1
|
Dehner CA, Pearson H, Almohsen SS, Lo YC, Thangaiah JJ, Torres-Mora J, Guo RR, Baker JC, Folpe AL, Alomari AK, Dickson BC, Billings SD, Michal M, Demicco EG, Fritchie KJ, Chrisinger JSA. Acral Fibrochondromyxoid Tumor: A Clinicopathologic and Molecular Genetic Study of 37 Cases. Mod Pathol 2024; 37:100599. [PMID: 39181449 DOI: 10.1016/j.modpat.2024.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Acral fibrochondromyxoid tumor (AFCMT) is a recently described likely benign mesenchymal neoplasm arising in the distal extremities with distinctive histologic features and a recurrent THBS1::ADGRF5 fusion. We studied an additional 37 cases of AFCMT and expanded on the so-far reported clinicopathologic and molecular findings. Tumors occurred in 21 females and 16 males, ranging in age from 17 to 78 years (median age: 47), and solely involved the hands (24/37, 65%) or feet (13/37, 35%). Histologic examination revealed well-delineated uni- or multinodular tumors with prominent vasculature-rich septa and bland, chondrocyte-like tumor cells set within abundant chondromyxoid stroma. Immunohistochemical studies showed that tumor cells were positive for CD34 (25/27; 93%) and ERG (27/27; 100%), whereas negative for S100 protein (0/31). Molecular analysis revealed evidence of a THBS1::ADGRF5 fusion in 17 of 19 (89%) successfully tested tumors. Clinical follow-up was available in 8 cases (median: 97 months), with multiple local recurrences in 1 case at 276, 312, and 360 months. We conclude that AFCMT is a distinct entity with reproducible morphologic, immunohistochemical, and molecular genetic features that should be differentiated from other similar appearing acral mesenchymal neoplasms.
Collapse
Affiliation(s)
- Carina A Dehner
- Department of Anatomic Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana
| | - Hadley Pearson
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Shahd S Almohsen
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ying-Chun Lo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Jorge Torres-Mora
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Ruifeng Ray Guo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida; Department of Dermatology, Mayo Clinic, Jacksonville, Florida
| | - Jonathan C Baker
- Mallinckrodt Institute of Radiology, Musculoskeletal Section, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew L Folpe
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Ahmed K Alomari
- Department of Anatomic Pathology and Laboratory Medicine, Indiana University, Indianapolis, Indiana
| | - Brendan C Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Steven D Billings
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Michael Michal
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Bioptical Laboratory Ltd, Pilsen, Czech Republic
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Karen J Fritchie
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - John S A Chrisinger
- Department of Pathology and Immunology, Division of Anatomic and Molecular Pathology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
2
|
Adams JC. Thrombospondins: Conserved mediators and modulators of metazoan extracellular matrix. Int J Exp Pathol 2024; 105:136-169. [PMID: 39267379 PMCID: PMC11574667 DOI: 10.1111/iep.12517] [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: 06/30/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 09/17/2024] Open
Abstract
This review provides a personal overview of significant scientific developments in the thrombospondin field during the course of my career. Thrombospondins are multidomain, multimeric, calcium-binding extracellular glycoproteins with context-specific roles in tissue organisation. They act at cell surfaces and within ECM to regulate cell phenotype and signalling, differentiation and assembly of collagenous ECM, along with tissue-specific roles in cartilage, angiogenesis and synaptic function. More recently, intracellular, homeostatic roles have also been identified. Resolution of structures for the major domains of mammalian thrombospondins has facilitated major advances in understanding thrombospondin biology from molecule to tissue; for example, in illuminating molecular consequences of disease-causing coding mutations in human pseudoachrondroplasia. Although principally studied in vertebrates, thrombospondins are amongst the most ancient of animal ECM proteins, with many invertebrates encoding a single thrombospondin and the thrombospondin gene family of vertebrates originating through gene duplications. Moreover, thrombospondins form one branch of a thrombospondin superfamily that debuted at the origin of metazoans. The super-family includes additional sub-groups, present only in invertebrates, that differ in N-terminal domain organisation, share the distinctive TSP C-terminal region domain architecture and, to the limited extent studied to date, apparently contribute to tissue development and organisation. Finally, major lines of translational research are discussed, related to fibrosis; TSP1, TSP2 and inhibition of angiogenesis; and the alleviation of chronic cartilage tissue pathologies in pseudoachrondroplasia.
Collapse
|
3
|
Ji Y, Chen Y, Tan X, Huang X, Gao Q, Ma Y, Yang S, Yin M, Yu M, Fang C, Wang Y, Shi Z, Chang J. Integrated transcriptomic and proteomic profiling reveals the key molecular signatures of brain endothelial reperfusion injury. CNS Neurosci Ther 2024; 30:e14483. [PMID: 37789643 PMCID: PMC11017417 DOI: 10.1111/cns.14483] [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: 12/20/2022] [Revised: 08/25/2023] [Accepted: 09/18/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Reperfusion therapy after ischemic stroke often causes brain microvascular injury. However, the underlying mechanisms are unclear. METHODS Transcriptomic and proteomic analyses were performed on human cerebral microvascular endothelial cells following oxygen-glucose deprivation (OGD) or OGD plus recovery (OGD/R) to identify molecules and signaling pathways dysregulated by reperfusion. Major findings were further validated in a mouse model of cerebral ischemia and reperfusion. RESULTS Transcriptomic analysis identified 390 differentially expressed genes (DEGs) between the OGD/R and OGD group. Pathway analysis indicated that these genes were mostly associated with inflammation, including the TNF signaling pathway, TGF-β signaling pathway, cytokine-cytokine receptor interaction, NOD-like receptor signaling pathway, and NF-κB signaling pathway. Proteomic analysis identified 201 differentially expressed proteins (DEPs), which were primarily associated with extracellular matrix destruction and remodeling, impairment of endothelial transport function, and inflammatory responses. Six genes (DUSP1, JUNB, NFKBIA, NR4A1, SERPINE1, and THBS1) were upregulated by OGD/R at both the mRNA and protein levels. In mice with cerebral ischemia and reperfusion, brain TNF signaling pathway was activated by reperfusion, and inhibiting TNF-α with adalimumab significantly attenuated reperfusion-induced brain endothelial inflammation. In addition, the protein level of THBS1 was substantially upregulated upon reperfusion in brain endothelial cells and the peri-endothelial area in mice receiving cerebral ischemia. CONCLUSION Our study reveals the key molecular signatures of brain endothelial reperfusion injury and provides potential therapeutic targets for the treatment of brain microvascular injury after reperfusion therapy in ischemic stroke.
Collapse
Affiliation(s)
- Yabin Ji
- Department of NeurologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Yiman Chen
- Department of NeurologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Xixi Tan
- Department of NeurologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
- Department of NeurologyYangjiang People's HospitalYangjiangChina
| | - Xiaowen Huang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Qiang Gao
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Yinzhong Ma
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Shilun Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Meifang Yin
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Cheng Fang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Zhu Shi
- Department of Neurology10th Affiliated Hospital, Southern Medical University (Dongguan People's Hospital)DongguanChina
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| |
Collapse
|
4
|
Montero E, Isenberg JS. The TSP1-CD47-SIRPα interactome: an immune triangle for the checkpoint era. Cancer Immunol Immunother 2023; 72:2879-2888. [PMID: 37217603 PMCID: PMC10412679 DOI: 10.1007/s00262-023-03465-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
The use of treatments, such as programmed death protein 1 (PD1) or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies, that loosen the natural checks upon immune cell activity to enhance cancer killing have shifted clinical practice and outcomes for the better. Accordingly, the number of antibodies and engineered proteins that interact with the ligand-receptor components of immune checkpoints continue to increase along with their use. It is tempting to view these molecular pathways simply from an immune inhibitory perspective. But this should be resisted. Checkpoint molecules can have other cardinal functions relevant to the development and use of blocking moieties. Cell receptor CD47 is an example of this. CD47 is found on the surface of all human cells. Within the checkpoint paradigm, non-immune cell CD47 signals through immune cell surface signal regulatory protein alpha (SIRPα) to limit the activity of the latter, the so-called trans signal. Even so, CD47 interacts with other cell surface and soluble molecules to regulate biogas and redox signaling, mitochondria and metabolism, self-renewal factors and multipotency, and blood flow. Further, the pedigree of checkpoint CD47 is more intricate than supposed. High-affinity interaction with soluble thrombospondin-1 (TSP1) and low-affinity interaction with same-cell SIRPα, the so-called cis signal, and non-SIRPα ectodomains on the cell membrane suggests that multiple immune checkpoints converge at and through CD47. Appreciation of this may provide latitude for pathway-specific targeting and intelligent therapeutic effect.
Collapse
Affiliation(s)
- Enrique Montero
- Department of Diabetes Immunology, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA
| | - Jeffrey S Isenberg
- Department of Diabetes Complications and Metabolism, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA.
- Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
5
|
Carminati L, Carlessi E, Longhi E, Taraboletti G. Controlled extracellular proteolysis of thrombospondins. Matrix Biol 2023; 119:82-100. [PMID: 37003348 DOI: 10.1016/j.matbio.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Limited proteolysis of thrombospondins is a powerful mechanism to ensure dynamic tuning of their activities in the extracellular space. Thrombospondins are multifunctional matricellular proteins composed of multiple domains, each with a specific pattern of interactions with cell receptors, matrix components and soluble factors (growth factors, cytokines and proteases), thus with different effects on cell behavior and responses to changes in the microenvironment. Therefore, the proteolytic degradation of thrombospondins has multiple functional consequences, reflecting the local release of active fragments and isolated domains, exposure or disruption of active sequences, altered protein location, and changes in the composition and function of TSP-based pericellular interaction networks. In this review current data from the literature and databases is employed to provide an overview of cleavage of mammalian thrombospondins by different proteases. The roles of the fragments generated in specific pathological settings, with particular focus on cancer and the tumor microenvironment, are discussed.
Collapse
Affiliation(s)
- Laura Carminati
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Elena Carlessi
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Elisa Longhi
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy
| | - Giulia Taraboletti
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 24126 Bergamo, Italy.
| |
Collapse
|
6
|
Fu H, Siggs OM, Knight LS, Staffieri SE, Ruddle JB, Birsner AE, Collantes ER, Craig JE, Wiggs JL, D’Amato RJ. Thrombospondin 1 missense alleles induce extracellular matrix protein aggregation and TM dysfunction in congenital glaucoma. J Clin Invest 2022; 132:e156967. [PMID: 36453543 PMCID: PMC9711877 DOI: 10.1172/jci156967] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 10/11/2022] [Indexed: 12/03/2022] Open
Abstract
Glaucoma is a highly heritable disease that is a leading cause of blindness worldwide. Here, we identified heterozygous thrombospondin 1 (THBS1) missense alleles altering p.Arg1034, a highly evolutionarily conserved amino acid, in 3 unrelated and ethnically diverse families affected by congenital glaucoma, a severe form of glaucoma affecting children. Thbs1R1034C-mutant mice had elevated intraocular pressure (IOP), reduced ocular fluid outflow, and retinal ganglion cell loss. Histology revealed an abundant, abnormal extracellular accumulation of THBS1 with abnormal morphology of juxtacanalicular trabecular meshwork (TM), an ocular tissue critical for aqueous fluid outflow. Functional characterization showed that the THBS1 missense alleles found in affected individuals destabilized the THBS1 C-terminus, causing protein misfolding and extracellular aggregation. Analysis using a range of amino acid substitutions at position R1034 showed that the extent of aggregation was correlated with the change in protein-folding free energy caused by variations in amino acid structure. Extracellular matrix (ECM) proteins, especially fibronectin, which bind to THBS1, also accumulated within THBS1 deposits. These results show that missense variants altering THBS1 p.Arg1034 can cause elevated IOP through a mechanism involving impaired TM fluid outflow in association with accumulation of aggregated THBS1 in the ECM of juxtacanalicular meshwork with altered morphology.
Collapse
Affiliation(s)
- Haojie Fu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Owen M. Siggs
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Lachlan S.W. Knight
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Sandra E. Staffieri
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
- Department of Ophthalmology, University of Melbourne, Department of Surgery, Parkville, Victoria, Australia
- Department of Ophthalmology, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Jonathan B. Ruddle
- Department of Ophthalmology, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Amy E. Birsner
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Robert J. D’Amato
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Feng QL, Gu JJ, Chen JY, Zheng WY, Pan HH, Xu XY, Deng CC, Yang B. TSP1 promotes fibroblast proliferation and extracellular matrix deposition via the IL6/JAK2/STAT3 signalling pathway in keloids. Exp Dermatol 2022; 31:1533-1542. [PMID: 35661430 DOI: 10.1111/exd.14623] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
Keloids are benign fibroproliferative diseases with abnormally proliferated bulges beyond the edge of the skin lesions, and they are characterized by uncontrolled fibroblast proliferation and excessive extracellular matrix deposition in the dermis. However, the definite mechanisms that increase fibroblast proliferation and collagen deposition in keloids remain unclear. Thrombospondin 1 (TSP1) has been suggested to play an important role in wound healing and fibrotic disorders, but its role in keloids is unknown. In this study, we aimed to clarify the specific role of TSP1 in keloids and explore the potential mechanism. Our results demonstrated that TSP1 was highly expressed in keloid lesions compared to normal skin. Knockdown of TSP1 in keloid fibroblasts decreased cell proliferation and collagen I deposition. Exogenous TSP1 treatment increased cell proliferation and collagen I deposition in normal fibroblasts. We further investigated the underlying mechanism and found that TSP1 promoted fibroblast proliferation and extracellular matrix deposition by upregulating the IL6/JAK2/STAT3 pathway. Moreover, we verified that TSP1 expression was positively correlated with IL6/STAT3 signalling activity in keloids. Taken together, our findings indicate that TSP1 promotes keloid development via the IL6/JAK2/STAT3 signalling pathway and blocking TSP1 may represent a potential strategy for keloid therapy.
Collapse
Affiliation(s)
- Qing-Lan Feng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Jing Gu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Yi Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Yue Zheng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Hui-Hui Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xue-Yan Xu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Cheng-Cheng Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Murphy-Ullrich JE. Thrombospondin-1 Signaling Through the Calreticulin/LDL Receptor Related Protein 1 Axis: Functions and Possible Roles in Glaucoma. Front Cell Dev Biol 2022; 10:898772. [PMID: 35693935 PMCID: PMC9185677 DOI: 10.3389/fcell.2022.898772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Thrombospondin-1 (TSP-1) is a matricellular extracellular matrix protein. Matricellular proteins are components of the extracellular matrix (ECM) that regulate key cellular functions and impact ECM organization, but which lack direct primary structural roles in the ECM. TSP-1 expression is upregulated in response to injury, hypoxia, growth factor stimulation, inflammation, glucose, and by reactive oxygen species. Relevant to glaucoma, TSP-1 is also a mechanosensitive molecule upregulated by mechanical stretch. TSP-1 expression is increased in ocular remodeling in glaucoma in both the trabecular meshwork and in the optic nerve head. The exact roles of TSP-1 in glaucoma remain to be defined, however. It plays important roles in cell behavior and in ECM remodeling during wound healing, fibrosis, angiogenesis, and in tumorigenesis and metastasis. At the cellular level, TSP-1 can modulate cell adhesion and migration, protease activity, growth factor activity, anoikis resistance, apoptosis, and collagen secretion and matrix assembly and cross-linking. These multiple functions and macromolecular and receptor interactions have been ascribed to specific domains of the TSP-1 molecule. In this review, we will focus on the cell regulatory activities of the TSP-1 N-terminal domain (NTD) sequence that binds to cell surface calreticulin (Calr) and which regulates cell functions via signaling through Calr complexed with LDL receptor related protein 1 (LRP1). We will describe TSP-1 actions mediated through the Calr/LRP1 complex in regulating focal adhesion disassembly and cytoskeletal reorganization, cell motility, anoikis resistance, and induction of collagen secretion and matrix deposition. Finally, we will consider the relevance of these TSP-1 functions to the pathologic remodeling of the ECM in glaucoma.
Collapse
Affiliation(s)
- Joanne E. Murphy-Ullrich
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Joanne E. Murphy-Ullrich,
| |
Collapse
|
9
|
Hellewell AL, Heesom KJ, Jepson MA, Adams JC. PDIA3/ERp57 promotes a matrix-rich secretome that stimulates fibroblast adhesion through CCN2. Am J Physiol Cell Physiol 2022; 322:C624-C644. [PMID: 35196163 PMCID: PMC8977143 DOI: 10.1152/ajpcell.00258.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The matricellular glycoprotein thrombospondin1 (TSP1) has complex roles in the extracellular matrix and at cell surfaces, but relatively little is known about its intracellular associations prior to secretion. To search for novel intracellular interactions of TSP1 in situ, we carried out a biotin ligase-based TSP1 interactome screen and identified protein disulphide isomerase A3 (PDIA3/ERp57) as a novel candidate binding protein. In validation, TSP1 and PDIA3 were established to bind in vitro and to colocalise in the endoplasmic reticulum of human dermal fibroblasts (HDF). Loss of PDIA3 function, either by pharmacological inhibition in HDF or in Pdia3-/- mouse embryo fibroblasts (Pdia3-/-MEF), led to alterations in the composition of cell-derived ECM, involving changed abundance of fibronectin and TSP1, and was correlated with reduced cell spreading, altered organisation of F-actin and reduced focal adhesions. These cellular phenotypes of Pdia3-/-MEF were normalised by exposure to conditioned medium (WTCM) or extracellular matrix (WTECM) from wild-type (WT)-MEF. Rescue depended on PDIA3 activity in WT-MEF, and was not prevented by immunodepletion of fibronectin. Heparin-binding proteins in WTCM were found to be necessary for rescue. Comparative quantitative tandem-mass-tag proteomics and functional assays on the heparin-binding secretomes of WT-MEF and Pdia3-/- MEF identified multiple ECM and growth factor proteins to be down-regulated in the CM of Pdia3-/- MEF. Of these, CCN2 was identified to be necessary for the adhesion-promoting activity of WTCM on Pdia3-/- MEF and to bind TSP1. Thus, PDIA3 coordinates fibroblast production of an ECM-rich, pro-adhesive microenvironment, with implications for PDIA3 as a translational target.
Collapse
Affiliation(s)
| | - Kate J Heesom
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Mark A Jepson
- Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
10
|
Hsieh LY, Chiang AWT, Duong LD, Kuo CC, Dong SX, Dohil R, Kurten R, Lewis NE, Aceves SS. A unique esophageal extracellular matrix proteome alters normal fibroblast function in severe eosinophilic esophagitis. J Allergy Clin Immunol 2021; 148:486-494. [PMID: 33556465 PMCID: PMC8342625 DOI: 10.1016/j.jaci.2021.01.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is a chronic TH2 disorder complicated by tissue fibrosis and loss of esophageal luminal patency. The fibrostenotic esophagus does not respond well to therapy, but profibrotic therapeutic targets are largely unclear. OBJECTIVE Our aim was to utilize proteomics and primary cells as a novel approach to determine relevant profibrotic factors. METHODS We utilized primary esophageal EoE and normal fibroblasts, their derivative extracellular matrixes (ECMs), an approach of fibroblast culture on autologous versus nonautologous ECM, and proteomics to elucidate EoE ECM proteins that dysregulate cellular function. RESULTS We cultured esophageal fibroblasts from normal esophagi and esophagi from patients with severe EoE on autologous versus nonautologous ECM. The EoE ECM proteome shifted normal esophageal fibroblast protein expression. Proteomic analysis demonstrated that thrombospondin-1 is detected only in the EoE ECM, is central in the EoE ECM protein-protein interactome, is found at significantly elevated levels in biopsy specimens from patients with active EoE, and induces fibroblast collagen I production. CONCLUSION Fibroblasts from patients with EoE secrete a unique ECM proteome that reflects their in vivo state and induces collagen I and α-smooth muscle actin protein expression from normal fibroblasts. Thrombospondin-1 is a previously unappreciated profibrotic molecule in EoE.
Collapse
Affiliation(s)
- Lance Y Hsieh
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Loan D Duong
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Chih-Chung Kuo
- Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Stephanie X Dong
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Gastroenterology, University of California, San Diego, La Jolla, Calif; Rady Children's Hospital San Diego, Calif, San Diego, Calif
| | - Richard Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Ark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Department of Bioengineering, University of California, San Diego, La Jolla, Calif
| | - Seema S Aceves
- Department of Pediatrics, University of California, San Diego, La Jolla, Calif; Division of Allergy Immunology, University of California, San Diego, La Jolla, Calif; Rady Children's Hospital San Diego, Calif, San Diego, Calif; Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
11
|
Kaur S, Bronson SM, Pal-Nath D, Miller TW, Soto-Pantoja DR, Roberts DD. Functions of Thrombospondin-1 in the Tumor Microenvironment. Int J Mol Sci 2021; 22:4570. [PMID: 33925464 PMCID: PMC8123789 DOI: 10.3390/ijms22094570] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
The identification of thrombospondin-1 as an angiogenesis inhibitor in 1990 prompted interest in its role in cancer biology and potential as a therapeutic target. Decreased thrombospondin-1 mRNA and protein expression are associated with progression in several cancers, while expression by nonmalignant cells in the tumor microenvironment and circulating levels in cancer patients can be elevated. THBS1 is not a tumor suppressor gene, but the regulation of its expression in malignant cells by oncogenes and tumor suppressor genes mediates some of their effects on carcinogenesis, tumor progression, and metastasis. In addition to regulating angiogenesis and perfusion of the tumor vasculature, thrombospondin-1 limits antitumor immunity by CD47-dependent regulation of innate and adaptive immune cells. Conversely, thrombospondin-1 is a component of particles released by immune cells that mediate tumor cell killing. Thrombospondin-1 differentially regulates the sensitivity of malignant and nonmalignant cells to genotoxic stress caused by radiotherapy and chemotherapy. The diverse activities of thrombospondin-1 to regulate autophagy, senescence, stem cell maintenance, extracellular vesicle function, and metabolic responses to ischemic and genotoxic stress are mediated by several cell surface receptors and by regulating the functions of several secreted proteins. This review highlights progress in understanding thrombospondin-1 functions in cancer and the challenges that remain in harnessing its therapeutic potential.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Steven M. Bronson
- Department of Internal Medicine, Section of Molecular Medicine, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Thomas W. Miller
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, 13273 Marseille, France
| | - David R. Soto-Pantoja
- Department of Surgery and Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| |
Collapse
|
12
|
Thrombospondin-1 CD47 Signalling: From Mechanisms to Medicine. Int J Mol Sci 2021; 22:ijms22084062. [PMID: 33920030 PMCID: PMC8071034 DOI: 10.3390/ijms22084062] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Recent advances provide evidence that the cellular signalling pathway comprising the ligand-receptor duo of thrombospondin-1 (TSP1) and CD47 is involved in mediating a range of diseases affecting renal, vascular, and metabolic function, as well as cancer. In several instances, research has barely progressed past pre-clinical animal models of disease and early phase 1 clinical trials, while for cancers, anti-CD47 therapy has emerged from phase 2 clinical trials in humans as a crucial adjuvant therapeutic agent. This has important implications for interventions that seek to capitalize on targeting this pathway in diseases where TSP1 and/or CD47 play a role. Despite substantial progress made in our understanding of this pathway in malignant and cardiovascular disease, knowledge and translational gaps remain regarding the role of this pathway in kidney and metabolic diseases, limiting identification of putative drug targets and development of effective treatments. This review considers recent advances reported in the field of TSP1-CD47 signalling, focusing on several aspects including enzymatic production, receptor function, interacting partners, localization of signalling, matrix-cellular and cell-to-cell cross talk. The potential impact that these newly described mechanisms have on health, with a particular focus on renal and metabolic disease, is also discussed.
Collapse
|
13
|
Anastasi C, Rousselle P, Talantikite M, Tessier A, Cluzel C, Bachmann A, Mariano N, Dussoyer M, Alcaraz LB, Fortin L, Aubert A, Delolme F, El Kholti N, Armengaud J, Fournié P, Auxenfans C, Valcourt U, Goff SVL, Moali C. BMP-1 disrupts cell adhesion and enhances TGF-β activation through cleavage of the matricellular protein thrombospondin-1. Sci Signal 2020; 13:13/639/eaba3880. [PMID: 32636307 DOI: 10.1126/scisignal.aba3880] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bone morphogenetic protein 1 (BMP-1) is an important metalloproteinase that synchronizes growth factor activation with extracellular matrix assembly during morphogenesis and tissue repair. The mechanisms by which BMP-1 exerts these effects are highly context dependent. Because BMP-1 overexpression induces marked phenotypic changes in two human cell lines (HT1080 and 293-EBNA cells), we investigated how BMP-1 simultaneously affects cell-matrix interactions and growth factor activity in these cells. Increasing BMP-1 led to a loss of cell adhesion that depended on the matricellular glycoprotein thrombospondin-1 (TSP-1). BMP-1 cleaved TSP-1 between the VWFC/procollagen-like domain and the type 1 repeats that mediate several key TSP-1 functions. This cleavage induced the release of TSP-1 C-terminal domains from the extracellular matrix and abolished its previously described multisite cooperative interactions with heparan sulfate proteoglycans and CD36 on HT1080 cells. In addition, BMP-1-dependent proteolysis potentiated the TSP-1-mediated activation of latent transforming growth factor-β (TGF-β), leading to increased signaling through the canonical SMAD pathway. In primary human corneal stromal cells (keratocytes), endogenous BMP-1 cleaved TSP-1, and the addition of exogenous BMP-1 enhanced cleavage, but this had no substantial effect on cell adhesion. Instead, processed TSP-1 promoted the differentiation of keratocytes into myofibroblasts and stimulated production of the myofibroblast marker α-SMA, consistent with the presence of processed TSP-1 in human corneal scars. Our results indicate that BMP-1 can both trigger the disruption of cell adhesion and stimulate TGF-β signaling in TSP-1-rich microenvironments, which has important potential consequences for wound healing and tumor progression.
Collapse
Affiliation(s)
- Cyril Anastasi
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Patricia Rousselle
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Maya Talantikite
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Agnès Tessier
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Caroline Cluzel
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Alice Bachmann
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Natacha Mariano
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Mélissa Dussoyer
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Lindsay B Alcaraz
- University of Lyon, Centre Léon Bérard, INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), F-69373 Lyon, France
| | - Laëtitia Fortin
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Alexandre Aubert
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Frédéric Delolme
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,University of Lyon, ENS de Lyon, INSERM US8, CNRS UMS3444, SFR Biosciences, F-69366 Lyon, France
| | - Naïma El Kholti
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Jean Armengaud
- CEA Marcoule, Innovative Technologies for Detection and Diagnostics Laboratory (DRF/Joliot/DMTS/SPI/Li2D), F-30200 Bagnols-sur-Cèze, France
| | - Pierre Fournié
- Purpan University Hospital, Ophthalmology Department, F-31059 Toulouse, France.,University of Toulouse, CNRS UMR 5165, INSERM U1056, Epithelial Differentiation and Rheumatoid Autoimmunity Unit (UDEAR), F-31059 Toulouse, France
| | - Céline Auxenfans
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,Hospices Civils de Lyon, Tissue and Cell Bank, F-69437 Lyon, France
| | - Ulrich Valcourt
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,University of Lyon, Centre Léon Bérard, INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), F-69373 Lyon, France
| | - Sandrine Vadon-Le Goff
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Catherine Moali
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.
| |
Collapse
|
14
|
Isenberg JS, Roberts DD. Thrombospondin-1 in maladaptive aging responses: a concept whose time has come. Am J Physiol Cell Physiol 2020; 319:C45-C63. [PMID: 32374675 DOI: 10.1152/ajpcell.00089.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous age-dependent alterations at the molecular, cellular, tissue and organ systems levels underlie the pathophysiology of aging. Herein, the focus is upon the secreted protein thrombospondin-1 (TSP1) as a promoter of aging and age-related diseases. TSP1 has several physiological functions in youth, including promoting neural synapse formation, mediating responses to ischemic and genotoxic stress, minimizing hemorrhage, limiting angiogenesis, and supporting wound healing. These acute functions of TSP1 generally require only transient expression of the protein. However, accumulating basic and clinical data reinforce the view that chronic diseases of aging are associated with accumulation of TSP1 in the extracellular matrix, which is a significant maladaptive contributor to the aging process. Identification of the relevant cell types that chronically produce and respond to TSP1 and the molecular mechanisms that mediate the resulting maladaptive responses could direct the development of therapeutic agents to delay or revert age-associated maladies.
Collapse
Affiliation(s)
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
15
|
Denèfle T, Pramil E, Gómez-Morales L, Levasseur MD, Lardé E, Newton C, Herry K, Herbi L, Lamotte Y, Odile E, Ancellin N, Grondin P, Martinez-Torres AC, Viviani F, Merle-Beral H, Lequin O, Susin SA, Karoyan P. Homotrimerization Approach in the Design of Thrombospondin-1 Mimetic Peptides with Improved Potency in Triggering Regulated Cell Death of Cancer Cells. J Med Chem 2019; 62:7656-7668. [PMID: 31403795 DOI: 10.1021/acs.jmedchem.9b00024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In order to optimize the potency of the first serum-stable peptide agonist of CD47 (PKHB1) in triggering regulated cell death of cancer cells, we designed a maturation process aimed to mimic the trimeric structure of the thrombospondin-1/CD47 binding epitope. For that purpose, an N-methylation scan of the PKHB1 sequence was realized to prevent peptide aggregation. Structural and pharmacological analyses were conducted in order to assess the conformational impact of these chemical modifications on the backbone structure and the biological activity. This structure-activity relationship study led to the discovery of a highly soluble N-methylated peptide that we termed PKT16. Afterward, this monomer was used for the design of a homotrimeric peptide mimic that we termed [PKT16]3, which proved to be 10-fold more potent than its monomeric counterpart. A pharmacological evaluation of [PKT16]3 in inducing cell death of adherent (A549) and nonadherent (MEC-1) cancer cell lines was also performed.
Collapse
Affiliation(s)
- Thomas Denèfle
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Elodie Pramil
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Luis Gómez-Morales
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,Laboratory of Immunology and Virology , Autonomous University of Nuevo Leon , 66451 San Nicolas de los Garza , NL , Mexico
| | - Mikail D Levasseur
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Eva Lardé
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Clara Newton
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | - Kenny Herry
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Linda Herbi
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Yann Lamotte
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Estelle Odile
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France
| | | | - Pascal Grondin
- OncoDesign , 25 Avenue du Québec , 91140 Les Ulis , France
| | - Ana-Carolina Martinez-Torres
- Laboratory of Immunology and Virology , Autonomous University of Nuevo Leon , 66451 San Nicolas de los Garza , NL , Mexico
| | | | - Hélène Merle-Beral
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Olivier Lequin
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France
| | - Santos A Susin
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team , Centre de Recherche des Cordeliers, INSERM UMRS 1138 , 75006 Paris , France
| | - Philippe Karoyan
- Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM , 75005 Paris , France.,Sorbonne Université , Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, Site OncoDesign , 25-27 Avenue du Québec , 91140 Les Ulis , France.,SiRIC CURAMUS (CANCER UNITED RESEARCH ASSOCIATING MEDICINE, UNIVERSITY & SOCIETY, Site de Recherche Intégrée sur le Cancer) IUC, AP-HP.6 , Sorbonne Université 75005 Paris , France.,Kayvisa AG , Industriestrasse, 44 , 6300 Zug , Switzerland.,Kaybiotix GmbH , Zugerstrasse 32 , 6340 Baar , Switzerland
| |
Collapse
|
16
|
Gebauer JM, Köhler A, Dietmar H, Gompert M, Neundorf I, Zaucke F, Koch M, Baumann U. COMP and TSP-4 interact specifically with the novel GXKGHR motif only found in fibrillar collagens. Sci Rep 2018; 8:17187. [PMID: 30464261 PMCID: PMC6249252 DOI: 10.1038/s41598-018-35447-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/05/2018] [Indexed: 12/19/2022] Open
Abstract
COMP (cartilage oligomeric matrix protein) is a member of the thrombospondin family and forms homopentamers as well as mixed heterooligomers with its closely related family member TSP-4. COMP is long known to bind to collagens and to influence collagen fibril formation. Recent work indicates that already intracellular interaction with collagen is important for collagen secretion. However, the exact binding site of COMP on the collagen triple helix has not been described up to now. In this study we have identified a GXKGHR motif on the collagen II helix to bind to COMP, using a recombinantly expressed collagen II peptide library. This binding sequence is conserved throughout evolution and we demonstrate that TSP-4 binds to the same sequence. The identified binding motif overlaps with the recognition sites of many other collagen-binding partners (e.g. PEDF, Heparin) and also spans the lysine residues, which form collagen cross-links. COMP might thereby protect collagen helices from premature modification and cross-linking. Interestingly, this motif is only found in classical fibrillar collagens, although COMP is known to also bind other types. This might indicate that COMP has a unique interface for fibrillar collagens, thus making it an interesting target for the development of antifibrotic drugs.
Collapse
Affiliation(s)
- Jan M Gebauer
- Institute of Biochemistry, University of Cologne, Cologne, Germany.
| | - Anna Köhler
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Helen Dietmar
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Monika Gompert
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Ines Neundorf
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| | - Frank Zaucke
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Dr. Rolf Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim gGmbH, Frankfurt, Germany
| | - Manuel Koch
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Institute for Dental Research and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ulrich Baumann
- Institute of Biochemistry, University of Cologne, Cologne, Germany
| |
Collapse
|
17
|
Lommel M, Strompen J, Hellewell AL, Balasubramanian GP, Christofidou ED, Thomson AR, Boyle AL, Woolfson DN, Puglisi K, Hartl M, Holstein TW, Adams JC, Özbek S. Hydra Mesoglea Proteome Identifies Thrombospondin as a Conserved Component Active in Head Organizer Restriction. Sci Rep 2018; 8:11753. [PMID: 30082916 PMCID: PMC6079037 DOI: 10.1038/s41598-018-30035-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
Thrombospondins (TSPs) are multidomain glycoproteins with complex matricellular functions in tissue homeostasis and remodeling. We describe a novel role of TSP as a Wnt signaling target in the basal eumetazoan Hydra. Proteome analysis identified Hydra magnipapillata TSP (HmTSP) as a major component of the cnidarian mesoglea. In general, the domain organization of cnidarian TSPs is related to the pentameric TSPs of bilaterians, and in phylogenetic analyses cnidarian TSPs formed a separate clade of high sequence diversity. HmTSP expression in polyps was restricted to the hypostomal tip and tentacle bases that harbor Wnt-regulated organizer tissues. In the hypostome, HmTSP- and Wnt3-expressing cells were identical or in close vicinity to each other, and regions of ectopic tentacle formation induced by pharmacological β-Catenin activation (Alsterpaullone) corresponded to foci of HmTSP expression. Chromatin immunoprecipitation (ChIP) confirmed binding of Hydra TCF to conserved elements in the HmTSP promotor region. Accordingly, β-Catenin knockdown by siRNAs reduced normal HmTSP expression at the head organizer. In contrast, knockdown of HmTSP expression led to increased numbers of ectopic organizers in Alsterpaullone-treated animals, indicating a negative regulatory function. Our data suggest an unexpected role for HmTSP as a feedback inhibitor of Wnt signaling during Hydra body axis patterning and maintenance.
Collapse
Affiliation(s)
- Mark Lommel
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Jennifer Strompen
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Andrew L Hellewell
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Gnana Prakash Balasubramanian
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.,G200 Division of Applied Bioinformatics, German Cancer Research Institute (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Elena D Christofidou
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Andrew R Thomson
- School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK.,School of Chemistry, University of Glasgow, Joseph Black Building, University Avenue, Glasgow, G12 8QQ, Scotland
| | - Aimee L Boyle
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.,School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK.,Leiden Institute of Chemistry, Leiden University, POB 9502, NL-2300, RA Leiden, Netherlands
| | - Derek N Woolfson
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.,School of Chemistry, Cantock's Close, University of Bristol, Bristol, BS8 1TS, UK
| | - Kane Puglisi
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Markus Hartl
- Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, A-6020, Innsbruck, Austria
| | - Thomas W Holstein
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany
| | - Josephine C Adams
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Suat Özbek
- University of Heidelberg, Centre for Organismal Studies, Department of Molecular Evolution and Genomics, Im Neuenheimer Feld 230, 69120, Heidelberg, Germany.
| |
Collapse
|
18
|
Rogers NM, Ghimire K, Calzada MJ, Isenberg JS. Matricellular protein thrombospondin-1 in pulmonary hypertension: multiple pathways to disease. Cardiovasc Res 2018; 113:858-868. [PMID: 28472457 DOI: 10.1093/cvr/cvx094] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/03/2017] [Indexed: 12/24/2022] Open
Abstract
Matricellular proteins are secreted molecules that have affinities for both extracellular matrix and cell surface receptors. Through interaction with structural proteins and the cells that maintain the matrix these proteins can alter matrix strength. Matricellular proteins exert control on cell activity primarily through engagement of membrane receptors that mediate outside-in signaling. An example of this group is thrombospondin-1 (TSP1), first identified as a component of the secreted product of activated platelets. As a result, TSP1 was initially studied in relation to coagulation, growth factor signaling and angiogenesis. More recently, TSP1 has been found to alter the effects of the gaseous transmitter nitric oxide (NO). This latter capacity has provided motivation to study TSP1 in diseases associated with loss of NO signaling as observed in cardiovascular disease and pulmonary hypertension (PH). PH is characterized by progressive changes in the pulmonary vasculature leading to increased resistance to blood flow and subsequent right heart failure. Studies have linked TSP1 to pre-clinical animal models of PH and more recently to clinical PH. This review will provide analysis of the vascular and non-vascular effects of TSP1 that contribute to PH, the experimental and translational studies that support a role for TSP1 in disease promotion and frame the relevance of these findings to therapeutic strategies.
Collapse
Affiliation(s)
- Natasha M Rogers
- Medicine, Westmead Clinical School, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Kedar Ghimire
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maria J Calzada
- Department of Medicine, Universidad Autónoma of Madrid, Diego de León, Hospital Universitario of the Princesa, 62?28006 Madrid, Spain
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
19
|
Suppression of TGFβ-mediated conversion of endothelial cells and fibroblasts into cancer associated (myo)fibroblasts via HDAC inhibition. Br J Cancer 2018; 118:1359-1368. [PMID: 29695769 PMCID: PMC5959903 DOI: 10.1038/s41416-018-0072-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/05/2018] [Accepted: 03/09/2018] [Indexed: 01/05/2023] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) support tumour progression and invasion, and they secrete abundant extracellular matrix (ECM) that may shield tumour cells from immune checkpoint or kinase inhibitors. Targeting CAFs using drugs that revert their differentiation, or inhibit their tumour-supportive functions, has been considered as an anti-cancer strategy. Methods We have used human and murine cell culture models, atomic force microscopy (AFM), microarray analyses, CAF/tumour cell spheroid co-cultures and transgenic fibroblast reporter mice to study how targeting HDACs using small molecule inhibitors or siRNAs re-directs CAF differentiation and function in vitro and in vivo. Results From a small molecule screen, we identified Scriptaid, a selective inhibitor of HDACs 1/3/8, as a repressor of TGFβ-mediated CAF differentiation. Scriptaid inhibits ECM secretion, reduces cellular contraction and stiffness, and impairs collective cell invasion in CAF/tumour cell spheroid co-cultures. Scriptaid also reduces CAF abundance and delays tumour growth in vivo. Conclusions Scriptaid is a well-tolerated and effective HDACi that reverses many of the functional and phenotypic properties of CAFs. Impeding or reversing CAF activation/function by altering the cellular epigenetic regulatory machinery could control tumour growth and invasion, and be beneficial in combination with additional therapies that target cancer cells or immune cells directly.
Collapse
|
20
|
Hellewell AL, Rosini S, Adams JC. A Rapid, Scalable Method for the Isolation, Functional Study, and Analysis of Cell-derived Extracellular Matrix. J Vis Exp 2017. [PMID: 28117783 PMCID: PMC5351878 DOI: 10.3791/55051] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix (ECM) is recognized as a diverse, dynamic, and complex environment that is involved in multiple cell-physiological and pathological processes. However, the isolation of ECM, from tissues or cell culture, is complicated by the insoluble and cross-linked nature of the assembled ECM and by the potential contamination of ECM extracts with cell surface and intracellular proteins. Here, we describe a method for use with cultured cells that is rapid and reliably removes cells to isolate a cell-derived ECM for downstream experimentation. Through use of this method, the isolated ECM and its components can be visualized by in situ immunofluorescence microscopy. The dynamics of specific ECM proteins can be tracked by tracing the deposition of a tagged protein using fluorescence microscopy, both before and after the removal of cells. Alternatively, the isolated ECM can be extracted for biochemical analysis, such as sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting. At larger scales, a full proteomics analysis of the isolated ECM by mass spectrometry can be conducted. By conducting ECM isolation under sterile conditions, sterile ECM layers can be obtained for functional or phenotypic studies with any cell of interest. The method can be applied to any adherent cell type, is relatively easy to perform, and can be linked to a wide repertoire of experimental designs.
Collapse
|
21
|
Starlinger P, Assinger A, Gruenberger T, Brostjan C. The role of platelets and portal venous pressure fluctuations in postoperative liver regeneration. Eur Surg 2015. [DOI: 10.1007/s10353-015-0352-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Abstract
OBJECTIVES Autism is a severe neurodevelopmental disorder. Many susceptible or causative genes have been identified, and most of them are related to synaptogenesis. The THBS1 gene encodes thrombospondin 1, which plays a critical role in synaptogenesis of the central nervous system in the developing brain. However, no study has been carried out revealing that THBS1 is an autism risk gene. METHODS We analyzed the whole coding region and the 5'-untranslated region of the THBS1 gene in 313 autistic patients by Sanger sequencing, which was also used to analyze the identified variants in 350 normal controls. Association analysis was carried out using PLINK or R. Haplotype analysis was carried out using Haploview. Functional prediction and conservation analysis of missense variants were carried out using ANNOVAR. RESULTS Twelve variants, including five common variants and seven rare variants, were identified in the THBS1 coding region and the 5'-untranslated region. Among them, one common variant (c.1567A>G:p.T523A) was significantly associated with autism (P<0.05). Two rare variants (c.2429G>A:p.R810Q, c.3496G>C:p.E1166Q) were absent in the 350 controls and were not reported in the single nucleotide polymorphism database (dbSNP). Combined association analysis of the rare variants (minor allele frequency<0.01) in patients and Asian samples in the 1000 genome project revealed a significant association between these rare variants and autism (P=0.039). CONCLUSION Our data revealed that both common and rare variants of the THBS1 gene are associated with risk for autism, suggesting that THBS1 is a novel susceptible gene for autism.
Collapse
|
23
|
Modulation of the extracellular matrix patterning of thrombospondins by actin dynamics and thrombospondin oligomer state. Biosci Rep 2015; 35:BSR20140168. [PMID: 26182380 PMCID: PMC4613707 DOI: 10.1042/bsr20140168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/21/2015] [Indexed: 01/01/2023] Open
Abstract
Thrombospondins (TSPs) are evolutionarily-conserved, secreted glycoproteins that interact with cell surfaces and extracellular matrix (ECM) and have complex roles in cell interactions. Unlike the structural components of the ECM that form networks or fibrils, TSPs are deposited into ECM as arrays of nanoscale puncta. The cellular and molecular mechanisms for the patterning of TSPs in ECM are poorly understood. In the present study, we investigated whether the mechanisms of TSP patterning in cell-derived ECM involves actin cytoskeletal pathways or TSP oligomer state. From tests of a suite of pharmacological inhibitors of small GTPases, actomyosin-based contractility, or actin microfilament integrity and dynamics, cytochalasin D and jasplakinolide treatment of cells were identified to result in altered ECM patterning of a model TSP1 trimer. The strong effect of cytochalasin D indicated that mechanisms controlling puncta patterning depend on global F-actin dynamics. Similar spatial changes were obtained with endogenous TSPs after cytochalasin D treatment, implicating physiological relevance. Under matched experimental conditions with ectopically-expressed TSPs, the magnitude of the effect was markedly lower for pentameric TSP5 and Drosophila TSP, than for trimeric TSP1 or dimeric Ciona TSPA. To distinguish between the variables of protein sequence or oligomer state, we generated novel, chimeric pentamers of TSP1. These proteins accumulated within ECM at higher levels than TSP1 trimers, yet the effect of cytochalasin D on the spatial distribution of puncta was reduced. These findings introduce a novel concept that F-actin dynamics modulate the patterning of TSPs in ECM and that TSP oligomer state is a key determinant of this process.
Collapse
|
24
|
Kim DJ, Christofidou ED, Keene DR, Hassan Milde M, Adams JC. Intermolecular interactions of thrombospondins drive their accumulation in extracellular matrix. Mol Biol Cell 2015; 26:2640-54. [PMID: 25995382 PMCID: PMC4501361 DOI: 10.1091/mbc.e14-05-0996] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 05/12/2015] [Indexed: 02/01/2023] Open
Abstract
A novel mechanism of intermolecular interactions in trans is identified by which thrombospondin molecules accumulate as puncta within the extracellular matrix. This process depends on a novel, conserved, surface-exposed site on the thrombospondin L-type lectin domain. Thrombospondins participate in many aspects of tissue organization in adult tissue homeostasis, and their dysregulation contributes to pathological processes such as fibrosis and tumor progression. The incorporation of thrombospondins into extracellular matrix (ECM) as discrete puncta has been documented in various tissue and cell biological contexts, yet the underlying mechanisms remain poorly understood. We find that collagen fibrils are disorganized in multiple tissues of Thbs1−/− mice. In investigating how thrombospondins become retained within ECM and thereby affect ECM organization, we find that accumulation of thrombospondin-1 or thrombospondin-5 puncta within cell-derived ECM is controlled by a novel, conserved, surface-exposed site on the thrombospondin L-type lectin domain. This site acts to recruit thrombospondin molecules into ECM by intermolecular interactions in trans. This mechanism is fibronectin independent, can take place extracellularly, and is demonstrated to be direct in vitro. The trans intermolecular interactions can also be heterotypic—for example, between thrombospondin-1 and thrombospondin-5. These data identify a novel concept of concentration-dependent, intermolecular “matrix trapping” as a conserved mechanism that controls the accumulation and thereby the functionality of thrombospondins in ECM.
Collapse
Affiliation(s)
- Dae Joong Kim
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195
| | | | - Douglas R Keene
- Micro-Imaging Center, Shriners Hospital for Children, Portland, OR 97239
| | - Marwah Hassan Milde
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195
| | - Josephine C Adams
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195 School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
25
|
Starlinger P, Haegele S, Wanek D, Zikeli S, Schauer D, Alidzanovic L, Fleischmann E, Gruenberger B, Gruenberger T, Brostjan C. Plasma thrombospondin 1 as a predictor of postoperative liver dysfunction. Br J Surg 2015; 102:826-36. [PMID: 25871570 DOI: 10.1002/bjs.9814] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/15/2015] [Accepted: 02/26/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Liver regeneration following liver resection involves a complex interplay of growth factors and their antagonists. Thrombospondin 1 has recently been identified as a critical inhibitor of liver regeneration by the activation of transforming growth factor β1 in mice, and preliminary data seem to confirm its relevance in humans. This study aimed to confirm these observations in an independent validation cohort. METHODS Perioperative circulating levels of thrombospondin 1 were measured in patients undergoing liver resection between January 2012 and September 2013. Postoperative liver dysfunction was defined according to the International Study Group of Liver Surgery and classification of morbidity was based on the criteria by Dindo et al. RESULTS In 85 patients (44 major and 41 minor liver resections), plasma levels of thrombospondin 1 increased 1 day after liver resection (mean 51·6 ng/ml before surgery and 68·3 ng/ml on postoperative day 1; P = 0·001). Circulating thrombospondin 1 concentration on the first postoperative day specifically predicted liver dysfunction (area under the receiver operating characteristic (ROC) curve 0·818, P = 0·003) and was confirmed as a significant predictor in multivariable analysis (Exp(B) 1·020, 95 per cent c.i. 1·005 to 1·035; P = 0·009). Patients with a high thrombospondin 1 concentration (over 80 ng/ml) on postoperative day 1 more frequently had postoperative liver dysfunction than those with a lower level (28 versus 2 per cent) and severe morbidity (44 versus 15 per cent), and their length of hospital stay was more than doubled (19·7 versus 9·9 days). CONCLUSION Thrombospondin 1 may prove a helpful clinical marker to predict postoperative liver dysfunction as early as postoperative day 1.
Collapse
Affiliation(s)
- P Starlinger
- Departments of Surgery, General Hospital, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Soto-Pantoja DR, Kaur S, Roberts DD. CD47 signaling pathways controlling cellular differentiation and responses to stress. Crit Rev Biochem Mol Biol 2015; 50:212-30. [PMID: 25708195 DOI: 10.3109/10409238.2015.1014024] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD47 is a widely expressed integral membrane protein that serves as the counter-receptor for the inhibitory phagocyte receptor signal-regulatory protein-α (SIRPα) and as a signaling receptor for the secreted matricellular protein thrombospondin-1. Recent studies employing mice and somatic cells lacking CD47 have revealed important pathophysiological functions of CD47 in cardiovascular homeostasis, immune regulation, resistance of cells and tissues to stress and chronic diseases of aging including cancer. With the emergence of experimental therapeutics targeting CD47, a more thorough understanding of CD47 signal transduction is essential. CD47 lacks a substantial cytoplasmic signaling domain, but several cytoplasmic binding partners have been identified, and lateral interactions of CD47 with other membrane receptors play important roles in mediating signaling resulting from the binding of thrombospondin-1. This review addresses recent advances in identifying the lateral binding partners, signal transduction pathways and downstream transcription networks regulated through CD47 in specific cell lineages. Major pathways regulated by CD47 signaling include calcium homeostasis, cyclic nucleotide signaling, nitric oxide and hydrogen sulfide biosynthesis and signaling and stem cell transcription factors. These pathways and other undefined proximal mediators of CD47 signaling regulate cell death and protective autophagy responses, mitochondrial biogenesis, cell adhesion and motility and stem cell self-renewal. Although thrombospondin-1 is the best characterized agonist of CD47, the potential roles of other members of the thrombospondin family, SIRPα and SIRPγ binding and homotypic CD47 interactions as agonists or antagonists of signaling through CD47 should also be considered.
Collapse
Affiliation(s)
- David R Soto-Pantoja
- a Laboratory of Pathology , Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | | | | |
Collapse
|
27
|
Leclair P, Lim CJ. CD47-independent effects mediated by the TSP-derived 4N1K peptide. PLoS One 2014; 9:e98358. [PMID: 24848268 PMCID: PMC4029904 DOI: 10.1371/journal.pone.0098358] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 05/01/2014] [Indexed: 11/18/2022] Open
Abstract
4N1K is a peptide fragment derived from the C-terminal, globular domain of thrombospondin which has been shown to mediate integrin-dependent cell adhesion and promote integrin activation acting via the cell-surface receptor, CD47. However, some studies found that 4N1K could act independently of CD47, putting in question the specificity of 4N1K for CD47. This led us to characterize the cellular and non-cellular effects of 4N1K. We found that 4N1K stimulated a potent increase in binding of a variety of non-specific IgG antibodies to cells in suspension. We also found that these same antibodies, as well as CD47-deficient cells, could bind substrate-immobilized 4N1K significantly better than a control peptide, 4NGG. Furthermore, we found that cells treated with 4N1K at higher concentrations inhibited, while lower concentrations promoted cell adhesion to immobilized fibronectin as an integrin substrate. Importantly, both the stimulatory and the inhibitory activity of 4N1K occurred as efficiently in the CD47-deficient JinB8 cells, as it did in the CD47-expressing parental or in JinB8 cells reconstituted with CD47 expression. Given these results, we suggest that 4N1K interacts non-specifically with epitopes commonly found on the cell surface, and conclude that it is not a suitable peptide for use to study the consequences of CD47 receptor ligation.
Collapse
Affiliation(s)
- Pascal Leclair
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cell and Developmental Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
- Child and Family Research Institute, BC Children's Hospital, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
28
|
Guerriero G. Putative chitin synthases from Branchiostoma floridae show extracellular matrix-related domains and mosaic structures. GENOMICS PROTEOMICS & BIOINFORMATICS 2012; 10:197-207. [PMID: 23084775 PMCID: PMC4937987 DOI: 10.1016/j.gpb.2012.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/27/2012] [Accepted: 04/23/2012] [Indexed: 01/04/2023]
Abstract
The transition from unicellular to multicellular life forms requires the development of a specialized structural component, the extracellular matrix (ECM). In Metazoans, there are two main supportive systems, which are based on chitin and collagen/hyaluronan, respectively. Chitin is the major constituent of fungal cell walls and arthropod exoskeleton. However, presence of chitin/chitooligosaccharides has been reported in lower chordates and during specific stages of vertebrate development. In this study, the occurrence of chitin synthases (CHSs) was investigated with a bioinformatics approach in the cephalochordate Branchiostoma floridae, in which the presence of chitin was initially reported in the skeletal rods of the pharyngeal gill basket. Twelve genes coding for proteins containing conserved amino acid residues of processive glycosyltransferases from GT2 family were found and 10 of them display mosaic structures with novel domains never reported previously in a chitin synthase. In particular, the presence of a discoidin (DS) and a sterile alpha motif (SAM) domain was found in nine identified proteins. Sequence analyses and homology modelling suggest that these domains might interact with the extracellular matrix and mediate protein–protein interaction. The multi-domain putative chitin synthases from B. floridae constitute an emblematic example of the explosion of domain innovation and shuffling which predate Metazoans.
Collapse
Affiliation(s)
- Gea Guerriero
- Fungal Genetics and Genomics Unit, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Science Vienna, University and Research Center Campus Tulln-Technopol, Tulln, Austria.
| |
Collapse
|
29
|
Sweetwyne MT, Murphy-Ullrich JE. Thrombospondin1 in tissue repair and fibrosis: TGF-β-dependent and independent mechanisms. Matrix Biol 2012; 31:178-86. [PMID: 22266026 DOI: 10.1016/j.matbio.2012.01.006] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 12/16/2022]
Abstract
Thrombospondin 1 (TSP1) plays major roles in both physiologic and pathologic tissue repair. TSP1 through its type 1 repeats is a known regulator of latent TGF-β activation and plays a role in wound healing and fibrosis. Binding of the TSP N-terminal domain to cell surface calreticulin in complex with LDL-receptor related protein 1 stimulates intermediate cell adhesion, cell migration, anoikis resistance, collagen expression and matrix deposition in an in vivo model of the foreign body response. There is also emerging evidence that TSP EGF-like repeats alter endothelial cell-cell interactions and stimulate epithelial migration through transactivation of EGF receptors. The mechanisms underlying these functions of TSP1 and the implications for physiologic and pathologic wound repair and fibrosis will be discussed.
Collapse
Affiliation(s)
- Mariya T Sweetwyne
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, United States
| | | |
Collapse
|
30
|
Sun Y, Ding W, Wei Q, Sheng WC. Disorder gene expression of extracellular matrix and adhesion molecules in saphenous vein conduits of diabetic patients. Interact Cardiovasc Thorac Surg 2011; 14:279-82. [PMID: 22171077 DOI: 10.1093/icvts/ivr053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Saphenous vein (SV) is the most commonly employed conduit in coronary surgery. However, the extracellular matrix (ECM) characteristics of SV in diabetic patients still remain unclear. This study was to survey the ECM gene expression profile of SV in diabetic patients. Thirty-five patients had type 2 diabetic mellitus; the non-diabetic (control) group comprised 49 patients. The expression profile of ECM genes was analysed by microarray. Tissue MMP/TIMP protein activities were evaluated by immunocytochemistry and western-blot. In this microarray, 25 genes demonstrated at least a 3-fold difference in expression. Upregulation was observed in 20 genes, while five genes appeared to be downregulated. SV exposed to DM conditions demonstrated a notable increase in MMP-2 and MMP-9 but a significant decrease in TIMP-2 and TIMP-3 in protein concentration compared with control group. This study suggests that native ECM gene expression profile of SV in diabetic patients has showed signs of the vein graft disease process before coronary surgery. Preoperative profiles of diabetic patients might provide some useful clues regarding vein graft quality and prompt adjustment in surgical strategy.
Collapse
Affiliation(s)
- Yongxin Sun
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
31
|
Abstract
Thrombospondins are evolutionarily conserved, calcium-binding glycoproteins that undergo transient or longer-term interactions with other extracellular matrix components. They share properties with other matrix molecules, cytokines, adaptor proteins, and chaperones, modulate the organization of collagen fibrils, and bind and localize an array of growth factors or proteases. At cell surfaces, interactions with an array of receptors activate cell-dependent signaling and phenotypic outcomes. Through these dynamic, pleiotropic, and context-dependent pathways, mammalian thrombospondins contribute to wound healing and angiogenesis, vessel wall biology, connective tissue organization, and synaptogenesis. We overview the domain organization and structure of thrombospondins, key features of their evolution, and their cell biology. We discuss their roles in vivo, associations with human disease, and ongoing translational applications. In many respects, we are only beginning to appreciate the important roles of these proteins in physiology and pathology.
Collapse
Affiliation(s)
- Josephine C Adams
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | | |
Collapse
|
32
|
Kaur S, Kuznetsova SA, Pendrak ML, Sipes JM, Romeo MJ, Li Z, Zhang L, Roberts DD. Heparan sulfate modification of the transmembrane receptor CD47 is necessary for inhibition of T cell receptor signaling by thrombospondin-1. J Biol Chem 2011; 286:14991-5002. [PMID: 21343308 DOI: 10.1074/jbc.m110.179663] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell surface proteoglycans on T cells contribute to retroviral infection, binding of chemokines and other proteins, and are necessary for some T cell responses to the matricellular glycoprotein thrombospondin-1. The major cell surface proteoglycans expressed by primary T cells and Jurkat T cells have an apparent M(r) > 200,000 and are modified with chondroitin sulfate and heparan sulfate chains. Thrombospondin-1 bound in a heparin-inhibitable manner to this proteoglycan and to a soluble form released into the medium. Based on mass spectrometry, knockdown, and immunochemical analyses, the proteoglycan contains two major core proteins as follows: amyloid precursor-like protein-2 (APLP2, apparent M(r) 230,000) and CD47 (apparent M(r) > 250,000). CD47 is a known thrombospondin-1 receptor but was not previously reported to be a proteoglycan. This proteoglycan isoform of CD47 is widely expressed on vascular cells. Mutagenesis identified glycosaminoglycan modification of CD47 at Ser(64) and Ser(79). Inhibition of T cell receptor signaling by thrombospondin-1 was lost in CD47-deficient T cells that express the proteoglycan isoform of APLP2, indicating that binding to APLP2 is not sufficient. Inhibition of CD69 induction was restored in CD47-deficient cells by re-expressing CD47 or an S79A mutant but not by the S64A mutant. Therefore, inhibition of T cell receptor signaling by thrombospondin-1 is mediated by CD47 and requires its modification at Ser(64).
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20982, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Starlinger P, Moll HP, Assinger A, Nemeth C, Hoetzenecker K, Gruenberger B, Gruenberger T, Kuehrer I, Schoppmann SF, Gnant M, Brostjan C. Thrombospondin-1: a unique marker to identify in vitro platelet activation when monitoring in vivo processes. J Thromb Haemost 2010; 8:1809-19. [PMID: 20492458 DOI: 10.1111/j.1538-7836.2010.03908.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Measuring platelet activation in patients has become a potent method to investigate pathophysiological processes. However, the commonly applied markers are sensitive to detrimental influences by in vitro platelet activation during blood analysis. OBJECTIVES Protein isoforms of platelet-derived thrombospondin-1 (TSP-1) were investigated for their potential to identify in vitro platelet activation when monitoring in vivo processes. METHODS TSP-1 was determined in plasma, serum or supernatant of purified platelets by ELISA and immunoblotting and was compared with standard markers of platelet activation. A collective of 20 healthy individuals and 30 cancer patients was analyzed. RESULTS While in vitro platelet degranulation led to a selective increase in the 200-kDa full-length molecule, an in vivo process involving platelet activation such as wound healing resulted in the predominant rise of the 140-kDa TSP-1 protein. The physiological ratio of circulating TSP-1 variants was determined and a cut-off level at 1.0 was defined to identify plasma samples with artificial in vitro platelet activation exceeding the cut-off level. In contrast, cancer patients known to frequently exhibit increased in vivo activation of platelets presented with a significantly decreased ratio of TSP-1 variants as compared with healthy volunteers. CONCLUSIONS In comparison to standard platelet markers, TSP-1 constitutes a sensitive and stable parameter suited to monitor in vitro platelet activation. The analysis of TSP-1 protein isoforms further offers a valuable tool to reliably discriminate between in vitro and in vivo effects, to exclude variability introduced during blood processing and improve clinical monitoring.
Collapse
Affiliation(s)
- P Starlinger
- Department of Surgery, General Hospital Institute of Physiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bentley AA, Adams JC. The evolution of thrombospondins and their ligand-binding activities. Mol Biol Evol 2010; 27:2187-97. [PMID: 20427418 DOI: 10.1093/molbev/msq107] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The extracellular matrix (ECM) is a complex, multiprotein network that has essential roles in tissue integrity and intercellular signaling in the metazoa. Thrombospondins (TSPs) are extracellular, calcium-binding glycoproteins that have biologically important roles in mammals in angiogenesis, vascular biology, connective tissues, immune response, and synaptogenesis. The evolution of these complex functional properties is poorly understood. We report here on the evolution of TSPs and their ligand-binding capacities, from comparative genomics of species representing the major phyla of metazoa and experimental analyses of the oligomerization properties of noncanonical TSPs of basal deuterostomes. Monomeric, dimeric, trimeric, and pentameric TSPs have arisen through separate evolutionary events involving gain, loss, or modification of a coiled-coil domain or distinct domains at the amino-terminus. The relative transience of monomeric forms under evolution implicates a biological importance for multivalency of the C-terminal region of TSPs. Most protostomes have a single TSP gene encoding a pentameric TSP. The pentameric form is also present in deuterostomes, and gene duplications at the origin of deuterostomes and gene loss and further gene duplication events in the vertebrate lineage gave rise to distinct forms and novel domain architectures. Parallel analysis of the major ligands of mammalian TSPs revealed that many binding activities are neofunctions representing either coevolutionary innovations in the deuterostome lineage or neofunctions of ancient molecules such as CD36. Contrasting widely conserved capacities include binding to heparan glycosaminoglycans, fibrillar collagen, or RGD-dependent integrins. These findings identify TSPs as fundamental components of the extracellular interaction systems of metazoa and thus impact understanding of the evolution of ECM networks. The widely conserved activities of TSPs in binding to ECM components or PS2 clade integrins will be relevant to use of TSPs in synthetic extracellular matrices or tissue engineering. In contrast, the neofunctions of vertebrate TSPs likely include interactions suitable for therapeutic targeting without general disruption of ECM.
Collapse
Affiliation(s)
- Amber A Bentley
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
35
|
Thrombospondin-1 as a Paradigm for the Development of Antiangiogenic Agents Endowed with Multiple Mechanisms of Action. Pharmaceuticals (Basel) 2010; 3:1241-1278. [PMID: 27713299 PMCID: PMC4034032 DOI: 10.3390/ph3041241] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 04/20/2010] [Accepted: 04/22/2010] [Indexed: 12/12/2022] Open
Abstract
Uncontrolled neovascularization occurs in several angiogenesis-dependent diseases, including cancer. Neovascularization is tightly controlled by the balance between angiogenic growth factors and antiangiogenic agents. The various natural angiogenesis inhibitors identified so far affect neovascularization by different mechanisms of action. Thrombospondin-1 (TSP-1) is a matricellular modular glycoprotein that acts as a powerful endogenous inhibitor of angiogenesis. It acts both indirectly, by sequestering angiogenic growth factors and effectors in the extracellular environment, and directly, by inducing an antiangiogenic program in endothelial cells following engagement of specific receptors including CD36, CD47, integrins and proteoglycans (all involved in angiogenesis ). In view of its central, multifaceted role in angiogenesis, TSP-1 has served as a source of antiangiogenic tools, including TSP-1 fragments, synthetic peptides and peptidomimetics, gene therapy strategies, and agents that up-regulate TSP-1 expression. This review discusses TSP-1-based inhibitors of angiogenesis, their mechanisms of action and therapeutic potential, drawing our experience with angiogenic growth factor-interacting TSP-1 peptides, and the possibility of exploiting them to design novel antiangiogenic agents.
Collapse
|
36
|
Rich RL, Myszka DG. Grading the commercial optical biosensor literature-Class of 2008: 'The Mighty Binders'. J Mol Recognit 2010; 23:1-64. [PMID: 20017116 DOI: 10.1002/jmr.1004] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Optical biosensor technology continues to be the method of choice for label-free, real-time interaction analysis. But when it comes to improving the quality of the biosensor literature, education should be fundamental. Of the 1413 articles published in 2008, less than 30% would pass the requirements for high-school chemistry. To teach by example, we spotlight 10 papers that illustrate how to implement the technology properly. Then we grade every paper published in 2008 on a scale from A to F and outline what features make a biosensor article fabulous, middling or abysmal. To help improve the quality of published data, we focus on a few experimental, analysis and presentation mistakes that are alarmingly common. With the literature as a guide, we want to ensure that no user is left behind.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
37
|
Tan K, Lawler J. The interaction of Thrombospondins with extracellular matrix proteins. J Cell Commun Signal 2009; 3:177-87. [PMID: 19830595 PMCID: PMC2778591 DOI: 10.1007/s12079-009-0074-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 09/30/2009] [Indexed: 02/06/2023] Open
Abstract
The thrombospondins (TSPs) are a family of five matricellular proteins that appear to function as adapter molecules to guide extracellular matrix synthesis and tissue remodeling in a variety of normal and disease settings. Various TSPs have been shown to bind to fibronectin, laminin, matrilins, collagens and other extracellular matrix (ECM) proteins. The importance of TSP-1 in this context is underscored by the fact that it is rapidly deposited at the sites of tissue damage by platelets. An association of TSPs with collagens has been known for over 25 years. The observation that the disruption of the TSP-2 gene in mice leads to collagen fibril abnormalities provided important in vivo evidence that these interactions are physiologically important. Recent biochemical studies have shown that TSP-5 promotes collagen fibril assembly and structural studies suggest that TSPs may interact with collagens through a highly conserved potential metal ion dependent adhesion site (MIDAS). These interactions are critical for normal tissue homeostasis, tumor progression and the etiology of skeletal dysplasias.
Collapse
Affiliation(s)
- Kemin Tan
- The Midwest Center for Structural Genomics and Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, IL USA
| | - Jack Lawler
- Division of Experimental Pathology, Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave., EC/CLS-503, Boston, MA 02215 USA
- Harvard Medical School, Boston, MA USA
| |
Collapse
|
38
|
Tan K, Duquette M, Joachimiak A, Lawler J. The crystal structure of the signature domain of cartilage oligomeric matrix protein: implications for collagen, glycosaminoglycan and integrin binding. FASEB J 2009; 23:2490-501. [PMID: 19276170 DOI: 10.1096/fj.08-128090] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cartilage oligomeric matrix protein (COMP), or thrombospondin-5 (TSP-5), is a secreted glycoprotein that is important for growth plate organization and function. Mutations in COMP cause two skeletal dysplasias, pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (EDM1). In this study, we determined the structure of a recombinant protein that contains the last epidermal growth factor repeat, the type 3 repeats and the C-terminal domain (CTD) of COMP to 3.15-A resolution limit by X-ray crystallography. The CTD is a beta-sandwich that is composed of 15 antiparallel beta-strands, and the type 3 repeats are a contiguous series of calcium binding sites that associate with the CTD at multiple points. The crystal packing reveals an exposed potential metal-ion-dependent adhesion site (MIDAS) on one edge of the beta-sandwich that is common to all TSPs and may serve as a binding site for collagens and other ligands. Disease-causing mutations in COMP disrupt calcium binding, disulfide bond formation, intramolecular interactions, or sites for potential ligand binding. The structure presented here and its unique molecular packing in the crystal identify potential interactive sites for glycosaminoglycans, integrins, and collagens, which are key to cartilage structure and function.
Collapse
Affiliation(s)
- Kemin Tan
- Midwest Center for Structural Genomics, Biosciences Division, Argonne National Laboratory, Argonne, Illinois, USA
| | | | | | | |
Collapse
|
39
|
Barclay AN. Signal regulatory protein alpha (SIRPalpha)/CD47 interaction and function. Curr Opin Immunol 2009; 21:47-52. [PMID: 19223164 PMCID: PMC3128989 DOI: 10.1016/j.coi.2009.01.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 01/20/2009] [Indexed: 12/12/2022]
Abstract
SIRPalpha is an inhibitory receptor present on myeloid cells that interacts with a widely distributed membrane protein CD47. The activating member SIRPbeta, despite extensive sequence similarity to SIRPalpha in the extracellular region, shows negligible binding to CD47. The SIRPalpha/CD47 interaction is unusual in that it can lead to bidirectional signalling through both SIRPalpha and CD47. This review concentrates on the interactions of SIRPalpha with CD47 where recent data have shed light on the structure of the proteins including determining why the activating SIRPbeta does not bind CD47, evidence of extensive polymorphisms and implication for the evolution and function of this protein and paired receptors in general. The interaction may be modified by endocytosis of the receptors, cleavage by proteolysis and through interactions of surfactant proteins.
Collapse
Affiliation(s)
- A Neil Barclay
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom.
| |
Collapse
|
40
|
Isenberg JS, Annis DS, Pendrak ML, Ptaszynska M, Frazier WA, Mosher DF, Roberts DD. Differential interactions of thrombospondin-1, -2, and -4 with CD47 and effects on cGMP signaling and ischemic injury responses. J Biol Chem 2008; 284:1116-25. [PMID: 19004835 DOI: 10.1074/jbc.m804860200] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Thrombospondin-1 regulates nitric oxide (NO) signaling in vascular cells via CD47. Because CD47 binding motifs are conserved in the C-terminal signature domains of all five thrombospondins and indirect evidence has implied CD47 interactions with other family members, we compared activities of recombinant signature domains of thrombospondin-1, -2, and -4 to interact with CD47 and modulate cGMP signaling. Signature domains of thrombospondin-2 and -4 were less active than that of thrombospondin-1 for inhibiting binding of radiolabeled signature domain of thrombospondin-1 or SIRPalpha (signal-regulatory protein) to cells expressing CD47. Consistent with this binding selectivity, the signature domain of thrombospondin-1 was more potent than those of thrombospondin-2 or -4 for inhibiting NO-stimulated cGMP synthesis in vascular smooth muscle cells and downstream effects on cell adhesion. In contrast to thrombospondin-1- and CD47-null cells, primary vascular cells from thrombospondin-2-null mice lack enhanced basal and NO-stimulated cGMP signaling. Effects of endogenous thrombospondin-2 on NO/cGMP signaling could be detected only in thrombospondin-1-null cells. Furthermore, tissue survival of ischemic injury and acute recovery of blood flow in thrombospondin-2-nulls resembles that of wild type mice. Therefore, thrombospondin-1 is the dominant regulator of NO/cGMP signaling via CD47, and its limiting role in acute ischemic injury responses is not shared by thrombospondin-2.
Collapse
Affiliation(s)
- Jeff S Isenberg
- Laboratory of Pathology, NCI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|