1
|
Aubert A, Liu A, Kao M, Goeres J, Richardson KC, Nierves L, Jung K, Nabai L, Zhao H, Orend G, Krawetz R, Lange PF, Younger A, Chan J, Granville DJ. Granzyme B cleaves tenascin-C to release its C-terminal domain in rheumatoid arthritis. JCI Insight 2024; 9:e181935. [PMID: 39475853 PMCID: PMC11623945 DOI: 10.1172/jci.insight.181935] [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: 04/11/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disorder characterized by exacerbated joint inflammation. Despite the well-documented accumulation of the serine protease granzyme B (GzmB) in RA patient biospecimens, little is understood pertaining to its role in pathobiology. In the present study, tenascin-C (TNC) - a large, pro-inflammatory extracellular matrix glycoprotein - was identified as a substrate for GzmB in RA. GzmB cleaves TNC to generate 3 fragments in vitro: a 130 kDa fragment that remains anchored to the matrix and 2 solubilized fragments of 70 and 30 kDa. Mass spectrometry results suggested that the 30 kDa fragment contained the pro-inflammatory TNC C-terminal fibrinogen-like domain. In the synovial fluids of patients with RA, soluble levels of GzmB and TNC were significantly elevated compared with healthy controls. Further, immunoblotting revealed soluble 70 and 30 kDa TNC fragments in the synovial fluids of patients with RA, matching TNC fragment sizes generated by GzmB cleavage in vitro. Granzyme K (GzmK), another serine protease of the granzyme family, also cleaves TNC in vitro; however, the molecular weights of GzmK-generated TNC fragments did not correspond to TNC fragment sizes detected in patients. Our data support that GzmB, but not GzmK, contributes to RA through the cleavage of TNC.
Collapse
Affiliation(s)
- Alexandre Aubert
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Liu
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Kao
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenna Goeres
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katlyn C. Richardson
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lorenz Nierves
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Cuccione Childhood Cancer Research Program and the BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
| | - Layla Nabai
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongyan Zhao
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d’Hématologie et d’Immunologie, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Roman Krawetz
- McCaig Institute for Bone and Joint Health, Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Philipp F. Lange
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Cuccione Childhood Cancer Research Program and the BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Alastair Younger
- Department of Orthopaedics, Foot & Ankle Research, St. Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Jonathan Chan
- Department of Medicine, Division of Rheumatology, University of British Columbia, Vancouver, British Columbia, Canada
- Arthritis Research Canada, Vancouver, British Columbia, Canada
| | - David J. Granville
- International Collaboration on Repair Discoveries (ICORD) Centre, British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute, and
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
2
|
Syx D, Malfait F. Pathogenic mechanisms in genetically defined Ehlers-Danlos syndromes. Trends Mol Med 2024; 30:824-843. [PMID: 39147618 DOI: 10.1016/j.molmed.2024.06.001] [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: 02/14/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 08/17/2024]
Abstract
The Ehlers-Danlos syndromes (EDS) are a group of rare heritable connective tissue disorders, common hallmarks of which are skin hyperextensibility, joint hypermobility, and generalized connective tissue fragility. Currently, 13 EDS types are recognized, caused by defects in 20 genes which consequently alter biosynthesis, organization, and/or supramolecular assembly of collagen fibrils in the extracellular matrix (ECM). Molecular analyses on patient samples (mostly dermal fibroblast cultures), combined with studies on animal models, have highlighted that part of EDS pathogenesis can be attributed to impaired cellular dynamics. Although our understanding of the full extent of (extra)cellular consequences is still limited, this narrative review aims to provide a comprehensive overview of our current knowledge on the extracellular, pericellular, and intracellular alterations implicated in EDS pathogenesis.
Collapse
Affiliation(s)
- Delfien Syx
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Fransiska Malfait
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
3
|
Liang G, Lv XF, Huang W, Jin YJ, Roquid KA, Kawase H, Offermanns S. Loss of Smooth Muscle Tenascin-X Inhibits Vascular Remodeling Through Increased TGF-β Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1748-1763. [PMID: 38934115 DOI: 10.1161/atvbaha.123.321067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) are highly plastic. Vessel injury induces a phenotypic transformation from differentiated to dedifferentiated VSMCs, which involves reduced expression of contractile proteins and increased production of extracellular matrix and inflammatory cytokines. This transition plays an important role in several cardiovascular diseases such as atherosclerosis, hypertension, and aortic aneurysm. TGF-β (transforming growth factor-β) is critical for VSMC differentiation and to counterbalance the effect of dedifferentiating factors. However, the mechanisms controlling TGF-β activity and VSMC phenotypic regulation under in vivo conditions are poorly understood. The extracellular matrix protein TN-X (tenascin-X) has recently been shown to bind TGF-β and to prevent it from activating its receptor. METHODS We studied the role of TN-X in VSMCs in various murine disease models using tamoxifen-inducible SMC-specific knockout and adeno-associated virus-mediated knockdown. RESULTS In hypertensive and high-fat diet-fed mice, after carotid artery ligation as well as in human aneurysmal aortae, expression of Tnxb, the gene encoding TN-X, was increased in VSMCs. Mice with smooth muscle cell-specific loss of TN-X (SMC-Tnxb-KO) showed increased TGF-β signaling in VSMCs, as well as upregulated expression of VSMC differentiation marker genes during vascular remodeling compared with controls. SMC-specific TN-X deficiency decreased neointima formation after carotid artery ligation and reduced vessel wall thickening during Ang II (angiotensin II)-induced hypertension. SMC-Tnxb-KO mice lacking ApoE showed reduced atherosclerosis and Ang II-induced aneurysm formation under high-fat diet. Adeno-associated virus-mediated SMC-specific expression of short hairpin RNA against Tnxb showed similar beneficial effects. Treatment with an anti-TGF-β antibody or additional SMC-specific loss of the TGF-β receptor reverted the effects of SMC-specific TN-X deficiency. CONCLUSIONS In summary, TN-X critically regulates VSMC plasticity during vascular injury by inhibiting TGF-β signaling. Our data indicate that inhibition of vascular smooth muscle TN-X may represent a strategy to prevent and treat pathological vascular remodeling.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/prevention & control
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/physiopathology
- Hypertension/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Signal Transduction
- Tenascin/metabolism
- Tenascin/genetics
- Tenascin/deficiency
- Transforming Growth Factor beta/metabolism
- Vascular Remodeling
Collapse
Affiliation(s)
- Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Xiao-Fei Lv
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (X.-F.L.)
| | - Wei Huang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Kenneth Anthony Roquid
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Haruya Kawase
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Center for Molecular Medicine, Goethe University Frankfurt, Germany (S.O.)
- Cardiopulmonary Institute, Bad Nauheim, Germany (S.O.)
- German Center for Cardiovascular Research, Bad Nauheim, Germany (S.O.)
| |
Collapse
|
4
|
Guerrero-Barberà G, Burday N, Costell M. Shaping Oncogenic Microenvironments: Contribution of Fibronectin. Front Cell Dev Biol 2024; 12:1363004. [PMID: 38660622 PMCID: PMC11039881 DOI: 10.3389/fcell.2024.1363004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and glycans, dynamically remodeled and specifically tailored to the structure/function of each organ. The malignant transformation of cancer cells is determined by both cell intrinsic properties, such as mutations, and extrinsic variables, such as the mixture of surrounding cells in the tumor microenvironment and the biophysics of the ECM. During cancer progression, the ECM undergoes extensive remodeling, characterized by disruption of the basal lamina, vascular endothelial cell invasion, and development of fibrosis in and around the tumor cells resulting in increased tissue stiffness. This enhanced rigidity leads to aberrant mechanotransduction and further malignant transformation potentiating the de-differentiation, proliferation and invasion of tumor cells. Interestingly, this fibrotic microenvironment is primarily secreted and assembled by non-cancerous cells. Among them, the cancer-associated fibroblasts (CAFs) play a central role. CAFs massively produce fibronectin together with type I collagen. This review delves into the primary interactions and signaling pathways through which fibronectin can support tumorigenesis and metastasis, aiming to provide critical molecular insights for better therapy response prediction.
Collapse
Affiliation(s)
| | | | - Mercedes Costell
- Departament of Biochemistry and Molecular Biology, Institut Universitari de Biotecnologia i Biomedicina, Universitat de València, Valencia, Spain
| |
Collapse
|
5
|
South AP, Laimer M, Gueye M, Sui JY, Eichenfield LF, Mellerio JE, Nyström A. Type VII Collagen Deficiency in the Oncogenesis of Cutaneous Squamous Cell Carcinoma in Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2023; 143:2108-2119. [PMID: 37327859 DOI: 10.1016/j.jid.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Dystrophic epidermolysis bullosa is a rare genetic skin disorder caused by COL7A1 sequence variations that result in type VII collagen deficits and cutaneous and extracutaneous manifestations. One serious complication of dystrophic epidermolysis bullosa is cutaneous squamous cell carcinoma, a leading driver of morbidity and mortality, especially among patients with recessive dystrophic epidermolysis bullosa. Type VII collagen deficits alter TGFβ signaling and evoke multiple other cutaneous squamous cell carcinoma progression-promoting activities within epidermal microenvironments. This review examines cutaneous squamous cell carcinoma pathophysiology in dystrophic epidermolysis bullosa with a focus on known oncogenesis pathways at play and explores the idea that therapeutic type VII collagen replacement may reduce cutaneous squamous cell carcinoma risk.
Collapse
Affiliation(s)
- Andrew P South
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | | | - Jennifer Y Sui
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, San Diego, California, USA; Division of Pediatric Dermatology, Rady Children's Hospital San Diego, San Diego, California, USA
| | - Jemima E Mellerio
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies, Freiburg, Germany
| |
Collapse
|
6
|
Vorobyev A, Ludwig RJ. Forschung für die Praxis: Ernährung und Mikrobiom bei Autoimmunkrankheiten. J Dtsch Dermatol Ges 2023; 21:958-963. [PMID: 37700405 DOI: 10.1111/ddg.15101_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/17/2023] [Indexed: 09/14/2023]
Abstract
ZusammenfassungDie Häufigkeit von Autoimmunerkrankungen in Industrieländern hat während der letzten Jahrzehnte ständig zugenommen. Diese Erkrankungen führen zu erhöhter Sterblichkeit sowie anhaltender Beeinträchtigung der Lebensqualität der Patienten und bedeuten eine große medizinische Belastung. Die Behandlung von Autoimmunkrankheiten beruht häufig auf unspezifischer Immunsuppression, was das Risiko von Infektionskrankheiten und Krebsmanifestationen erhöht. Die Pathogenese von Autoimmunerkrankungen ist komplex und umfasst nicht nur genetische Faktoren, sondern auch Umwelteinflüsse, die als Grund für die Zunahme von Autoimmunerkrankungen angesehen werden. Zahlreiche Umweltfaktoren wie Infektionen, Rauchen, Medikamente oder Ernährung können das Auftreten von Autoimmunität entweder fördern oder verhindern. Die Mechanismen der Beeinflussung durch Umwelteinflüsse sind jedoch komplex und derzeit noch nicht eindeutig geklärt. Die Entschlüsselung dieser Wechselwirkungen könnte unser Verständnis der Autoimmunität verbessern und neue Behandlungsmöglichkeiten für die Patienten eröffnen.
Collapse
Affiliation(s)
- Artem Vorobyev
- Abteilung für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck
- Lübecker Institut für experimentelle Dermatologie, Lübeck
| | - Ralf J Ludwig
- Abteilung für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck
- Lübecker Institut für experimentelle Dermatologie, Lübeck
| |
Collapse
|
7
|
Vorobyev A, Ludwig RJ. Research in practice: Diet and microbiome in autoimmune diseases. J Dtsch Dermatol Ges 2023; 21:958-962. [PMID: 37235511 DOI: 10.1111/ddg.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 05/28/2023]
Abstract
The incidence of autoimmune diseases in industrialized countries is constantly increasing over past decades. These diseases lead to increased mortality and persistent reduction in quality of life of the patients, posing a severe medical burden. Treatment of autoimmune diseases is often based on unspecific immune suppression, increasing the risk of infectious diseases as well as cancer manifestation. Pathogenesis of autoimmune conditions is complex and includes not only genetic factors, but also environmental influence, which is considered to be the reason for the rise of incidence of autoimmune diseases. Environmental factors comprise numerous elements, such as infections, smoking, medication, diet etc., which can either promote or prevent the onset of autoimmunity. However, the mechanisms of environmental influence are complex and for this moment not clearly understood. Deciphering of these interactions could enhance our comprehension of autoimmunity and provide some novel treatment options for the patients.
Collapse
Affiliation(s)
- Artem Vorobyev
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
- Lübeck Institute of Experimental Dermatology, Lübeck, Germany
| | - Ralf J Ludwig
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
- Lübeck Institute of Experimental Dermatology, Lübeck, Germany
| |
Collapse
|
8
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 181] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
9
|
Xia Y, Wang H, Yang R, Hou Y, Li Y, Zhu J, Fu C. Biomaterials delivery strategies to repair degenerated intervertebral discs by regulating the inflammatory microenvironment. Front Immunol 2023; 14:1051606. [PMID: 36756124 PMCID: PMC9900107 DOI: 10.3389/fimmu.2023.1051606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is one of the leading causes of lower back pain. Although IVDD cannot directly cause death, it can cause pain, psychological burdens, and economic burdens to patients. Current conservative treatments for IVDD can relieve pain but cannot reverse the disease. Patients who cannot tolerate pain usually resort to a strategy of surgical resection of the degenerated disc. However, the surgical removal of IVDD can affect the stability of adjacent discs. Furthermore, the probability of the reherniation of the intervertebral disc (IVD) after surgery is as high as 21.2%. Strategies based on tissue engineering to deliver stem cells for the regeneration of nucleus purposes (NP) and annulus fibrosus (AF) have been extensively studied. The developed biomaterials not only locally withstand the pressure of the IVD but also lay the foundation for the survival of stem cells. However, the structure of IVDs does not provide sufficient nutrients for delivered stem cells. The role of immune mechanisms in IVDD has recently become clear. In IVDD, the IVD that was originally in immune privilege prevents the attack of immune cells (mainly effector T cells and macrophages) and aggravates the disease. Immune regulatory and inflammatory factors released by effector T cells, macrophages, and the IVD further aggravate IVDD. Reversing IVDD by regulating the inflammatory microenvironment is a potential approach for the treatment of the disease. However, the biological factors modulating the inflammatory microenvironment easily degrade in vivo. It makes it possible for different biomaterials to modulate the inflammatory microenvironment to repair IVDD. In this review, we have discussed the structures of IVDs and the immune mechanisms underlying IVDD. We have described the immune mechanisms elicited by different biological factors, including tumor necrosis factors, interleukins, transforming growth factors, hypoxia-inducible factors, and reactive oxygen species in IVDs. Finally, we have discussed the biomaterials used to modulate the inflammatory microenvironment to repair IVDD and their development.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yulin Hou
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China,*Correspondence: Changfeng Fu,
| |
Collapse
|
10
|
Abstract
Tenascin-C is a large extracellular matrix glycoprotein with complex, not yet fully unveiled roles. Its context- and structure-dependent modus operandi renders tenascin-C a puzzling protein. Since its discovery ∼40 years ago, research into tenascin-C biology continues to reveal novel functions, the most recent of all being its immunomodulatory activity, especially its role in infection, which is just now beginning to emerge. Here, we explore the role of tenascin-C in the immune response to viruses, including SARS-CoV-2 and HIV-1. Recently, tenascin-C has emerged as a biomarker of disease severity during COVID-19 and other viral infections, and we highlight relevant RNA sequencing and proteomic analyses that suggest a correlation between tenascin-C levels and disease severity. Finally, we ask what the function of this protein during viral replication is and propose tenascin-C as an intercellular signal of inflammation shuttled to distal sites via exosomes, a player in the repair and remodeling of infected and damaged tissues during severe infectious disease, as well as a ligand for specific pathogens with distinct implications for the host.
Collapse
Affiliation(s)
- Lorena Zuliani-Alvarez
- 1QBI Coronavirus Research Group, San Francisco, California,2Quantitative Biosciences Institute, University of California, San Francisco, California,3Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California
| | - Anna M. Piccinini
- 4School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
11
|
Yi M, Li T, Niu M, Wu Y, Zhao Z, Wu K. TGF-β: A novel predictor and target for anti-PD-1/PD-L1 therapy. Front Immunol 2022; 13:1061394. [PMID: 36601124 PMCID: PMC9807229 DOI: 10.3389/fimmu.2022.1061394] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor-β (TGF-β) signaling regulates multiple physiological processes, such as cell proliferation, differentiation, immune homeostasis, and wound healing. Besides, TGF-β plays a vital role in diseases, including cancer. Accumulating evidence indicates that TGF-β controls the composition and behavior of immune components in the tumor microenvironment (TME). Advanced cancers leverage TGF-β to reshape the TME and escape immune surveillance. TGF-β-mediated immune evasion is an unfavorable factor for cancer immunotherapy, especially immune checkpoint inhibitors (ICI). Numerous preclinical and clinical studies have demonstrated that hyperactive TGF-β signaling is closely associated with ICI resistance. It has been validated that TGF-β blockade synergizes with ICI and overcomes treatment resistance. TGF-β-targeted therapies, including trap and bispecific antibodies, have shown immense potential for cancer immunotherapy. In this review, we summarized the predictive value of TGF-β signaling and the prospects of TGF-β-targeted therapies for cancer immunotherapy.
Collapse
Affiliation(s)
- Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenyu Zhao
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Kongming Wu, ; Zhenyu Zhao,
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Kongming Wu, ; Zhenyu Zhao,
| |
Collapse
|
12
|
Wang Y, Wang G, Liu H. Tenascin-C: A Key Regulator in Angiogenesis during Wound Healing. Biomolecules 2022; 12:1689. [PMID: 36421704 PMCID: PMC9687801 DOI: 10.3390/biom12111689] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Injury repair is a complex physiological process in which multiple cells and molecules are involved. Tenascin-C (TNC), an extracellular matrix (ECM) glycoprotein, is essential for angiogenesis during wound healing. This study aims to provide a comprehensive review of the dynamic changes and functions of TNC throughout tissue regeneration and to present an up-to-date synthesis of the body of knowledge pointing to multiple mechanisms of TNC at different restoration stages. (2) Methods: A review of the PubMed database was performed to include all studies describing the pathological processes of damage restoration and the role, structure, expression, and function of TNC in post-injury treatment; (3) Results: In this review, we first introduced the construction and expression signature of TNC. Then, the role of TNC during the process of damage restoration was introduced. We highlight the temporal heterogeneity of TNC levels at different restoration stages. Furthermore, we are surprised to find that post-injury angiogenesis is dynamically consistent with changes in TNC. Finally, we discuss the strategies for TNC in post-injury treatment. (4) Conclusions: The dynamic expression of TNC has a significant impact on angiogenesis and healing wounds and counters many negative aspects of poorly healing wounds, such as excessive inflammation, ischemia, scarring, and wound infection.
Collapse
Affiliation(s)
- Yucai Wang
- Department of Orthopaedic Surgery, Tangdu Hospital, AirForce Medical University, Xi’an 710000, China
| | - Guangfu Wang
- Vasculocardiology Department, The Fourth People’s Hospital of Jinan, Jinan 250000, China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
13
|
Yilmaz A, Loustau T, Salomé N, Poilil Surendran S, Li C, Tucker RP, Izzi V, Lamba R, Koch M, Orend G. Advances on the roles of tenascin-C in cancer. J Cell Sci 2022; 135:276631. [PMID: 36102918 PMCID: PMC9584351 DOI: 10.1242/jcs.260244] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The roles of the extracellular matrix molecule tenascin-C (TNC) in health and disease have been extensively reviewed since its discovery over 40 years ago. Here, we will describe recent insights into the roles of TNC in tumorigenesis, angiogenesis, immunity and metastasis. In addition to high levels of expression in tumors, and during chronic inflammation, and bacterial and viral infection, TNC is also expressed in lymphoid organs. This supports potential roles for TNC in immunity control. Advances using murine models with engineered TNC levels were instrumental in the discovery of important functions of TNC as a danger-associated molecular pattern (DAMP) molecule in tissue repair and revealed multiple TNC actions in tumor progression. TNC acts through distinct mechanisms on many different cell types with immune cells coming into focus as important targets of TNC in cancer. We will describe how this knowledge could be exploited for cancer disease management, in particular for immune (checkpoint) therapies.
Collapse
Affiliation(s)
- Alev Yilmaz
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Thomas Loustau
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Nathalie Salomé
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Suchithra Poilil Surendran
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Chengbei Li
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| | - Richard P. Tucker
- University of California at Davis 4 Department of Cell Biology and Human Anatomy , , 95616 Davis, CA , USA
| | - Valerio Izzi
- University of Oulu 5 Faculty of Biochemistry and Molecular Medicine , , FI-90014 Oulu , Finland
- University of Oulu 6 Faculty of Medicine , , FI-90014 Oulu , Finland
| | - Rijuta Lamba
- University of Oulu 5 Faculty of Biochemistry and Molecular Medicine , , FI-90014 Oulu , Finland
- University of Oulu 6 Faculty of Medicine , , FI-90014 Oulu , Finland
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Research, Center for Biochemistry, Center for Molecular Medicine Cologne (CMMC) 7 , Faculty of Medicine and , Joseph-Stelzmann-Str. 52, 50931 Cologne , Germany
- University Hospital Cologne, University of Cologne 7 , Faculty of Medicine and , Joseph-Stelzmann-Str. 52, 50931 Cologne , Germany
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie 1 , 1 Place de l'Hôpital, 67091 Strasbourg , France
- Université Strasbourg 2 , 67000 Strasbourg , France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) 3 , 67000 Strasbourg , France
| |
Collapse
|
14
|
Tenascin-C in fibrosis in multiple organs: Translational implications. Semin Cell Dev Biol 2022; 128:130-136. [PMID: 35400564 PMCID: PMC10119770 DOI: 10.1016/j.semcdb.2022.03.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 12/28/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex disease with a pathogenic triad of autoimmunity, vasculopathy, and fibrosis involving the skin and multiple internal organs [1]. Because fibrosis accounts for as much as 45% of all deaths worldwide and appears to be increasing in prevalence [2], understanding its pathogenesis and progression is an urgent scientific challenge. Fibroblasts and myofibroblasts are the key effector cells executing physiologic tissue repair on one hand, and pathological fibrogenesis leading to chronic fibrosing conditions on the other. Recent studies identify innate immune signaling via toll-like receptors (TLRs) as a key driver of persistent fibrotic response in SSc. Repeated injury triggers the in-situ generation of "damage-associated molecular patterns" (DAMPs) or danger signals. Sensing of these danger signals by TLR4 on resident cells elicits potent stimulatory effects on fibrotic gene expression and myofibroblast differentiation triggering the self-limited tissue repair response to self-sustained pathological fibrosis characteristic of SSc. Our unbiased survey for DAMPs associated with SSc identified extracellular matrix glycoprotein tenascin-C as one of the most highly up-regulated ECM proteins in SSc skin and lung biopsies [3,4]. Furthermore, tenascin C is responsible for driving sustained fibroblasts activation, thereby progression of fibrosis [3]. This review summarizes recent studies examining the regulation and complex functional role of tenascin C, presenting tenascin-TLR4 axis in pathological fibrosis, and novel anti-fibrotic approaches targeting their signaling.
Collapse
|
15
|
Tucker RP, Degen M. Revisiting the Tenascins: Exploitable as Cancer Targets? Front Oncol 2022; 12:908247. [PMID: 35785162 PMCID: PMC9248440 DOI: 10.3389/fonc.2022.908247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
For their full manifestation, tumors require support from the surrounding tumor microenvironment (TME), which includes a specific extracellular matrix (ECM), vasculature, and a variety of non-malignant host cells. Together, these components form a tumor-permissive niche that significantly differs from physiological conditions. While the TME helps to promote tumor progression, its special composition also provides potential targets for anti-cancer therapy. Targeting tumor-specific ECM molecules and stromal cells or disrupting aberrant mesenchyme-cancer communications might normalize the TME and improve cancer treatment outcome. The tenascins are a family of large, multifunctional extracellular glycoproteins consisting of four members. Although each have been described to be expressed in the ECM surrounding cancer cells, tenascin-C and tenascin-W are currently the most promising candidates for exploitability and clinical use as they are highly expressed in various tumor stroma with relatively low abundance in healthy tissues. Here, we review what is known about expression of all four tenascin family members in tumors, followed by a more thorough discussion on tenascin-C and tenascin-W focusing on their oncogenic functions and their potential as diagnostic and/or targetable molecules for anti-cancer treatment purposes.
Collapse
Affiliation(s)
- Richard P. Tucker
- Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, CA, United States
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- *Correspondence: Martin Degen,
| |
Collapse
|
16
|
Liang G, Wang S, Shao J, Jin Y, Xu L, Yan Y, Günther S, Wang L, Offermanns S. Tenascin-X Mediates Flow-Induced Suppression of EndMT and Atherosclerosis. Circ Res 2022; 130:1647-1659. [PMID: 35443807 DOI: 10.1161/circresaha.121.320694] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial-to-mesenchymal transition (EndMT) has been identified as a critical driver of vascular inflammation and atherosclerosis, and TGF-β (transforming growth factor β) is a key mediator of EndMT. Both EndMT and atherosclerosis are promoted by disturbed flow, whereas unidirectional laminar flow limits EndMT and is atheroprotective. How EndMT and endothelial TGF-β signaling are regulated by different flow patterns is, however, still poorly understood. METHODS Flow chamber experiments in vitro and endothelium-specific knockout mice were used to study the role of tenascin-X in the regulation of EndMT and atherosclerosis as well as the underlying mechanisms. RESULTS In human endothelial cells as well as in human and mouse aortae, unidirectional laminar flow but not disturbed flow strongly increased endothelial expression of the extracellular matrix protein TN-X (tenascin-X) in a KLF4 (Krüppel-like factor 4) dependent manner. Mice with endothelium-specific loss of TN-X (EC-Tnxb-KO) showed increased endothelial TGF-β signaling as well as increased endothelial expression of EndMT and inflammatory marker genes. When EC-Tnxb-KO mice were subjected to partial carotid artery ligation, we observed increased vascular remodeling. EC-Tnxb-KO mice crossed to low-density lipoprotein receptor-deficient mice showed advanced atherosclerotic lesions after being fed a high-fat diet. Treatment of EC-Tnxb-KO mice with an anti-TGF-beta antibody or additional endothelial loss of TGF-beta receptors 1 and 2 normalized endothelial TGF-beta signaling and prevented EndMT. In in vitro studies, we found that TN-X through its fibrinogen-like domain directly interacts with TGF-β and thereby interferes with its binding to the TGF-β receptor. CONCLUSIONS In summary, we show that TN-X is a central mediator of flow-induced inhibition of EndMT, endothelial inflammation and atherogenesis, which functions by binding to and by blocking the activity of TGF-β. Our data identify a novel mechanism of flow-dependent regulation of vascular TGF-β, which holds promise for generating new strategies to prevent vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - ShengPeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (S.W., L.X.)
| | - Jingchen Shao
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - YoungJune Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - Liran Xu
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (S.W., L.X.)
| | - Yang Yan
- Department of Cardiovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, China (Y.Y.)
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Germany (S.G.)
| | - Lei Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.)
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Germany (G.L., J.S., Y.J., L.W., S.O.).,Center for Molecular Medicine, Goethe University Frankfurt, Germany (S.O.).,Cardiopulmonary Institute (CPI), Frankfurt/Bad Nauheim, Germany (S.O.).,German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany (S.O.)
| |
Collapse
|
17
|
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298:101530. [PMID: 34953859 PMCID: PMC8784641 DOI: 10.1016/j.jbc.2021.101530] [Citation(s) in RCA: 147] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Various forms of fibrosis, comprising tissue thickening and scarring, are involved in 40% of deaths across the world. Since the discovery of scarless functional healing in fetuses prior to a certain stage of development, scientists have attempted to replicate scarless wound healing in adults with little success. While the extracellular matrix (ECM), fibroblasts, and inflammatory mediators have been historically investigated as separate branches of biology, it has become increasingly necessary to consider them as parts of a complex and tightly regulated system that becomes dysregulated in fibrosis. With this new paradigm, revisiting fetal scarless wound healing provides a unique opportunity to better understand how this highly regulated system operates mechanistically. In the following review, we navigate the four stages of wound healing (hemostasis, inflammation, repair, and remodeling) against the backdrop of adult versus fetal wound healing, while also exploring the relationships between the ECM, effector cells, and signaling molecules. We conclude by singling out recent findings that offer promising leads to alter the dynamics between the ECM, fibroblasts, and inflammation to promote scarless healing. One factor that promises to be significant is fibroblast heterogeneity and how certain fibroblast subpopulations might be predisposed to scarless healing. Altogether, reconsidering fetal wound healing by examining the interplay of the various factors contributing to fibrosis provides new research directions that will hopefully help us better understand and address fibroproliferative diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic sclerosis, progressive kidney disease, and cardiovascular fibrosis.
Collapse
Affiliation(s)
- Leandro Moretti
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jack Stalfort
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas Harrison Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
18
|
Matricellular proteins in intrahepatic cholangiocarcinoma. Adv Cancer Res 2022; 156:249-281. [DOI: 10.1016/bs.acr.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
Stuelten CH, Zhang YE. Transforming Growth Factor-β: An Agent of Change in the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:764727. [PMID: 34712672 PMCID: PMC8545984 DOI: 10.3389/fcell.2021.764727] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming Growth Factor-β (TGF-β) is a key regulator of embryonic development, adult tissue homeostasis, and lesion repair. In tumors, TGF-β is a potent inhibitor of early stage tumorigenesis and promotes late stage tumor progression and metastasis. Here, we review the roles of TGF-β as well as components of its signaling pathways in tumorigenesis. We will discuss how a core property of TGF-β, namely its ability to change cell differentiation, leads to the transition of epithelial cells, endothelial cells and fibroblasts to a myofibroblastoid phenotype, changes differentiation and polarization of immune cells, and induces metabolic reprogramming of cells, all of which contribute to the progression of epithelial tumors.
Collapse
Affiliation(s)
- Christina H. Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Ying E. Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|