1
|
Ghahtan N, Dehghan N, Ullah M, Khoradmehr A, Habibi H, Nabipour I, Baghban N. From seaweed to healing: the potential of fucoidan in wound therapy. Nat Prod Res 2025; 39:1345-1358. [PMID: 38804629 DOI: 10.1080/14786419.2024.2358387] [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: 09/26/2023] [Revised: 04/28/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
This bibliometric review examines the current state of research on fucoidan, a sulphated polysaccharide found in brown seaweed species, and its potential for wound healing. The review included 58 studies that investigated fucoidan's effects on wound healing, revealing that it possesses anti-inflammatory and antioxidant properties that could aid in the healing process. Fucoidan was also found to promote cell proliferation, migration, and angiogenesis, essential for wound healing. However, the optimal dosage, treatment duration, safety, and efficacy of fucoidan in various wound types and patient populations still require further investigation. Additionally, advanced wound dressings like hydrogels have garnered significant attention for their potential in wound healing. While this review indicates promise for fucoidan as a natural wound healing compound, it underscores the need for additional clinical trials to determine its optimal use as a commercial therapeutic agent in wound healing.
Collapse
Affiliation(s)
- Najmeh Ghahtan
- Department of Medicinal Chemistry, Faculty of Chemistry, Shiraz University of Technology, Shiraz, Iran
| | - Niloofar Dehghan
- Bushehr University of Medical Sciences, Bushehr, Iran
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
- Department of Cancer Immunology, Genentech Inc, South SanFrancisco, CA, USA
- Molecular Medicine Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hassan Habibi
- Department of Animal Sciences, Faculty of Agricultural and Natural Resources, Persian Gulf University, Bushehr, Iran
| | - Iraj Nabipour
- Bushehr University of Medical Sciences, Bushehr, Iran
| | - Neda Baghban
- Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
2
|
Forget A, Shastri VP. Sulfated and Phosphorylated Agarose as Biomaterials for a Biomimetic Paradigm for FGF-2 Release. Biomimetics (Basel) 2024; 10:12. [PMID: 39851728 PMCID: PMC11761575 DOI: 10.3390/biomimetics10010012] [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] [Received: 10/10/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/26/2025] Open
Abstract
Cardiovascular diseases such as myocardial infarction or limb ischemia are characterized by regression of blood vessels. Local delivery of growth factors (GFs) involved in angiogenesis such as fibroblast blast growth factor-2 (FGF-2) has been shown to trigger collateral neovasculature and might lead to a therapeutic strategy. In vivo, heparin, a sulfated polysaccharide present in abundance in the extracellular matrix (ECM), has been shown to function as a local reservoir for FGF-2 by binding FGF-2 and other morphogens and it plays a role in the evolution of GF gradients. To access injectable biomaterials that can mimic such natural electrostatic interactions between soluble signals and macromolecules and mechanically tunable environments, the backbone of agarose, a thermogelling marine-algae-derived polysaccharide, was modified with sulfate, phosphate, and carboxylic moieties and the interaction and release of FGF-2 from these functionalized hydrogels was assessed by ELISA in vitro and CAM assay in ovo. Our findings show that FGF-2 remains active after release, and FGF-2 release profiles can be influenced by sulfated and phosphorylated agarose, and in turn, promote varied blood vessel formation kinetics. These modified agaroses offer a simple approach to mimicking electrostatic interactions experienced by GFs in the extracellular environment and provide a platform to probe the role of these interactions in the modulation of growth factor activity and may find utility as an injectable gel for promoting angiogenesis and as bioinks in 3D bioprinting.
Collapse
Affiliation(s)
- Aurelien Forget
- Institute for Macromolecular Chemistry, Stefan-Meier-Strasse 31, 79104 Freiburg, Germany;
- BIOSS, Centre for Biological Signalling, Schanzelstrasse 18, 79104 Freiburg, Germany
| | - V. Prasad Shastri
- Institute for Macromolecular Chemistry, Stefan-Meier-Strasse 31, 79104 Freiburg, Germany;
- BIOSS, Centre for Biological Signalling, Schanzelstrasse 18, 79104 Freiburg, Germany
| |
Collapse
|
3
|
Whitehead AK, Wang Z, Boustany RJ, Vivès RR, Lazartigues E, Liu J, Siggins RW, Yue X. Myeloid deficiency of heparan sulfate 6-O-endosulfatases impairs bone marrow hematopoiesis. Matrix Biol 2024; 134:107-118. [PMID: 39368561 PMCID: PMC11585435 DOI: 10.1016/j.matbio.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/06/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The heparan sulfate (HS) 6-O-endosulfatases or the Sulfs (Sulf1 and Sulf2) are the only known enzymes that can modify HS sulfation status extracellularly and have been shown to regulate diverse biological processes. The role of the Sulfs in bone marrow (BM) hematopoiesis is not known. In this study, we generated a novel mouse line with myeloid-specific deletion of the Sulfs by crossing Sulf1/2 double floxed mice with the LysM-cre line. The LysM-Sulf knockout (KO) male mice exhibited age-dependent expansion of hematopoietic stem cells and the granulocyte-monocyte lineages in the BM, whereas common lymphoid progenitors and B lymphocyte populations were significantly reduced. Although megakaryocytic and erythroid progenitors were not reduced in the BM, the LysM-Sulf KO males suffered age-dependent reduction of red blood cells (RBCs) and platelets in the peripheral blood, suggesting that the production of RBCs and platelets was arrested at later stages. In addition, LysM-Sulf KO males displayed progressive splenomegaly with extramedullary hematopoiesis. Compared to males, LysM-Sulf KO females exhibited a much-reduced phenotype, and ovariectomy had little effect. Mechanistically, reduced TGF-β/Smad2 but enhanced p53/p21 signaling were observed in male but not female LysM-Sulf KO mice. Finally, HS disaccharide analysis via LC-MS/MS revealed increased HS 6-O-sulfation in the BM from both male and female LysM-Sulf KO mice, however, the distribution of 6-O-sulfated motifs were different between the sexes with compensatory increase in Sulf1 expression observed only in LysM-Sulf KO females. In conclusion, our study reveals that myeloid deficiency of the Sulfs leads to multilineage abnormalities in BM hematopoiesis in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Anna K Whitehead
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | | | - Romain R Vivès
- University of Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Eric Lazartigues
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Robert W Siggins
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Xinping Yue
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
4
|
Ahrens L, Forget A, Shastri VP. Modulation of Short-Term Delivery of Proteins from Hydrogels. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64568-64578. [PMID: 39552122 DOI: 10.1021/acsami.4c15185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
For modulation of cellular behavior, systems that can provide controlled delivery of proteins (soluble signals) over a few hours to a few days are highly desirable. Conventional erosion-controlled systems are inadequate as their degradation spans days to months. Conversely, hydrogels offer quicker release but are limited by a high burst release that can lead to cytotoxicity and rapid depletion of the permeant. To avoid burst release and achieve controlled diffusion of proteins, we propose exploiting electrostatic interactions between the hydrogel matrix and proteins. Here we demonstrate this concept using two disparate hydrogel systems: (1) a chemically cross-linked protein (gelatin) matrix and (2) a physically cross-linked polysaccharide (agarose) matrix and three proteins having different isoelectric points. By introducing fixed charges into the hydrogel matrix using carboxylated agarose (CA), the precise and controlled release of BSA, lactoferrin, and FGF2 over a few hours to days is demonstrated. Using electroendosmosis, we further provide evidence for a clear role for CA in modulating the release. Our findings suggest that the paradigm presented herein has the potential to significantly enhance the design of hydrogel systems for the delivery of proteins and RNA therapeutics for vaccines and biomedical applications ranging from tissue engineering to functional coatings for medical devices.
Collapse
Affiliation(s)
- Lucas Ahrens
- Institute for Macromolecular Chemistry, Hermann-Staudinger-Haus, University of Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg, Germany
- BIOSS Centre for Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Aurélien Forget
- Institute for Macromolecular Chemistry, Hermann-Staudinger-Haus, University of Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg, Germany
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, Hermann-Staudinger-Haus, University of Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg, Germany
- BIOSS Centre for Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| |
Collapse
|
5
|
Brown NE, Ellerbe LR, Hollister SJ, Temenoff JS. Development and Characterization of Heparin-Containing Hydrogel/3D-Printed Scaffold Composites for Craniofacial Reconstruction. Ann Biomed Eng 2024; 52:2287-2307. [PMID: 38734845 DOI: 10.1007/s10439-024-03530-z] [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: 01/11/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Regeneration of cartilage and bone tissues remains challenging in tissue engineering due to their complex structures, and the need for both mechanical support and delivery of biological repair stimuli. Therefore, the goal of this study was to develop a composite scaffold platform for anatomic chondral and osteochondral repair using heparin-based hydrogels to deliver small molecules within 3D-printed porous scaffolds that provide structure, stiffness, and controlled biologic delivery. We designed a mold-injection system to combine hydrolytically degradable hydrogels and 3D-printed scaffolds that could be employed rapidly (< 30 min) in operating room settings (~23 °C). Micro-CT analysis demonstrated the effectiveness of our injection system through homogeneously distributed hydrogel within the pores of the scaffolds. Hydrogels and composite scaffolds exhibited efficient loading (~94%) of a small positively charged heparin-binding molecule (crystal violet) with sustained release over 14 days and showed high viability of encapsulated porcine chondrocytes over 7 days. Compression testing demonstrated nonlinear viscoelastic behavior where tangent stiffness decreased with scaffold porosity (porous scaffold tangent stiffness: 70%: 4.9 MPa, 80%: 1.5 MPa, and 90%: 0.20 MPa) but relaxation was not affected. Lower-porosity scaffolds (70%) showed stiffness similar to lower ranges of trabecular bone (4-8 MPa) while higher-porosity scaffolds (80% and 90%) showed stiffness similar to auricular cartilage (0.16-2 MPa). Ultimately, this rapid composite scaffold fabrication method may be employed in the operating room and utilized to control biologic delivery within load-bearing scaffolds.
Collapse
Affiliation(s)
- Nettie E Brown
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Lela R Ellerbe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Scott J Hollister
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA.
| | - Johnna S Temenoff
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA.
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr, Atlanta, GA, 30332, USA.
| |
Collapse
|
6
|
Piszczatowski RT, Bülow HE, Steidl U. Heparan sulfates and heparan sulfate proteoglycans in hematopoiesis. Blood 2024; 143:2571-2587. [PMID: 38639475 PMCID: PMC11830984 DOI: 10.1182/blood.2023022736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
ABSTRACT From signaling mediators in stem cells to markers of differentiation and lineage commitment to facilitators for the entry of viruses, such as HIV-1, cell surface heparan sulfate (HS) glycans with distinct modification patterns play important roles in hematopoietic biology. In this review, we provide an overview of the importance of HS and the proteoglycans (HSPGs) to which they are attached within the major cellular subtypes of the hematopoietic system. We summarize the roles of HSPGs, HS, and HS modifications within each main hematopoietic cell lineage of both myeloid and lymphoid arms. Lastly, we discuss the biological advances in the detection of HS modifications and their potential to further discriminate cell types within hematopoietic tissue.
Collapse
Affiliation(s)
- Richard T. Piszczatowski
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Pediatrics, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
- Departments of Oncology, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY
- Blood Cancer Institute, Albert Einstein College of Medicine, Bronx, NY
- Ruth L. and David S. Gottesman Institute for Stem Cell Research and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
7
|
Messina JM, Luo M, Hossan MS, Gadelrab HA, Yang X, John A, Wilmore JR, Luo J. Unveiling cytokine charge disparity as a potential mechanism for immune regulation. Cytokine Growth Factor Rev 2024; 77:1-14. [PMID: 38184374 PMCID: PMC11923798 DOI: 10.1016/j.cytogfr.2023.12.002] [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: 11/20/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
Cytokines are small signaling proteins that regulate the immune responses to infection and tissue damage. Surface charges of cytokines determine their in vivo fate in immune regulation, e.g., half-life and distribution. The overall negative charges in the extracellular microenvironment and the acidosis during inflammation and infection may differentially impact cytokines with different surface charges for fine-tuned immune regulation via controlling tissue residential properties. However, the trend and role of cytokine surface charges has yet to be elucidated in the literature. Interestingly, we have observed that most pro-inflammatory cytokines have a negative charge, while most anti-inflammatory cytokines and chemokines have a positive charge. In this review, we extensively examined the surface charges of all cytokines and chemokines, summarized the pharmacokinetics and tissue adhesion of major cytokines, and analyzed the link of surface charge with cytokine biodistribution, activation, and function in immune regulation. Additionally, we identified that the general trend of charge disparity between pro- and anti-inflammatory cytokines represents a unique opportunity to develop precise immune modulation approaches, which can be applied to many inflammation-associated diseases including solid tumors, chronic wounds, infection, and sepsis.
Collapse
Affiliation(s)
- Jennifer M Messina
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Minghao Luo
- Department of Clinical Medicine, 2nd Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Md Shanewaz Hossan
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Hadil A Gadelrab
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiguang Yang
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Anna John
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Joel R Wilmore
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States; Upstate Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Juntao Luo
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States; Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States; Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY 13210, United States; Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States; Upstate Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States.
| |
Collapse
|
8
|
Schuurmans F, Wagemans KE, Adema GJ, Cornelissen LAM. Tumor glucose metabolism and the T cell glycocalyx: implication for T cell function. Front Immunol 2024; 15:1409238. [PMID: 38881904 PMCID: PMC11176483 DOI: 10.3389/fimmu.2024.1409238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The T cell is an immune cell subset highly effective in eliminating cancer cells. Cancer immunotherapy empowers T cells and occupies a solid position in cancer treatment. The response rate, however, remains relatively low (<30%). The efficacy of immunotherapy is highly dependent on T cell infiltration into the tumor microenvironment (TME) and the ability of these infiltrated T cells to sustain their function within the TME. A better understanding of the inhibitory impact of the TME on T cells is crucial to improve cancer immunotherapy. Tumor cells are well described for their switch into aerobic glycolysis (Warburg effect), resulting in high glucose consumption and a metabolically distinct TME. Conversely, glycosylation, a predominant posttranslational modification of proteins, also relies on glucose molecules. Proper glycosylation of T cell receptors influences the immunological synapse between T cells and tumor cells, thereby affecting T cell effector functions including their cytolytic and cytostatic activities. This review delves into the complex interplay between tumor glucose metabolism and the glycocalyx of T cells, shedding light on how the TME can induce alterations in the T cell glycocalyx, which can subsequently influence the T cell's ability to target and eliminate tumor cells.
Collapse
Affiliation(s)
| | | | | | - Lenneke A. M. Cornelissen
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
9
|
Fok SW, Gresham RCH, Ryan W, Osipov B, Bahney C, Leach JK. Macromolecular crowding and decellularization method increase the growth factor binding potential of cell-secreted extracellular matrices. Front Bioeng Biotechnol 2023; 11:1091157. [PMID: 36756385 PMCID: PMC9899907 DOI: 10.3389/fbioe.2023.1091157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Recombinant growth factors are used in tissue engineering to stimulate cell proliferation, migration, and differentiation. Conventional methods of growth factor delivery for therapeutic applications employ large amounts of these bioactive cues. Effective, localized growth factor release is essential to reduce the required dose and potential deleterious effects. The endogenous extracellular matrix (ECM) sequesters native growth factors through its negatively charged sulfated glycosaminoglycans. Mesenchymal stromal cells secrete an instructive extracellular matrix that can be tuned by varying culture and decellularization methods. In this study, mesenchymal stromal cell-secreted extracellular matrix was modified using λ-carrageenan as a macromolecular crowding (MMC) agent and decellularized with DNase as an alternative to previous decellularized extracellular matrices (dECM) to improve growth factor retention. Macromolecular crowding decellularized extracellular matrix contained 7.7-fold more sulfated glycosaminoglycans and 11.7-fold more total protein than decellularized extracellular matrix, with no significant difference in residual DNA. Endogenous BMP-2 was retained in macromolecular crowding decellularized extracellular matrix, whereas BMP-2 was not detected in other extracellular matrices. When implanted in a murine muscle pouch, we observed increased mineralized tissue formation with BMP-2-adsorbed macromolecular crowding decellularized extracellular matrix in vivo compared to conventional decellularized extracellular matrix. This study demonstrates the importance of decellularization method to retain endogenous sulfated glycosaminoglycans in decellularized extracellular matrix and highlights the utility of macromolecular crowding to upregulate sulfated glycosaminoglycan content. This platform has the potential to aid in the delivery of lower doses of BMP-2 or other heparin-binding growth factors in a tunable manner.
Collapse
Affiliation(s)
- Shierly W. Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Robert C. H. Gresham
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Weston Ryan
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Benjamin Osipov
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States
| | - Chelsea Bahney
- Steadman Philippon Research Institute, Vail, CO, United States
| | - J. Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, United States,Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States,*Correspondence: J. Kent Leach,
| |
Collapse
|
10
|
Regeneration of Osteochondral Defects by Combined Delivery of Synovium-Derived Mesenchymal Stem Cells, TGF-β1 and BMP-4 in Heparin-Conjugated Fibrin Hydrogel. Polymers (Basel) 2022; 14:polym14245343. [PMID: 36559710 PMCID: PMC9780905 DOI: 10.3390/polym14245343] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/08/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The regeneration of cartilage and osteochondral defects remains one of the most challenging clinical problems in orthopedic surgery. Currently, tissue-engineering techniques based on the delivery of appropriate growth factors and mesenchymal stem cells (MSCs) in hydrogel scaffolds are considered as the most promising therapeutic strategy for osteochondral defects regeneration. In this study, we fabricated a heparin-conjugated fibrin (HCF) hydrogel with synovium-derived mesenchymal stem cells (SDMSCs), transforming growth factor-β1 (TGF-β1) and bone morphogenetic protein-4 (BMP-4) to repair osteochondral defects in a rabbit model. An in vitro study showed that HCF hydrogel exhibited good biocompatibility, a slow degradation rate and sustained release of TGF-β1 and BMP-4 over 4 weeks. Macroscopic and histological evaluations revealed that implantation of HCF hydrogel with SDMSCs, TGF-β1 and BMP-4 significantly enhanced the regeneration of hyaline cartilage and the subchondral bone plate in osteochondral defects within 12 weeks compared to hydrogels with SDMSCs or growth factors alone. Thus, these data suggest that combined delivery of SDMSCs with TGF-β1 and BMP-4 in HCF hydrogel may synergistically enhance the therapeutic efficacy of osteochondral defect repair of the knee joints.
Collapse
|
11
|
Liu Z, Chen M, Sun Y, Li X, Cao L, Ma X. Transforming growth factor-β receptor type 2 is required for heparin-binding protein-induced acute lung injury and vascular leakage for transforming growth factor-β/Smad/Rho signaling pathway activation. FASEB J 2022; 36:e22580. [PMID: 36189652 DOI: 10.1096/fj.202200228rrrrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/11/2022]
Abstract
Heparin-binding protein (HBP), as a granule protein secreted by polymorphonuclear neutrophils, participates in the pathophysiological process of sepsis. It has been reported that HBP is a biomarker of sepsis related to the severity of septic shock and organ dysfunction. HBP binds to vascular endothelial cells as a primary target site. However, it is still unclear whether HBP-binding protein receptors exist on the surface of endothelial cells. The effect of HBP on vascular permeability in sepsis and its mechanism needs to be explored. We conducted in vivo and in vitro studies and demonstrated that HBP binds to transforming growth factor-β receptor type 2 (TGF-β-R2) as a ligand. Glutathione S-transferase pull-down analysis revealed that HBP mainly interacts with the extracellular domain of TGF-β-R2. HBP induces acute lung injury and vascular leakage via activation of the TGF-β/SMAD2/3 signaling pathway. A permeability assay suggested that TGF-β-R2 is necessary for HBP-induced increased permeability. We also defined the role of HBP and its potential membrane receptor TGF-β-R2 in the blood-gas barrier in the pathogenesis of HBP-related acute lung injury.
Collapse
Affiliation(s)
- Zixuan Liu
- Department of Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China.,Department of Critical Care Medicine, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China
| | - Mingming Chen
- Department of Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| | - Yini Sun
- Department of Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| | - Xu Li
- Department of Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| | - Liu Cao
- Institute of Translational Medicine, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, China Medical University, Shenyang, China
| | - Xiaochun Ma
- Department of Critical Care Medicine, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Wan HY, Shin RLY, Chen JCH, Assunção M, Wang D, Nilsson SK, Tuan RS, Blocki A. Dextran sulfate-amplified extracellular matrix deposition promotes osteogenic differentiation of mesenchymal stem cells. Acta Biomater 2022; 140:163-177. [PMID: 34875356 DOI: 10.1016/j.actbio.2021.11.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
The development of bone-like tissues in vitro that exhibit key features similar to those in vivo is needed to produce tissue models for drug screening and the study of bone physiology and disease pathogenesis. Extracellular matrix (ECM) is a predominant component of bone in vivo; however, as ECM assembly is sub-optimal in vitro, current bone tissue engineering approaches are limited by an imbalance in ECM-to-cell ratio. We amplified the deposition of osteoblastic ECM by supplementing dextran sulfate (DxS) into osteogenically induced cultures of human mesenchymal stem cells (MSCs). DxS, previously implicated to act as a macromolecular crowder, was recently demonstrated to aggregate and co-precipitate major ECM components, including collagen type I, thereby amplifying its deposition. This effect was re-confirmed for MSC cultures undergoing osteogenic induction, where DxS supplementation augmented collagen type I deposition, accompanied by extracellular osteocalcin accumulation. The resulting differentiated osteoblasts exhibited a more mature osteogenic gene expression profile, indicated by a strong upregulation of the intermediate and late osteogenic markers ALP and OCN, respectively. The associated cellular microenvironment was also enriched in bone morphogenetic protein 2 (BMP-2). Interestingly, the resulting decellularized matrices exhibited the strongest osteo-inductive effects on re-seeded MSCs, promoted cell proliferation, osteogenic marker expression and ECM calcification. Taken together, these findings suggest that DxS-mediated enhancement of osteogenic differentiation by MSCs is mediated by the amplified ECM, which is enriched in osteo-inductive factors. We have thus established a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with sequestration of osteo-inductive factors. STATEMENT OF SIGNIFICANCE: As extracellular matrix (ECM) assembly is significantly retarded in vitro, the imbalance in ECM-to-cell ratio hampers current in vitro bone tissue engineering approaches in their ability to faithfully resemble their in vivo counterpart. We addressed this limitation by leveraging a poly-electrolyte mediated co-assembly and amplified deposition of ECM during osteogenic differentiation of human mesenchymal stem cells (MSCs). The resulting pericelluar space in culture was enriched in organic and inorganic bone ECM components, as well as osteo-inductive factors, which promoted the differentiation of MSCs towards a more mature osteoblastic phenotype. These findings thus demonstrated a simple and reproducible approach to generate ECM-rich bone-like tissue in vitro with a closer recapitulation of the in vivo tissue niche.
Collapse
|
13
|
Poosti F, Soebadi MA, Crijns H, De Zutter A, Metzemaekers M, Berghmans N, Vanheule V, Albersen M, Opdenakker G, Van Damme J, Sprangers B, Proost P, Struyf S. Inhibition of renal fibrosis with a human CXCL9‐derived glycosaminoglycan‐binding peptide. Clin Transl Immunology 2022; 11:e1370. [PMID: 35140938 PMCID: PMC8810938 DOI: 10.1002/cti2.1370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 05/18/2021] [Accepted: 01/05/2022] [Indexed: 11/09/2022] Open
Abstract
Objectives Renal fibrosis accompanies all chronic kidney disorders, ultimately leading to end‐stage kidney disease and the need for dialysis or even renal replacement. As such, renal fibrosis poses a major threat to global health and the search for effective therapeutic strategies to prevent or treat fibrosis is highly needed. We evaluated the applicability of a highly positively charged human peptide derived from the COOH‐terminal domain of the chemokine CXCL9, namely CXCL9(74–103), for therapeutic intervention. Because of its high density of net positive charges at physiological pH, CXCL9(74–103) competes with full‐length chemokines for glycosaminoglycan (GAG) binding. Consequently, CXCL9(74–103) prevents recruitment of inflammatory leucocytes to sites of inflammation. Methods CXCL9(74–103) was chemically synthesised and tested in vitro for anti‐fibrotic properties on human fibroblasts and in vivo in the unilateral ureteral obstruction (UUO) mouse model. Results CXCL9(74–103) significantly reduced the mRNA and/or protein expression of connective tissue growth factor (CTGF), alpha‐smooth muscle actin (α‐SMA) and collagen III by transforming growth factor (TGF)‐β1‐stimulated human fibroblasts. In addition, administration of CXCL9(74–103) inhibited fibroblast migration towards platelet‐derived growth factor (PDGF), without affecting cell viability. In the UUO model, CXCL9(74–103) treatment significantly decreased renal α‐SMA, vimentin, and fibronectin mRNA and protein expression. Compared with vehicle, CXCL9(74–103) attenuated mRNA expression of TGF‐β1 and the inflammatory markers/mediators MMP‐9, F4/80, CCL2, IL‐6 and TNF‐α. Finally, CXCL9(74–103) treatment resulted in reduced influx of leucocytes in the UUO model and preserved tubular morphology. The anti‐fibrotic and anti‐inflammatory effects of CXCL9(74–103) were mediated by competition with chemokines and growth factors for GAG binding. Conclusions Our findings provide a scientific rationale for targeting GAG–protein interactions in renal fibrotic disease.
Collapse
Affiliation(s)
- Fariba Poosti
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Mohammad Ayodhia Soebadi
- Laboratory of Experimental Urology University Hospitals Leuven Leuven Belgium
- Department of Urology Faculty of Medicine Universitas Airlangga Surabaya Indonesia
| | - Helena Crijns
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Alexandra De Zutter
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Mieke Metzemaekers
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Vincent Vanheule
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Maarten Albersen
- Laboratory of Experimental Urology University Hospitals Leuven Leuven Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Ben Sprangers
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
- Department of Nephrology University Hospitals Leuven Leuven Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgium
| |
Collapse
|
14
|
Marcisz M, Zacharias M, Samsonov SA. Modeling Protein-Glycosaminoglycan Complexes: Does the Size Matter? J Chem Inf Model 2021; 61:4475-4485. [PMID: 34494837 PMCID: PMC8479808 DOI: 10.1021/acs.jcim.1c00664] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Docking glycosaminoglycans (GAGs) has been challenging because
of the complex nature of these long periodic linear and negatively
charged polysaccharides. Although standard docking tools like Autodock3
are successful when docking GAGs up to hexameric length, they experience
challenges to properly dock longer GAGs. Similar limitations concern
other docking approaches typically developed for docking ligands of
limited size to proteins. At the same time, most of more advanced
docking approaches are challenging for a user who is inexperienced
with complex in silico methodologies. In this work,
we evaluate the binding energies of complexes with different lengths
of GAGs using all-atom molecular dynamics simulations. Based on this
analysis, we propose a new docking protocol for long GAGs that consists
of conventional docking of short GAGs and further elongation with
the use of a coarse-grained representation of the GAG parts not being
in direct contact with its protein receptor. This method automated
by a simple script is straightforward to use within the Autodock3
framework but also useful in combination with other standard docking
tools. We believe that this method with some minor case-specific modifications
could also be used for docking other linear charged polymers.
Collapse
Affiliation(s)
- Mateusz Marcisz
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.,Intercollegiate Faculty of Biotechnology of UG and MUG, ul. Abrahama 58, 80-307 Gdańsk, Poland
| | - Martin Zacharias
- Center of Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748 Garching, Germany
| | - Sergey A Samsonov
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
15
|
Marcisz M, Huard B, Lipska AG, Samsonov SA. Further analyses of APRIL/APRIL-Receptor/Glycosaminoglycan interactions by biochemical assays linked to computational studies. Glycobiology 2021; 31:772-786. [PMID: 33682874 DOI: 10.1093/glycob/cwab016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
A proliferation-inducing ligand (APRIL) is a member of the tumor necrosis factor superfamily. APRIL is quite unique in this superfamily for at least for two reasons: i) it binds to glycosaminoglycans (GAGs) via its positively charged N-terminus; ii) one of its signaling receptor, the transmembrane activator CAML interactor (TACI) was also reported to bind GAGs. Here, as provided by biochemical evidences with the use of an APRIL deletion mutant linked to computational studies, APRIL-GAG interaction involved other regions than the APRIL N-terminus. Preferential interaction of APRIL with heparin followed by chondroitin sulfate E were confirmed by in silico analysis. Both computational and experimental approaches did not reveal heparan sulfate binding to TACI. Together, computational results corroborated experiments contributing with atomistic details to the knowledge on this biologically relevant trimolecular system. Additionally, a high-throughput rigorous analysis of the free energy calculations data was performed to critically evaluate the applied computational methodologies.
Collapse
Affiliation(s)
- Mateusz Marcisz
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.,Intercollegiate Faculty of Biotechnology of UG and MUG, ul. Abrahama 58, 80-307 Gdańsk, Poland
| | - Bertrand Huard
- TIMC-IMAG, university Grenoble-Alpes, CNRS UMR 5525, La Tronche, France
| | - Agnieszka G Lipska
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Sergey A Samsonov
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
16
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
17
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|
18
|
Abstract
Endoglin, known to be expressed in proliferating vessels, is of worth when evaluating microvessel density as a prognostic factor in many types of malignancies, including some subtypes of leukemia cells. In childhood acute lymphoblastic leukemia, endoglin is associated with adverse outcome. In bone marrow, endoglin identifies the repopulating hematopoietic stem cells. Mast cells are a component of normal tissue and play an important role in the regulation of several processes, including inflammation and neoplasia. The aim of this study was to evaluate the use of endoglin as a biological marker of mast cells compared with the gold standard stains. We studied 15 specimens of neurofibroma, 9 of mastocytosis, and 6 of fibrous scar tissue through immunohistochemistry (for endoglin and mast cell tryptase) and histochemical staining using toluidine blue. Quantitative analysis of the cells was performed by counting 5 hotspots. The validity of endoglin as a mast cell marker was assessed by intraclass correlation coefficient. The Kruskal-Wallis test was used to compare mast cell count for each marker. A strong endoglin expression was found in the cytoplasmic granules of mast cells within the 3 groups. Similar results were observed with mast cell tryptase as well as toluidine blue. The intraclass correlation coefficient revealed that endoglin is a highly reliable biomarker of mast cells when compared with mast cell tryptase and toluidine blue. In conclusion, endoglin may assist in the diagnosis and pathogenesis study of various processes associated with mast cells. An endoglin-neutralizing treatment for solid cancers and leukemia could also affect mastocytes and the immunologic system.
Collapse
|
19
|
Jensen MM, Karring H. The origins and developments of sulfation-prone tyrosine-rich and acidic N- and C-terminal extensions of class ll and lll small leucine-rich repeat proteins shed light on connective tissue evolution in vertebrates. BMC Evol Biol 2020; 20:73. [PMID: 32576155 PMCID: PMC7310474 DOI: 10.1186/s12862-020-01634-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Small leucine-rich repeat protein (SLRP) family members contain conserved leucine-rich repeat motifs flanked by highly variable N- and C-terminal regions. Most class II and III SLRPs have tyrosine-rich N-terminal regions and some of these are sulfated. However, the evolutionary origin and conservation of the tyrosine-rich and acidic terminal regions remain undetermined. In this study, we present the most comprehensive multiple sequence alignment (MSA) analyses of all eight class II and III SLRPs to date. Based on the level of conservation of tyrosine residues and adjacent sequences, we predict which tyrosine residues are most likely to be sulfated in the terminal regions of human class II and III SLRPs. Results Using this novel approach, we predict a total of 22 tyrosine sulfation sites in human SLRPs, of which only 8 sites had been experimentally identified in mammals. Our analyses suggest that sulfation-prone, tyrosine-rich and acidic terminal regions of the class II and III SLRPs emerged via convergent evolution at different stages of vertebrate evolution, coinciding with significant evolutionary events including the development of endochondral bones and articular cartilage, the aquatic to terrestrial transition, and the formation of an amnion. Conclusions Our study suggests that selective pressures due to changes in life conditions led to the formation of sulfotyrosine-rich and acidic terminal regions. We believe the independent emergence and evolution of sulfotyrosine-rich and acidic N- and C-terminal regions have provided each class II and III SLRP member with novel vital functions required to develop new specialized extracellular matrices and tissues in vertebrate species.
Collapse
Affiliation(s)
- Morten M Jensen
- Department of Chemical Engineering, Biotechnology and Environmental Technology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Henrik Karring
- Department of Chemical Engineering, Biotechnology and Environmental Technology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark.
| |
Collapse
|
20
|
Gulberti S, Mao X, Bui C, Fournel-Gigleux S. The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs). Semin Cancer Biol 2020; 62:68-85. [DOI: 10.1016/j.semcancer.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
|
21
|
Ishihara M, Nakamura S, Sato Y, Takayama T, Fukuda K, Fujita M, Murakami K, Yokoe H. Heparinoid Complex-Based Heparin-Binding Cytokines and Cell Delivery Carriers. Molecules 2019; 24:molecules24244630. [PMID: 31861225 PMCID: PMC6943580 DOI: 10.3390/molecules24244630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Heparinoid is the generic term that is used for heparin, heparan sulfate (HS), and heparin-like molecules of animal or plant origin and synthetic derivatives of sulfated polysaccharides. Various biological activities of heparin/HS are attributed to their specific interaction and regulation with various heparin-binding cytokines, antithrombin (AT), and extracellular matrix (ECM) biomolecules. Specific domains with distinct saccharide sequences in heparin/HS mediate these interactions are mediated and require different highly sulfated saccharide sequences with different combinations of sulfated groups. Multivalent and cluster effects of the specific sulfated sequences in heparinoids are also important factors that control their interactions and biological activities. This review provides an overview of heparinoid-based biomaterials that offer novel means of engineering of various heparin-binding cytokine-delivery systems for biomedical applications and it focuses on our original studies on non-anticoagulant heparin-carrying polystyrene (NAC-HCPS) and polyelectrolyte complex-nano/microparticles (N/MPs), in addition to heparin-coating devices.
Collapse
Affiliation(s)
- Masayuki Ishihara
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
- Correspondence: ; Tel.: +81-429-95-1211 (ext. 2610)
| | - Shingo Nakamura
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Yoko Sato
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Tomohiro Takayama
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| | - Koichi Fukuda
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Masanori Fujita
- Division of Environmental Medicine, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-1324, Japan;
| | - Kaoru Murakami
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| | - Hidetaka Yokoe
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| |
Collapse
|
22
|
Levinson C, Lee M, Applegate LA, Zenobi-Wong M. An injectable heparin-conjugated hyaluronan scaffold for local delivery of transforming growth factor β1 promotes successful chondrogenesis. Acta Biomater 2019; 99:168-180. [PMID: 31536840 DOI: 10.1016/j.actbio.2019.09.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023]
Abstract
Cartilage lacks basic repair mechanisms and thus surgical interventions are necessary to treat lesions. Minimally-invasive arthroscopic procedures require the development of injectable biomaterials to support chondrogenesis of implanted cells. However, most cartilage tissue engineering approaches rely on pre-culture of scaffolds in media containing growth factors (GFs) such as transforming growth factor (TGF)-β1, which are crucial for cartilage formation and homeostasis. GFs media-supplementation is incompatible with injectable approaches and has led to a knowledge gap about optimal dose of GFs and release profiles needed to achieve chondrogenesis. This study aims to determine the optimal loading and release kinetics of TGF-β1 bound to an engineered GAG hydrogel to promote optimal cartilaginous matrix production in absence of TGF-β1 media-supplementation. We show that heparin, a GAG known to bind a wide range of GFs, covalently conjugated to a hyaluronan hydrogel, leads to a sustained release of TGF-β1. Using this heparin-conjugated hyaluronan hydrogel, 0.25 to 50 ng TGF-β1 per scaffold was loaded and cell viability, proliferation and cartilaginous matrix deposition of the encapsulated chondroprogenitor cells were measured. Excellent chondrogenesis was found when 5 ng TGF-β1 per scaffold and higher were used. We also demonstrate the necessity of a sustained release of TGF-β1, as no matrix deposition is observed upon a burst release. In conclusion, our biomaterial loaded with an optimal initial dose of 5 ng/scaffold TGF-β1 is a promising injectable material for cartilage repair, with potentially increased safety due to the low, locally administered GF dose. STATEMENT OF SIGNIFICANCE: Cartilage cell-based products are dependent on exogenous growth factor supplementation in order for proper tissue maturation. However, for a one-step repair of defects without need for expensive tissue maturation, an injectable, growth factor loaded formulation is required. Here we show development of an injectable hyaluronan hydrogel, which achieves a sustained release of TGF-β1 due to covalent conjugation of heparin. These grafts matured into cartilaginous tissue in the absence of growth factor supplementation. Additionally, this system allowed us to screen TGF-β1 concentrations to determine the mimimum amount of growth factor required for chondrogenesis. This study represents a critical step towards development of a minimally-invasive, arthroscopic treatment for cartilage lesions.
Collapse
|
23
|
Comparison of the Interactions of Different Growth Factors and Glycosaminoglycans. Molecules 2019; 24:molecules24183360. [PMID: 31527407 PMCID: PMC6767211 DOI: 10.3390/molecules24183360] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 01/24/2023] Open
Abstract
Most growth factors are naturally occurring proteins, which are signaling molecules implicated in cellular multiple functions such as proliferation, migration and differentiation under patho/physiological conditions by interacting with cell surface receptors and other ligands in the extracellular microenvironment. Many of the growth factors are heparin-binding proteins (HBPs) that have a high affinity for cell surface heparan sulfate proteoglycans (HSPG). In the present study, we report the binding kinetics and affinity of heparin interacting with different growth factors, including fibroblast growth factor (FGF) 2,7,10, hepatocyte growth factor (HGF) and transforming growth factor (TGF β-1), using a heparin chip. Surface plasmon resonance studies revealed that all the tested growth factors bind to heparin with high affinity (with KD ranging from ~0.1 to 59 nM) and all the interactions are oligosaccharide size dependent except those involving TGF β-1. These heparin-binding growth factors also interact with other glycosaminoglycans (GAGs), as well as various chemically modified heparins. Other GAGs, including heparan sulfate, chondroitin sulfates A, B, C, D, E and keratan sulfate, showed different inhibition activities for the growth factor-heparin interactions. FGF2, FGF7, FGF10 and HGF bind heparin but the 2-O-sulfo and 6-O-sulfo groups on heparin have less impact on these interactions than do the N-sulfo groups. All the three sulfo groups (N-, 2-O and 6-O) on heparin are important for TGFβ-1-heparin interaction.
Collapse
|
24
|
Li X, Wang J, Liu J, Tang J, Wang J, Guo J, Wang Y, Huang L, Aleem AR, Kipper MJ, Belfiore LA. Strong luminescence and sharp heavy metal ion sensitivity of water-soluble hybrid polysaccharide nanoparticles with Eu3+ and Tb3+ inclusions. APPLIED NANOSCIENCE 2019. [DOI: 10.1007/s13204-019-01048-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
25
|
Cavalli E, Levinson C, Hertl M, Broguiere N, Brück O, Mustjoki S, Gerstenberg A, Weber D, Salzmann G, Steinwachs M, Barreto G, Zenobi-Wong M. Characterization of polydactyly chondrocytes and their use in cartilage engineering. Sci Rep 2019; 9:4275. [PMID: 30862915 PMCID: PMC6414529 DOI: 10.1038/s41598-019-40575-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/19/2019] [Indexed: 01/22/2023] Open
Abstract
Treating cartilage injuries and degenerations represents an open surgical challenge. The recent advances in cell therapies have raised the need for a potent off-the-shelf cell source. Intra-articular injections of TGF-β transduced polydactyly chondrocytes have been proposed as a chronic osteoarthritis treatment but despite promising results, the use of gene therapy still raises safety concerns. In this study, we characterized infant, polydactyly chondrocytes during in vitro expansion and chondrogenic re-differentiation. Polydactyly chondrocytes have a steady proliferative rate and re-differentiate in 3D pellet culture after up to five passages. Additionally, we demonstrated that polydactyly chondrocytes produce cartilage-like matrix in a hyaluronan-based hydrogel, namely transglutaminase cross-linked hyaluronic acid (HA-TG). We utilized the versatility of TG cross-linking to augment the hydrogels with heparin moieties. The heparin chains allowed us to load the scaffolds with TGF-β1, which induced cartilage-like matrix deposition both in vitro and in vivo in a subcutaneous mouse model. This strategy introduces the possibility to use infant, polydactyly chondrocytes for the clinical treatment of joint diseases.
Collapse
Affiliation(s)
- Emma Cavalli
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland
| | - Clara Levinson
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland
| | - Matthias Hertl
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland
| | - Nicolas Broguiere
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland
| | - Oscar Brück
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Anja Gerstenberg
- Division of Hand Surgery, University Children's Hospital, Steinwiesstrasse 75, 8032, Zürich, Switzerland
| | - Daniel Weber
- Division of Hand Surgery, University Children's Hospital, Steinwiesstrasse 75, 8032, Zürich, Switzerland
| | - Gian Salzmann
- Schulthess Clinic, Lengghalde 2, 8008, Zürich, Switzerland
| | - Matthias Steinwachs
- Sport Clinic Zürich Hirslanden, Witellikerstrasse 40, 8032, Zürich, Switzerland
| | - Gonçalo Barreto
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication, Institute for Biomechanics, Swiss Federal Institute of Technology Zürich (ETH Zürich), Otto-Stern-Weg 7, CH-8093, Zürich, Switzerland.
| |
Collapse
|
26
|
Zykwinska A, Marquis M, Sinquin C, Marchand L, Colliec-Jouault S, Cuenot S. Investigation of interactions between the marine GY785 exopolysaccharide and transforming growth factor-β1 by atomic force microscopy. Carbohydr Polym 2018; 202:56-63. [DOI: 10.1016/j.carbpol.2018.08.104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/13/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022]
|
27
|
Xie M, Li JP. Heparan sulfate proteoglycan - A common receptor for diverse cytokines. Cell Signal 2018; 54:115-121. [PMID: 30500378 DOI: 10.1016/j.cellsig.2018.11.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/04/2023]
Abstract
Heparan sulfate proteoglycans (HSPG) are macromolecular glyco-conjugates expressed ubiquitously on the cell surface and in the extracellular matrix where they interact with a wide range of ligands to regulate many aspects of cellular function. The capacity of the side glycosaminoglycan chain heparan sulfate (HS) being able to interact with diverse protein ligands relies on its complex structure that is generated by a controlled biosynthesis process, involving the actions of glycosyl-transferases, sulfotransferases and the glucuronyl C5-epimerase. It is believed that activities of the modification enzymes control the HS structures that are designed to serve the biological functions in a given cell or biological status. In this review, we briefly discuss recent understandings on the roles of HSPG in cytokine stimulated cellular signaling, focusing on FGF, TGF-β, Wnt, Hh, HGF and VEGF.
Collapse
Affiliation(s)
- Meng Xie
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
28
|
Kunisch E, Knauf AK, Hesse E, Freudenberg U, Werner C, Bothe F, Diederichs S, Richter W. StarPEG/heparin-hydrogel based in vivo engineering of stable bizonal cartilage with a calcified bottom layer. Biofabrication 2018; 11:015001. [PMID: 30376451 DOI: 10.1088/1758-5090/aae75a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Repaired cartilage tissue lacks the typical zonal structure of healthy native cartilage needed for appropriate function. Current grafts for treatment of full thickness cartilage defects focus primarily on a nonzonal design and this may be a reason why inferior nonzonal regeneration tissue developed in vivo. No biomaterial-based solutions have been developed so far to induce a proper zonal architecture into a non-mineralized and a calcified cartilage layer. The objective was to grow bizonal cartilage with a calcified cartilage bottom zone wherein main tissue development is occurring in vivo. We hypothesized that starPEG/heparin-hydrogel owing to the glycosaminoglycan heparin contained as a building-block would prevent mineralization of the upper cartilage zone and be beneficial in inhibiting long-term progression of calcified cartilage into bone. MSCs were pre-cultured as self-assembling non-mineralized cell discs before a chondrocyte-seeded fibrin- or starPEG/heparin-hydrogel layer was cast on top directly before ectopic implantation. Bizonal cartilage with a calcified bottom-layer developed in vivo showing stronger mineralization compared to in vitro samples, but the hydrogel strongly determined outcome. Zonal fibrin-constructs lost volume and allowed non-organized expansion of collagen type X, ALP-activity and mineralization from the bottom-layer into upper regions, whereas zonal starPEG/heparin-constructs were of stable architecture. While non-zonal MSCs-derived discs formed bone over 12 weeks, the starPEG/heparin-chondrocyte layer prevented further progression of calcified cartilage into bone tissue. Conclusively, starPEG/heparin-hydrogel-controlled and cell-type mediated spatiotemporal regulation allowed in vivo growth of bizonal cartilage with a stable calcified cartilage layer. Altogether our work is an important milestone encouraging direct in vivo growth of organized cartilage after biofabrication.
Collapse
Affiliation(s)
- Elke Kunisch
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Frevert CW, Felgenhauer J, Wygrecka M, Nastase MV, Schaefer L. Danger-Associated Molecular Patterns Derived From the Extracellular Matrix Provide Temporal Control of Innate Immunity. J Histochem Cytochem 2018; 66:213-227. [PMID: 29290139 DOI: 10.1369/0022155417740880] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It is evident that components of the extracellular matrix (ECM) act as danger-associated molecular patterns (DAMPs) through direct interactions with pattern recognition receptors (PRRs) including Toll-like receptors (TLRs) and inflammasomes. Through these interactions, ECM-derived DAMPs autonomously trigger sterile inflammation or prolong pathogen-induced responses through the production of proinflammatory mediators and the recruitment of leukocytes to sites of injury and infection. Recent research, however, suggests that ECM-derived DAMPs are additionally involved in the resolution and fine-tuning of inflammation by orchestrating the production of anti-inflammatory mediators that are required for the resolution of tissue inflammation and the transition to acquired immunity. Thus, in this review, we discuss the current knowledge of the interplay between ECM-derived DAMPs and the innate immune signaling pathways that are activated to provide temporal control of innate immunity.
Collapse
Affiliation(s)
- Charles W Frevert
- Center for Lung Biology, University of Washington, Seattle, Washington
| | | | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Madalina V Nastase
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany.,National Institute for Chemical-Pharmaceutical Research and Development, Bucharest, Romania
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany
| |
Collapse
|
31
|
Ansorge M, Sapudom J, Chkolnikov M, Wilde M, Anderegg U, Möller S, Schnabelrauch M, Pompe T. Mimicking Paracrine TGFβ1 Signals during Myofibroblast Differentiation in 3D Collagen Networks. Sci Rep 2017; 7:5664. [PMID: 28720779 PMCID: PMC5515936 DOI: 10.1038/s41598-017-05912-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/05/2017] [Indexed: 01/06/2023] Open
Abstract
TGFβ1 is a key regulator for induction of tissue remodeling after dermal wounding. We present a model of paracrine delivery of TGFβ1 for differentiation of dermal fibroblasts based on a fibrillar 3D collagen matrix and embedded TGFβ1 releasing microparticles. We found differentiation into myofibroblasts was achieved in a TGFβ1 dependent manner at much lower doses than systemic delivery. This effect is accounted to the slow and sustained TGFβ1 release mimicking paracrine cell signals.
Collapse
Affiliation(s)
- Michael Ansorge
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Jiranuwat Sapudom
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Marina Chkolnikov
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Martin Wilde
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Ulf Anderegg
- Universitätsklinikum Leipzig, Department of Dermatology, Venereology and Allergology, 04103, Leipzig, Germany
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e. V., Prüssingstr. 27B, 07745, Jena, Germany
| | | | - Tilo Pompe
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany.
| |
Collapse
|
32
|
Rider CC, Mulloy B. Heparin, Heparan Sulphate and the TGF-β Cytokine Superfamily. Molecules 2017; 22:molecules22050713. [PMID: 28468283 PMCID: PMC6154108 DOI: 10.3390/molecules22050713] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Of the circa 40 cytokines of the TGF-β superfamily, around a third are currently known to bind to heparin and heparan sulphate. This includes TGF-β1, TGF-β2, certain bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), as well as GDNF and two of its close homologues. Experimental studies of their heparin/HS binding sites reveal a diversity of locations around the shared cystine-knot protein fold. The activities of the TGF-β cytokines in controlling proliferation, differentiation and survival in a range of cell types are in part regulated by a number of specific, secreted BMP antagonist proteins. These vary in structure but seven belong to the CAN or DAN family, which shares the TGF-β type cystine-knot domain. Other antagonists are more distant members of the TGF-β superfamily. It is emerging that the majority, but not all, of the antagonists are also heparin binding proteins. Any future exploitation of the TGF-β cytokines in the therapy of chronic diseases will need to fully consider their interactions with glycosaminoglycans and the implications of this in terms of their bioavailability and biological activity.
Collapse
Affiliation(s)
- Chris C Rider
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK.
| | - Barbara Mulloy
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK.
| |
Collapse
|
33
|
Koehler L, Samsonov S, Rother S, Vogel S, Köhling S, Moeller S, Schnabelrauch M, Rademann J, Hempel U, Pisabarro MT, Scharnweber D, Hintze V. Sulfated Hyaluronan Derivatives Modulate TGF-β1:Receptor Complex Formation: Possible Consequences for TGF-β1 Signaling. Sci Rep 2017; 7:1210. [PMID: 28446792 PMCID: PMC5430790 DOI: 10.1038/s41598-017-01264-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/24/2017] [Indexed: 12/15/2022] Open
Abstract
Glycosaminoglycans are known to bind biological mediators thereby modulating their biological activity. Sulfated hyaluronans (sHA) were reported to strongly interact with transforming growth factor (TGF)-β1 leading to impaired bioactivity in fibroblasts. The underlying mechanism is not fully elucidated yet. Examining the interaction of all components of the TGF-β1:receptor complex with sHA by surface plasmon resonance, we could show that highly sulfated HA (sHA3) blocks binding of TGF-β1 to its TGF-β receptor-I (TβR-I) and -II (TβR-II). However, sequential addition of sHA3 to the TβR-II/TGF-β1 complex led to a significantly stronger recruitment of TβR-I compared to a complex lacking sHA3, indicating that the order of binding events is very important. Molecular modeling suggested a possible molecular mechanism in which sHA3 could potentially favor the association of TβR-I when added sequentially. For the first time bioactivity of TGF-β1 in conjunction with sHA was investigated at the receptor level. TβR-I and, furthermore, Smad2 phosphorylation were decreased in the presence of sHA3 indicating the formation of an inactive signaling complex. The results contribute to an improved understanding of the interference of sHA3 with TGF-β1:receptor complex formation and will help to further improve the design of functional biomaterials that interfere with TGF-β1-driven skin fibrosis.
Collapse
Affiliation(s)
- Linda Koehler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Sergey Samsonov
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Sarah Vogel
- Medical Department, Institute of Physiological Chemistry, TU Dresden, Fiedlerstraße 42, 01307, Dresden, Germany
| | - Sebastian Köhling
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195, Berlin, Germany
| | - Stephanie Moeller
- Biomaterials Department, INNOVENT e.V., Prüssingstraße 27 B, 07745, Jena, Germany
| | | | - Jörg Rademann
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195, Berlin, Germany
| | - Ute Hempel
- Medical Department, Institute of Physiological Chemistry, TU Dresden, Fiedlerstraße 42, 01307, Dresden, Germany
| | - M Teresa Pisabarro
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, 01307, Dresden, Germany
| | - Dieter Scharnweber
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, 01069, Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Straße 27, 01069, Dresden, Germany.
| |
Collapse
|
34
|
Romero R, Travers JK, Asbury E, Pennybaker A, Chubb L, Rose R, Ehrhart NP, Kipper MJ. Combined delivery of FGF-2, TGF-β1, and adipose-derived stem cells from an engineered periosteum to a critical-sized mouse femur defect. J Biomed Mater Res A 2016; 105:900-911. [PMID: 27874253 DOI: 10.1002/jbm.a.35965] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/15/2016] [Accepted: 11/17/2016] [Indexed: 12/18/2022]
Abstract
Critical-sized long bone defects suffer from complications including impaired healing and non-union due to substandard healing and integration of devitalized bone allograft. Removal of the periosteum contributes to the limited healing of bone allografts. Restoring a periosteum on bone allografts may provide improved allograft healing and integration. This article reports a polysaccharide-based tissue engineered periosteum that delivers basic fibroblast growth factor (FGF-2), transforming growth factor-β1 (TGF-β1), and adipose-derived mesenchymal stem cells (ASCs) to a critical-sized mouse femur defect. The tissue engineered periosteum was evaluated for improving bone allograft healing and incorporation by locally delivering FGF-2, TGF-β1, and supporting ASCs transplantation. ASCs were successfully delivered and longitudinally tracked at the defect site for at least 7 days post operation with delivered FGF-2 and TGF-β1 showing a mitogenic effect on the ASCs. At 6 weeks post implantation, data showed a non-significant increase in normalized bone callus volume. However, union ratio analysis showed a significant inhibition in allograft incorporation, confirmed by histological analysis, due to loosening of the nanofiber coating from the allograft surface. Ultimately, this investigation shows our tissue engineered periosteum can deliver FGF-2, TGF-β1, and ASCs to a mouse critical-sized femur defect and further optimization may yield improved bone allograft healing. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 900-911, 2017.
Collapse
Affiliation(s)
- Raimundo Romero
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523
| | - John K Travers
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523
| | - Emilie Asbury
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523
| | - Attie Pennybaker
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523
| | - Laura Chubb
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, 80523
| | - Ruth Rose
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, 80523
| | - Nicole P Ehrhart
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523.,Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, 80523
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, 80523.,Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, 80523
| |
Collapse
|
35
|
TGF-β1 autocrine signalling and enamel matrix components. Sci Rep 2016; 6:33644. [PMID: 27633089 PMCID: PMC5025654 DOI: 10.1038/srep33644] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/31/2016] [Indexed: 01/03/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is present in porcine enamel extracts and is critical for proper mineralization of tooth enamel. Here, we show that the mRNA of latent TGF-β1 is expressed throughout amelogenesis. Latent TGF-β1 is activated by matrix metalloproteinase 20 (MMP20), coinciding with amelogenin processing by the same proteinase. Activated TGF-β1 binds to the major amelogenin cleavage products, particularly the neutral-soluble P103 amelogenin, to maintain its activity. The P103 amelogenin-TGF-β1 complex binds to TGFBR1 to induce TGF-β1 signalling. The P103 amelogenin-TGF-β1 complex is slowly cleaved by kallikrein 4 (KLK4), which is secreted into the transition- and maturation-stage enamel matrix, thereby reducing TGF-β1 activity. To exert the multiple biological functions of TGF-β1 for amelogenesis, we propose that TGF-β1 is activated or inactivated by MMP20 or KLK4 and that the amelogenin cleavage product is necessary for the in-solution mobility of TGF-β1, which is necessary for binding to its receptor on ameloblasts and retention of its activity.
Collapse
|
36
|
Choi SH, Lee JY, Suh JS, Park YS, Chung CP, Park YJ. Dual-function synthetic peptide derived from BMP4 for highly efficient tumor targeting and antiangiogenesis. Int J Nanomedicine 2016; 11:4643-4656. [PMID: 27695323 PMCID: PMC5028098 DOI: 10.2147/ijn.s115044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Angiogenesis plays a critical role in the growth and metastasis of cancer, and growth factors released from cancer promote blood-vessel formation in the tumor microenvironment. The angiogenesis is accelerated via interactions of growth factors with the high-affinity receptors on cancer cells. In particular, heparan sulfate proteoglycans (HSPGs) on the surface of cancer cells have been shown to be important in many aspects of determining a tumor's phenotype and development. Specifically, the regulation of the interactions between HSPGs and growth factors results in changes in tumor progression. A peptide with heparin-binding (HBP) activity has been developed and synthesized to inhibit tumor growth via the prevention of angiogenesis. We hypothesized that HBP could inhibit the interaction of growth factors and HSPGs on the surface of cancer cells, decrease paracrine signaling in endothelial cells (ECs), and finally decrease angiogenesis in the tumor microenvironment. In this study, we found that HBP had antiangiogenic effects in vitro and in vivo. The conditioned media obtained from a breast cancer cell line treated with HBP were used to culture human umbilical vein ECs (HUVECs) to evaluate the antiangiogenic effect of HBP on ECs. HBP effectively inhibited the migration, invasion, and tube formation of HUVECs in vitro. In addition, the expressions of angiogenesis-mediating factors, including ERK, FAK, and Akt, were considerably decreased. HBP also decreased the levels of invasive factors, including MMP2 and MMP9, secreted by the HUVECs. We demonstrated significant suppression of tumor growth in a breast cancer xenograft model and enhanced distribution of HBP at the site of tumors. Taken together, our results show that HBP has antiangiogenic effects on ECs, and suggest that it may serve as a potential antitumor agent through control of the tumor microenvironment.
Collapse
Affiliation(s)
- Suk Hyun Choi
- Department of Dental Regenerative Biotechnology, Dental Research Institute
| | - Jue Yeon Lee
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), School of Dentistry, Seoul National University, Seoul
| | - Jin Sook Suh
- Department of Dental Regenerative Biotechnology, Dental Research Institute
| | - Yoon Shin Park
- Department in Microbiology, School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, South Korea
| | - Chong Pyoung Chung
- Central Research Institute, Nano Intelligent Biomedical Engineering Corporation (NIBEC), School of Dentistry, Seoul National University, Seoul
| | - Yoon Jeong Park
- Department of Dental Regenerative Biotechnology, Dental Research Institute
| |
Collapse
|
37
|
Habuchi H, Ushida T, Habuchi O. Mice deficient in N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase exhibit enhanced liver fibrosis and delayed recovery from fibrosis in carbon tetrachloride-treated mice. Heliyon 2016; 2:e00138. [PMID: 27547834 PMCID: PMC4983273 DOI: 10.1016/j.heliyon.2016.e00138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/01/2016] [Indexed: 01/18/2023] Open
Abstract
Background Chondroitin/dermatan sulfate (CS/DS) rich in N-acetylgalactosamine 4,6-bissulfate (GalNAc(4,6SO4)) residues is present as decorin and/or biglycan in mouse liver, and GalNAc(4,6SO4) residues disappeared completely in N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase (GalNAc4S-6ST) knockout (KO) mice. The aim of this study was to investigate whether CS/DS rich in GalNAc(4,6SO4) residues participate in the progression or resolution of liver fibrosis. Methods Wild type (WT) and GalNAc4S-6ST KO mice were treated with CCl4 for 5 weeks. After discontinuation of CCl4 administration, histochemical and biochemical changes and expression of genes related to matrix components were compared between WT and GalNAc4S-6ST KO mice. Results and conclusion On 2 days after cessation of CCl4 administration, higher fibrosis was observed in KO mice than in WT mice by Sirius Red staining. Serum alanine aminotransferase activity was higher in KO mice than in WT mice. Hydroxyproline contents and Sirius Red staining showed that repair of liver fibrosis in the recovery stages appeared to be delayed in KO mice. Expression of mRNA of matrix metalloproteinase (MMP)-2, MMP-13 and versican peaked at 2 days after cessation of CCl4 administration and was higher in KO mice than in WT mice. Expression of MMP-9 in the recovery stage was lower in KO mice than in WT mice. Our findings demonstrate that defect in GalNAc4S-6ST, which resulted in disappearance of CS/DS containing GalNAc(4,6SO4), appear to contribute to progression of liver fibrosis, delayed recovery from fibrosis, and various changes in the expression of proteoglycans and MMPs in carbon tetrachloride–treated mice.
Collapse
Affiliation(s)
- Hiroko Habuchi
- Advanced Medical Research Center, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Multidisciplinary Pain Center, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Takahiro Ushida
- Multidisciplinary Pain Center, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| | - Osami Habuchi
- Advanced Medical Research Center, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Multidisciplinary Pain Center, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
38
|
Insights into the molecular roles of heparan sulfate proteoglycans (HSPGs—syndecans) in autocrine and paracrine growth factor signaling in the pathogenesis of Hodgkin’s lymphoma. Tumour Biol 2016; 37:11573-11588. [DOI: 10.1007/s13277-016-5118-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
|
39
|
Xiong GM, Yap YZ, Choong C. Single-step synthesis of heparin-doped polypyrrole nanoparticles for delivery of angiogenic factor. Nanomedicine (Lond) 2016; 11:749-65. [DOI: 10.2217/nnm.16.13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To perform one-pot synthesis of heparin-immobilized polypyrrole (PPy) nanoparticles and evaluate the use of these nanoparticles for the delivery of VEGF. Materials & methods: Heparin-stabilized synthesis of PPy nanoparticles was performed via oxidative polymerization. VEGF-bound PPy-heparin nanoparticles were delivered to endothelial cells and bioactivity of VEGF was assessed by Matrigel tube formation. Results: Size-controllable synthesis of heparin-doped PPy nanoparticles was achieved, and heparin promoted the conjugation of VEGF. Angiogenic activity of the VEGF-conjugated PPy nanoparticles was verified. Conclusion: Heparin-doped PPy nanoparticles can be synthesized using one-pot reaction and provide a delivery platform by which VEGF can be conjugated onto.
Collapse
Affiliation(s)
- Gordon M Xiong
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Yi Zhen Yap
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Cleo Choong
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
- KK Research Centre, KK Women's & Children Hospital, 100 Bukit Timah Road, 229899 Singapore
| |
Collapse
|
40
|
Place LW, Sekyi M, Taussig J, Kipper MJ. Two-Phase Electrospinning to Incorporate Polyelectrolyte Complexes and Growth Factors into Electrospun Chitosan Nanofibers. Macromol Biosci 2015; 16:371-80. [DOI: 10.1002/mabi.201500288] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/20/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Laura W. Place
- School of Biomedical Engineering; 1370 Campus Delivery Fort Collins CO 80523-1370 USA
| | - Maria Sekyi
- Department of Chemical and Biological Engineering; 1370 Campus Delivery Fort Collins CO 80523-1370 USA
| | - Julia Taussig
- Department of Chemical and Biological Engineering; 1370 Campus Delivery Fort Collins CO 80523-1370 USA
| | - Matt J. Kipper
- School of Biomedical Engineering; 1370 Campus Delivery Fort Collins CO 80523-1370 USA
| |
Collapse
|
41
|
Watarai A, Schirmer L, Thönes S, Freudenberg U, Werner C, Simon JC, Anderegg U. TGFβ functionalized starPEG-heparin hydrogels modulate human dermal fibroblast growth and differentiation. Acta Biomater 2015. [PMID: 26219861 DOI: 10.1016/j.actbio.2015.07.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Hydrogels are promising biomaterials that can adapt easily to complex tissue entities. Furthermore, chemical modifications enable these hydrogels to become an instructive biomaterial to a variety of cell types. Human dermal fibroblasts play a pivotal role during wound healing, especially for the synthesis of novel dermal tissue replacing the primary fibrin clot. Thus, the control of growth and differentiation of dermal fibroblasts is important to modulate wound healing. In here, we utilized a versatile starPEG-heparin hydrogel platform that can be independently adjusted with respect to mechanical and biochemical properties for cultivating human dermal fibroblasts. Cell-based remodeling of the artificial matrix was ensured by using matrix metalloprotease (MMP) cleavable crosslinker peptides. Attachment and proliferation of fibroblasts on starPEG-heparin hydrogels of differing stiffness, density of pro-adhesive RGD peptides and MMP cleavable peptide linkers were tested. Binding and release of human TGFβ1 as well as biological effect of the pre-adsorbed growth factor on fibroblast gene expression and myofibroblast differentiation were investigated. Hydrogels containing RGD peptides supported fibroblast attachment, spreading, proliferation matrix deposition and remodeling compared to hydrogels without any modifications. Reversibly conjugated TGFβ1 was demonstrated to be constantly released from starPEG-heparin hydrogels for several days and capable of inducing myofibroblast differentiation of fibroblasts as determined by induction of collagen type I, ED-A-Fibronectin expression and incorporation of alpha smooth muscle actin and palladin into F-actin stress fibers. Taken together, customized starPEG-heparin hydrogels could be of value to promote dermal wound healing by stimulating growth and differentiation of human dermal fibroblasts. STATEMENT OF SIGNIFICANCE The increasing number of people of advanced age within the population results in an increasing demand for the treatment of non-healing wounds. Hydrogels are promising biomaterials for the temporary closure of large tissue defects: They can adapt to complex tissue geometry and can be engineered for specific tissue needs. We used a starPEG-heparin hydrogel platform that can be independently adjusted to mechanical and biochemical characteristics. We investigated how these hydrogels can support attachment, proliferation and differentiation of dermal fibroblasts. After introducing adhesive peptides these hydrogels support cell attachment and proliferation. Moreover, TGFβ - an essential growth and differentiation factor for fibroblasts - can be immobilized reversibly and functionally on these hydrogels. Thus, starPEG-heparin hydrogels could be developed to bioactive temporary wound dressings.
Collapse
|
42
|
Lee J, Wee S, Gunaratne J, Chua RJE, Smith RAA, Ling L, Fernig DG, Swaminathan K, Nurcombe V, Cool SM. Structural determinants of heparin-transforming growth factor-β1 interactions and their effects on signaling. Glycobiology 2015; 25:1491-504. [PMID: 26306634 DOI: 10.1093/glycob/cwv064] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/08/2015] [Indexed: 12/28/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1, Uniprot: P01137) is a heparin-binding protein that has been implicated in a number of physiological processes, including the initiation of chondrogenesis by human mesenchymal stem cells (hMSCs). Here, we identify the molecular features in the protein and in heparin required for binding and their effects on the potentiation of TGF-β1's activity on hMSCs. Using a proteomics "Protect and Label" approach, lysines K291, K304, K309, K315, K338, K373, K375 and K388 were identified as being directly involved in binding heparin (Data are available via ProteomeXchange with identifier PXD002772). Competition assays in an optical biosensor demonstrated that TGF-β1 does require N- and 6-O-sulfate groups for binding but that 2-O-sulfate groups are unlikely to underpin the interaction. Heparin-derived oligosaccharides as short as degree of polymerization (dp) 4 have a weak ability to compete for TGF-β1 binding to heparin, which increases with the length of the oligosaccharide to reach a maximum between dp18 and dp24. In cell-based assays, heparin, 2-O-, 6-O- and N-desulfated re-N-acetylated heparin and oligosaccharides 14-24 saccharides (dp14-24) in length all increased the phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) after 6 h of stimulation with TGF-β1. The results provide the structural basis for a model of heparin/heparan sulfate binding to TGF-β1 and demonstrate that the features in the polysaccharide required for binding are not identical to those required for sustaining the signaling by TGF-β1 in hMSCs.
Collapse
Affiliation(s)
- Jonathan Lee
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore 117456 Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648
| | - Sheena Wee
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore 138673
| | - Jayantha Gunaratne
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore 138673
| | - R J E Chua
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648
| | - Raymond A A Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648
| | - Ling Ling
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648
| | - David G Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648 Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College, Singapore, Singapore 639798
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore, Singapore 138648 Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore 119228
| |
Collapse
|
43
|
Heparin/Heparan sulfate proteoglycans glycomic interactome in angiogenesis: biological implications and therapeutical use. Molecules 2015; 20:6342-88. [PMID: 25867824 PMCID: PMC6272510 DOI: 10.3390/molecules20046342] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis, the process of formation of new blood vessel from pre-existing ones, is involved in various intertwined pathological processes including virus infection, inflammation and oncogenesis, making it a promising target for the development of novel strategies for various interventions. To induce angiogenesis, angiogenic growth factors (AGFs) must interact with pro-angiogenic receptors to induce proliferation, protease production and migration of endothelial cells (ECs). The action of AGFs is counteracted by antiangiogenic modulators whose main mechanism of action is to bind (thus sequestering or masking) AGFs or their receptors. Many sugars, either free or associated to proteins, are involved in these interactions, thus exerting a tight regulation of the neovascularization process. Heparin and heparan sulfate proteoglycans undoubtedly play a pivotal role in this context since they bind to almost all the known AGFs, to several pro-angiogenic receptors and even to angiogenic inhibitors, originating an intricate network of interaction, the so called "angiogenesis glycomic interactome". The decoding of the angiogenesis glycomic interactome, achievable by a systematic study of the interactions occurring among angiogenic modulators and sugars, may help to design novel antiangiogenic therapies with implications in the cure of angiogenesis-dependent diseases.
Collapse
|
44
|
Zhang Z, Gupte MJ, Jin X, Ma PX. Injectable Peptide Decorated Functional Nanofibrous Hollow Microspheres to Direct Stem Cell Differentiation and Tissue Regeneration. ADVANCED FUNCTIONAL MATERIALS 2015; 25:350-360. [PMID: 26069467 PMCID: PMC4459759 DOI: 10.1002/adfm.201402618] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Injectable microspheres are attractive stem cell carriers for minimally invasive procedures. For tissue regeneration, the microspheres need to present the critical cues to properly direct stem cell differentiation. In natural extracellular matrix (ECM), growth factors (GFs) and collagen nanofibers provide critical chemical and physical cues. However, there have been no reported technologies that integrate synthetic nanofibers and GFs into injectable microspheres. In this study, we synthesized functional nanofibrous hollow microspheres (FNF-HMS), which can covalently bind GF-mimicking peptides. Two different GF-mimicking peptides, Transforming Growth Factor-β1 mimicking peptide Cytomodulin (CM) and Bone Morphogenetic Protein-2 mimicking peptide P24, were separately conjugated onto the FNF-HMS to induce distinct differentiation pathways of rabbit bone marrow-derived mesenchymal stem cells (BMSCs). While no existing biomaterials were reported to successfully deliver CM to induce chondrogenesis, the developed FNF-HMS were shown to effectively present CM to BMSCs and successfully induced their chondrogenesis for cartilage formation in both in vitro and in vivo studies. In addition, P24 was conjugated onto the newly developed FNF-HMS and was capable of retaining its bioactivity and inducing ectopic bone formation in nude mice. These results demonstrate that the novel FNF-HMS can effectively deliver GF-mimicking peptides to modulate stem cell fate and tissue regeneration.
Collapse
Affiliation(s)
- Zhanpeng Zhang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109-1078
| | - Melanie J. Gupte
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109-1078
| | - Xiaobing Jin
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109-1078
| | - Peter X. Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109-1078
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI, 48109-1078
- Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI, 48109-1078
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, 48109-1078
- Peter X. Ma, Ph.D., Professor, Department of Biologic and Materials Sciences, 1011 North University Ave., Room 2211, The University of Michigan, Ann Arbor, MI 48109-1078, Tel: (734) 764-2209, Fax: (734) 647-2110,
| |
Collapse
|
45
|
Kim J, Lin B, Kim S, Choi B, Evseenko D, Lee M. TGF-β1 conjugated chitosan collagen hydrogels induce chondrogenic differentiation of human synovium-derived stem cells. J Biol Eng 2015; 9:1. [PMID: 25745515 PMCID: PMC4350967 DOI: 10.1186/1754-1611-9-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/24/2014] [Indexed: 12/27/2022] Open
Abstract
Background Unlike bone tissue, articular cartilage regeneration has not been very successful and has many challenges ahead. We have previously developed injectable hydrogels using photopolymerizable chitosan (MeGC) that supported growth of chondrocytes. In this study, we demonstrate a biofunctional hydrogel for specific use in cartilage regeneration by conjugating transforming growth factor-β1 (TGF-β1), a well-documented chondrogenic factor, to MeGC hydrogels impregnating type II collagen (Col II), one of the major cartilaginous extracellular matrix (ECM) components. Results TGF-β1 was delivered from MeGC hydrogels in a controlled manner with reduced burst release by chemically conjugating the protein to MeGC. The hydrogel system did not compromise viability of encapsulated human synovium-derived mesenchymal stem cells (hSMSCs). Col II impregnation and TGF-β1 delivery significantly enhanced cellular aggregation and deposition of cartilaginous ECM by the encapsulated cells, compared with pure MeGC hydrogels. Conclusions This study demonstrates successful engineering of a biofunctional hydrogel with a specific microenvironment tailored to promote chondrogenesis. This hydrogel system can provide promising efficacious therapeutics in the treatment of cartilage defects. Electronic supplementary material The online version of this article (doi:10.1186/1754-1611-9-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, 339-701 South Korea
| | - Brian Lin
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Soyon Kim
- Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| | - Bogyu Choi
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095 USA
| | - Min Lee
- Division of Advanced Prosthodontics, University of California, Los Angeles, CA 90095 USA ; Department of Bioengineering, University of California, Los Angeles, CA 90095 USA
| |
Collapse
|
46
|
Billings PC, Pacifici M. Interactions of signaling proteins, growth factors and other proteins with heparan sulfate: mechanisms and mysteries. Connect Tissue Res 2015; 56:272-80. [PMID: 26076122 PMCID: PMC4785798 DOI: 10.3109/03008207.2015.1045066] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heparan sulfate (HS) is a component of cell surface and matrix-associated proteoglycans (HSPGs) that, collectively, play crucial roles in many physiologic processes including cell differentiation, organ morphogenesis and cancer. A key function of HS is to bind and interact with signaling proteins, growth factors, plasma proteins, immune-modulators and other factors. In doing so, the HS chains and HSPGs are able to regulate protein distribution, bio-availability and action on target cells and can also serve as cell surface co-receptors, facilitating ligand-receptor interactions. These proteins contain an HS/heparin-binding domain (HBD) that mediates their association and contacts with HS. HBDs are highly diverse in sequence and predicted structure, contain clusters of basic amino acids (Lys and Arg) and possess an overall net positive charge, most often within a consensus Cardin-Weintraub (CW) motif. Interestingly, other domains and residues are now known to influence protein-HS interactions, as well as interactions with other glycosaminoglycans, such as chondroitin sulfate. In this review, we provide a description and analysis of HBDs in proteins including amphiregulin, fibroblast growth factor family members, heparanase, sclerostin and hedgehog protein family members. We discuss HBD structural and functional features and important roles carried out by other protein domains, and also provide novel conformational insights into the diversity of CW motifs present in Sonic, Indian and Desert hedgehogs. Finally, we review progress in understanding the pathogenesis of a rare pediatric skeletal disorder, Hereditary Multiple Exostoses (HME), characterized by HS deficiency and cartilage tumor formation. Advances in understanding protein-HS interactions will have broad implications for basic biology and translational medicine as well as for the development of HS-based therapeutics.
Collapse
Affiliation(s)
- Paul C. Billings
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA 19104
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, 3615 Civic Center Boulevard, Philadelphia, PA 19104
| |
Collapse
|
47
|
Cuellar A, Reddi AH. Stimulation of Superficial Zone Protein/Lubricin/PRG4 by Transforming Growth Factor-β in Superficial Zone Articular Chondrocytes and Modulation by Glycosaminoglycans. Tissue Eng Part A 2014; 21:1973-81. [PMID: 25398329 DOI: 10.1089/ten.tea.2014.0381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Superficial zone protein (SZP), also known as lubricin and proteoglycan 4 (PRG4), plays an important role in the boundary lubrication of articular cartilage and is regulated by transforming growth factor (TGF)-β. Here, we evaluate the role of cell surface glycosaminoglycans (GAGs) during TGF-β1 stimulation of SZP/lubricin/PRG4 in superficial zone articular chondrocytes. We utilized primary monolayer superficial zone articular chondrocyte cultures and treated them with various concentrations of TGF-β1, in the presence or absence of heparan sulfate (HS), heparin, and chondroitin sulfate (CS). The cell surface GAGs were removed by pretreatment with either heparinase I or chondroitinase-ABC before TGF-β1 stimulation. Accumulation of SZP/lubricin/PRG4 in the culture medium in response to stimulation with TGF-β1 and various exogenous GAGs was demonstrated by immunoblotting and quantitated by enzyme-linked immunosorbent assay. We show that TGF-β1 and exogenous HS enhanced SZP accumulation of superficial zone chondrocytes in the presence of surface GAGs. At the dose of 1 ng/mL of TGF-β1, the presence of exogenous heparin inhibited SZP accumulation whereas the presence of exogenous CS stimulated SZP accumulation in the culture medium. Enzymatic depletion of GAGs on the surface of superficial zone chondrocytes enhanced the ability of TGF-β1 to stimulate SZP accumulation in the presence of both exogenous heparin and CS. Collectively, these results suggest that GAGs at the surface of superficial zone articular chondrocytes influence the response to TGF-β1 and exogenous GAGs to stimulate SZP accumulation. Cell surface GAGs modulate superficial zone chondrocytes' response to TGF-β1 and exogenous HS.
Collapse
Affiliation(s)
- Araceli Cuellar
- Department of Orthopedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Sacramento, California
| | - A Hari Reddi
- Department of Orthopedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Sacramento, California
| |
Collapse
|
48
|
Mercier F, Douet V. Bone morphogenetic protein-4 inhibits adult neurogenesis and is regulated by fractone-associated heparan sulfates in the subventricular zone. J Chem Neuroanat 2014; 57-58:54-61. [PMID: 24681169 DOI: 10.1016/j.jchemneu.2014.03.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 03/12/2014] [Accepted: 03/12/2014] [Indexed: 01/24/2023]
Abstract
Fractones are extracellular matrix structures that display a fractal ultrastructure and that are visualized as puncta after immunolabeling for laminin or heparan sulfate proteoglycans. In the adult brain, fractones are found throughout the subventricular zone (SVZ). The role of fractones is just emerging. We have recently shown that fractones sequester fibroblast growth factor-2 and bone morphogenetic protein-7 from the brain ventricles to regulate cell proliferation in the SVZ of the lateral ventricle, the primary neural stem cell niche and neurogenic zone in adulthood. Here, we have examined in vivo the effect of bone morphogenetic protein-4 (BMP-4) on cell proliferation in the SVZ and we have determined whether BMP-4 interacts with fractones to promote this effect. To examine BMP-4 effect on cell proliferation, BMP-4 was intracerebroventricularly injected, and bromodeoxyuridine immunolabeling was performed on frozen sections of the adult mouse brain. To identify the location of BMP-4 binding, biotinylated-BMP-4 was injected, and its binding localized post-mortem with streptavidin, Texas red conjugate. Injection of heparitinase-1 was used to desulfate fractones and determine whether the binding and the effect of BMP-4 on cell proliferation are heparan sulfate-dependent. BMP-4 inhibited cell proliferation in the SVZ neurogenic zone. Biotinylated-BMP-4 bound to fractones and some adjacent blood vessels. Co-injection of heparitinase-1 and biotinylated-BMP-4 resulted in the absence of signal for biotinylated-BMP-4, indicating that the binding was heparan sulfate dependent. Moreover, preventing the binding of BMP-4 to fractones by heparitinase-1 reinforced the inhibitory effect of BMP-4 on cell proliferation in the SVZ. These results show that BMP-4 inhibits cell proliferation in the SVZ neurogenic zone and that the binding of BMP-4 to fractone-associated heparan sulfates moderates this inhibitory effect. Together with our previous results, these data support the view that fractones capture growth factors and modulate their activity in the neural tissues lining the ventricles.
Collapse
Affiliation(s)
- Frederic Mercier
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Biomed T401, 1960 East-West Road, Honolulu, HI 96822, USA.
| | - Vanessa Douet
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Biomed T401, 1960 East-West Road, Honolulu, HI 96822, USA.
| |
Collapse
|
49
|
Place LW, Sekyi M, Kipper MJ. Aggrecan-Mimetic, Glycosaminoglycan-Containing Nanoparticles for Growth Factor Stabilization and Delivery. Biomacromolecules 2014; 15:680-9. [DOI: 10.1021/bm401736c] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Laura W. Place
- School of Biomedical Engineering and ‡Department of Chemical
and Biological Engineering, Colorado State University, 1370 Campus
Delivery, Fort
Collins, Colorado, United States
| | - Maria Sekyi
- School of Biomedical Engineering and ‡Department of Chemical
and Biological Engineering, Colorado State University, 1370 Campus
Delivery, Fort
Collins, Colorado, United States
| | - Matt J. Kipper
- School of Biomedical Engineering and ‡Department of Chemical
and Biological Engineering, Colorado State University, 1370 Campus
Delivery, Fort
Collins, Colorado, United States
| |
Collapse
|
50
|
Tortelli F, Pisano M, Briquez PS, Martino MM, Hubbell JA. Fibronectin binding modulates CXCL11 activity and facilitates wound healing. PLoS One 2013; 8:e79610. [PMID: 24205388 PMCID: PMC3808276 DOI: 10.1371/journal.pone.0079610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/24/2013] [Indexed: 11/18/2022] Open
Abstract
Engineered biomatrices offer the potential to recapitulate the regenerative microenvironment, with important implications in tissue repair. In this context, investigation of the molecular interactions occurring between growth factors, cytokines and extracellular matrix (ECM) has gained increasing interest. Here, we sought to investigate the possible interactions between the ECM proteins fibronectin (FN) and fibrinogen (Fg) with the CXCR3 ligands CXCL9, CXCL10 and CXCL11, which are expressed during wound healing. New binding interactions were observed and characterized. Heparin-binding domains within Fg (residues 15-66 of the β chain, Fg β15-66) and FN (FNI1-5, but not FNIII12-14) were involved in binding to CXCL10 and CXCL11 but not CXCL9. To investigate a possible influence of FN and Fg interactions with CXCL11 in mediating its role during re-epithelialization, we investigated human keratinocyte migration in vitro and wound healing in vivo in diabetic db/db mice. A synergistic effect on CXCL11-induced keratinocyte migration was observed when cells were treated with CXCL11 in combination with FN in a transmigration assay. Moreover, wound healing was enhanced in full thickness excisional wounds treated with fibrin matrices functionalized with FN and containing CXCL11. These findings highlight the importance of the interactions occurring between cytokines and ECM and point to design concepts to develop functional matrices for regenerative medicine.
Collapse
Affiliation(s)
- Federico Tortelli
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marco Pisano
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Priscilla S. Briquez
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mikaël M. Martino
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jeffrey A. Hubbell
- Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|