1
|
Zondervan RL, Capobianco CA, Jenkins DC, Reicha JD, Fredrick L, Lam C, Schmanski JT, Isenberg JS, Ahn J, Marcucio RS, Hankenson KD. CD47 is required for mesenchymal progenitor proliferation and fracture repair. Bone Res 2025; 13:29. [PMID: 40025005 PMCID: PMC11873311 DOI: 10.1038/s41413-025-00409-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 03/04/2025] Open
Abstract
CD47 is a ubiquitous and pleiotropic cell-surface receptor. Disrupting CD47 enhances injury repair in various tissues but the role of CD47 has not been studied in bone injuries. In a murine closed-fracture model, CD47-null mice showed decreased callus bone formation as assessed by microcomputed tomography 10 days post-fracture and increased fibrous volume as determined by histology. To understand the cellular basis for this phenotype, mesenchymal progenitors (MSC) were harvested from bone marrow. CD47-null MSC showed decreased large fibroblast colony formation (CFU-F), significantly less proliferation, and fewer cells in S-phase, although osteoblast differentiation was unaffected. However, consistent with prior research, CD47-null endothelial cells showed increased proliferation relative to WT cells. Similarly, in a murine ischemic fracture model, CD47-null mice showed reduced fracture callus size due to a reduction in bone relative to WT 15 days-post fracture. Consistent with our in vitro results, in vivo EdU labeling showed decreased cell proliferation in the callus of CD47-null mice, while staining for CD31 and endomucin demonstrated increased endothelial cell density. Finally, WT mice with ischemic fracture that were administered a CD47 morpholino, which blocks CD47 protein production, showed a callus phenotype similar to that of ischemic fractures in CD47-null mice, suggesting the phenotype was not due to developmental changes in the knockout mice. Thus, inhibition of CD47 during bone healing reduces both non-ischemic and ischemic fracture healing, in part, by decreasing MSC proliferation. Furthermore, the increase in endothelial cell proliferation and early blood vessel density caused by CD47 disruption is not sufficient to overcome MSC dysfunction.
Collapse
Affiliation(s)
- Robert L Zondervan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Christina A Capobianco
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Daniel C Jenkins
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - John D Reicha
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Livia Fredrick
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Charles Lam
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Jeanna T Schmanski
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jeffery S Isenberg
- Department of Diabetes Complications and Metabolism and Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Jaimo Ahn
- Department of Orthopaedics, Grady Memorial Hospital and Emory School of Medicine, Atlanta, GA, USA
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
3
|
Marin-Lopez A, Huck JD, Esterly AT, Azcutia V, Rosen C, Garcia-Milian R, Sefik E, Vidal-Pedrola G, Raduwan H, Chen TY, Arora G, Halene S, Shaw AC, Palm NW, Flavell RA, Parkos CA, Thangamani S, Ring AM, Fikrig E. The human CD47 checkpoint is targeted by an immunosuppressive Aedes aegypti salivary factor to enhance arboviral skin infectivity. Sci Immunol 2024; 9:eadk9872. [PMID: 39121194 PMCID: PMC11924945 DOI: 10.1126/sciimmunol.adk9872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/02/2024] [Accepted: 07/16/2024] [Indexed: 08/11/2024]
Abstract
The Aedes aegypti mosquito is a vector of many infectious agents, including flaviviruses such as Zika virus. Components of mosquito saliva have pleomorphic effects on the vertebrate host to enhance blood feeding, and these changes also create a favorable niche for pathogen replication and dissemination. Here, we demonstrate that human CD47, which is known to be involved in various immune processes, interacts with a 34-kilodalton mosquito salivary protein named Nest1. Nest1 is up-regulated in blood-fed female A. aegypti and facilitates Zika virus dissemination in human skin explants. Nest1 has a stronger affinity for CD47 than its natural ligand, signal regulatory protein α, competing for binding at the same interface. The interaction between Nest1 with CD47 suppresses phagocytosis by human macrophages and inhibits proinflammatory responses by white blood cells, thereby suppressing antiviral responses in the skin. This interaction elucidates how an arthropod protein alters the human response to promote arbovirus infectivity.
Collapse
Affiliation(s)
- Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John D Huck
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Allen T Esterly
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Connor Rosen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gemma Vidal-Pedrola
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Saravanan Thangamani
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Wilson ZS, Raya-Sandino A, Miranda J, Fan S, Brazil JC, Quiros M, Garcia-Hernandez V, Liu Q, Kim CH, Hankenson KD, Nusrat A, Parkos CA. Critical role of thrombospondin-1 in promoting intestinal mucosal wound repair. JCI Insight 2024; 9:e180608. [PMID: 39078701 PMCID: PMC11385097 DOI: 10.1172/jci.insight.180608] [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: 02/23/2024] [Accepted: 07/18/2024] [Indexed: 09/11/2024] Open
Abstract
Thrombospondin-1 (TSP1) is a matricellular protein associated with the regulation of cell migration through direct binding interactions with integrin proteins and by associating with other receptors known to regulate integrin function, including CD47 and CD36. We previously demonstrated that deletion of an epithelial TSP1 receptor, CD47, attenuates epithelial wound repair following intestinal mucosal injury. However, the mechanisms by which TSP1 contributes to intestinal mucosal repair remain poorly understood. Our results show upregulated TSP1 expression in colonic mucosal wounds and impaired intestinal mucosal wound healing in vivo upon intestinal epithelium-specific loss of TSP1 (VillinCre/+ Thbs1fl/fl or Thbs1ΔIEC mice). We report that exposure to exogenous TSP1 enhanced migration of intestinal epithelial cells in a CD47- and TGF-β1-dependent manner and that deficiency of TSP1 in primary murine colonic epithelial cells resulted in impaired wound healing. Mechanistically, TSP1 modulated epithelial actin cytoskeletal dynamics through suppression of RhoA activity, activation of Rho family small GTPase (Rac1), and changes in filamentous-actin bundling. Overall, TSP1 was found to regulate intestinal mucosal wound healing via CD47 and TGF-β1, coordinate integrin-containing cell-matrix adhesion dynamics, and remodel the actin cytoskeleton in migrating epithelial cells to enhance cell motility and promote wound repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingyang Liu
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Chang H. Kim
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
5
|
Moon S, Han S, Jang IH, Ryu J, Rha MS, Cho HJ, Yoon SS, Nam KT, Kim CH, Park MS, Seong JK, Lee WJ, Yoon JH, Chung YW, Ryu JH. Airway epithelial CD47 plays a critical role in inducing influenza virus-mediated bacterial super-infection. Nat Commun 2024; 15:3666. [PMID: 38693120 PMCID: PMC11063069 DOI: 10.1038/s41467-024-47963-5] [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: 02/01/2023] [Accepted: 04/16/2024] [Indexed: 05/03/2024] Open
Abstract
Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.
Collapse
Affiliation(s)
- Sungmin Moon
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seunghan Han
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - In-Hwan Jang
- National Creative Research Initiative Center for Hologenomics and School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaechan Ryu
- Microenvironment and Immunity Unit, Institut Pasteur, INSERM U1224, Paris, France
| | - Min-Seok Rha
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyung-Ju Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Airway Mucus Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Chang-Hoon Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Airway Mucus Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Vaccine Innovation Center, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea
- Laboratory of Developmental Biology and Genomics, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Won-Jae Lee
- National Creative Research Initiative Center for Hologenomics and School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Joo-Heon Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Airway Mucus Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Youn Wook Chung
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Airway Mucus Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
6
|
Lu YZ, Nayer B, Singh SK, Alshoubaki YK, Yuan E, Park AJ, Maruyama K, Akira S, Martino MM. CGRP sensory neurons promote tissue healing via neutrophils and macrophages. Nature 2024; 628:604-611. [PMID: 38538784 PMCID: PMC11023938 DOI: 10.1038/s41586-024-07237-y] [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: 08/04/2023] [Accepted: 02/26/2024] [Indexed: 04/06/2024]
Abstract
The immune system has a critical role in orchestrating tissue healing. As a result, regenerative strategies that control immune components have proved effective1,2. This is particularly relevant when immune dysregulation that results from conditions such as diabetes or advanced age impairs tissue healing following injury2,3. Nociceptive sensory neurons have a crucial role as immunoregulators and exert both protective and harmful effects depending on the context4-12. However, how neuro-immune interactions affect tissue repair and regeneration following acute injury is unclear. Here we show that ablation of the NaV1.8 nociceptor impairs skin wound repair and muscle regeneration after acute tissue injury. Nociceptor endings grow into injured skin and muscle tissues and signal to immune cells through the neuropeptide calcitonin gene-related peptide (CGRP) during the healing process. CGRP acts via receptor activity-modifying protein 1 (RAMP1) on neutrophils, monocytes and macrophages to inhibit recruitment, accelerate death, enhance efferocytosis and polarize macrophages towards a pro-repair phenotype. The effects of CGRP on neutrophils and macrophages are mediated via thrombospondin-1 release and its subsequent autocrine and/or paracrine effects. In mice without nociceptors and diabetic mice with peripheral neuropathies, delivery of an engineered version of CGRP accelerated wound healing and promoted muscle regeneration. Harnessing neuro-immune interactions has potential to treat non-healing tissues in which dysregulated neuro-immune interactions impair tissue healing.
Collapse
Affiliation(s)
- Yen-Zhen Lu
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Bhavana Nayer
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Shailendra Kumar Singh
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yasmin K Alshoubaki
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Elle Yuan
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Anthony J Park
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Kenta Maruyama
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Pharmacology, School of Medicine, Aichi Medical University, Aichi, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia.
- Laboratory of Host Defense, World Premier International Research Center, Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Hankenson K, Zondervan R, Capobianco C, Jenkins D, Reicha J, Frederick L, Lam C, Isenberg J, Ahn J, Marcucio RS. CD47 is Required for Mesenchymal Progenitor Proliferation and Fracture Repair. RESEARCH SQUARE 2024:rs.3.rs-4022423. [PMID: 38562718 PMCID: PMC10984034 DOI: 10.21203/rs.3.rs-4022423/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
CD47 is a ubiquitous and pleiotropic cell-surface receptor. Disrupting CD47 enhances injury repair in various tissues but the role of CD47 has not been studied in bone injuries. In a murine closed-fracture model, CD47-null mice showed decreased callus bone volume, bone mineral content, and tissue mineral content as assessed by microcomputed tomography 10 days post-fracture, and increased fibrous volume as determined by histology. To understand the cellular basis for this phenotype, mesenchymal progenitors (MSC) were harvested from bone marrow. CD47-null MSC showed decreased large fibroblast colony formation (CFU-F), significantly less proliferation, and fewer cells in S-phase, although osteoblast differentiation was unaffected. However, consistent with prior research, CD47-null endothelial cells showed increased proliferation relative to WT cells. Similarly, in a murine ischemic fracture model, CD47-null mice showed reduced fracture callus bone volume and bone mineral content relative to WT. Consistent with our In vitro results, in vivo EdU labeling showed decreased cell proliferation in the callus of CD47-null mice, while staining for CD31 and endomucin demonstrated increased endothelial cell mass. Finally, WT mice administered a CD47 morpholino, which blocks CD47 protein production, showed a callus phenotype similar to that of non-ischemic and ischemic fractures in CD47-null mice, suggesting the phenotype was not due to developmental changes in the knockout mice. Thus, inhibition of CD47 during bone healing reduces both non-ischemic and ischemic fracture healing, in part, by decreasing MSC proliferation. Furthermore, the increase in endothelial cell proliferation and early blood vessel density caused by CD47 disruption is not sufficient to overcome MSC dysfunction.
Collapse
|
8
|
Zondervan RL, Capobianco CA, Jenkins DC, Reicha JD, Fredrick LM, Lam C, Isenberg JS, Ahn J, Marcucio RS, Hankenson KD. CD47 is Required for Mesenchymal Progenitor Proliferation and Fracture Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583756. [PMID: 38496546 PMCID: PMC10942414 DOI: 10.1101/2024.03.06.583756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
CD47 is a ubiquitous and pleiotropic cell-surface receptor. Disrupting CD47 enhances injury repair in various tissues but the role of CD47 has not been studied in bone injuries. In a murine closed-fracture model, CD47-null mice showed decreased callus bone volume, bone mineral content, and tissue mineral content as assessed by microcomputed tomography 10 days post-fracture, and increased fibrous volume as determined by histology. To understand the cellular basis for this phenotype, mesenchymal progenitors (MSC) were harvested from bone marrow. CD47-null MSC showed decreased large fibroblast colony formation (CFU-F), significantly less proliferation, and fewer cells in S-phase, although osteoblast differentiation was unaffected. However, consistent with prior research, CD47-null endothelial cells showed increased proliferation relative to WT cells. Similarly, in a murine ischemic fracture model, CD47-null mice showed reduced fracture callus bone volume and bone mineral content relative to WT. Consistent with our in vitro results, in vivo EdU labeling showed decreased cell proliferation in the callus of CD47-null mice, while staining for CD31 and endomucin demonstrated increased endothelial cell mass. Finally, WT mice administered a CD47 morpholino, which blocks CD47 protein production, showed a callus phenotype similar to that of non-ischemic and ischemic fractures in CD47-null mice, suggesting the phenotype was not due to developmental changes in the knockout mice. Thus, inhibition of CD47 during bone healing reduces both non-ischemic and ischemic fracture healing, in part, by decreasing MSC proliferation. Furthermore, the increase in endothelial cell proliferation and early blood vessel density caused by CD47 disruption is not sufficient to overcome MSC dysfunction.
Collapse
Affiliation(s)
- Robert L. Zondervan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States, 48824
| | - Christina A. Capobianco
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor Michigan, United States, 48109
| | - Daniel C. Jenkins
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
| | - John D. Reicha
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
| | - Livia M. Fredrick
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
| | - Charles Lam
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California, United States, 94142
| | - Jeffery S. Isenberg
- Department of Diabetes Complications and Metabolism and Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States, 91010
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California, United States, 94142
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, United States, 48109
| |
Collapse
|
9
|
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
|
10
|
Erdem N, Chen KT, Qi M, Zhao Y, Wu X, Garcia I, Ku HT, Montero E, Al-Abdullah IH, Kandeel F, Roep BO, Isenberg JS. Thrombospondin-1, CD47, and SIRPα display cell-specific molecular signatures in human islets and pancreata. Am J Physiol Endocrinol Metab 2023; 324:E347-E357. [PMID: 36791324 PMCID: PMC11967708 DOI: 10.1152/ajpendo.00221.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Thrombospondin-1 (TSP1) is a secreted protein minimally expressed in health but increased in disease and age. TSP1 binds to the cell membrane receptor CD47, which itself engages signal regulatory protein α (SIRPα), and the latter creates a checkpoint for immune activation. Individuals with cancer administered checkpoint-blocking molecules developed insulin-dependent diabetes. Relevant to this, CD47 blocking antibodies and SIRPα fusion proteins are in clinical trials. We characterized the molecular signature of TSP1, CD47, and SIRPα in human islets and pancreata. Fresh islets and pancreatic tissue from nondiabetic individuals were obtained. The expression of THBS1, CD47, and SIRPA was determined using single-cell mRNA sequencing, immunofluorescence microscopy, Western blot, and flow cytometry. Islets were exposed to diabetes-affiliated inflammatory cytokines and changes in protein expression were determined. CD47 mRNA was expressed in all islet cell types. THBS1 mRNA was restricted primarily to endothelial and mesenchymal cells, whereas SIRPA mRNA was found mostly in macrophages. Immunofluorescence staining showed CD47 protein expressed by β cells and present in the exocrine pancreas. TSP1 and SIRPα proteins were not seen in islets or the exocrine pancreas. Western blot and flow cytometry confirmed immunofluorescent expression patterns. Importantly, human islets produced substantial quantities of secreted TSP1. Human pancreatic exocrine and endocrine tissue expressed CD47, whereas fresh islets displayed cell surface CD47 and secreted TSP1 at baseline and in inflammation. These findings suggest unexpected effects on islets from agents that intersect TSP1-CD47-SIRPα.NEW & NOTEWORTHY CD47 is a cell surface receptor with two primary ligands, soluble thrombospondin-1 (TSP1) and cell surface signal regulatory protein alpha (SIRPα). Both interactions provide checkpoints for immune cell activity. We determined that fresh human islets display CD47 and secrete TSP1. However, human islet endocrine cells lack SIRPα. These gene signatures are likely important given the increasing use of CD47 and SIRPα blocking molecules in individuals with cancer.
Collapse
Affiliation(s)
- Neslihan Erdem
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Kuan-Tsen Chen
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Meirigeng Qi
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Isaac Garcia
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Hsun Teresa Ku
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Enrique Montero
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Ismail H Al-Abdullah
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Bart O Roep
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Jeffrey S Isenberg
- Department of Diabetes Complications & Metabolism, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| |
Collapse
|
11
|
Wang JM, Li H, Xu L, Kim H, Qiu Y, Zhang K. Boosting UPR transcriptional activator XBP1 accelerates acute wound healing. PNAS NEXUS 2023; 2:pgad050. [PMID: 36959909 PMCID: PMC10028334 DOI: 10.1093/pnasnexus/pgad050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
Patients' suffering from large or deep wounds caused by traumatic and/or thermal injuries have significantly lower chances of recapitulating lost skin function through natural healing. We tested whether enhanced unfolded protein response (UPR) by expression of a UPR transcriptional activator, X-box-binding protein 1 (XBP1) can significantly promote wound repair through stimulating growth factor production and promoting angiogenesis. In mouse models of a second-degree thermal wound, a full-thickness traumatic wound, and a full-thickness diabetic wound, the topical gene transfer of the activated form of XBP1 (spliced XBP1, XBP1s) can significantly enhance re-epithelialization and increase angiogenesis, leading to rapid, nearly complete wound closure with intact regenerated epidermis and dermis. Overexpression of XBP1s stimulated the transcription of growth factors in fibroblasts critical to proliferation and remodeling during wound repair, including platelet-derived growth factor BB, basic fibroblast growth factor, and transforming growth factor beta 3. Meanwhile, the overexpression of XBP1s boosted the migration and tube formation of dermal microvascular endothelial cells in vitro. Our functional and mechanistic investigations of XBP1-mediated regulation of wound healing processes provide novel insights into the previously undermined physiological role of the UPR in skin injuries. The finding opens an avenue to developing potential XBP1-based therapeutic strategies in clinical wound care protocols.
Collapse
Affiliation(s)
- Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, 540 Canfield Street, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, 540 Canfield Street, Detroit, MI 48201, USA
| | - Hainan Li
- Center for Molecular Medicine and Genetics, Wayne State University, 540 Canfield Street, Detroit, MI 48201, USA
| | - Liping Xu
- Center for Molecular Medicine and Genetics, Wayne State University, 540 Canfield Street, Detroit, MI 48201, USA
| | - Hyunbae Kim
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201, USA
| | - Yining Qiu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201, USA
| | - Kezhong Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, 540 Canfield Street, Detroit, MI 48201, USA
- Karmanos Cancer Institute, 4100 John R, Detroit, MI 48201, USA
| |
Collapse
|
12
|
Du Y, Liu X, Du K, Zhang W, Li R, Yang L, Cheng L, He W, Zhang W. Decorin inhibits the formation of hard nodules after microwave ablation by inhibiting the TGF-β1/SMAD and MAPK signaling pathways: in a Bama miniature pig model of mammary gland hyperplasia. Int J Hyperthermia 2023; 40:2188151. [PMID: 36919520 DOI: 10.1080/02656736.2023.2188151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Benign breast lesions are often associated with hard nodule formation after microwave ablation (MWA), which persists for a long time and causes problems in patients. The aim of this study was to evaluate the efficacy of decorin in the treatment of hard nodule formation and its potential mechanism of action. METHODS Using a Bama miniature pig model of mammary gland hyperplasia, immunohistochemistry, Masson's trichrome and western blotting were firstly applied to compare the extent of fibrosis and activation of key members of the TGF-β1/SMAD and MAPK signaling pathways of hard nodule in the control and MWA groups, and then the extent of fibrosis and expression of signaling pathways in hard nodule were examined after application of decorin. RESULTS The results showed that the MWA group had increased levels of TGF-β1, p-SMAD2/3, p-ERK1/2, and collagen I proteins and increased fibrosis at 2 weeks, 4 weeks, and 3 months after MWA. After decorin treatment, the expression levels of each protein were significantly downregulated, and the degree of fibrosis was reduced at 2 weeks, 4 weeks, and 3 months after MWA compared with the MWA group. CONCLUSION In conclusion, these results suggest that activation of TGF-β1 may play an important role in hard nodule formation and that decorin may reduce hard nodule formation after MWA in a model of mammary gland hyperplasia by inhibiting the TGF-β1/SMAD and MAPK signaling pathways.
Collapse
Affiliation(s)
- Yue Du
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xinyao Liu
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kai Du
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenkai Zhang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rui Li
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lizhi Yang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Linggang Cheng
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen He
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
14
|
Park JM, Han YM, Hwang SJ, Kim SJ, Hahm KB. Therapeutic effects of placenta derived-, umbilical cord derived-, and adipose tissue derived-mesenchymal stem cells in chronic Helicobacter pylori infection: comparison and novel mechanisms. J Clin Biochem Nutr 2021; 69:188-202. [PMID: 34616110 PMCID: PMC8482378 DOI: 10.3164/jcbn.20-151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022] Open
Abstract
Supported with significant rejuvenating and regenerating actions of mesenchymal stem cells (MSCs) in various gastrointestinal diseases including Helicobacter pylori (H. pylori)-associated gastric diseases, we have compared these actions among placenta derived-MSCs (PD-MSCs), umbilical cord derived-MSCs (UC-MSCs), and adipose tissue derived-MSCs (AD-MSCs) and explored contributing genes implicated in rejuvenation of H. pylori-chronic atrophic gastritis (CAG) and tumorigenesis. In this study adopting H. pylori-initiated, high salt diet-promoted gastric carcinogenesis model, we have administered three kinds of MSCs around 15-18 weeks in H. pylori infected C57BL/6 mice and sacrificed at 24 and 48 weeks, respectively, in order to either assess the rejuvenating capability or anti-tumorigenesis. At 24 weeks, MSCs all led to significantly mitigated atrophic gastritis, for which significant inductions of autophagy, preservation of tumor suppressive 15-PGDH, attenuated apoptosis, and efficient efferocytosis was imposed with MSCs administration during atrophic gastritis. At 48 weeks, MSCs administered during H. pylori-associated atrophic gastritis afforded significant blocking the progression of CAG, as evidenced with statistically significant reduction in H. pylori-associated gastric tumor (p<0.05) accompanied with significant decreases in IL-1β, COX-2, STAT3, and NF-κB. Combined together with the changes of stanniocalcin-1 (STC-1), thrombospondin-1 (TSP-1), and IL-10 known as biomarkers reflecting stem cell activities at 48 weeks after H. pylori, PD-MSCs among MSCs afforded the best rejuvenating action against H. pylori-associated CAG via additional actions of efferocytosis, autophagy, and anti-apoptosis at 24 weeks. In conclusion, MSCs, especially PD-MSCs, exerted rejuvenating actions against H. pylori-associated CAG via anti-mutagenesis of IL-10, CD-36, ATG5 and cancer suppressive influences of STC-1, TSP-1, and 15-PGDH.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daehak-ro 62, Dong-gu, Daejeon 34520, Korea
| | - Young Min Han
- Western Seoul Center, Korea Basic Science Institute, University-Industry Cooperate Building, 150 Bugahyeon-ro, Seodaemun-gu, Seoul 03759, Korea
| | - Sun Jin Hwang
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
| | - Seong Jin Kim
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea
| | - Ki Baik Hahm
- Medpacto Research Institute, Medpacto, Myungdal-ro 92, Seocho-gu, Seoul 06668, Korea.,CHA Cancer Preventive Research Center, CHA Bio Complex, CHA University, 330 Pangyo-dong, Bundang-gu, Seongnam 13497, Korea
| |
Collapse
|
15
|
Rada M, Kapelanski-Lamoureux A, Petrillo S, Tabariès S, Siegel P, Reynolds AR, Lazaris A, Metrakos P. Runt related transcription factor-1 plays a central role in vessel co-option of colorectal cancer liver metastases. Commun Biol 2021; 4:950. [PMID: 34376784 PMCID: PMC8355374 DOI: 10.1038/s42003-021-02481-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 06/17/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer liver metastasis (CRCLM) has two major histopathological growth patterns: angiogenic desmoplastic and non-angiogenic replacement. The replacement lesions obtain their blood supply through vessel co-option, wherein the cancer cells hijack pre-existing blood vessels of the surrounding liver tissue. Consequentially, anti-angiogenic therapies are less efficacious in CRCLM patients with replacement lesions. However, the mechanisms which drive vessel co-option in the replacement lesions are unknown. Here, we show that Runt Related Transcription Factor-1 (RUNX1) overexpression in the cancer cells of the replacement lesions drives cancer cell motility via ARP2/3 to achieve vessel co-option. Furthermore, overexpression of RUNX1 in the cancer cells is mediated by Transforming Growth Factor Beta-1 (TGFβ1) and thrombospondin 1 (TSP1). Importantly, RUNX1 knockdown impaired the metastatic capability of colorectal cancer cells in vivo and induced the development of angiogenic lesions in liver. Our results confirm that RUNX1 may be a potential target to overcome vessel co-option in CRCLM.
Collapse
Affiliation(s)
- Miran Rada
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | | | - Stephanie Petrillo
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Sébastien Tabariès
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Peter Siegel
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | | | - Anthoula Lazaris
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Peter Metrakos
- Cancer Research Program, McGill University Health Centre Research Institute, Montreal, QC, Canada.
| |
Collapse
|
16
|
Roberts DD, Isenberg JS. CD47 and thrombospondin-1 regulation of mitochondria, metabolism, and diabetes. Am J Physiol Cell Physiol 2021; 321:C201-C213. [PMID: 34106789 DOI: 10.1152/ajpcell.00175.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thrombospondin-1 (TSP1) is the prototypical member of a family of secreted proteins that modulate cell behavior by engaging with molecules in the extracellular matrix and with receptors on the cell surface. CD47 is widely displayed on many, if not all, cell types and is a high-affinity TSP1 receptor. CD47 is a marker of self that limits innate immune cell activities, a feature recently exploited to enhance cancer immunotherapy. Another major role for CD47 in health and disease is to mediate TSP1 signaling. TSP1 acting through CD47 contributes to mitochondrial, metabolic, and endocrine dysfunction. Studies in animal models found that elevated TSP1 expression, acting in part through CD47, causes mitochondrial and metabolic dysfunction. Clinical studies established that abnormal TSP1 expression positively correlates with obesity, fatty liver disease, and diabetes. The unabated increase in these conditions worldwide and the availability of CD47 targeting drugs justify a closer look into how TSP1 and CD47 disrupt metabolic balance and the potential for therapeutic intervention.
Collapse
Affiliation(s)
- David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
17
|
Kaur S, Isenberg JS, Roberts DD. CD47 (Cluster of Differentiation 47). ATLAS OF GENETICS AND CYTOGENETICS IN ONCOLOGY AND HAEMATOLOGY 2021; 25:83-102. [PMID: 34707698 PMCID: PMC8547767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
CD47, also known as integrin-associated protein, is a constitutively and ubiquitously expressed transmembrane receptor. CD47 is conserved across amniotes including mammals, reptiles, and birds. Expression is increased in many cancers and, in non-malignant cells, by stress and with aging. The up-regulation of CD47 expression is generally epigenetic, whereas gene amplification occurs with low frequency in some cancers. CD47 is a high affinity signaling receptor for the secreted protein thrombospondin-1 (THBS1) and the counter-receptor for signal regulatory protein-α (SIRPA, SIRPα) and SIRPγ (SIRPG). CD47 interaction with SIRPα serves as a marker of self to innate immune cells and thereby protects cancer cells from phagocytic clearance. Consequently, higher CD47 correlates with a poor prognosis in some cancers, and therapeutic blockade can suppress tumor growth by enhancing innate antitumor immunity. CD47 expressed on cytotoxic T cells, dendritic cells, and NK cells mediates inhibitory THBS1 signaling that further limits antitumor immunity. CD47 laterally associates with several integrins and thereby regulates cell adhesion and migration. CD47 has additional lateral binding partners in specific cell types, and ligation of CD47 in some cases modulates their function. THBS1-CD47 signaling in non-malignant cells inhibits nitric oxide/cGMP, calcium, and VEGF signaling, mitochondrial homeostasis, stem cell maintenance, protective autophagy, and DNA damage response, and promotes NADPH oxidase activity. CD47 signaling is a physiological regulator of platelet activation, angiogenesis and blood flow. THBS1/CD47 signaling is frequently dysregulated in chronic diseases.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892, USA
| | | | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
18
|
Julovi SM, Sanganeria B, Minhas N, Ghimire K, Nankivell B, Rogers NM. Blocking thrombospondin-1 signaling via CD47 mitigates renal interstitial fibrosis. J Transl Med 2020; 100:1184-1196. [PMID: 32366943 DOI: 10.1038/s41374-020-0434-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 03/31/2020] [Accepted: 04/11/2020] [Indexed: 11/10/2022] Open
Abstract
Acute kidney injury triggers a complex cascade of molecular responses that can culminate in maladaptive repair and fibrosis. We have previously reported that the matrix protein thrombospondin-1 (TSP1), binding its high affinity its receptor CD47, promotes acute kidney injury. However, the role of this pathway in promoting fibrosis is less clear. Hypothesizing that limiting TSP1-CD47 signaling is protective against fibrosis, we interrogated this pathway in a mouse model of chronic ischemic kidney injury. Plasma and renal parenchymal expression of TSP1 in patients with chronic kidney disease was also assessed. We found that CD47-/- mice or wild-type mice treated with a CD47 blocking antibody showed clear amelioration of fibrotic histological changes compared to control animals. Wild-type mice showed upregulated TSP1 and pro-fibrotic markers which were significantly abrogated in CD47-/- and antibody-treated cohorts. Renal tubular epithelial cells isolated from WT mice showed robust upregulation of pro-fibrotic markers following hypoxic stress or exogenous TSP1, which was mitigated in CD47-/- cells. Patient sera showed a proportionate correlation between TSP1 levels and worsening glomerular filtration rate. Immunohistochemistry of human kidney tissue demonstrated tubular and glomerular matrix localization of TSP1 expression in patients with CKD. These data suggest that renal tubular epithelial cells contribute to fibrosis by activating TSP1-CD47 signaling, and point to CD47 as a potential target to limit fibrosis following ischemic injury.
Collapse
Affiliation(s)
- Sohel M Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Barkha Sanganeria
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Nikita Minhas
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia
| | - Brian Nankivell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia.,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia.,Renal Division, Westmead Hospital, Camperdown, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Camperdown, NSW, Australia. .,Westmead Clinical Medical School, University of Sydney, Camperdown, NSW, Australia. .,Renal Division, Westmead Hospital, Camperdown, NSW, Australia. .,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
19
|
Vinaik R, Barayan D, Jeschke MG. NLRP3 Inflammasome in Inflammation and Metabolism: Identifying Novel Roles in Postburn Adipose Dysfunction. Endocrinology 2020; 161:5868467. [PMID: 32790834 PMCID: PMC7426001 DOI: 10.1210/endocr/bqaa116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Inflammasomes are multiprotein complexes that respond to pathogen or host associated damage markers, leading to caspase-1 maturation and processing of pro-inflammatory cytokines. Initially, inflammasomes were implicated primarily in inflammatory and infectious conditions. However, increasing evidence demonstrates broader roles beyond inflammation, including regulation of adipose tissue metabolism after burns. Here, we conducted a search for articles on PubMed, Web of Science, Embase, Scopus, and UpToDate with applied search strategies including a combination of "burns," "trauma," "(NLRP3) inflammasome," "metabolic conditions," "white adipose tissue," "macrophages," "browning," and "lipolysis" and included papers from 2000 to 2020. We discuss unexpected roles for NLRP3, the most characterized inflammasome to date, as a key metabolic driver in a variety of conditions. In particular, we highlight the function of NLRP3 inflammasome in burn trauma, which is characterized by both hyperinflammation and hypermetabolism. We identify a critical part for NLRP3 activation in macrophage dynamics and delineate a novel role in postburn white adipose tissue remodeling, a pathological response associated with hypermetabolism and poor clinical outcomes. Mechanistically, how inflammation and inflammasome activation is linked to postburn hypermetabolism is a novel concept to contemplate, and herein we provide evidence of an immunometabolic crosstalk between adipocytes and infiltrating macrophages.
Collapse
Affiliation(s)
| | | | - Marc G Jeschke
- Department of Surgery, Division of Plastic Surgery, University of Toronto, Canada
- Department of Immunology, University of Toronto, Canada
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
- Correspondence: Marc G. Jeschke, MD, PhD, Director Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre; Division of Plastic Surgery, Department of Surgery, Department of Immunology, University of Toronto; Sunnybrook Research Institute, 2075 Bayview Ave., Rm. D704, Toronto, ON, CANADA, M4N 3M5. E-mail:
| |
Collapse
|
20
|
Duan FM, Fu LJ, Wang YH, Adu-Gyamfi EA, Ruan LL, Xu ZW, Xiao SQ, Chen XM, Wang YX, Liu TH, Ding YB. THBS1 regulates trophoblast fusion through a CD36-dependent inhibition of cAMP, and its upregulation participates in preeclampsia. Genes Dis 2020; 8:353-363. [PMID: 33997182 PMCID: PMC8093648 DOI: 10.1016/j.gendis.2020.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/11/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Preeclampsia is a pregnancy complication which threatens the survival of mothers and fetuses. It originates from abnormal placentation, especially insufficient fusion of the cytotrophoblast cells to form the syncytiotrophoblast. In this study, we found that THBS1, a matricellular protein that mediates cell-to-cell and cell-to-matrix interactions, is downregulated during the fusion of primary cytotrophoblast and BeWo cells, but upregulated in the placenta of pregnancies complicated by preeclampsia. Also, THBS1 was observed to interact with CD36, a membrane signal receptor and activator of the cAMP signaling pathway, to regulate the fusion of cytotrophoblast cells. Overexpression of THBS1 inhibited the cAMP signaling pathway and reduced the BeWo cells fusion ratio, while the effects of THBS1 were abolished by a CD36-blocking antibody. Our results suggest that THBS1 signals through a CD36-mediated cAMP pathway to regulate syncytialization of the cytotrophoblast cells, and that its upregulation impairs placental formation to cause preeclampsia. Thus, THBS1 can serve as a therapeutic target regarding the mitigation of abnormal syncytialization and preeclampsia.
Collapse
Affiliation(s)
- Fu-Mei Duan
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China
| | - Li-Juan Fu
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yong-Heng Wang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Enoch Appiah Adu-Gyamfi
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Ling-Ling Ruan
- Department of Herbal Medicine, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, PR China
| | - Zeng-Wei Xu
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China
| | - Shi-Quan Xiao
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China.,Department of Reproductive Medicine, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 401120, PR China
| | - Xue-Mei Chen
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Ying-Xiong Wang
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Tai-Hang Liu
- Department of Bioinformatics, The School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| | - Yu-Bin Ding
- School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China.,The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, Chongqing, 400016, PR China
| |
Collapse
|
21
|
Liu Y, Li M, Wei C, Tang L, Sheng Y, Liu Y, Li D, Ding D, Qiu J, Zhu X. TSP1-CD47-SIRPα signaling facilitates the development of endometriosis by mediating the survival of ectopic endometrium. Am J Reprod Immunol 2020; 83:e13236. [PMID: 32196807 DOI: 10.1111/aji.13236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/28/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
PROBLEM To explore whether the thrombospondin-1(TSP1)-CD47-signal regulatory protein alpha (SIRPα) signaling pathway has impacts on the development of endometriosis. METHOD OF STUDY Endometrial stromal cells (ESCs) originated from ectopic and eutopic endometrial tissues with or without endometriosis. Monocytes (Macrophages) were isolated from peripheral blood and peritoneal fluids with or without endometriosis. The expression levels of molecules were investigated by flow cytometry (FCM), immunohistochemistry (IHC), and RT-qPCR. The concentration of TSP1 was assessed via ELISA. The capacities of angiogenesis and phagocytosis were measured via tube formation assay and phagocytic assay, respectively. RESULTS We confirmed the up-regulation of critical molecules within the pathway in endometriosis patients. TSP1 can encourage normal ESCs (NESCs) growth and fibrosis. It simultaneously promotes the secretion of inflammatory factors and inhibits the phagocytic abilities of macrophages. Moreover, the proliferation of vascular endothelial cells (VECs) may be improved by TSP1. These effects may be offset by CD47 blocking antibodies. In addition, ectopic ESCs (EESCs) directly improve SIRPα expression on macrophages, which may further exhaust their phagocytic ability. Phagocytosis efficiency of macrophages on EESCs significantly improves by blocking CD47-SIRPα pathway. CONCLUSION TSP1-CD47-SIRPα signaling pathway not only improves the viability of NESCs per se but also promotes their survival circumstances by affecting the function of macrophages and VECs, which are mutually reinforcing and jointly promote the development of endometriosis.
Collapse
Affiliation(s)
- Yukai Liu
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Chunyan Wei
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingli Tang
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanran Sheng
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuyin Liu
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding Ding
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianping Qiu
- Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xiaoyong Zhu
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
22
|
Sorkin M, Huber AK, Hwang C, Carson WF, Menon R, Li J, Vasquez K, Pagani C, Patel N, Li S, Visser ND, Niknafs Y, Loder S, Scola M, Nycz D, Gallagher K, McCauley LK, Xu J, James AW, Agarwal S, Kunkel S, Mishina Y, Levi B. Regulation of heterotopic ossification by monocytes in a mouse model of aberrant wound healing. Nat Commun 2020; 11:722. [PMID: 32024825 PMCID: PMC7002453 DOI: 10.1038/s41467-019-14172-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 12/13/2019] [Indexed: 11/08/2022] Open
Abstract
Heterotopic ossification (HO) is an aberrant regenerative process with ectopic bone induction in response to musculoskeletal trauma, in which mesenchymal stem cells (MSC) differentiate into osteochondrogenic cells instead of myocytes or tenocytes. Despite frequent cases of hospitalized musculoskeletal trauma, the inflammatory responses and cell population dynamics that regulate subsequent wound healing and tissue regeneration are still unclear. Here we examine, using a mouse model of trauma-induced HO, the local microenvironment of the initial post-injury inflammatory response. Single cell transcriptome analyses identify distinct monocyte/macrophage populations at the injury site, with their dynamic changes over time elucidated using trajectory analyses. Mechanistically, transforming growth factor beta-1 (TGFβ1)-producing monocytes/macrophages are associated with HO and aberrant chondrogenic progenitor cell differentiation, while CD47-activating peptides that reduce systemic macrophage TGFβ levels and help ameliorate HO. Our data thus implicate CD47 activation as a therapeutic approach for modulating monocyte/macrophage phenotypes, MSC differentiation and HO formation during wound healing.
Collapse
Affiliation(s)
- Michael Sorkin
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Amanda K Huber
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Charles Hwang
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - William F Carson
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - John Li
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaetlin Vasquez
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chase Pagani
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nicole Patel
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shuli Li
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Noelle D Visser
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yashar Niknafs
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shawn Loder
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Melissa Scola
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dylan Nycz
- Division of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Katherine Gallagher
- Division of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Shailesh Agarwal
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yuji Mishina
- Department of Biologic and Material Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin Levi
- Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Choi MR, Han JS, Chai YG, Jin YB, Lee SR, Kim DJ. Gene expression profiling in the hippocampus of adolescent rats after chronic alcohol administration. Basic Clin Pharmacol Toxicol 2019; 126:389-398. [PMID: 31628824 DOI: 10.1111/bcpt.13342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/15/2019] [Indexed: 11/29/2022]
Abstract
In South Korea, the average age of onset of alcohol drinking is 13.3 years and half of adolescents drink alcohol more than once a month; 8.45% of the Korean adolescent population become future high-risk alcohol drinkers. Chronic alcohol abuse causes physical and psychiatric health problems such as alcohol addiction, liver disease, stroke and cognitive impairments. This study aimed to investigate the effect of alcohol on gene expression and their function in the hippocampus of adolescent rats. After chronic alcohol administration in male (control, n = 6; alcohol, n = 6) Sprague-Dawley rats for 6 weeks, we analysed up- or down-regulated genes using RNA-sequencing technology. We found 83 genes more than 1.5-fold up- or down-regulated in the alcohol-treated group. Among them, genes (Dnai1, Cfap206 and Dnah1) associated with cilium movement were up-regulated in the alcohol-treated group. Mlf1, related to cell cycle arrest, was also up-regulated in the alcohol-treated group. On the other hand, genes (Smad3 and Plk5) involved in negative regulation of cell proliferation were down-regulated in the hippocampus by chronic alcohol administration. In addition, expression levels of genes associated with oxidative stress (Krt8 and Car3) and migration (Vim) were changed by chronic alcohol administration. These results pave a path for a better understanding of the neuromolecular mechanisms mediated by chronic alcohol exposure in the hippocampus of adolescents and negative pathology due to chronic alcohol abuse.
Collapse
Affiliation(s)
- Mi Ran Choi
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jasmin Sanghyun Han
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Young Gyu Chai
- Department of Molecular and Life Sciences, Hanyang University, Ansan, Korea
| | - Yeung-Bae Jin
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Sang-Rae Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Korea
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Epithelial CD47 is critical for mucosal repair in the murine intestine in vivo. Nat Commun 2019; 10:5004. [PMID: 31676794 PMCID: PMC6825175 DOI: 10.1038/s41467-019-12968-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/07/2019] [Indexed: 02/07/2023] Open
Abstract
CD47 is a ubiquitously expressed transmembrane glycoprotein that regulates inflammatory responses and tissue repair. Here, we show that normal mice treated with anti-CD47 antibodies, and Cd47-null mice have impaired intestinal mucosal wound healing. Furthermore, intestinal epithelial cell (IEC)-specific loss of CD47 does not induce spontaneous immune-mediated intestinal barrier disruption but results in defective mucosal repair after biopsy-induced colonic wounding or Dextran Sulfate Sodium (DSS)-induced mucosal damage. In vitro analyses using primary cultures of CD47-deficient murine colonic IEC or human colonoid-derived IEC treated with CD47-blocking antibodies demonstrate impaired epithelial cell migration in wound healing assays. Defective wound repair after CD47 loss is linked to decreased epithelial β1 integrin and focal adhesion signaling, as well as reduced thrombospondin-1 and TGF-β1. These results demonstrate a critical role for IEC-expressed CD47 in regulating mucosal repair and raise important considerations for possible alterations in wound healing secondary to therapeutic targeting of CD47. The role of the transmembrane glycoprotein CD47 in healing injured intestinal mucosa is unclear. Here, the authors show that selective loss of CD47 in the murine intestinal epithelium results in defective mucosal repair after colonic wounding, with suggested impaired cell migration in vitro.
Collapse
|
25
|
Zhou S, Hokugo A, McClendon M, Zhang Z, Bakshi R, Wang L, Segovia LA, Rezzadeh K, Stupp SI, Jarrahy R. Bioactive peptide amphiphile nanofiber gels enhance burn wound healing. Burns 2019; 45:1112-1121. [DOI: 10.1016/j.burns.2018.06.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/08/2018] [Accepted: 06/16/2018] [Indexed: 12/18/2022]
|
26
|
Nath PR, Pal-Nath D, Mandal A, Cam MC, Schwartz AL, Roberts DD. Natural Killer Cell Recruitment and Activation Are Regulated by CD47 Expression in the Tumor Microenvironment. Cancer Immunol Res 2019; 7:1547-1561. [PMID: 31362997 DOI: 10.1158/2326-6066.cir-18-0367] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 03/29/2019] [Accepted: 07/26/2019] [Indexed: 12/19/2022]
Abstract
Elevated CD47 expression in some cancers is associated with decreased survival and limited clearance by phagocytes expressing the CD47 counterreceptor SIRPα. In contrast, elevated CD47 mRNA expression in human melanomas was associated with improved survival. Gene-expression data were analyzed to determine a potential mechanism for this apparent protective function and suggested that high CD47 expression increases recruitment of natural killer (NK) cells into the tumor microenvironment. The CD47 ligand thrombospondin-1 inhibited NK cell proliferation and CD69 expression in vitro Cd47 -/- NK cells correspondingly displayed augmented effector phenotypes, indicating an inhibitory function of CD47 on NK cells. Treating human NK cells with a CD47 antibody that blocks thrombospondin-1 binding abrogated its inhibitory effect on NK cell proliferation. Similarly, treating wild-type mice with a CD47 antibody that blocks thrombospondin-1 binding delayed B16 melanoma growth, associating with increased NK cell recruitment and increased granzyme B and interferon-γ levels in intratumoral NK but not CD8+ T cells. However, B16 melanomas grew faster in Cd47 -/- than in wild-type mice. Melanoma-bearing Cd47 -/- mice exhibited decreased splenic NK cell numbers, with impaired effector protein expression and elevated exhaustion markers. Proapoptotic gene expression in Cd47-/- NK cells was associated with stress-mediated increases in mitochondrial proton leak, reactive oxygen species, and apoptosis. Global gene-expression profiling in NK cells from tumor-bearing mice identified CD47-dependent transcriptional responses that regulate systemic NK activation and exhaustion. Therefore, CD47 positively and negatively regulates NK cell function, and therapeutic antibodies that block inhibitory CD47 signaling can enhance NK immune surveillance of melanomas.
Collapse
Affiliation(s)
- Pulak Ranjan Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ajeet Mandal
- Human Brain Collection Core, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Margaret C Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute, and Leidos Biomedical Research, Inc., National Institutes of Health, Bethesda, Maryland
| | - Anthony L Schwartz
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
27
|
Nath PR, Gangaplara A, Pal-Nath D, Mandal A, Maric D, Sipes JM, Cam M, Shevach EM, Roberts DD. CD47 Expression in Natural Killer Cells Regulates Homeostasis and Modulates Immune Response to Lymphocytic Choriomeningitis Virus. Front Immunol 2018; 9:2985. [PMID: 30643501 PMCID: PMC6320676 DOI: 10.3389/fimmu.2018.02985] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/04/2018] [Indexed: 01/08/2023] Open
Abstract
CD47 is a ubiquitous cell surface receptor that directly regulates T cell immunity by interacting with its inhibitory ligand thrombospondin-1 and limits clearance of cells by phagocytes that express its counter-receptor signal-regulatory protein-α. Murine natural killer (NK) cells express higher levels of CD47 than other lymphocytes, but the role of CD47 in regulating NK cell homeostasis and immune function remains unclear. Cd47 -/- mice exhibited depletion of NK precursors in bone marrow, consistent with the antiphagocytic function of CD47. In contrast, antisense CD47 knockdown or gene disruption resulted in a dose dependent accumulation of immature and mature NK cells in spleen. Mature Cd47 -/- NK cells exhibited increased expression of NK effector and interferon gene signatures and an increased proliferative response to interleukin-15 in vitro. Cd47 -/- mice showed no defect in their early response to acute Armstrong lymphocytic choriomeningitis virus (LCMV) infection but were moderately impaired in controlling chronic Clone-13 LCMV infection, which was associated with depletion of splenic NK cells and loss of effector cytokine and interferon response gene expression in Cd47 -/- NK cells. Broad CD47-dependent differences in NK activation, survival, and exhaustion pathways were observed in NK cell transcriptional signatures in LCMV infected mice. These data identify CD47 as a cell-intrinsic and systemic regulator of NK cell homeostasis and NK cell function in responding to a viral infection.
Collapse
Affiliation(s)
- Pulak Ranjan Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Arunakumar Gangaplara
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ajeet Mandal
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute and Leidos Biomedical Research, Inc., National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - John M Sipes
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, National Cancer Institute and Leidos Biomedical Research, Inc., National Institutes of Health, Bethesda, MD, United States
| | - Ethan M Shevach
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [PMID: 30204432 DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
29
|
Murphy-Ullrich JE, Suto MJ. Thrombospondin-1 regulation of latent TGF-β activation: A therapeutic target for fibrotic disease. Matrix Biol 2018; 68-69:28-43. [PMID: 29288716 PMCID: PMC6015530 DOI: 10.1016/j.matbio.2017.12.009] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/14/2017] [Accepted: 12/16/2017] [Indexed: 12/12/2022]
Abstract
Transforming growth factor-β (TGF-β) is a central player in fibrotic disease. Clinical trials with global inhibitors of TGF-β have been disappointing, suggesting that a more targeted approach is warranted. Conversion of the latent precursor to the biologically active form of TGF-β represents a novel approach to selectively modulating TGF-β in disease, as mechanisms employed to activate latent TGF-β are typically cell, tissue, and/or disease specific. In this review, we will discuss the role of the matricellular protein, thrombospondin 1 (TSP-1), in regulation of latent TGF-β activation and the use of an antagonist of TSP-1 mediated TGF-β activation in a number of diverse fibrotic diseases. In particular, we will discuss the TSP-1/TGF-β pathway in fibrotic complications of diabetes, liver fibrosis, and in multiple myeloma. We will also discuss emerging evidence for a role for TSP-1 in arterial remodeling, biomechanical modulation of TGF-β activity, and in immune dysfunction. As TSP-1 expression is upregulated by factors induced in fibrotic disease, targeting the TSP-1/TGF-β pathway potentially represents a more selective approach to controlling TGF-β activity in disease.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Departments of Pathology, Cell Developmental and Integrative Biology, and Ophthalmology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | - Mark J Suto
- Southern Research, 2000 Ninth Avenue South, Birmingham, AL 35205, United States
| |
Collapse
|
30
|
Brassolatti P, Kido HW, Bossini PS, Gabbai-Armelin PR, Otterço AN, Almeida-Lopes L, Zanardi LM, Napolitano MA, de Avó LRDS, Forato LA, Araújo-Moreira FM, Parizotto NA. Bacterial cellulose membrane used as biological dressings on third-degree burns in rats. Biomed Mater Eng 2018; 29:29-42. [PMID: 29254071 DOI: 10.3233/bme-171710] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Burn injuries represent a high risk of morbidity and mortality. The wound healing process is complex and requires the participation of different types of cells. Therefore, new biomaterials, which innovate the wound healing process, are being investigated. OBJECTIVE The aim of this study was to investigate the use of bacterial cellulose both in its pure state and enriched with lidocaine in full-thickness burns in rats. METHODS Thirty rats (Wistar) (260 ± 20 gramas) divided into control group (CG), bacterial cellulose membrane group (MG) and bacterial cellulose membrane enriched with lidocaine group (MLG) were used. The burns were induced using a 150°C heated soldering iron, held on the animal neck for 10 seconds. The biomaterial was applied immediately after injury and skin samples were collected on the tenth day of the treatment. The level of significance of p⩽0.05 was used for the conclusion of the statistical analysis. RESULTS The groups treated with the biomaterials, a histological pattern compatible with a more advanced repair stage showing skin appendages, mild inflammatory infiltrate, better collagen fiber organization and mild immunostaining COX-2 and MMP-9 was observed, when compared to the control group that did not receive any type of treatment. CONCLUSION Thus, was concluded that the bacterial cellulose-based biomaterial both in its pure state and enriched with lidocaine optimizing the full-thickness burn wound healing in rats.
Collapse
Affiliation(s)
- Patricia Brassolatti
- Department of Physiotherapy, Post-Graduate Program in Biotechnology, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil. E-mail:
| | - Hueliton Wilian Kido
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Paulo Sérgio Bossini
- Research and Education Center for Photo Therapy in Health Science (NUPEN), DMC Equipment Import and Export-Co. Ltda, São Carlos, SP, Brazil
| | - Paulo R Gabbai-Armelin
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Albaiza Nicoletti Otterço
- Department of Physiotherapy, Post-Graduate Program in Biotechnology, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil. E-mail:
| | - Luciana Almeida-Lopes
- Research and Education Center for Photo Therapy in Health Science (NUPEN), DMC Equipment Import and Export-Co. Ltda, São Carlos, SP, Brazil
| | - Lisinéia Maria Zanardi
- Research and Education Center for Photo Therapy in Health Science (NUPEN), DMC Equipment Import and Export-Co. Ltda, São Carlos, SP, Brazil
| | - Marcos Aurélio Napolitano
- Research and Education Center for Photo Therapy in Health Science (NUPEN), DMC Equipment Import and Export-Co. Ltda, São Carlos, SP, Brazil
| | | | | | - Fernando M Araújo-Moreira
- Department of Physics, Post-Graduate Program of Biotechnology, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil
| | - Nivaldo Antonio Parizotto
- Department of Physiotherapy, Post-Graduate Program in Biotechnology, Federal University of São Carlos (UFSCar), São Carlos, SP, Brazil. E-mail:
| |
Collapse
|
31
|
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
|
32
|
Bazzazi H, Zhang Y, Jafarnejad M, Isenberg JS, Annex BH, Popel AS. Computer Simulation of TSP1 Inhibition of VEGF-Akt-eNOS: An Angiogenesis Triple Threat. Front Physiol 2018; 9:644. [PMID: 29899706 PMCID: PMC5988849 DOI: 10.3389/fphys.2018.00644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/11/2018] [Indexed: 01/08/2023] Open
Abstract
The matricellular protein thrombospondin-1 (TSP1) is a potent inhibitor of angiogenesis. Specifically, TSP1 has been experimentally shown to inhibit signaling downstream of vascular endothelial growth factor (VEGF). The molecular mechanism of this inhibition is not entirely clear. We developed a detailed computational model of VEGF signaling to Akt-endothelial nitric oxide synthase (eNOS) to investigate the quantitative molecular mechanism of TSP1 inhibition. The model demonstrated that TSP1 acceleration of VEGFR2 degradation is sufficient to explain the inhibition of VEGFR2 and eNOS phosphorylation. However, Akt inhibition requires TSP1-induced phosphatase recruitment to VEGFR2. The model was then utilized to test various strategies for the rescue of VEGF signaling to Akt and eNOS. Inhibiting TSP1 was predicted to be not as effective as CD47 depletion in rescuing signaling to Akt. The model further predicts that combination strategy involving depletion of CD47 and inhibition of TSP1 binding to CD47 is necessary for effective recovery of signaling to eNOS. In all, computational modeling offers insight to molecular mechanisms involving TSP1 interaction with VEGF signaling and provides strategies for rescuing angiogenesis by targeting TSP1-CD47 axis.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jeffrey S Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian H Annex
- Division of Cardiovascular Medicine, Department of Medicine, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
33
|
Giusti I, Di Francesco M, D'Ascenzo S, Palumbo P, Rughetti A, Dell'Orso L, Varasano PA, Pressanti GL, Dolo V. Leukocyte depletion does not affect the in vitro healing ability of platelet rich plasma. Exp Ther Med 2018; 15:4029-4038. [PMID: 29556269 DOI: 10.3892/etm.2018.5887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/19/2018] [Indexed: 01/15/2023] Open
Abstract
The clinical use of platelet-rich plasma (PRP) containing or deprived of leukocytes remains a subject of debate and a controversial issue. It is not yet clear whether leukocyte content has a positive or negative effect on tissue healing processes. Several studies, conducted mainly in the orthopedic field, support the use of leukocyte-poor (LP) PRP, whereas other studies have not identified any significant differences between the use of LP and leukocyte-rich PRP. In the present study, the role of leukocytes contained in PRP was assessed to verify their in vitro effect on fibroblasts and endothelial cells, which have a leading role in the biological processes associated with wound healing (including angiogenesis and matrix remodeling). The original sample of PRP was divided into two aliquots, one of which remained unaltered, while the other was deprived of leukocytes. The two aliquots were used in in vitro tests in order to verify the effects of leukocytes on proliferation, wound healing and tube formation, and in molecular analyses of growth factor and enzyme content. The present results highlighted a substantial overlap between the two formulations. This may be explained by similar levels of growth factors (vascular endothelial growth factor, thrombospondin-1, interferon-γ, platelet-derived growth factor-BB, -AA and -B, tumor growth factor-β1, fibroblast growth factor 7 and tumor necrosis factor-α) and enzymes (gelatinases and plasminogen activators) in the two formulations. These results support the hypothesis that the ability of the PRP to affect the in vitro biological response of endothelial cells and fibroblasts does not rely on the presence of leukocytes.
Collapse
Affiliation(s)
- Ilaria Giusti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy
| | - Marianna Di Francesco
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy
| | - Sandra D'Ascenzo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy
| | - Paola Palumbo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy
| | - Anna Rughetti
- Immunotransfusion Medicine Unit, 'San Salvatore' Hospital, I-67100 L'Aquila, Italy
| | - Luigi Dell'Orso
- Immunotransfusion Medicine Unit, 'San Salvatore' Hospital, I-67100 L'Aquila, Italy
| | | | | | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, I-67100 L'Aquila, Italy
| |
Collapse
|
34
|
Cook KL, Soto-Pantoja DR. "UPRegulation" of CD47 by the endoplasmic reticulum stress pathway controls anti-tumor immune responses. Biomark Res 2017; 5:26. [PMID: 28815041 PMCID: PMC5557514 DOI: 10.1186/s40364-017-0105-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
We recently demonstrated that targeting the unfolded protein response (UPR) protein GRP78 down-regulates CD47 expression, resulting in increased tumor macrophage infiltration and inhibited resistance to anti-estrogen therapy. We now show new data indicating that anti-estrogen therapy regulates CD47 expression and implicates its ligand, thrombospondin-1, in regulation of tumor macrophage infiltration. Moreover, GRP78 and CD47 co-expression is associated with poor prognosis in breast cancer patients, suggesting the existence of crosstalk between UPR and immunity that regulates therapeutic responses in breast cancer.
Collapse
Affiliation(s)
- Katherine L Cook
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA.,Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA.,Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA.,Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|
35
|
Zhang S, Yeap XY, DeBerge M, Naresh NK, Wang K, Jiang Z, Wilcox JE, White SM, Morrow JP, Burridge PW, Procissi D, Scott EA, Frazier W, Thorp EB. Acute CD47 Blockade During Ischemic Myocardial Reperfusion Enhances Phagocytosis-Associated Cardiac Repair. JACC Basic Transl Sci 2017; 2:386-397. [PMID: 28920097 PMCID: PMC5595371 DOI: 10.1016/j.jacbts.2017.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our data suggest that, after a myocardial infarction, integrin-associated protein CD47 on cardiac myocytes is elevated. In culture, increased CD47 on the surface of dying cardiomyocytes impairs phagocytic removal by immune cell macrophages. After myocardial ischemia and reperfusion, acute CD47 inhibition with blocking antibodies enhanced dead myocyte clearance by cardiac phagocytes and also improved the resolution of cardiac inflammation, reduced infarct size, and preserved cardiac contractile function. Early targeting of CD47 in the myocardium after reperfusion may be a new strategy to enhance wound repair in the ischemic heart.
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xin-Yi Yeap
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Matthew DeBerge
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Nivedita K Naresh
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kevin Wang
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Zhengxin Jiang
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jane E Wilcox
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Steven M White
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Daniel Procissi
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Evan A Scott
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - William Frazier
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri
| | - Edward B Thorp
- Department of Pathology and Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
36
|
TGF-β activation by bone marrow-derived thrombospondin-1 causes Schistosoma- and hypoxia-induced pulmonary hypertension. Nat Commun 2017; 8:15494. [PMID: 28555642 PMCID: PMC5459967 DOI: 10.1038/ncomms15494] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 04/03/2017] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an obstructive disease of the precapillary pulmonary arteries. Schistosomiasis-associated PAH shares altered vascular TGF-β signalling with idiopathic, heritable and autoimmune-associated etiologies; moreover, TGF-β blockade can prevent experimental pulmonary hypertension (PH) in pre-clinical models. TGF-β is regulated at the level of activation, but how TGF-β is activated in this disease is unknown. Here we show TGF-β activation by thrombospondin-1 (TSP-1) is both required and sufficient for the development of PH in Schistosoma-exposed mice. Following Schistosoma exposure, TSP-1 levels in the lung increase, via recruitment of circulating monocytes, while TSP-1 inhibition or knockout bone marrow prevents TGF-β activation and protects against PH development. TSP-1 blockade also prevents the PH in a second model, chronic hypoxia. Lastly, the plasma concentration of TSP-1 is significantly increased in subjects with scleroderma following PAH development. Targeting TSP-1-dependent activation of TGF-β could thus be a therapeutic approach in TGF-β-dependent vascular diseases. Thrombospondin-1 (TSP-1) activates latent TGF-β in the extracellular matrix. Here the authors show that inappropriate activation of latent TGF-β in murine, bovine and human lung by monocyte-produced TSP-1 causes pulmonary hypertension, and that interference with the activation process prevents disease development.
Collapse
|
37
|
Bazzazi H, Isenberg JS, Popel AS. Inhibition of VEGFR2 Activation and Its Downstream Signaling to ERK1/2 and Calcium by Thrombospondin-1 (TSP1): In silico Investigation. Front Physiol 2017; 8:48. [PMID: 28220078 PMCID: PMC5292565 DOI: 10.3389/fphys.2017.00048] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
VEGF signaling through VEGFR2 is a central regulator of the angiogenic response. Inhibition of VEGF signaling by the stress-induced matricellular protein TSP1 plays a role in modulating the angiogenic response to VEGF in both health and disease. TSP1 binding to CD47 inhibits VEGFR2 activation. The full implications of this inhibitory interaction are unknown. We developed a detailed rule-based computational model to inquire if TSP1-CD47 signaling through VEGF had downstream effects upon ERK1/2 and calcium. Our Simulations suggest that enhanced degradation of VEGFR2 initiated by the binding of TSP1 to CD47 is sufficient to explain the inhibition of VEGFR2 phosphorylation, calcium elevation, and ERK1/2 activation downstream of VEGF. A complementary mechanism involving the recruitment of phosphatases to the VEGFR2 complex with consequent increase in the rate of receptor dephosphorylation may augment the inhibition of the VEGF signal. The model was then utilized to simulate the effect of inhibiting external TSP1 or the depletion of CD47 as potential therapeutic strategies in restoring VEGF signaling. Results suggest that depleting CD47 is a more efficient strategy in inhibiting the effects of TSP1/CD47 on VEGF signaling. Our results highlight the utility of in silico investigations in elucidating and clarifying molecular mechanisms at the intersection of TSP1 and VEGF biology and in differentiating between competing pro-angiogenic therapeutic strategies relevant to peripheral arterial disease (PAD) and wound healing.
Collapse
Affiliation(s)
- Hojjat Bazzazi
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| | - Jeffery S Isenberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University Baltimore, MD, USA
| |
Collapse
|
38
|
Li Y, Turpin CP, Wang S. Role of thrombospondin 1 in liver diseases. Hepatol Res 2017; 47:186-193. [PMID: 27492250 PMCID: PMC5292098 DOI: 10.1111/hepr.12787] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/13/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023]
Abstract
Thrombospondin 1 (TSP1) is a matricellular glycoprotein that can be secreted by many cell types. Through binding to extracellular proteins and/or cell surface receptors, TSP1 modulates a variety of cellular functions. Since its discovery in 1971, TSP1 has been found to play important roles in multiple biological processes including angiogenesis, apoptosis, latent transforming growth factor-β activation, and immune regulation. Thrombospondin 1 is also involved in regulating many organ functions. However, the role of TSP1 in liver diseases has not been extensively addressed. In this review, we summarize the findings about the possible role that TSP1 plays in chronic liver diseases focusing on non-alcoholic fatty liver diseases, liver fibrosis, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yanzhang Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Medical College of Henan University, Kaifeng, Henan 475004, China
| | - Courtney P Turpin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
39
|
Magdaleno F, Arriazu E, Ruiz de Galarreta M, Chen Y, Ge X, Conde de la Rosa L, Nieto N. Cartilage oligomeric matrix protein participates in the pathogenesis of liver fibrosis. J Hepatol 2016; 65:963-971. [PMID: 27318326 PMCID: PMC5831373 DOI: 10.1016/j.jhep.2016.06.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 05/30/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Liver fibrosis is characterized by significant accumulation of extracellular matrix (ECM) proteins, mainly fibrillar collagen-I, as a result of persistent liver injury. Cartilage oligomeric matrix protein (COMP) is largely found in the ECM of skeletal tissue. Increased COMP expression has been associated with fibrogenesis in systemic sclerosis, lung fibrosis, chronic pancreatitis, cirrhosis and hepatocellular carcinoma. We hypothesized that COMP could induce fibrillar collagen-I deposition and participate in matrix remodeling thus contributing to the pathophysiology of liver fibrosis. METHODS Thioacetamide (TAA) and carbon tetrachloride (CCl4) were used to induce liver fibrosis in wild-type (WT) and Comp-/- mice. In vitro experiments were performed with primary hepatic stellate cells (HSCs). RESULTS COMP expression was detected in livers from control WT mice and was upregulated in response to either TAA or CCl4-induced liver fibrosis. TAA-treated or CCl4-injected Comp-/- mice showed less liver injury, inflammation and fibrosis compared to their corresponding control WT mice. Challenge of HSCs with recombinant COMP (rCOMP) induced intra- plus extracellular collagen-I deposition and increased matrix metalloproteinases (MMPs) 2, 9 and 13, albeit similar expression of transforming growth factor beta (TGFβ) protein, in addition to Tgfβ, tumour necrosis factor alpha (Tnfα) and tissue inhibitor of metalloproteinases-1 (Timp1) mRNAs. We demonstrated that COMP binds collagen-I; yet, it does not prevent collagen-I cleavage by MMP1. Last, rCOMP induced collagen-I expression in HSCs via CD36 receptor signaling and activation of the MEK1/2-pERK1/2 pathway. CONCLUSION These results suggest that COMP contributes to liver fibrosis by regulating collagen-I deposition. LAY SUMMARY Cartilage oligomeric matrix protein (COMP) induces fibrillar collagen-I deposition via the CD36 receptor signaling and activation of the MEK1/2-pERK1/2 pathway, and participates in extracellular matrix remodeling contributing to the pathophysiology of liver fibrosis.
Collapse
Affiliation(s)
- Fernando Magdaleno
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA; Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Elena Arriazu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA
| | - Marina Ruiz de Galarreta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA
| | - Yu Chen
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Xiaodong Ge
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA; Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Laura Conde de la Rosa
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA
| | - Natalia Nieto
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Avenue, Room 11-70, New York, NY 10029, USA; Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA.
| |
Collapse
|
40
|
Gubbiotti MA, Vallet SD, Ricard-Blum S, Iozzo RV. Decorin interacting network: A comprehensive analysis of decorin-binding partners and their versatile functions. Matrix Biol 2016; 55:7-21. [PMID: 27693454 DOI: 10.1016/j.matbio.2016.09.009] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decorin, a prototype small leucine-rich proteoglycan, regulates a vast array of cellular processes including collagen fibrillogenesis, wound repair, angiostasis, tumor growth, and autophagy. This functional versatility arises from a wide array of decorin/protein interactions also including interactions with its single glycosaminoglycan side chain. The decorin-binding partners encompass numerous categories ranging from extracellular matrix molecules to cell surface receptors to growth factors and enzymes. Despite the diversity of the decorin interacting network, two main roles emerge as prominent themes in decorin function: maintenance of cellular structure and outside-in signaling, culminating in anti-tumorigenic effects. Here we present contemporary knowledge regarding the decorin interacting network and discuss in detail the biological relevance of these pleiotropic interactions, some of which could be targeted by therapeutic interventions.
Collapse
Affiliation(s)
- Maria A Gubbiotti
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sylvain D Vallet
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Sylvie Ricard-Blum
- Pericellular and Extracellular Supramolecular Assemblies, Institute of Molecular and Supramolecular Chemistry and Biochemistry, University Claude Bernard, Lyon, France
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
41
|
Breitenbach JS, Rinnerthaler M, Trost A, Weber M, Klausegger A, Gruber C, Bruckner D, Reitsamer HA, Bauer JW, Breitenbach M. Transcriptome and ultrastructural changes in dystrophic Epidermolysis bullosa resemble skin aging. Aging (Albany NY) 2016; 7:389-411. [PMID: 26143532 PMCID: PMC4505166 DOI: 10.18632/aging.100755] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aging process of skin has been investigated recently with respect to mitochondrial function and oxidative stress. We have here observed striking phenotypic and clinical similarity between skin aging and recessive dystrophic Epidermolysis bullosa (RDEB), which is caused by recessive mutations in the gene coding for collagen VII, COL7A1. Ultrastructural changes, defects in wound healing, and inflammation markers are in part shared with aged skin. We have here compared the skin transcriptomes of young adults suffering from RDEB with that of sex‐ and age‐matched healthy probands. In parallel we have compared the skin transcriptome of healthy young adults with that of elderly healthy donors. Quite surprisingly, there was a large overlap of the two gene lists that concerned a limited number of functional protein families. Most prominent among the proteins found are a number of proteins of the cornified envelope or proteins mechanistically involved in cornification and other skin proteins. Further, the overlap list contains a large number of genes with a known role in inflammation. We are documenting some of the most prominent ultrastructural and protein changes by immunofluorescence analysis of skin sections from patients, old individuals, and healthy controls.
Collapse
Affiliation(s)
- Jenny S Breitenbach
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | - Mark Rinnerthaler
- Fachbereich Zellbiologie der Universität Salzburg, Salzburg, Austria
| | - Andrea Trost
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Manuela Weber
- Fachbereich Zellbiologie der Universität Salzburg, Salzburg, Austria
| | - Alfred Klausegger
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | - Christina Gruber
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | - Daniela Bruckner
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Herbert A Reitsamer
- University Clinic of Ophthalmology and Optometry, Research Program for Ophthalmology and Glaucoma Research, Paracelsus Medical University, Salzburg, Austria
| | - Johann W Bauer
- Department of Dermatology and EB House Austria, Paracelsus Medical University, Salzburg, Austria
| | | |
Collapse
|
42
|
Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev 2016; 97:4-27. [PMID: 26562801 DOI: 10.1016/j.addr.2015.11.001] [Citation(s) in RCA: 1554] [Impact Index Per Article: 172.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM) is a non-cellular three-dimensional macromolecular network composed of collagens, proteoglycans/glycosaminoglycans, elastin, fibronectin, laminins, and several other glycoproteins. Matrix components bind each other as well as cell adhesion receptors forming a complex network into which cells reside in all tissues and organs. Cell surface receptors transduce signals into cells from ECM, which regulate diverse cellular functions, such as survival, growth, migration, and differentiation, and are vital for maintaining normal homeostasis. ECM is a highly dynamic structural network that continuously undergoes remodeling mediated by several matrix-degrading enzymes during normal and pathological conditions. Deregulation of ECM composition and structure is associated with the development and progression of several pathologic conditions. This article emphasizes in the complex ECM structure as to provide a better understanding of its dynamic structural and functional multipotency. Where relevant, the implication of the various families of ECM macromolecules in health and disease is also presented.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece
| | - Chrysostomi Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece; Division of Medical Protein Chemistry, Department of Translational Medicine Malmö, Lund University, S-20502 Malmö, Sweden
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
43
|
Neill T, Schaefer L, Iozzo RV. Decorin as a multivalent therapeutic agent against cancer. Adv Drug Deliv Rev 2016; 97:174-85. [PMID: 26522384 DOI: 10.1016/j.addr.2015.10.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 12/22/2022]
Abstract
Decorin is a prototypical small leucine-rich proteoglycan that epitomizes the multifunctional nature of this critical gene family. Soluble decorin engages multiple receptor tyrosine kinases within the target-rich environment of the tumor stroma and tumor parenchyma. Upon receptor binding, decorin initiates signaling pathways within endothelial cells downstream of VEGFR2 that ultimately culminate in a Peg3/Beclin 1/LC3-dependent autophagic program. Concomitant with autophagic induction, decorin blunts capillary morphogenesis and endothelial cell migration, thereby significantly compromising tumor angiogenesis. In parallel within the tumor proper, decorin binds multiple RTKs with high affinity, including Met, for a multitude of oncosuppressive functions including growth inhibition, tumor cell mitophagy, and angiostasis. Decorin is also pro-inflammatory by modulating macrophage function and cytokine secretion. Decorin suppresses tumorigenic growth, angiogenesis, and prevents metastatic lesions in a variety of in vitro and in vivo tumor models. Therefore, decorin would be an ideal therapeutic candidate for combating solid malignancies.
Collapse
|
44
|
|
45
|
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
|
46
|
Murphy-Ullrich JE, Sage EH. Revisiting the matricellular concept. Matrix Biol 2014; 37:1-14. [PMID: 25064829 PMCID: PMC4379989 DOI: 10.1016/j.matbio.2014.07.005] [Citation(s) in RCA: 303] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/16/2022]
Abstract
The concept of a matricellular protein was first proposed by Paul Bornstein in the mid-1990s to account for the non-lethal phenotypes of mice with inactivated genes encoding thrombospondin-1, tenascin-C, or SPARC. It was also recognized that these extracellular matrix proteins were primarily counter or de-adhesive. This review reappraises the matricellular concept after nearly two decades of continuous investigation. The expanded matricellular family as well as the diverse and often unexpected functions, cellular location, and interacting partners/receptors of matricellular proteins are considered. Development of therapeutic strategies that target matricellular proteins are discussed in the context of pathology and regenerative medicine.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | | |
Collapse
|