alginates, hyaluronic acid, and chondroitin sulfate. The emphasis lies on innovative strategies such as sulphated and cross-linked polysaccharides, with injectable polysaccharide hydrogels offering adjustable mechanical properties and easy administration. Growth factor and cellular incorporation into hydrogels enhance their therapeutic potential. At the same time, biofabrication techniques, such as filamented light biofabrication, cartilage spheroid generation, and 3D printing, offer precise control over cartilage architecture, with bio-inks comprising alginate, gelatin, and hyaluronic acid showing promise. These advancements underscore the potential of polysaccharides to revolutionize cartilage regeneration strategies, offering hope for improved patient outcomes in the future. The article concludes by addressing regulatory hurdles and the future perspective of polysaccharide-based approaches in clinical translation for cartilage regeneration.
Collapse
Affiliation(s)
- Noella Abraham
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mansi Negi
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
2
|
Hu X, Xie D, Li Y, Niu Y, Tan R, She Z, Wang C. A dual-modified glucomannan polysaccharide selectively sequesters growth factors for skin tissue repair. J Control Release 2025; 380:185-198. [PMID: 39894264 DOI: 10.1016/j.jconrel.2025.01.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Artificial dermal matrixes (ADMs) are valuable clinical options for treating large soft tissue defects, but their suboptimal bioactivities compared with the real tissue limit their therapeutic potential. For example, glycosaminoglycan (GAG) polysaccharides in the native skin vitally and differentially regulate endogenous growth factors (GFs) to maintain tissue homeostasis. However, the GAG used in the current ADMs has often lost such delicate regulation. Here, we developed a novel polysaccharide-based ADM that can promote skin tissue repair through selective modulation of specific pro-healing GFs. First, we prepared a plant-derived backbone of glucomannan (named BSP) - representing the two dominant monosaccharide components in the human body - in mass and homogenic quality. Then, we modified this backbone with sulfate and acetyl groups in a controlled manner to yield an optimized BSP derivative (SMAL-BSP) as a main composition to generate a new ADM. In vitro, SMAL-BSP enabled the ADM to selectively sequester pro-angiogenic GFs of VEGF-A and FGF-2 in situ for stimulating endothelial cell growth. Moreover, the addition of the acetyl group induced macrophages to secrete nitric oxide (NO) with antibacterial activities. Further in vivo tests in a rat model of full-thickness skin wounds indicated that SMAL-BSP ADM could sequester GFs in situ to promote angiogenesis and thus tissue regeneration, with superior effects than conventional chondroitin sulfate-based ADM, while showing no adverse effects often associated with animal-derived products. Our study represents a novel strategy for ADM design, targeting selective GF sequestration towards optimal skin tissue regeneration.
Collapse
Affiliation(s)
- Xiaotong Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; Guangdong Engineering Technology Research Center of Implantable Medical Polymer, Shenzhen Lando Biomaterials, Shenzhen, China
| | - Daping Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yuwei Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; Zhuhai UM Science and Technology Research Institute (ZUMRI), University of Macau, Hengqin, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Rongwei Tan
- Guangdong Engineering Technology Research Center of Implantable Medical Polymer, Shenzhen Lando Biomaterials, Shenzhen, China
| | - Zhending She
- Guangdong Engineering Technology Research Center of Implantable Medical Polymer, Shenzhen Lando Biomaterials, Shenzhen, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Taipa, Macau SAR, China; Zhuhai UM Science and Technology Research Institute (ZUMRI), University of Macau, Hengqin, China.
| |
Collapse
|
3
|
Ma M, Wang C, Wu M, Gu S, Yang J, Zhang Y, Cheng S, Xu S, Zhang M, Wu Y, Zhao Y, Tian X, Voon DCC, Takahashi C, Sheng J, Wang Y. CSGALNACT2 restricts ovarian cancer migration and invasion by modulating MAPK/ERK pathway through DUSP1. Cell Oncol (Dordr) 2024; 47:897-915. [PMID: 38082211 PMCID: PMC11219422 DOI: 10.1007/s13402-023-00903-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Ovarian cancer is one of the leading causes of cancer-related death among women. CSGALNACT2 is a vital Golgi transferase and is related to a variety of human diseases. However, its expression pattern and function in ovarian cancer remain uncertain. METHODS The Cancer Genome Atlas and GEPIA databases were used to assess the expression of CSGALNACT2 in ovarian cancer patients. RNA-seq, qRT-PCR, and IHC were used to verify the expression of CSGALNACT2 in ovarian cancer tissues. Then, in vivo and in vitro experiments were conducted to evaluate the role of CSGALNACT2 in the progression of ovarian cancer. RNA-seq and GSEA were used to reveal the potential biological function and oncogenic pathways of CSGALNACT2. RESULTS We demonstrated that the mRNA expression and protein level of CSGALNACT2 were significantly downregulated in ovarian cancer and ovarian cancer metastatic tissues. CSGALNACT2 can significantly inhibit the migration, invasion, and clonogenic growth of ovarian cancer in vitro and is progressively lost during ovarian cancer progression in vivo. CSGALNACT2 suppresses ovarian cancer migration and invasion via DUSP1 modulation of the MAPK/ERK pathway through RNA-seq, KEGG analysis, and Western blotting. Moreover, CSGALNACT2 expression was correlated with immune cell infiltration and had prognostic value in different immune cell-enriched or decreased ovarian cancer. In addition, patients with CSGALNACT2 downregulation are less likely to benefit from immunotherapy. CONCLUSION As an ovarian cancer suppressor gene, CSGALNACT2 inhibits the development of ovarian cancer, and it might be used as a prognostic biomarker in patients with ovarian cancer.
Collapse
Affiliation(s)
- Mingjun Ma
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chao Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Meixuan Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiani Yang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yue Zhang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shanshan Cheng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shilin Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Minghai Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongsong Wu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yaqian Zhao
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiu Tian
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | | | - Chiaki Takahashi
- Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, 920-1192, Japan
| | - Jindan Sheng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Yu Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No.2699, Gaoke West Rd, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
4
|
Haryono A, Ikeda K, Nugroho DB, Ogata T, Tsuji Y, Matoba S, Moriwaki K, Kitagawa H, Igarashi M, Hirata KI, Emoto N. ChGn-2 Plays a Cardioprotective Role in Heart Failure Caused by Acute Pressure Overload. J Am Heart Assoc 2022; 11:e023401. [PMID: 35322673 PMCID: PMC9075488 DOI: 10.1161/jaha.121.023401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cardiac extracellular matrix is critically involved in cardiac homeostasis, and accumulation of chondroitin sulfate glycosaminoglycans (CS-GAGs) was previously shown to exacerbate heart failure by augmenting inflammation and fibrosis at the chronic phase. However, the mechanism by which CS-GAGs affect cardiac functions remains unclear, especially at the acute phase. Methods and Results We explored a role of CS-GAG in heart failure using mice with target deletion of ChGn-2 (chondroitin sulfate N-acetylgalactosaminyltransferase-2) that elongates CS chains of glycosaminoglycans. Heart failure was induced by transverse aortic constriction in mice. The role of CS-GAG derived from cardiac fibroblasts in cardiomyocyte death was analyzed. Cardiac fibroblasts were subjected to cyclic mechanical stretch that mimics increased workload in the heart. Significant CS-GAGs accumulation was detected in the heart of wild-type mice after transverse aortic constriction, which was substantially reduced in ChGn-2-/- mice. Loss of ChGn-2 deteriorated the cardiac dysfunction caused by pressure overload, accompanied by augmented cardiac hypertrophy and increased cardiomyocyte apoptosis. Cyclic mechanical stretch increased ChGn-2 expression and enhanced glycosaminoglycan production in cardiac fibroblasts. Conditioned medium derived from the stretched cardiac fibroblasts showed cardioprotective effects, which was abolished by CS-GAGs degradation. We found that CS-GAGs elicits cardioprotective effects via dual pathway; direct pathway through interaction with CD44, and indirect pathway through binding to and activating insulin-like growth factor-1. Conclusions Our data revealed the cardioprotective effects of CS-GAGs; therefore, CS-GAGs may play biphasic role in the development of heart failure; cardioprotective role at acute phase despite its possible unfavorable role in the advanced phase.
Collapse
Affiliation(s)
- Andreas Haryono
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan.,Department of Epidemiology for Longevity and Regional Health Kyoto Prefectural University of Medicine Kyoto Japan.,Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Dhite Bayu Nugroho
- Department of Internal Medicine Faculty of Medicine, Public Health, and Nursing Gadjah Mada University Indonesia
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation Kyoto Prefectural University of Medicine Kyoto Japan
| | - Yumika Tsuji
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Satoaki Matoba
- Department of Cardiology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Kensuke Moriwaki
- Comprehensive Unit for Health Economic Evidence Review and Decision Support (CHEERS) Research Organization of Science and TechnologyRitsumeikan University Kyoto Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry Kobe Pharmaceutical University Kobe Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology Graduate School of Medical and Dental Sciences and Trans-disciplinary Program Niigata University Niigata Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan
| | - Noriaki Emoto
- Division of Cardiovascular Medicine Department of Internal Medicine Kobe University Graduate School of Medicine Kobe Japan.,Laboratory of Clinical Pharmaceutical Science Kobe Pharmaceutical University Kobe Japan
| |
Collapse
|
5
|
Balamurugan K, Koehler L, Dürig JN, Hempel U, Rademann J, Hintze V, Pisabarro MT. Structural insights into the modulation of PDGF/PDGFR-β complexation by hyaluronan derivatives. Biol Chem 2021; 402:1441-1452. [PMID: 34280958 DOI: 10.1515/hsz-2021-0173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022]
Abstract
Angiogenesis is an important physiological process playing a crucial role in wound healing and cancer progression. Vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) are key players in angiogenesis. Based on previous findings regarding the modulation of VEGF activity by glycosaminoglycans (GAG), here we explore the interaction of hyaluronan (HA)-based GAG with PDGF and its receptor PDGFR-β by applying molecular modeling and dynamics simulations in combination with surface plasmon resonance (SPR). Computational analysis on the interaction of oligo-hyaluronan derivatives with different sulfation pattern and functionalization shows that these GAG interact with PDGF in relevant regions for receptor recognition, and that high sulfation as well as modification with the TAMRA group convey stronger binding. On the other hand, the studied oligo-hyaluronan derivatives are predicted to scarcely recognize PDGFR-β. SPR results are in line with the computational predictions regarding the binding pattern of HA tetrasaccharide (HA4) derivatives to PDGF and PDGFR-β. Furthermore, our experimental results also show that the complexation of PDGF to PDGFR-β can be modulated by HA4 derivatives. The results found open the path for considering HA4 derivatives as potential candidates to be exploited for modulation of the PDGF/PDGFR-β signaling system in angiogenesis and related disease conditions.
Collapse
Affiliation(s)
- Kanagasabai Balamurugan
- Structural Bioinformatics, BIOTEC Technische Universität Dresden, Tatzberg 47-51, D-01307Dresden, Germany
| | - Linda Koehler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069Dresden, Germany
| | - Jan-Niklas Dürig
- Medicinal Chemistry Department, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, D-14195Berlin, Germany
| | - Ute Hempel
- Institute of Physiological Chemistry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Fiedlerstraße 42, D-01307Dresden, Germany
| | - Jörg Rademann
- Medicinal Chemistry Department, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, D-14195Berlin, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, Technische Universität Dresden, Budapester Str. 27, D-01069Dresden, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC Technische Universität Dresden, Tatzberg 47-51, D-01307Dresden, Germany
| |
Collapse
|
6
|
Snigireva AV, Morenkov OS, Skarga YY, Lisov AV, Lisova ZA, Leontievsky AA, Zhmurina MA, Petrenko VS, Vrublevskaya VV. A 2,5-Dihydroxybenzoic Acid-Gelatin Conjugate Inhibits the Basal and Hsp90-Stimulated Migration and Invasion of Tumor Cells. J Funct Biomater 2020; 11:jfb11020039. [PMID: 32503118 PMCID: PMC7353502 DOI: 10.3390/jfb11020039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
The extracellular cell surface-associated and soluble heat shock protein 90 (Hsp90) is known to participate in the migration and invasion of tumor cells. Earlier, we demonstrated that plasma membrane-associated heparan sulfate proteoglycans (HSPGs) bind the extracellular Hsp90 and thereby promote the Hsp90-mediated motility of tumor cells. Here, we showed that a conjugate of 2,5-dihydroxybenzoic acid with gelatin (2,5-DHBA–gelatin), a synthetic polymer with heparin-like properties, suppressed the basal (unstimulated) migration and invasion of human glioblastoma A-172 and fibrosarcoma HT1080 cells, which was accompanied by the detachment of a fraction of Hsp90 from cell surface HSPGs. The polymeric conjugate also inhibited the migration/invasion of cells stimulated by exogenous soluble native Hsp90, which correlated with the inhibition of the attachment of soluble Hsp90 to cell surface HSPGs. The action of the 2,5-DHBA–gelatin conjugate on the motility of A-172 and HT1080 cells was similar to that of heparin. The results demonstrate a potential of the 2,5-DHBA–gelatin polymer for the development of antimetastatic drugs targeting cell motility and a possible role of extracellular Hsp90 in the suppression of the migration and invasion of tumor cells mediated by the 2,5-DHBA–gelatin conjugate and heparin.
Collapse
Affiliation(s)
- Anastasiya V. Snigireva
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
| | - Oleg S. Morenkov
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
| | - Yuri Y. Skarga
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
| | - Alexander V. Lisov
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.L.); (Z.A.L.); (A.A.L.)
| | - Zoya A. Lisova
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.L.); (Z.A.L.); (A.A.L.)
| | - Alexey A. Leontievsky
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.L.); (Z.A.L.); (A.A.L.)
| | - Mariya A. Zhmurina
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
| | - Viktoria S. Petrenko
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
| | - Veronika V. Vrublevskaya
- Institute of Cell Biophysics, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Moscow Region, Russia; (A.V.S.); (O.S.M.); (Y.Y.S.); (M.A.Z.); (V.S.P.)
- Correspondence: ; Tel.: +7-4967-739221
| |
Collapse
|
7
|
Pan H, Xue W, Zhao W, Schachner M. Expression and function of chondroitin 4-sulfate and chondroitin 6-sulfate in human glioma. FASEB J 2020; 34:2853-2868. [PMID: 31908019 DOI: 10.1096/fj.201901621rrr] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 02/05/2023]
Abstract
Key molecules promoting migration and invasion exist in the extracellular matrix, and include chondroitin 4-sulfate (C4S) and chondroitin 6-sulfate (C6S), functionally important carbohydrate chains of chondroitin sulfate proteoglycans that participate in regulating cancer development. Here, we show that C4S and C6S expression is upregulated in human glioma tissues, when compared to normal brain tissue, and that the extent of upregulation positively correlated with glioma malignancy. Treatment of cultured glioma cells with C4S and C6S enhanced cell viability, migration, and invasion, increased MMP-2 and MMP-9 levels, enhanced N-cadherin, but reduced E-cadherin expression. Inhibition of expression of the two CS synthetic enzymes chondroitin 4-O-sulfotransferase-1 (C4ST-1/CHST11) and chondroitin 6-O-sulfotransferase-1 (C6ST-1/CHST3) suppressed cell viability, migration and invasion, reduced MMP-2 and MMP-9 expression, and reduced N-cadherin expression, but increased E-cadherin levels. The C4S- and C6S-enhanced epithelial-to-mesenchymal transition and expression of MMP-2 occurred via activation of the PI3K/AKT signaling pathway, known to be involved in promoting cell migration and invasion. In immune-deficient larval zebrafish, C4S and C6S increased the numbers of viable tumor cells, thereby promoting glioma cell proliferation. The present observations point to a novel role of C4S and C6S in human glioma cell functions, thus possibly representing targets in glioma therapy.
Collapse
Affiliation(s)
- Hongchao Pan
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Weikang Xue
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
8
|
Morla S. Glycosaminoglycans and Glycosaminoglycan Mimetics in Cancer and Inflammation. Int J Mol Sci 2019; 20:ijms20081963. [PMID: 31013618 PMCID: PMC6514582 DOI: 10.3390/ijms20081963] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/22/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
Glycosaminoglycans (GAGs) are a class of biomolecules expressed virtually on all mammalian cells and usually covalently attached to proteins, forming proteoglycans. They are present not only on the cell surface, but also in the intracellular milieu and extracellular matrix. GAGs interact with multiple ligands, both soluble and insoluble, and modulate an important role in various physiological and pathological processes including cancer, bacterial and viral infections, inflammation, Alzheimer’s disease, and many more. Considering their involvement in multiple diseases, their use in the development of drugs has been of significant interest in both academia and industry. Many GAG-based drugs are being developed with encouraging results in animal models and clinical trials, showcasing their potential for development as therapeutics. In this review, the role GAGs play in both the development and inhibition of cancer and inflammation is presented. Further, advancements in the development of GAGs and their mimetics as anti-cancer and anti-inflammatory agents are discussed.
Collapse
Affiliation(s)
- Shravan Morla
- Department of Medicinal Chemistry, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA.
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA.
| |
Collapse
|
9
|
Abstract
Glycosaminoglycans (GAGs) are a class of biomolecules expressed virtually on all mammalian cells and usually covalently attached to proteins, forming proteoglycans. They are present not only on the cell surface, but also in the intracellular milieu and extracellular matrix. GAGs interact with multiple ligands, both soluble and insoluble, and modulate an important role in various physiological and pathological processes including cancer, bacterial and viral infections, inflammation, Alzheimer's disease, and many more. Considering their involvement in multiple diseases, their use in the development of drugs has been of significant interest in both academia and industry. Many GAG-based drugs are being developed with encouraging results in animal models and clinical trials, showcasing their potential for development as therapeutics. In this review, the role GAGs play in both the development and inhibition of cancer and inflammation is presented. Further, advancements in the development of GAGs and their mimetics as anti-cancer and anti-inflammatory agents are discussed.
Collapse
|
10
|
Mytilinaiou M, Nikitovic D, Berdiaki A, Papoutsidakis A, Papachristou DJ, Tsatsakis A, Tzanakakis GN. IGF-I regulates HT1080 fibrosarcoma cell migration through a syndecan-2/Erk/ezrin signaling axis. Exp Cell Res 2017; 361:9-18. [PMID: 28962916 DOI: 10.1016/j.yexcr.2017.09.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/15/2017] [Accepted: 09/25/2017] [Indexed: 11/17/2022]
Abstract
Fibrosarcoma is a tumor of mesenchymal origin, originating from fibroblasts. IGF-I is an anabolic growth factor which exhibits significant involvement in cancer progression. In this study, we investigated the possible participation of syndecan-2 (SDC-2), a cell membrane heparan sulfate (HS) proteoglycan on IGF-I dependent fibrosarcoma cell motility. Our results demonstrate that SDC-2-deficient HT1080 cells exhibit attenuated IGF-I-dependent chemotactic migration (p < 0.001). SDC-2 was found to co-localize to IGF-I receptor (IGF-IR) in a manner dependent on IGF-I activity (P ≤ 0.01). In parallel, the downregulation of SDC-2 significantly inhibited both basal and due to IGF-I action ERK1/2 activation, (p < 0.001). The phosphorylation levels of ezrin (Thr567), which is suggested to act as a signaling bridge between the cellular membrane receptors and actin cytoskeleton, were strongly enhanced by IGF-I at both 1h and 24h (p < 0.05; p < 0.01). The formation of an immunoprecipitative complex revealed an association between SDC2 and ezrin which was enhanced through IGF-I action (p < 0.05). Immunoflourescence demonstrated a co-localization of IGF-IR, SDC2 and ezrin upregulated by IGF-I action. IGF-I enhanced actin polymerization and ezrin/actin specific localization to cell membranes. Finally, treatment with IGF-I strongly increased SDC2 expression at both the mRNA and protein level (p < 0.001). Therefore, we propose a novel SDC2-dependent mechanism, where SDC2 is co-localized with IGF-IR and enhances its' IGFI-dependent downstream signaling. SDC2 mediates directly IGFI-induced ERK1/2 activation, it recruits ezrin, contributes to actin polymerization and ezrin/actin specific localization to cell membranes, ultimately facilitating the progression of IGFI-dependent fibrosarcoma cell migration.
Collapse
Affiliation(s)
- Maria Mytilinaiou
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Antonis Papoutsidakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | | | - Aristidis Tsatsakis
- Laboratory of Anatomy-Histology-Embryology, Unit of Bone and Soft Tissue Studies, School of Medicine, University of Patras, Patras, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, 71003 Heraklion, Greece.
| |
Collapse
|
11
|
Molecular characterization and transcriptional analysis of the female-enriched chondroitin proteoglycan 2 of Toxocara canis. J Helminthol 2017; 92:154-160. [PMID: 28434412 DOI: 10.1017/s0022149x17000359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Toxocara canis is an important but neglected zoonotic parasite, and is the causative agent of human toxocariasis. Chondroitin proteoglycans are biological macromolecules, widely distributed in extracellular matrices, with a great diversity of functions in mammals. However, there is limited information regarding chondroitin proteoglycans in nematode parasites. In the present study, a female-enriched chondroitin proteoglycan 2 gene of T. canis (Tc-cpg-2) was cloned and characterized. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to measure the transcription levels of Tc-cpg-2 among tissues of male and female adult worms. A 485-amino-acid (aa) polypeptide was predicted from a continuous 1458-nuleotide open reading frame and designated as TcCPG2, which contains a 21-aa signal peptide. Conserved domain searching indicated three chitin-binding peritrophin-A (CBM_14) domains in the amino acid sequence of TcCPG2. Multiple alignment with the inferred amino acid sequences of Caenorhabditis elegans and Ascaris suum showed that CBM_14 domains were well conserved among these species. Phylogenetic analysis suggested that TcCPG2 was closely related to the sequence of chondroitin proteoglycan 2 of A. suum. Interestingly, a high level of Tc-cpg-2 was detected in female germline tissues, particularly in the oviduct, suggesting potential roles of this gene in reproduction (e.g. oogenesis and embryogenesis) of adult T. canis. The functional roles of Tc-cpg-2 in reproduction and development in this parasite and related parasitic nematodes warrant further functional studies.
Collapse
|
12
|
Voudouri K, Nikitovic D, Berdiaki A, Papachristou DJ, Tsiaoussis J, Spandidos DA, Tsatsakis AM, Tzanakakis GN. Heparin regulates B6FS cell motility through a FAK/actin cytoskeleton axis. Oncol Rep 2016; 36:2471-2480. [PMID: 27572115 PMCID: PMC5055209 DOI: 10.3892/or.2016.5057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/11/2016] [Indexed: 02/06/2023] Open
Abstract
Soft tissue sarcomas are rare, heterogeneous tumors of mesenchymal origin with an aggressive behavior. Heparin is a mixture of heavily sulfated, linear glycosaminoglycan (GAG) chains, which participate in the regulation of various cell biological functions. Heparin is considered to have significant anticancer capabilities, although the mechanisms involved have not been fully defined. In the present study, the effects of unfractionated heparin (UFH) and low-molecular-weight heparin (LMWH) on B6FS fibrosarcoma cell motility were examined. Both preparations of heparin were shown to both enhance B6FS cell adhesion (p<0.01 and p<0.05), and migration (p<0.05), the maximal effect being evident at the concentration of 10 µg/ml. The utilization of FAK-deficient cells demonstrated that the participation of FAK was obligatory for heparin-dependent fibrosarcoma cell adhesion (p<0.05). The results of confocal microscopy indicated that heparin was taken up by the B6FS cells, and that UFH and LMWH induced F-actin polymerization. Heparitinase digestion demonstrated that the endogenous heparan sulfate (HS) chains did not affect the motility of the B6FS cells (p>0.05, not significant). In conclusion, both UFH and LMWH, through a FAK/actin cytoskeleton axis, promoted the adhesion and migration of B6FS fibrosarcoma cells. Thus, our findings indicate that the responsiveness of fibrosarcoma cells to the exogenous heparin/HS content of the cancer microenvironment may play a role in their ability to become mobile and metastasize.
Collapse
Affiliation(s)
- Kallirroi Voudouri
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Dionysios J Papachristou
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Patras, Patras 23001, Greece
| | - John Tsiaoussis
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Demetrios A Spandidos
- Laboratory of Virology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Aristides M Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy‑Histology‑Embryology, School of Medicine, University of Crete, Heraklion 71003, Greece
| |
Collapse
|
13
|
Organotypic Cultures of Intervertebral Disc Cells: Responses to Growth Factors and Signaling Pathways Involved. BIOMED RESEARCH INTERNATIONAL 2015; 2015:427138. [PMID: 26583105 PMCID: PMC4637029 DOI: 10.1155/2015/427138] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/23/2015] [Indexed: 01/07/2023]
Abstract
Intervertebral disc (IVD) degeneration is strongly associated with low back pain, a major cause of disability worldwide. An in-depth understanding of IVD cell physiology is required for the design of novel regenerative therapies. Accordingly, aim of this work was the study of IVD cell responses to mitogenic growth factors in a three-dimensional (3D) organotypic milieu, comprising characteristic molecules of IVD's extracellular matrix. In particular, annulus fibrosus (AF) cells were cultured inside collagen type-I gels, while nucleus pulposus (NP) cells in chondroitin sulfate A (CSA) supplemented collagen gels, and the effects of Platelet-Derived Growth Factor (PDGF), basic Fibroblast Growth Factor (bFGF), and Insulin-Like Growth Factor-I (IGF-I) were assessed. All three growth factors stimulated DNA synthesis in both AF and NP 3D cell cultures, with potencies similar to those observed previously in monolayers. CSA supplementation inhibited basal DNA synthesis rates, without affecting the response to growth factors. ERK and Akt were found to be phosphorylated following growth factor stimulation. Blockade of these two signaling pathways using pharmacologic inhibitors significantly, though not completely, inhibited growth factor-induced DNA synthesis. The proposed culture systems may prove useful for further in vitro studies aiming at future interventions for IVD regeneration.
Collapse
|
14
|
Kouvidi K, Nikitovic D, Berdiaki A, Tzanakakis GN. Hyaluronan/RHAMM interactions in mesenchymal tumor pathogenesis: role of growth factors. Adv Cancer Res 2015; 123:319-49. [PMID: 25081535 DOI: 10.1016/b978-0-12-800092-2.00012-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosarcoma belongs to the sarcoma cancer group, which are spindle cell malignancies of mesenchymal origin, and owe their name to the predominant cell line that is present within the tumor. The extracellular matrix (ECM) is a complicated structure that surrounds and supports cells within tissues. Its main components are proteoglycans, collagens, glycoproteins, hyaluronan (HA), and several matrix-degrading enzymes. During cancer progression, significant changes can be observed in the structural and mechanical properties of ECM components. The ECM provides a physical scaffold to which tumor cells attach and migrate. Thus, it is required for key cellular events such as cell motility, adhesion, proliferation, invasion, and metastasis. Importantly, fibrosarcomas were shown to have a high content and turnover of ECM components including HA, proteoglycans, collagens, fibronectin, and laminin. In this review, we will focus on the HA component of fibrosarcoma ECM and critically discuss its role and involved mechanisms during fibrosarcoma pathogenesis.
Collapse
Affiliation(s)
- Katerina Kouvidi
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece.
| |
Collapse
|
15
|
Vallen MJ, van der Steen SC, van Tilborg AA, Massuger LF, van Kuppevelt TH. Sulfated sugars in the extracellular matrix orchestrate ovarian cancer development: ‘When sweet turns sour’. Gynecol Oncol 2014; 135:371-81. [DOI: 10.1016/j.ygyno.2014.08.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 08/15/2014] [Accepted: 08/16/2014] [Indexed: 01/14/2023]
|
16
|
Koźma EM, Wisowski G, Latocha M, Kusz D, Olczyk K. Complex influence of dermatan sulphate on breast cancer cells. Exp Biol Med (Maywood) 2014; 239:1575-88. [PMID: 24912503 DOI: 10.1177/1535370214538590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Tumor transformation and progression both lead to extracellular matrix remodeling, which is also reflected in an alteration in the proportion of dermatan sulphate (DS) and chondroitin sulphate (CS) and an accumulation of the latter. In addition, a significant increase in the 6-O-sulphated disaccharide contribution to the structure of both glycosaminoglycans has been observed. It is commonly accepted that CS is more permissive for tumor growth than DS. However, the detailed role of DS in tumor progression is poorly known. We tested the effects of structurally different DSs on the behavior of cultured breast cancer cells. At a high dose (10 µg/mL), all of the DSs significantly reduced cancer cell growth, although some differences in the efficiency of action were apparent. In contrast, when used at a concentration of 1 µg/mL, the examined DSs evoked different responses ranging from the stimulation to the inhibition of cancer cell proliferation. The highest stimulatory activity was associated with fibrosis-affected fascia decorin DS, which is characterized by a particularly high content of 6-O-sulphated disaccharides. Further reduction in DS concentration to 0.5 µg/mL preserved majority of biological effects which were apparent at a dose of 1 µg/mL. The enzymatic fragmentation of the DSs, particularly by chondroitinase AC I, abolished the impact exerted by 1 µg/mL of the intact DS chains and sometimes resulted in the opposite effect. In contrast to DSs, highly sulphated C-6-S exhibited no effect on the cancer cells. Our data revealed the complexity of the effects of DSs on breast cancer cells, which include both co-receptor activity and the prevention of vascular endothelial growth factor action. In addition, the biological effect of DSs is strongly dependent not only on the glycosaminoglycan structure but also on its content in the cancer environment.
Collapse
Affiliation(s)
- Ewa M Koźma
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Silesia, Sosnowiec 41-200, ul. Jedności 8, Poland
| | - Grzegorz Wisowski
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Silesia, Sosnowiec 41-200, ul. Jedności 8, Poland
| | - Małgorzata Latocha
- Department of Cell Biology, Medical University of Silesia, Sosnowiec 41-200, ul. Jedności 8, Poland
| | - Damian Kusz
- Department of Orthopaedics and Traumatology, Medical University of Silesia, Katowice 40-635, ul. Ziołowa 45/47, Poland
| | - Krystyna Olczyk
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Silesia, Sosnowiec 41-200, ul. Jedności 8, Poland
| |
Collapse
|
17
|
Affiliation(s)
- Vitor H. Pomin
- Program of
Glycobiology, Institute of Medical Biochemistry,
and University Hospital Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-913,
Brazil
| |
Collapse
|
18
|
Afratis N, Gialeli C, Nikitovic D, Tsegenidis T, Karousou E, Theocharis AD, Pavão MS, Tzanakakis GN, Karamanos NK. Glycosaminoglycans: key players in cancer cell biology and treatment. FEBS J 2012; 279:1177-97. [DOI: 10.1111/j.1742-4658.2012.08529.x] [Citation(s) in RCA: 380] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
19
|
Boddohi S, Kipper MJ. Engineering nanoassemblies of polysaccharides. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2010; 22:2998-3016. [PMID: 20593437 DOI: 10.1002/adma.200903790] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Polysaccharides offer a wealth of biochemical and biomechanical functionality that can be used to develop new biomaterials. In mammalian tissues, polysaccharides often exhibit a hierarchy of structure, which includes assembly at the nanometer length scale. Furthermore, their biochemical function is determined by their nanoscale organization. These biological nanostructures provide the inspiration for developing techniques to tune the assembly of polysaccharides at the nanoscale. These new polysaccharide nanostructures are being used for the stabilization and delivery of drugs, proteins, and genes, the engineering of cells and tissues, and as new platforms on which to study biochemistry. In biological systems polysaccharide nanostructures are assembled via bottom-up processes. Many biologically derived polysaccharides behave as polyelectrolytes, and their polyelectrolyte nature can be used to tune their bottom-up assembly. New techniques designed to tune the structure and composition of polysaccharides at the nanoscale are enabling researchers to study in detail the emergent biological properties that arise from the nanoassembly of these important biological macromolecules.
Collapse
Affiliation(s)
- Soheil Boddohi
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
20
|
Conrad AH, Zhang Y, Tasheva ES, Conrad GW. Proteomic analysis of potential keratan sulfate, chondroitin sulfate A, and hyaluronic acid molecular interactions. Invest Ophthalmol Vis Sci 2010; 51:4500-15. [PMID: 20375348 DOI: 10.1167/iovs.09-4914] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Corneal stroma extracellular matrix (ECM) glycosaminoglycans (GAGs) include keratan sulfate (KS), chondroitin sulfate A (CSA), and hyaluronic acid (HA). Embryonic corneal keratocytes and sensory nerve fibers grow and differentiate according to chemical cues they receive from the ECM. This study asked which of the proteins that may regulate keratocytes or corneal nerve growth cone immigration interact with corneal GAGs. METHODS Biotinylated KS (bKS), CSA (bCSA), and HA (bHA) were prepared and used in microarray protocols to assess their interactions with 8268 proteins and a custom microarray of 85 extracellular epitopes of nerve growth-related proteins. Surface plasmon resonance (SPR) was performed with bKS and SLIT2, and their ka, kd, and KD were determined. RESULTS Highly sulfated KS interacted with 217 microarray proteins, including 75 kinases, several membrane or secreted proteins, many cytoskeletal proteins, and many nerve function proteins. CSA interacted with 24 proteins, including 10 kinases and 2 cell surface proteins. HA interacted with 6 proteins, including several ECM-related structural proteins. Of 85 ECM nerve-related epitopes, KS bound 40 proteins, including SLIT, 2 ROBOs, 9 EPHs, 8 Ephrins (EFNs), 8 semaphorins (SEMAs), and 2 nerve growth factor receptors. CSA bound nine proteins, including ROBO2, 2 EPHs, 1 EFN, two SEMAs, and netrin 4. HA bound no ECM nerve-related epitopes. SPR confirmed that KS binds SLIT2 strongly. The KS core protein mimecan/osteoglycin bound 15 proteins. CONCLUSIONS Corneal stromal GAGs bind, and thus could alter the availability or conformation of, many proteins that may influence keratocyte and nerve growth cone behavior in the cornea.
Collapse
Affiliation(s)
- Abigail H Conrad
- Division of Biology, Kansas State University, Manhattan, Kansas 66506-4901, USA.
| | | | | | | |
Collapse
|
21
|
Bret C, Hose D, Reme T, Sprynski AC, Mahtouk K, Schved JF, Quittet P, Rossi JF, Goldschmidt H, Klein B. Expression of genes encoding for proteins involved in heparan sulphate and chondroitin sulphate chain synthesis and modification in normal and malignant plasma cells. Br J Haematol 2009; 145:350-68. [PMID: 19298595 DOI: 10.1111/j.1365-2141.2009.07633.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Syndecan-1 is a proteoglycan that concentrates heparin-binding factors on the surface of multiple myeloma cells, and probably plays a major role in multiple myeloma biology. As heparan sulphate and chondroitin sulphate are the bioactive components of syndecan-1, we analysed the signature of genes encoding 100 proteins involved in synthesis of these chains, i.e. from precursor uptake to post-translational modifications, using Affymetrix microarrays. The expression of enzymes required for heparan sulphate and chondroitin sulphate biosynthesis was shown to increase in parallel with syndecan-1 expression, throughout the differentiation of memory B cells into plasmablasts and normal bone marrow plasma cells. Sixteen genes were significantly different between normal and malignant plasma cells, nine of these genes -EXT2, CHSY3, CSGALNACT1, HS3ST2, HS2ST1, CHST11, CSGALNACT2, HPSE, SULF2 - encode proteins involved in glycosaminoglycan chain synthesis or modifications. Kaplan-Meier analysis was performed in two independent series of patients: B4GALT7, CSGALNACT1, HS2ST1 were associated with a good prognosis whereas EXT1 was linked to a bad prognosis. This study provides an overall picture of the major genes encoding for proteins involved in heparan sulphate and chondroitin sulphate synthesis and modifications that can be implicated in normal and malignant plasma cells.
Collapse
Affiliation(s)
- Caroline Bret
- INSERM U847, Equipe Labellisée LIGUE 2006, Université Montpellier, UFR Méldecine, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Increased expression of non-sulfated chondroitin correlates with adverse clinicopathological parameters in prostate cancer. Mod Pathol 2008; 21:893-901. [PMID: 18487997 DOI: 10.1038/modpathol.2008.70] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Chondroitin sulfate is a structurally diverse glycosaminoglycan, which contains a variable degree of sulfation that helps to determine its biological function. It is involved in the regulation of cellular activity and has been implicated in carcinogenesis. To determine if the non-sulfated chondroitin backbone has a functional role in prostate cancer, we analyzed its expression by immunohistochemistry using the 1B5 monoclonal antibody and a set of tissue microarrays constructed with 227 prostate specimen cores from 81 cases of benign prostate tissue and 77 cases of prostate cancer, of which 69 of these cases are matched. Non-sulfated chondroitin was found in the secretory epithelial cells and stromal regions of both prostatic adenocarcinoma and benign prostatic tissues, as well as in the basal cells of benign glands. A higher percentage of cancerous cells were stained positively for non-sulfated chondroitin as compared with benign secretory cells of the same patient. Cancerous cells stained more intensely for non-sulfated chondroitin. This increase in percentage of cells stained and increase in staining intensity were associated with higher pathological T stage and extraprostatic extension. Non-sulfated chondroitin expression (either staining intensity or percentage of cells stained) in adenocarcinoma and its peritumoral stroma correlated significantly with several clinicopathological parameters of unfavorable outcome, including higher pathological T stage and Gleason score, presence of tumor in both prostatic lobes, extraprostatic extension, seminal vesicle involvement and preoperative prostate-specific antigen levels. These data suggest that non-sulfated chondroitin is a potentially useful biomarker for prostate cancer, and may be involved in regulating prostate cancer behavior.
Collapse
|
23
|
Fthenou E, Zafiropoulos A, Katonis P, Tsatsakis A, Karamanos N, Tzanakakis G. Chondroitin sulfate prevents platelet derived growth factor-mediated phosphorylation of PDGF-Rβ in normal human fibroblasts severely impairing mitogenic responses. J Cell Biochem 2008; 103:1866-76. [DOI: 10.1002/jcb.21570] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
24
|
Zafiropoulos A, Fthenou E, Chatzinikolaou G, Tzanakakis GN. Glycosaminoglycans and PDGF signaling in mesenchymal cells. Connect Tissue Res 2008; 49:153-6. [PMID: 18661332 DOI: 10.1080/03008200802148702] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Platelet derived growth factor (PDGF) is involved in the autocrine growth stimulation of normal and malignant cells, the stimulation of angiogenesis, and the recruitment and regulation of tumor fibroblasts. PDGF has been shown to physically interact with glycosaminoglycans which are abundant in the extracellular microenvironment. The present review discusses the effects of glycosaminoglycans on the functions mediated by the PDGF on cells of mesenchymal origin. Recent studies have demonstrated that both soluble and surface bound glycosaminoglycan chains can modulate PDGF-BB isoform signaling depending on the cell type. These data demonstrated that the microenvironment rich in GAGs/PGs is able to significantly modify the cellular response to PDGF-BB signaling in a critical way for cell growth and differentiation.
Collapse
|
25
|
Berdiaki A, Zafiropoulos A, Fthenou E, Katonis P, Tsatsakis A, Karamanos NK, Tzanakakis GN. Regulation of hyaluronan and versican deposition by growth factors in fibrosarcoma cell lines. Biochim Biophys Acta Gen Subj 2007; 1780:194-202. [PMID: 17980161 DOI: 10.1016/j.bbagen.2007.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/14/2007] [Accepted: 10/10/2007] [Indexed: 10/22/2022]
Abstract
Versican, a large chondroitin sulphate proteoglycan and hyaluronan (HA), a non-sulphated glycosaminoglycan are major constituents of the pericellular matrix. In many neoplastic tissues, changes in the expression of versican and HA affect tumour progression. Here, we analyse the synthesis of versican and hyaluronan by fibrosarcoma cells, and document how the latter is affected by PDGF-BB, bFGF and TGFB2, growth factors endogenously produced by these cells. Fibrosarcoma cell lines B6FS and HT1080 were utilised and compared with normal lung fibroblasts (DLF). The major versican isoforms expressed by DLF and B6FS cells were V0 and V1. Treatment of B6FS cells with TGFB2 showed a significant increase of V0 and V1 mRNAs. Versican expression in HT1080 cells was not significantly affected by any of the growth factors. In addition, TGFB2 treatment increased versican protein in DLF cells. HA, showed approximately a 2-fold and a 9-fold higher production in DLF cells compared to B6FS and HT1080 cells, respectively. In HT1080 cells, HA biosynthesis was significantly increased by bFGF, whereas, in B6FS cells it was increased by TGFB2 and PDGF-BB. Furthermore, analysis of HA synthases (HAS) expression indicated that HT1080 expressed similar levels of all three HAS isoforms in the following order: HAS2> HAS3> HAS1. bFGF shifted that balance by increasing the abundance of HAS1. The major HAS isoform expressed by B6FS cells was HAS2. PDGF-BB and TGFB2 showed the most prominent effects by increasing both HAS2 and HAS1 isoforms. In conclusion, these growth factors modulated, through upregulation of specific HAS isoforms, HA synthesis, secretion and net deposition to the pericellular matrix.
Collapse
Affiliation(s)
- A Berdiaki
- Department of Histology, Medical School, University of Crete, 71003, Heraklion, Greece
| | | | | | | | | | | | | |
Collapse
|
26
|
Yu F, Wolff JJ, Amster IJ, Prestegard JH. Conformational Preferences of Chondroitin Sulfate Oligomers Using Partially Oriented NMR Spectroscopy of 13C-Labeled Acetyl Groups. J Am Chem Soc 2007; 129:13288-97. [DOI: 10.1021/ja075272h] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Fei Yu
- Contribution from the Complex Carbohydrate Research Center and Department of Chemistry, University of Georgia, Athens, Georgia 30602-4712
| | - Jeremy J. Wolff
- Contribution from the Complex Carbohydrate Research Center and Department of Chemistry, University of Georgia, Athens, Georgia 30602-4712
| | - I. Jonathan Amster
- Contribution from the Complex Carbohydrate Research Center and Department of Chemistry, University of Georgia, Athens, Georgia 30602-4712
| | - James H. Prestegard
- Contribution from the Complex Carbohydrate Research Center and Department of Chemistry, University of Georgia, Athens, Georgia 30602-4712
| |
Collapse
|