251
|
Hu B, Wang J, Chen J, Zhao L, Li X. The heterogeneity of fibroblasts in laryngotracheal stenosis and skin hypertrophic scar in pediatric patients. Int J Pediatr Otorhinolaryngol 2021; 145:110709. [PMID: 33910042 DOI: 10.1016/j.ijporl.2021.110709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/07/2021] [Accepted: 04/05/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To investigate the heterogeneity between the laryngotracheal stenosis and hypertrophic scar derived fibroblasts. METHODS Human laryngotracheal stenosis (LTS) and skin hypertrophic scar (HTS) specimens were obtained during the tracheal resection and T-shaped tracheal stent implantation surgery. Fibroblasts were isolated and cultured. Cell proliferation and migration were analyzed by cell count, EdU proliferation and wound-healing assays. The expressions of COL1a1, α-SMA, TGF-β1 signaling pathway, chemokines and receptors were analyzed by qRT-PCR, western blotting, and immunohistochemistry. RESULTS Cell proliferation and migration of LTS derived fibroblasts were significantly faster than HTS fibroblasts, with no significant difference of the percentage of apoptotic cells. COL1a1, α-SMA, and Integrins were down-regulated in LTS fibroblasts, but TGFB1 and chemokine receptor CXCR7 were up-regulated in LTS fibroblasts. However, the expressions of SMAD4 and phospho-SMAD2/3 were not significantly different. CONCLUSIONS Human LTS and HTS derived fibroblasts differ in cell proliferation and migration. Different expressions of COL1a1, α-SMA, and CXCR7 were found between the two fibroblasts.
Collapse
Affiliation(s)
- Bin Hu
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, People's Republic of China.
| | - Jing Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, People's Republic of China.
| | - Jiarui Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, People's Republic of China.
| | - Limin Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, People's Republic of China.
| | - Xiaoyan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200062, People's Republic of China.
| |
Collapse
|
252
|
Monika P, Waiker PV, Chandraprabha MN, Rangarajan A, Murthy KNC. Myofibroblast progeny in wound biology and wound healing studies. Wound Repair Regen 2021; 29:531-547. [PMID: 34009713 DOI: 10.1111/wrr.12937] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 12/19/2022]
Abstract
Fibroblasts and myofibroblasts play a myriad of important roles in human tissue function, especially in wound repair and healing. Among all cells, fibroblasts are group of cells that decide the status of wound as they maintain tissue homeostasis. Currently, the increase in the deleterious effects of chronic wound and their morbidity rate has necessitated the need to understand the influence of fibroblasts and myofibroblasts, which chiefly originate locally from tissue-resident fibroblasts to address the same. Wound pathophysiology is complex, herein, we have discussed fibroblast and myofibroblast heterogeneity in skin and different organs by understanding the phenotypical and functional properties of each of its sub-populations in the process of wound healing. Recent advancements in fibroblast activation, differentiation to myofibroblasts, proliferation and migration are discussed in detail. Fibroblasts and myofibroblasts are key players in wound healing and wound remodelling, respectively, and their significance in wound repair is discussed. An increased understanding of their biology during wound healing also gives an opportunity to explore more of fibroblast and myofibroblast focused therapies to treat chronic wounds which are clinical challenges. In this regard, in the current review, we have described the different methods for isolation of primary fibroblasts and myofibroblasts from both animal models and humans, and their characterization. Additionally, we have also provided details on possible molecular targets for better understanding of prognosis, diagnosis and treatment of chronic wounds. Information will help both researchers and clinicians in providing molecular insight that enable them for effective chronic wound management. The knowledge of intimate dialogue between the fibroblast, sub-populations like, myofibroblast and their microenvironment, will serve useful in determining novel, efficient and specific therapeutic targets to treat pathological wound conditions.
Collapse
Affiliation(s)
- Prakash Monika
- Department of Biotechnology, M. S. Ramaiah Institute of Technology, Bangalore, India
| | | | | | - Annapoorni Rangarajan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | | |
Collapse
|
253
|
Goss G, Rognoni E, Salameti V, Watt FM. Distinct Fibroblast Lineages Give Rise to NG2+ Pericyte Populations in Mouse Skin Development and Repair. Front Cell Dev Biol 2021; 9:675080. [PMID: 34124060 PMCID: PMC8194079 DOI: 10.3389/fcell.2021.675080] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
We have examined the developmental origins of Ng2+ perivascular cell populations that adhere to the basement membrane of blood vessels, and their contribution to wound healing. Neural/glial antigen 2 (Ng2) labeled most perivascular cells (70-80%) in developing and adult mouse back skin, a higher proportion than expressed by other pericyte markers Tbx18, Nestin and Pdgfrβ. In adult mouse back skin Ng2+ perivascular cells could be categorized into 4 populations based on whether they expressed Pdgfrα and Pdgfrβ individually or in combination or were Pdgfr-negative. Lineage tracing demonstrated that although Ng2+ cells in embryonic and neonatal back skin contributed to multiple cell types they did not give rise to interfollicular fibroblasts within the dermis. Lineage tracing of distinct fibroblast populations during skin development showed that papillary fibroblasts (Lrig1+) gave rise to Ng2+ perivascular cells in the upper dermis, whilst Ng2+ perivascular cells in the lower dermis were primarily derived from reticular Dlk1+ fibroblasts. Following wounding of adult skin, Ng2+ dermal cells only give rise to Ng2+ blood vessel associated cells and did not contribute to other fibroblast lineages. The relative abundance of Ng2+ Pdgfrβ+ perivascular populations was comparable in wounded and non-wounded skin, indicating that perivascular heterogeneity was maintained during full thickness skin repair. In the wound bed Ng2+ perivascular populations were primarily derived from Lrig1+ papillary or Dlk1+ reticular fibroblast lineages, according to the location of the regenerating blood vessels. We conclude that Ng2+ perivascular cells represent a heterogeneous lineage restricted population that is primarily recruited from the papillary or reticular fibroblast lineages during tissue regeneration.
Collapse
Affiliation(s)
| | | | | | - Fiona M. Watt
- Centre for Stem Cells and Regenerative Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| |
Collapse
|
254
|
Guimarães GR, Almeida PP, de Oliveira Santos L, Rodrigues LP, de Carvalho JL, Boroni M. Hallmarks of Aging in Macrophages: Consequences to Skin Inflammaging. Cells 2021; 10:cells10061323. [PMID: 34073434 PMCID: PMC8228751 DOI: 10.3390/cells10061323] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/22/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is our largest organ and the outermost protective barrier. Its aging reflects both intrinsic and extrinsic processes resulting from the constant insults it is exposed to. Aging in the skin is accompanied by specific epigenetic modifications, accumulation of senescent cells, reduced cellular proliferation/tissue renewal, altered extracellular matrix, and a proinflammatory environment favoring undesirable conditions, including disease onset. Macrophages (Mφ) are the most abundant immune cell type in the skin and comprise a group of heterogeneous and plastic cells that are key for skin homeostasis and host defense. However, they have also been implicated in orchestrating chronic inflammation during aging. Since Mφ are related to innate and adaptive immunity, it is possible that age-modified skin Mφ promote adaptive immunity exacerbation and exhaustion, favoring the emergence of proinflammatory pathologies, such as skin cancer. In this review, we will highlight recent findings pertaining to the effects of aging hallmarks over Mφ, supporting the recognition of such cell types as a driving force in skin inflammaging and age-related diseases. We will also present recent research targeting Mφ as potential therapeutic interventions in inflammatory skin disorders and cancer.
Collapse
Affiliation(s)
- Gabriela Rapozo Guimarães
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Palloma Porto Almeida
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Leandro de Oliveira Santos
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
| | - Leane Perim Rodrigues
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasilia 70790-160, Brazil; (L.P.R.); (J.L.d.C.)
| | - Juliana Lott de Carvalho
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasilia 70790-160, Brazil; (L.P.R.); (J.L.d.C.)
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Mariana Boroni
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil; (G.R.G.); (P.P.A.); (L.d.O.S.)
- Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas 13083-970, Brazil
- Correspondence:
| |
Collapse
|
255
|
Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev 2021; 302:147-162. [PMID: 34036608 DOI: 10.1111/imr.12972] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022]
Abstract
Fibroblastic stromal cells are as diverse, in origin and function, as the niches they fashion in the mammalian body. This cellular variety impacts the spectrum of responses elicited by the immune system. Fibroblast influence on the immune system keeps evolving our perspective on fibroblast roles and functions beyond just a passive structural part of organs. This review discusses the foundations of fibroblastic stromal-immune crosstalk, under the scope of stromal heterogeneity as a basis for tissue-specific tutoring of the immune system. Focusing on the skin as a relevant immunological organ, we detail the complex interactions between distinct fibroblast populations and immune cells that occur during homeostasis, injury repair, scarring, and disease. We further review the relevance of fibroblastic stromal cell heterogeneity and how this heterogeneity is central to regulate the immune system from its inception during embryonic development into adulthood.
Collapse
Affiliation(s)
- Donovan Correa-Gallegos
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Dongsheng Jiang
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
256
|
Mechanical and Immunological Regulation in Wound Healing and Skin Reconstruction. Int J Mol Sci 2021; 22:ijms22115474. [PMID: 34067386 PMCID: PMC8197020 DOI: 10.3390/ijms22115474] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
In the past decade, a new frontier in scarless wound healing has arisen because of significant advances in the field of wound healing realised by incorporating emerging concepts from mechanobiology and immunology. The complete integumentary organ system (IOS) regeneration and scarless wound healing mechanism, which occurs in specific species, body sites and developmental stages, clearly shows that mechanical stress signals and immune responses play important roles in determining the wound healing mode. Advances in tissue engineering technology have led to the production of novel human skin equivalents and organoids that reproduce cell–cell interactions with tissue-scale tensional homeostasis, and enable us to evaluate skin tissue morphology, functionality, drug response and wound healing. This breakthrough in tissue engineering has the potential to accelerate the understanding of wound healing control mechanisms through complex mechanobiological and immunological interactions. In this review, we present an overview of recent studies of biomechanical and immunological wound healing and tissue remodelling mechanisms through comparisons of species- and developmental stage-dependent wound healing mechanisms. We also discuss the possibility of elucidating the control mechanism of wound healing involving mechanobiological and immunological interaction by using next-generation human skin equivalents.
Collapse
|
257
|
Distinct fibroblasts in scars and regeneration. Curr Opin Genet Dev 2021; 70:7-14. [PMID: 34022662 DOI: 10.1016/j.gde.2021.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/12/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022]
Abstract
The skin is home to a collection of fibroblastic cell types from varying embryonic origins. These varying fibroblastic lineages display unique genetic programs and in vivo functions. Studying the diversity of fibroblastic cells is emerging as an important area for cutaneous biology, wound repair and regenerative medicine. In this mini-review we discuss the distinct embryonic origins, microenvironments, and transcriptomic profiles of fibroblastic lineages, and how these varying lineages shape the skin's wound response across injury depths, anatomic locations, and developmental time to promote either scarring or regeneration. We outline how the development of single cell sequencing has led to our improved understanding of fibroblastic lineages at the molecular level and discuss existing challenges and future outlook on developing regenerative therapies that are based on this emerging field of eclectic fibroblasts.
Collapse
|
258
|
An integrated analysis of human myeloid cells identifies gaps in in vitro models of in vivo biology. Stem Cell Reports 2021; 16:1629-1643. [PMID: 33989517 PMCID: PMC8190595 DOI: 10.1016/j.stemcr.2021.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
The Stemformatics myeloid atlas is an integrated transcriptome atlas of human macrophages and dendritic cells that systematically compares freshly isolated tissue-resident, cultured, and pluripotent stem cell–derived myeloid cells. Three classes of tissue-resident macrophage were identified: Kupffer cells and microglia; monocyte-associated; and tumor-associated macrophages. Culture had a major impact on all primary cell phenotypes. Pluripotent stem cell–derived macrophages were characterized by atypical expression of collagen and a highly efferocytotic phenotype. Myeloid subsets, and phenotypes associated with derivation, were reproducible across experimental series including data projected from single-cell studies, demonstrating that the atlas provides a robust reference for myeloid phenotypes. Implementation in Stemformatics.org allows users to visualize patterns of sample grouping or gene expression for user-selected conditions and supports temporary upload of your own microarray or RNA sequencing samples, including single-cell data, to benchmark against the atlas. A reference transcriptome atlas for human macrophage biology Culture alters primary myeloid phenotypes Pluripotent stem cell–derived macrophages retain a common stromal signature FLT3L-derived cord blood DCs lack expression of key pattern recognition receptors
Collapse
|
259
|
Schuster R, Rockel JS, Kapoor M, Hinz B. The inflammatory speech of fibroblasts. Immunol Rev 2021; 302:126-146. [PMID: 33987902 DOI: 10.1111/imr.12971] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Activation of fibroblasts is a key event during normal tissue repair after injury and the dysregulated repair processes that result in organ fibrosis. To most researchers, fibroblasts are rather unremarkable spindle-shaped cells embedded in the fibrous collagen matrix of connective tissues and/or deemed useful to perform mechanistic studies with adherent cells in culture. For more than a century, fibroblasts escaped thorough classification due to the lack of specific markers and were treated as the leftovers after all other cells have been identified from a tissue sample. With novel cell lineage tracing and single cell transcriptomics tools, bona fide fibroblasts emerge as only one heterogeneous sub-population of a much larger group of partly overlapping cell types, including mesenchymal stromal cells, fibro-adipogenic progenitor cells, pericytes, and/or perivascular cells. All these cells are activated to contribute to tissue repair after injury and/or chronic inflammation. "Activation" can entail various functions, such as enhanced proliferation, migration, instruction of inflammatory cells, secretion of extracellular matrix proteins and organizing enzymes, and acquisition of a contractile myofibroblast phenotype. We provide our view on the fibroblastic cell types and activation states playing a role during physiological and pathological repair and their crosstalk with inflammatory macrophages. Inflammation and fibrosis of the articular synovium during rheumatoid arthritis and osteoarthritis are used as specific examples to discuss inflammatory fibroblast phenotypes. Ultimately, delineating the precursors and functional roles of activated fibroblastic cells will contribute to better and more specific intervention strategies to treat fibroproliferative and fibrocontractive disorders.
Collapse
Affiliation(s)
- Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,PhenomicAI, MaRS Centre, Toronto, ON, Canada
| | - Jason S Rockel
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
260
|
Mascharak S, desJardins-Park HE, Davitt MF, Griffin M, Borrelli MR, Moore AL, Chen K, Duoto B, Chinta M, Foster DS, Shen AH, Januszyk M, Kwon SH, Wernig G, Wan DC, Lorenz HP, Gurtner GC, Longaker MT. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 2021; 372:372/6540/eaba2374. [PMID: 33888614 DOI: 10.1126/science.aba2374] [Citation(s) in RCA: 327] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 10/01/2020] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Skin scarring, the end result of adult wound healing, is detrimental to tissue form and function. Engrailed-1 lineage-positive fibroblasts (EPFs) are known to function in scarring, but Engrailed-1 lineage-negative fibroblasts (ENFs) remain poorly characterized. Using cell transplantation and transgenic mouse models, we identified a dermal ENF subpopulation that gives rise to postnatally derived EPFs by activating Engrailed-1 expression during adult wound healing. By studying ENF responses to substrate mechanics, we found that mechanical tension drives Engrailed-1 activation via canonical mechanotransduction signaling. Finally, we showed that blocking mechanotransduction signaling with either verteporfin, an inhibitor of Yes-associated protein (YAP), or fibroblast-specific transgenic YAP knockout prevents Engrailed-1 activation and promotes wound regeneration by ENFs, with recovery of skin appendages, ultrastructure, and mechanical strength. This finding suggests that there are two possible outcomes to postnatal wound healing: a fibrotic response (EPF-mediated) and a regenerative response (ENF-mediated).
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heather E desJardins-Park
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael F Davitt
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alessandra L Moore
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bryan Duoto
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Malini Chinta
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deshka S Foster
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Abra H Shen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sun Hyung Kwon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gerlinde Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Peter Lorenz
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
261
|
Arai H, Sato Y, Yanagita M. Fibroblast heterogeneity and tertiary lymphoid tissues in the kidney. Immunol Rev 2021; 302:196-210. [PMID: 33951198 PMCID: PMC8360208 DOI: 10.1111/imr.12969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Fibroblasts reside in various organs and support tissue structure and homeostasis under physiological conditions. Phenotypic alterations of fibroblasts underlie the development of diverse pathological conditions, including organ fibrosis. Recent advances in single‐cell biology have revealed that fibroblasts comprise heterogeneous subpopulations with distinct phenotypes, which exert both beneficial and detrimental effects on the host organs in a context‐dependent manner. In the kidney, phenotypic alterations of resident fibroblasts provoke common pathological conditions of chronic kidney disease (CKD), such as renal anemia and peritubular capillary loss. Additionally, in aged injured kidneys, fibroblasts provide functional and structural supports for tertiary lymphoid tissues (TLTs), which serve as the ectopic site of acquired immune reactions in various clinical contexts. TLTs are closely associated with aging and CKD progression, and the developmental stages of TLTs reflect the severity of renal injury. In this review, we describe the current understanding of fibroblast heterogeneity both under physiological and pathological conditions, with special emphasis on fibroblast contribution to TLT formation in the kidney. Dissecting the heterogeneous characteristics of fibroblasts will provide a promising therapeutic option for fibroblast‐related pathological conditions, including TLT formation.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Sato
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Medical Innovation Center, TMK Project, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
262
|
Abstract
Tissue-resident macrophages are present in most tissues with developmental, self-renewal, or functional attributes that do not easily fit into a textbook picture of a plastic and multifunctional macrophage originating from hematopoietic stem cells; nor does it fit a pro- versus anti-inflammatory paradigm. This review presents and discusses current knowledge on the developmental biology of macrophages from an evolutionary perspective focused on the function of macrophages, which may aid in study of developmental, inflammatory, tumoral, and degenerative diseases. We also propose a framework to investigate the functions of macrophages in vivo and discuss how inherited germline and somatic mutations may contribute to the roles of macrophages in diseases.
Collapse
Affiliation(s)
- Nehemiah Cox
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
263
|
Miah M, Goh I, Haniffa M. Prenatal Development and Function of Human Mononuclear Phagocytes. Front Cell Dev Biol 2021; 9:649937. [PMID: 33898444 PMCID: PMC8060508 DOI: 10.3389/fcell.2021.649937] [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: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
The human mononuclear phagocyte (MP) system, which includes dendritic cells, monocytes, and macrophages, is a critical regulator of innate and adaptive immune responses. During embryonic development, MPs derive sequentially in yolk sac progenitors, fetal liver, and bone marrow haematopoietic stem cells. MPs maintain tissue homeostasis and confer protective immunity in post-natal life. Recent evidence - primarily in animal models - highlight their critical role in coordinating the remodeling, maturation, and repair of target organs during embryonic and fetal development. However, the molecular regulation governing chemotaxis, homeostasis, and functional diversification of resident MP cells in their respective organ systems during development remains elusive. In this review, we summarize the current understanding of the development and functional contribution of tissue MPs during human organ development and morphogenesis and its relevance to regenerative medicine. We outline how single-cell multi-omic approaches and next-generation ex-vivo organ-on-chip models provide new experimental platforms to study the role of human MPs during development and disease.
Collapse
Affiliation(s)
- Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
264
|
Culley OJ, Louis B, Philippeos C, Oulès B, Tihy M, Segal JM, Hyliands D, Jenkins G, Bhogal RK, Siow RC, Watt FM. Differential Expression of Insulin-Like Growth Factor 1 and Wnt Family Member 4 Correlates With Functional Heterogeneity of Human Dermal Fibroblasts. Front Cell Dev Biol 2021; 9:628039. [PMID: 33889572 PMCID: PMC8056032 DOI: 10.3389/fcell.2021.628039] [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: 11/10/2020] [Accepted: 03/09/2021] [Indexed: 11/22/2022] Open
Abstract
Although human dermis contains distinct fibroblast subpopulations, the functional heterogeneity of fibroblast lines from different donors is under-appreciated. We identified one commercially sourced fibroblast line (c64a) that failed to express α-smooth muscle actin (α-SMA), a marker linked to fibroblast contractility, even when treated with transforming growth factor-β1 (TGF-β1). Gene expression profiling identified insulin-like growth factor 1 (IGF1) as being expressed more highly, and Asporin (ASPN) and Wnt family member 4 (WNT4) expressed at lower levels, in c64a fibroblasts compared to three fibroblast lines that had been generated in-house, independent of TGF-β1 treatment. TGF-β1 increased expression of C-X-C motif chemokine ligand 1 (CXCL1) in c64a cells to a greater extent than in the other lines. The c64a gene expression profile did not correspond to any dermal fibroblast subpopulation identified by single-cell RNAseq of freshly isolated human skin cells. In skin reconstitution assays, c64a fibroblasts did not support epidermal stratification as effectively as other lines tested. In fibroblast lines generated in-house, shRNA-mediated knockdown of IGF1 increased α-SMA expression without affecting epidermal stratification. Conversely, WNT4 knockdown had no consistent effect on α-SMA expression, but increased the ability of fibroblasts to support epidermal stratification. Thus, by comparing the properties of different lines of cultured dermal fibroblasts, we have identified IGF1 and WNT4 as candidate mediators of two distinct dermal functions: myofibroblast formation and epidermal maintenance.
Collapse
Affiliation(s)
- Oliver J Culley
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Blaise Louis
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Christina Philippeos
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Bénédicte Oulès
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Matthieu Tihy
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Joe M Segal
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| | - Della Hyliands
- Unilever R&D Colworth, Colworth Science Park, Bedford, United Kingdom
| | - Gail Jenkins
- Unilever R&D Colworth, Colworth Science Park, Bedford, United Kingdom
| | - Ranjit K Bhogal
- Unilever R&D Colworth, Colworth Science Park, Bedford, United Kingdom
| | - Richard C Siow
- Cardiovascular Division, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
265
|
Ramos R, Plikus MV. Moving On after Trauma: Fibroblasts Thrive in the Right Environment. Cell Stem Cell 2021; 27:349-351. [PMID: 32888421 DOI: 10.1016/j.stem.2020.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Large skin wounds in adult mice can heal by regenerating new hair follicles and adipocytes in their center. In this issue of Cell Stem Cell, Abbasi et al. (2020) report how typically scar-forming wound fibroblasts enter a transcriptionally distinct single-cell state of competence during regeneration of new hair follicle mesenchyme.
Collapse
Affiliation(s)
- Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
266
|
Cooper PO, Haas MR, Noonepalle SKR, Shook BA. Dermal Drivers of Injury-Induced Inflammation: Contribution of Adipocytes and Fibroblasts. Int J Mol Sci 2021; 22:1933. [PMID: 33669239 PMCID: PMC7919834 DOI: 10.3390/ijms22041933] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
Irregular inflammatory responses are a major contributor to tissue dysfunction and inefficient repair. Skin has proven to be a powerful model to study mechanisms that regulate inflammation. In particular, skin wound healing is dependent on a rapid, robust immune response and subsequent dampening of inflammatory signaling. While injury-induced inflammation has historically been attributed to keratinocytes and immune cells, a vast body of evidence supports the ability of non-immune cells to coordinate inflammation in numerous tissues and diseases. In this review, we concentrate on the active participation of tissue-resident adipocytes and fibroblasts in pro-inflammatory signaling after injury, and how altered cellular communication from these cells can contribute to irregular inflammation associated with aberrant wound healing. Furthering our understanding of how tissue-resident mesenchymal cells contribute to inflammation will likely reveal new targets that can be manipulated to regulate inflammation and repair.
Collapse
Affiliation(s)
| | | | | | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA; (P.O.C.); (M.R.H.); (S.k.R.N.)
| |
Collapse
|
267
|
Spatz LB, Jin RU, Mills JC. Cellular plasticity at the nexus of development and disease. Development 2021; 148:148/3/dev197392. [PMID: 33547203 DOI: 10.1242/dev.197392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
In October 2020, the Keystone Symposia Global Health Series hosted a Keystone eSymposia entitled 'Tissue Plasticity: Preservation and Alteration of Cellular Identity'. The event synthesized groundbreaking research from unusually diverse fields of study, presented in various formats, including live and virtual talks, panel discussions and interactive e-poster sessions. The meeting focused on cell identity changes and plasticity in multiple tissues, species and developmental contexts, both in homeostasis and during injury. Here, we review the key themes of the meeting: (1) cell-extrinsic drivers of plasticity; (2) epigenomic regulation of cell plasticity; and (3) conserved mechanisms governing plasticity. A salient take-home conclusion was that there may be conserved mechanisms used by cells to execute plasticity, with autodegradative activity (autophagy and lysosomes) playing a crucial initial step in diverse organs and organisms.
Collapse
Affiliation(s)
- Lillian B Spatz
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Ramon U Jin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jason C Mills
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA .,Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
268
|
Witherel CE, Sao K, Brisson BK, Han B, Volk SW, Petrie RJ, Han L, Spiller KL. Regulation of extracellular matrix assembly and structure by hybrid M1/M2 macrophages. Biomaterials 2021; 269:120667. [PMID: 33450585 PMCID: PMC7870567 DOI: 10.1016/j.biomaterials.2021.120667] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
Aberrant extracellular matrix (ECM) assembly surrounding implanted biomaterials is the hallmark of the foreign body response, in which implants become encapsulated in thick fibrous tissue that prevents their proper function. While macrophages are known regulators of fibroblast behavior, how their phenotype influences ECM assembly and the progression of the foreign body response is poorly understood. In this study, we used in vitro models with physiologically relevant macrophage phenotypes, as well as controlled release of macrophage-modulating cytokines from gelatin hydrogels implanted subcutaneously in vivo to investigate the role of macrophages in ECM assembly. Primary human macrophages were polarized to four distinct phenotypes, which have each been associated with fibrosis, including pro-inflammatory M1, pro-healing M2, and a hybrid M1/M2, generated by exposing macrophages to M1-and M2-promoting stimuli simultaneously. Additionally, macrophages were first polarized to M1 and then to M2 (M1→M2) to generate a phenotype typically observed during normal wound healing. Human dermal fibroblasts that were cultured in macrophage-conditioned media upregulated numerous genes involved in regulation of ECM assembly, especially in M2-conditioned media. Hybrid M1/M2 macrophage-conditioned media caused fibroblasts to produce a matrix with thicker and less aligned fibers, while M2 macrophage-conditioned media caused the formation of a more aligned matrix with thinner fibers. Gelatin methacrylate hydrogels containing interleukin-4 (IL4) and IL13-loaded poly(lactic-co-glycolic acid) (PLGA) microparticles were designed to promote the M2 phenotype in a murine subcutaneous in vivo model. NanoString multiplex gene expression analysis of hydrogel explants showed that hydrogels without cytokines caused mostly M1 phenotype markers to be highly expressed at an early time point (3 days), but the release of IL4+IL13 promoted upregulation of M2 markers and genes associated with regulation of ECM assembly, such as Col5a1 and Col6a1. Biochemical analysis and second harmonic generation microscopy showed that the release of IL4+IL13 increased total sulfated glycosaminoglycan content and decreased fibril alignment, which is typically associated with less fibrotic tissue. Together, these results show that hybrid M1/M2 macrophages regulate ECM assembly, and that shifting the balance towards M2 may promote architectural and compositional changes in ECM with enhanced potential for downstream remodeling.
Collapse
Affiliation(s)
- Claire E Witherel
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kimheak Sao
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Becky K Brisson
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Biao Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Susan W Volk
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Ryan J Petrie
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
269
|
Luo G, Sun Y, Zhang J, Xu Z, Lu W, Wang H, Zhang Y, Li H, Mao Z, Ye S, Cheng B, Fang X. Nanodefensin-encased hydrogel with dual bactericidal and pro-regenerative functions for advanced wound therapy. Am J Cancer Res 2021; 11:3642-3660. [PMID: 33664853 PMCID: PMC7914350 DOI: 10.7150/thno.53089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/31/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Host defense peptides (HDPs) have emerged as a novel therapeutic paradigm for wound management; however, their clinical applications remain a challenge owing to their poor pharmacological properties and lack of suitable pharmaceutical formulations. Nanodefensin (ND), a nanoengineered human α-defensin 5 (HD5), has shown improved pharmacological properties relative to the parent compound. In this study, we engineered a nanodefensin-encased hydrogel (NDEFgel), investigated the effects of NDEFgel on wound healing, and elucidated underlying mechanisms. Method: ND was chemically synthesized and tested functions by in vitro antimicrobial and scratch assays and western blotting. Different NDEFgels were evaluated by in vitro characterizations including degradation, drug release and antimicrobial activity. In full-thickness excisional murine models, the optimal NDEFgel was directly applied onto wound sites, and the efficacy was assessed. Moreover, the underlying mechanisms of pro-regenerative effect developed by NDEFgel were also explored. Results: Apart from bactericidal effects, ND modulated fibroblast behaviors by promoting migration and differentiation. Among the tested hydrogels, the Pluronic F127 (Plu) hydrogel represented the most desirable carrier for ND delivery owing to its favorable controlled release and compatibility with ND. Local treatment of NDEFgel on the wound bed resulted in accelerated wound regeneration and attenuated bacterial burden. We further demonstrated that NDEFgel therapy significantly upregulated genes related to collagen deposition and fibroblasts, and increased the expression of myofibroblasts and Rac1. We therefore found that Rac1 is a critical factor in the ND-induced modulation of fibroblast behaviors in vitro through a Rac1-dependent cytoskeletal rearrangement. Conclusion: Our results indicate that NDEFgel may be a promising dual-action therapeutic option for advanced wound management in the future.
Collapse
|
270
|
Best KT, Korcari A, Mora KE, Nichols AE, Muscat SN, Knapp E, Buckley MR, Loiselle AE. Scleraxis-lineage cell depletion improves tendon healing and disrupts adult tendon homeostasis. eLife 2021; 10:62203. [PMID: 33480357 PMCID: PMC7850622 DOI: 10.7554/elife.62203] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Despite the requirement for Scleraxis-lineage (ScxLin) cells during tendon development, the function of ScxLin cells during adult tendon repair, post-natal growth, and adult homeostasis have not been defined. Therefore, we inducibly depleted ScxLin cells (ScxLinDTR) prior to tendon injury and repair surgery and hypothesized that ScxLinDTR mice would exhibit functionally deficient healing compared to wild-type littermates. Surprisingly, depletion of ScxLin cells resulted in increased biomechanical properties without impairments in gliding function at 28 days post-repair, indicative of regeneration. RNA sequencing of day 28 post-repair tendons highlighted differences in matrix-related genes, cell motility, cytoskeletal organization, and metabolism. We also utilized ScxLinDTR mice to define the effects on post-natal tendon growth and adult tendon homeostasis and discovered that adult ScxLin cell depletion resulted in altered tendon collagen fibril diameter, density, and dispersion. Collectively, these findings enhance our fundamental understanding of tendon cell localization, function, and fate during healing, growth, and homeostasis.
Collapse
Affiliation(s)
- Katherine T Best
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Antonion Korcari
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Department of Biomedical Engineering, University of Rochester, New York, United States
| | - Keshia E Mora
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Department of Biomedical Engineering, University of Rochester, New York, United States
| | - Anne Ec Nichols
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Samantha N Muscat
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Emma Knapp
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Mark R Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Department of Biomedical Engineering, University of Rochester, New York, United States
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Department of Biomedical Engineering, University of Rochester, New York, United States
| |
Collapse
|
271
|
Multi-faceted enhancement of full-thickness skin wound healing by treatment with autologous micro skin tissue columns. Sci Rep 2021; 11:1688. [PMID: 33462350 PMCID: PMC7814113 DOI: 10.1038/s41598-021-81179-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Impaired wound healing is an immense medical challenge, and while autologous skin grafting remains the "gold-standard" therapeutic option for repairing wounds that cannot be closed by primary or secondary intention, it is limited by substantial donor site morbidity. We previously developed the alternative approach of harvesting full-thickness skin tissue in the form of "micro skin tissue columns" (MSTCs), without causing scarring or any other long-term morbidity. In this study we investigated how MSTC treatment affects the different cellular processes involved in wound healing. We found that MSTC-derived cells were able to remodel and repopulate the wound volume, and positively impact multiple aspects of the wound healing process, including accelerating re-epithelialization by providing multiple cell sources throughout the wound area, increasing collagen deposition, enhancing dermal remodeling, and attenuating the inflammatory response. These effects combined to enhance both epidermal and dermal wound healing. This MSTC treatment approach was designed for practical clinical use, could convey many benefits of autologous skin grafting, and avoids the major drawback of donor site morbidity.
Collapse
|
272
|
Zhang J, Liu N, Wu X, Wu P, Song N, Ma J. Identification of differentially expressed circular RNAs in keloid and normal skin tissue by high-throughput sequencing. Dermatol Ther 2021; 34:e14745. [PMID: 33405341 DOI: 10.1111/dth.14745] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/18/2023]
Abstract
Keloid is a kind of pathological skin scar with unclear molecular pathology. Circular RNAs (circRNAs) are involved in the occurrence and development of many diseases; however, their relationship with keloid is not well understood. To investigate the involvement of dysregulated circRNAs in keloid. Thirty-seven keloids and 37 normal skin tissues were collected, and the changes of circRNAs, microRNAs (miRNAs) and mRNAs in 3 keloids and 3 normal samples by high-throughput sequencing were detected first. Based on the circRNA-miRNA-mRNA interaction network construction, gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis combining several signaling pathways associated with keloid formation and progression, the circRNAs required further verification were screened out. The expression levels of the selected circRNAs were verified in 37 keloids and 37 normal skin tissues using quantitative real-time polymerase chain reaction (QPCR). The interaction of candidate circRNA and its predicted binding miRNA was tested by dual-luciferase reporter gene experiment. Compared with normal controls, there was an average of 120 and 12 circRNAs, 44 and 63 miRNAs, 656 and 156 mRNAs were upregulated and downregulated, respectively, in keloids. According to the analysis of bioinformation, six circRNAs were picked out. The QPCR validation results of two upregulated circRNAs (hsa_circ_0001320 and circCOL5A1) were consistent with previous sequencing results. The interaction between hsa_circ_0001320 and miR-574-5p was confirmed. This study makes it clear that the abnormal expression of circRNAs may be related to the pathological process of keloid.
Collapse
Affiliation(s)
- Jing Zhang
- Departments of Facial Plastic Surgery, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Ninghua Liu
- Departments of Facial Plastic Surgery, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Xiufa Wu
- Otolaryngology Research Institute,Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Peixuan Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nan Song
- Departments of Facial Plastic Surgery, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Jing Ma
- Departments of Facial Plastic Surgery, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.,Otolaryngology Research Institute,Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| |
Collapse
|
273
|
Muñoz-Rojas AR, Kelsey I, Pappalardo JL, Chen M, Miller-Jensen K. Co-stimulation with opposing macrophage polarization cues leads to orthogonal secretion programs in individual cells. Nat Commun 2021; 12:301. [PMID: 33436596 PMCID: PMC7804107 DOI: 10.1038/s41467-020-20540-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Macrophages are innate immune cells that contribute to fighting infections, tissue repair, and maintaining tissue homeostasis. To enable such functional diversity, macrophages resolve potentially conflicting cues in the microenvironment via mechanisms that are unclear. Here, we use single-cell RNA sequencing to explore how individual macrophages respond when co-stimulated with inflammatory stimuli LPS and IFN-γ and the resolving cytokine IL-4. These co-stimulated macrophages display a distinct global transcriptional program. However, variable negative cross-regulation between some LPS + IFN-γ-specific and IL-4-specific genes results in cell-to-cell heterogeneity in transcription. Interestingly, negative cross-regulation leads to mutually exclusive expression of the T-cell-polarizing cytokine genes Il6 and Il12b versus the IL-4-associated factors Arg1 and Chil3 in single co-stimulated macrophages, and single-cell secretion measurements show that these specialized functions are maintained for at least 48 h. This study suggests that increasing functional diversity in the population is one strategy macrophages use to respond to conflicting environmental cues.
Collapse
Affiliation(s)
- Andrés R Muñoz-Rojas
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ilana Kelsey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jenna L Pappalardo
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Meibin Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA.
- Systems Biology Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
274
|
Gao R, Peng X, Perry C, Sun H, Ntokou A, Ryu C, Gomez JL, Reeves BC, Walia A, Kaminski N, Neumark N, Ishikawa G, Black KE, Hariri LP, Moore MW, Gulati M, Homer RJ, Greif DM, Eltzschig HK, Herzog EL. Macrophage-derived netrin-1 drives adrenergic nerve-associated lung fibrosis. J Clin Invest 2021; 131:136542. [PMID: 33393489 PMCID: PMC7773383 DOI: 10.1172/jci136542] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is a macrophage-driven process of uncontrolled extracellular matrix accumulation. Neuronal guidance proteins such as netrin-1 promote inflammatory scarring. We found that macrophage-derived netrin-1 stimulates fibrosis through its neuronal guidance functions. In mice, fibrosis due to inhaled bleomycin engendered netrin-1-expressing macrophages and fibroblasts, remodeled adrenergic nerves, and augmented noradrenaline. Cell-specific knockout mice showed that collagen accumulation, fibrotic histology, and nerve-associated endpoints required netrin-1 of macrophage but not fibroblast origin. Adrenergic denervation; haploinsufficiency of netrin-1's receptor, deleted in colorectal carcinoma; and therapeutic α1 adrenoreceptor antagonism improved collagen content and histology. An idiopathic pulmonary fibrosis (IPF) lung microarray data set showed increased netrin-1 expression. IPF lung tissues were enriched for netrin-1+ macrophages and noradrenaline. A longitudinal IPF cohort showed improved survival in patients prescribed α1 adrenoreceptor blockade. This work showed that macrophages stimulate lung fibrosis via netrin-1-driven adrenergic processes and introduced α1 blockers as a potentially new fibrotic therapy.
Collapse
Affiliation(s)
- Ruijuan Gao
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xueyan Peng
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carrighan Perry
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Huanxing Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Aglaia Ntokou
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jose L. Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Benjamin C. Reeves
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anjali Walia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nir Neumark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine, and
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Meagan W. Moore
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Robert J. Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| | - Daniel M. Greif
- Section of Cardiology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, University of Texas at Houston Medical School, Houston, Texas, USA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, and
| |
Collapse
|
275
|
Bio-artificial pleura using autologous dermal fibroblast sheets to mitigate air leaks during thoracoscopic lung resection. NPJ Regen Med 2021; 6:2. [PMID: 33398054 PMCID: PMC7782673 DOI: 10.1038/s41536-020-00113-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 11/20/2020] [Indexed: 11/09/2022] Open
Abstract
Lung air leaks (LALs) due to visceral pleura injury during surgery are a difficult-to-avoid complication in thoracic surgery (TS). Reliable LAL closure is an important patient management issue after TS. We demonstrated both safeties of transplantation of a cultured human autologous dermal fibroblast sheet (DFS) to LALs. From May 2016 to March 2018, five patients who underwent thoracoscopic lung resection met all the inclusion criteria. Skin biopsies were acquired from each patient to source autologous dermal cells for DFS fabrication. During the primary culture, fibroblasts migrated from the dermal tissue pieces and proliferated to form cell monolayers. These fibroblasts were subcultured to confluence. Transplantable DFSs were fabricated from these subcultured fibroblasts that were trypsinized and seeded onto temperature-responsive culture dishes. After 10 days of fabrication culture, intact patient-specific DFS were harvested. DFSs were analyzed for fibroblast cell content and tissue contaminants prior to application. For closing intraoperative LAL, mean number of transplanted autologous DFS per patient was 6 ± 2 sheets. Mean chest drainage duration was 5.0 ± 4.8 days. The two patients with major LAL had a drainage duration of more than 7 days. All patients currently have no LAL recurrence after discharge. DFSs effectively maintain LAL closure via remodeling of the deposited extracellular matrix. The use of autologous DFSs to permanently close air leaks using a patient-derived source is expected to reduce surgical complications during high-risk lung resections.
Collapse
|
276
|
Biffi G, Tuveson DA. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol Rev 2021; 101:147-176. [PMID: 32466724 PMCID: PMC7864232 DOI: 10.1152/physrev.00048.2019] [Citation(s) in RCA: 720] [Impact Index Per Article: 180.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023] Open
Abstract
Efforts to develop anti-cancer therapies have largely focused on targeting the epithelial compartment, despite the presence of non-neoplastic stromal components that substantially contribute to the progression of the tumor. Indeed, cancer cell survival, growth, migration, and even dormancy are influenced by the surrounding tumor microenvironment (TME). Within the TME, cancer-associated fibroblasts (CAFs) have been shown to play several roles in the development of a tumor. They secrete growth factors, inflammatory ligands, and extracellular matrix proteins that promote cancer cell proliferation, therapy resistance, and immune exclusion. However, recent work indicates that CAFs may also restrain tumor progression in some circumstances. In this review, we summarize the body of work on CAFs, with a particular focus on the most recent discoveries about fibroblast heterogeneity, plasticity, and functions. We also highlight the commonalities of fibroblasts present across different cancer types, and in normal and inflammatory states. Finally, we present the latest advances regarding therapeutic strategies targeting CAFs that are undergoing preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York; and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York; and Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
277
|
Moulin VJ. Three-Dimensional Model of Hypertrophic Scar Using a Tissue-Engineering Approach. Methods Mol Biol 2021; 2299:419-434. [PMID: 34028758 DOI: 10.1007/978-1-0716-1382-5_28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Following wound healing, skin is replaced by a specialized tissue called scar. Sometime, this scar can become pathologic, called hypertrophic scar, with a high amount of extracellular matrix, capillaries, and myofibroblast persistence. To understand the mechanisms at the origin of the fibrosis is paramount to treat patients, but despite few animal models and in vitro studies using mainly human pathological cells cultured on plastic on monolayer, the treatment of these fibrotic scars remains unsatisfactory. As in tissue, cells are most often imbedded in extracellular matrix, we have developed, using a tissue engineering method, new in vitro models to study human fibrotic skin pathologies as hypertrophic scars. Human cells isolated from hypertrophic scars are used to reconstitute a three-dimensional fibrotic skin comprising both dermal and epidermal parts. This method called the self-assembly approach is based on the cell capacity to reconstitute their own environment as in vivo. In this chapter, the described methods include extraction and culture of human scar keratinocytes and fibroblasts from cutaneous biopsies as well as the protocols to produce fibrotic skin that can be used to study pathological process.
Collapse
Affiliation(s)
- Veronique J Moulin
- Centre LOEX de l'Université Laval, Research Center CHU de Québec-Université Laval and Faculty of Medicine, Surgery Department, Université Laval, Québec, QC, Canada.
| |
Collapse
|
278
|
miR-127-3p Is an Epigenetic Activator of Myofibroblast Senescence Situated within the MicroRNA-Enriched Dlk1-Dio3‒Imprinted Domain on Mouse Chromosome 12. J Invest Dermatol 2020; 141:1076-1086.e3. [PMID: 33279585 DOI: 10.1016/j.jid.2020.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/14/2023]
Abstract
During wound healing, fibroblasts differentiate into nonproliferative contractile myofibroblasts, contribute to skin repair, and eventually undergo apoptosis or become senescent. MicroRNAs are post-transcriptional regulators of gene expression networks that control cell fate and survival and may also regulate senescence. In this study, we determined the regulated microRNAs in myofibroblasts isolated from wounds and analyzed their role in senescent myofibroblast formation. Transcriptome profiling showed that a 200 kilobase pair region of the Dlk1-Dio3‒imprinted domain on mouse chromosome 12 encodes for most of the upregulated microRNAs in the entire genome of mouse myofibroblasts. Among those, miR-127-3p induced a myofibroblast-like phenotype associated with a block in proliferation. Molecular analysis revealed that miR-127-3p induced a prolonged cell cycle arrest with unique molecular features of senescence, including the activation of the senescence-associated β-galactosidase, increase in p53 and p21 levels, inhibition of lamin B1, proliferation factors, and the production of senescence-associated inflammatory and extracellular matrix‒remodeling components. Hence, miR-127-3p emerges as an epigenetic activator regulating the transition from repair to remodeling during skin wound healing but may also induce age-related defects, pathological scarring, and fibrosis, all linked to myofibroblast senescence.
Collapse
|
279
|
Usansky I, Jaworska P, Asti L, Kenny FN, Hobbs C, Sofra V, Song H, Logan M, Graham A, Shaw TJ. A developmental basis for the anatomical diversity of dermis in homeostasis and wound repair. J Pathol 2020; 253:315-325. [PMID: 33197044 PMCID: PMC7898902 DOI: 10.1002/path.5589] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/27/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
The dermis has disparate embryonic origins; abdominal dermis develops from lateral plate mesoderm, dorsal dermis from paraxial mesoderm and facial dermis from neural crest. However, the cell and molecular differences and their functional implications have not been described. We hypothesise that the embryonic origin of the dermis underpins regional characteristics of skin, including its response to wounding. We have compared abdomen, back and cheek, three anatomical sites representing the distinct embryonic tissues from which the dermis can arise, during homeostasis and wound repair using RNA sequencing, histology and fibroblast cultures. Our transcriptional analyses demonstrate differences between body sites that reflect their diverse origins. Moreover, we report histological and transcriptional variations during a wound response, including site differences in ECM composition, cell migration and proliferation, and re‐enactment of distinct developmental programmes. These findings reveal profound regional variation in the mechanisms of tissue repair. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ivy Usansky
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| | - Patrycja Jaworska
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| | - Ludovica Asti
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| | - Fiona N Kenny
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Carl Hobbs
- Wolfson Centre for Age-Related Disease, King's College London, London, UK
| | - Vasiliki Sofra
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| | - Hanfei Song
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| | - Malcolm Logan
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Anthony Graham
- Department of Developmental Neurobiology, King's College London, London, UK
| | - Tanya J Shaw
- Centre for Inflammation Biology & Cancer Immunology, King's College London, London, UK
| |
Collapse
|
280
|
Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020; 587:555-566. [PMID: 33239795 DOI: 10.1038/s41586-020-2938-9] [Citation(s) in RCA: 1050] [Impact Index Per Article: 210.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Fibrosis can affect any organ and is responsible for up to 45% of all deaths in the industrialized world. It has long been thought to be relentlessly progressive and irreversible, but both preclinical models and clinical trials in various organ systems have shown that fibrosis is a highly dynamic process. This has clear implications for therapeutic interventions that are designed to capitalize on this inherent plasticity. However, despite substantial progress in our understanding of the pathobiology of fibrosis, a translational gap remains between the identification of putative antifibrotic targets and conversion of this knowledge into effective treatments in humans. Here we discuss the transformative experimental strategies that are being leveraged to dissect the key cellular and molecular mechanisms that regulate fibrosis, and the translational approaches that are enabling the emergence of precision medicine-based therapies for patients with fibrosis.
Collapse
Affiliation(s)
- Neil C Henderson
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Thomas A Wynn
- Inflammation & Immunology Research Unit, Pfizer Worldwide Research, Development & Medical, Cambridge, MA, USA.
| |
Collapse
|
281
|
Hosio M, Jaks V, Lagus H, Vuola J, Ogawa R, Kankuri E. Primary Ciliary Signaling in the Skin-Contribution to Wound Healing and Scarring. Front Cell Dev Biol 2020; 8:578384. [PMID: 33282860 PMCID: PMC7691485 DOI: 10.3389/fcell.2020.578384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/08/2020] [Indexed: 12/21/2022] Open
Abstract
Primary cilia (PC) are solitary, post-mitotic, microtubule-based, and membrane-covered protrusions that are found on almost every mammalian cell. PC are specialized cellular sensory organelles that transmit environmental information to the cell. Signaling through PC is involved in the regulation of a variety of cellular processes, including proliferation, differentiation, and migration. Conversely, defective, or abnormal PC signaling can contribute to the development of various pathological conditions. Our knowledge of the role of PC in organ development and function is largely based on ciliopathies, a family of genetic disorders with mutations affecting the structure and function of PC. In this review, we focus on the role of PC in their major signaling pathways active in skin cells, and their contribution to wound healing and scarring. To provide comprehensive insights into the current understanding of PC functions, we have collected data available in the literature, including evidence across cell types, tissues, and animal species. We conclude that PC are underappreciated subcellular organelles that significantly contribute to both physiological and pathological processes of the skin development and wound healing. Thus, PC assembly and disassembly and PC signaling may serve as attractive targets for antifibrotic and antiscarring therapies.
Collapse
Affiliation(s)
- Mayu Hosio
- Faculty of Medicine, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Jyrki Vuola
- Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Esko Kankuri
- Faculty of Medicine, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
282
|
Leavitt T, Hu MS, Borrelli MR, Januszyk M, Garcia JT, Ransom RC, Mascharak S, desJardins-Park HE, Litzenburger UM, Walmsley GG, Marshall CD, Moore AL, Duoto B, Adem S, Foster DS, Salhotra A, Shen AH, Griffin M, Shen EZ, Barnes LA, Zielins ER, Maan ZN, Wei Y, Chan CKF, Wan DC, Lorenz HP, Chang HY, Gurtner GC, Longaker MT. Prrx1 Fibroblasts Represent a Pro-fibrotic Lineage in the Mouse Ventral Dermis. Cell Rep 2020; 33:108356. [PMID: 33176144 PMCID: PMC7742512 DOI: 10.1016/j.celrep.2020.108356] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/27/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
Fibroblast heterogeneity has been shown within the unwounded mouse dorsal dermis, with fibroblast subpopulations being identified according to anatomical location and embryonic lineage. Using lineage tracing, we demonstrate that paired related homeobox 1 (Prrx1)-expressing fibroblasts are responsible for acute and chronic fibroses in the ventral dermis. Single-cell transcriptomics further corroborated the inherent fibrotic characteristics of Prrx1 fibroblasts during wound repair. In summary, we identify and characterize a fibroblast subpopulation in the mouse ventral dermis with intrinsic scar-forming potential.
Collapse
Affiliation(s)
- Tripp Leavitt
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael S Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Julia T Garcia
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Shamik Mascharak
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Heather E desJardins-Park
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ulrike M Litzenburger
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Graham G Walmsley
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Clement D Marshall
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Alessandra L Moore
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Bryan Duoto
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sandeep Adem
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Deshka S Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ankit Salhotra
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Abra H Shen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ethan Z Shen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Leandra A Barnes
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Elizabeth R Zielins
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yuning Wei
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Charles K F Chan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Hermann P Lorenz
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
283
|
Jiang D, Christ S, Correa-Gallegos D, Ramesh P, Kalgudde Gopal S, Wannemacher J, Mayr CH, Lupperger V, Yu Q, Ye H, Mück-Häusl M, Rajendran V, Wan L, Liu J, Mirastschijski U, Volz T, Marr C, Schiller HB, Rinkevich Y. Injury triggers fascia fibroblast collective cell migration to drive scar formation through N-cadherin. Nat Commun 2020; 11:5653. [PMID: 33159076 PMCID: PMC7648088 DOI: 10.1038/s41467-020-19425-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
Scars are more severe when the subcutaneous fascia beneath the dermis is injured upon surgical or traumatic wounding. Here, we present a detailed analysis of fascia cell mobilisation by using deep tissue intravital live imaging of acute surgical wounds, fibroblast lineage-specific transgenic mice, and skin-fascia explants (scar-like tissue in a dish - SCAD). We observe that injury triggers a swarming-like collective cell migration of fascia fibroblasts that progressively contracts the skin and form scars. Swarming is exclusive to fascia fibroblasts, and requires the upregulation of N-cadherin. Both swarming and N-cadherin expression are absent from fibroblasts in the upper skin layers and the oral mucosa, tissues that repair wounds with minimal scar. Impeding N-cadherin binding inhibits swarming and skin contraction, and leads to reduced scarring in SCADs and in animals. Fibroblast swarming and N-cadherin thus provide therapeutic avenues to curtail fascia mobilisation and pathological fibrotic responses across a range of medical settings.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Simon Christ
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Donovan Correa-Gallegos
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Pushkar Ramesh
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Shruthi Kalgudde Gopal
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Juliane Wannemacher
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Christoph H Mayr
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Systems Medicine of Chronic Lung Disease, Munich, Germany
| | - Valerio Lupperger
- Helmholtz Zentrum München, Institute of Computational Biology, Munich, Germany
| | - Qing Yu
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Haifeng Ye
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Martin Mück-Häusl
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Vijayanand Rajendran
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Li Wan
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Juan Liu
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany
| | - Ursula Mirastschijski
- Mira-Beau gender esthetics, Berlin, Germany
- Wound Repair Unit, CBIB, Faculty of Biology and Biochemistry, University of Bremen, Bremen, Germany
| | - Thomas Volz
- Department of Dermatology and Allergology, Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Munich, Germany
| | - Carsten Marr
- Helmholtz Zentrum München, Institute of Computational Biology, Munich, Germany
| | - Herbert B Schiller
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Systems Medicine of Chronic Lung Disease, Munich, Germany
- German Centre for Lung Research (DZL), Munich, Germany
| | - Yuval Rinkevich
- Helmholtz Zentrum München, Institute of Lung Biology and Disease, Group Regenerative Biology and Medicine, Munich, Germany.
- German Centre for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
284
|
Shen T, Dai K, Yu Y, Wang J, Liu C. Sulfated chitosan rescues dysfunctional macrophages and accelerates wound healing in diabetic mice. Acta Biomater 2020; 117:192-203. [PMID: 33007486 DOI: 10.1016/j.actbio.2020.09.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Emerging evidence suggests that dysfunctional macrophages can cause chronic inflammation and impair tissue regeneration in diabetic wounds. Therefore, improving macrophage behaviors and functions may improve therapeutic outcomes of current treatments in diabetic wounds. Herein, we present a sulfated chitosan (SCS)-doped Collagen type I (Col I/SCS) hydrogel as a candidate for diabetic wound treatments, and assess its efficacy using streptozocin (STZ)-induced diabetic wound model. Results showed that Col I/SCS hydrogel significantly improved wound closure rate, collagen deposition, and revascularization in diabetic wounds. Flow cytometry analysis and immunofluorescent staining analysis showed that the Col I/SCS hydrogel accelerated the resolution of excessive inflammation by reducing the polarization of M1-like macrophages in chronic diabetic wounds. In addition, ELISA analysis revealed that the Col I/SCS hydrogel reduced the production of pro-inflammatory interleukin (IL)-6 and increased the production of anti-inflammatory cytokines including IL-4 and transforming growth factor-beta 1 (TGF-β1) during wound healing. Moreover, the Col I/SCS hydrogel enhanced the transdifferentiation of macrophages into fibroblasts, which enhanced the formation of collagen and the extracellular matrix (ECM) in wound tissue. We highlight a potential application of manipulating macrophages behaviors in the pathological microenvironment via materials strategy. STATEMENT OF SIGNIFICANCE: Improving the chronic inflammatory microenvironment of diabetic wounds by regulating macrophage behaviors has been of wide concern in recent years. We designed a Col I/SCS hydrogel based on Collagen type I and sulfated chitosan (SCS) without exogenous cells or cytokines, which could significantly improve angiogenesis and resolve chronic inflammation in diabetic wounds, and hence accelerate diabetic wound healing. The Col I/SCS hydrogel could facilitate the polarization of M1-to-M2 macrophages and activate the transdifferentiation of macrophages to fibroblasts. Additionally, the Col I/SCS hydrogel also equilibrated the content of pro-inflammatory and anti-inflammatory cytokines. This strategy may afford a new avenue to improve macrophage functions and accelerate diabetic chronic wound healing.
Collapse
Affiliation(s)
- Tong Shen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR. China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR. China
| | - Kai Dai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR. China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR. China
| | - Yuanman Yu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR. China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR. China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR. China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR. China.
| | - Changsheng Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR. China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, PR. China.
| |
Collapse
|
285
|
Abstract
Abstract
Purpose of Review
In this brief review, we will highlight important observational and experimental data in the literature that address the origin of scar-forming cells in lung fibrosis.
Recent Findings
Several cellular sources of activated scar-forming cells (myofibroblasts) have been postulated including alveolar epithelial cells; circulating fibrocytes; and lung stromal cell subpopulations including resident fibroblasts, pericytes, and resident mesenchymal stem cells. Recent advances in lineage-tracing models, however, fail to provide experimental evidence for epithelial and fibrocyte origins of lung myofibroblasts. Resident mesenchymal cells of the lung, which include various cell types including resident fibroblasts, pericytes, and resident mesenchymal stem cells, appear to be important sources of myofibroblasts in murine models of lung injury and fibrosis.
Summary
Lung myofibroblasts likely originate from multiple sources of lung-resident mesenchymal cells. Their relative contributions may vary depending on the type of injury. Although lineage-tracing experiments have failed to show significant contribution from epithelial cells or fibrocytes, they may play important functional roles in myofibroblast activation through paracrine signaling.
Collapse
|
286
|
Wilgus TA. Inflammation as an orchestrator of cutaneous scar formation: a review of the literature. PLASTIC AND AESTHETIC RESEARCH 2020; 7:54. [PMID: 33123623 PMCID: PMC7592345 DOI: 10.20517/2347-9264.2020.150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a key phase in the cutaneous wound repair process. The activation of inflammatory cells is critical for preventing infection in contaminated wounds and results in the release of an array of mediators, some of which stimulate the activity of keratinocytes, endothelial cells, and fibroblasts to aid in the repair process. However, there is an abundance of data suggesting that the strength of the inflammatory response early in the healing process correlates directly with the amount of scar tissue that will eventually form. This review will summarize the literature related to inflammation and cutaneous scar formation, highlight recent discoveries, and discuss potential treatment modalities that target inflammation to minimize scarring.
Collapse
Affiliation(s)
- Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
287
|
Hu M, Wu Y, Yang C, Wang X, Wang W, Zhou L, Zeng T, Zhou J, Wang C, Lao G, Yan L, Ren M. Novel Long Noncoding RNA lnc-URIDS Delays Diabetic Wound Healing by Targeting Plod1. Diabetes 2020; 69:2144-2156. [PMID: 32801140 DOI: 10.2337/db20-0147] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022]
Abstract
Impaired wound healing is one of the main causes of diabetic foot ulcerations. However, the exact mechanism of delayed wound healing in diabetes is not fully understood. Long noncoding RNAs (lncRNAs) are widely involved in a variety of biological processes and diseases, including diabetes and its associated complications. In this study, we identified a novel lncRNA, MRAK052872, named lncRNA UpRegulated in Diabetic Skin (lnc-URIDS), which regulates wound healing in diabetes. lnc-URIDS was highly expressed in diabetic skin and dermal fibroblasts treated with advanced glycation end products (AGEs). lnc-URIDS knockdown promoted migration of dermal fibroblasts under AGEs treatment in vitro and accelerated diabetic wound healing in vivo. Mechanistically, lnc-URIDS interacts with procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1 (Plod1), a critical enzyme responsible for collagen cross-linking. The binding of lnc-URIDS to Plod1 results in a decreased protein stability of Plod1, which ultimately leads to the dysregulation of collagen production and deposition and delays wound healing. Collectively, this study identifies a novel lncRNA that regulates diabetic wound healing by targeting Plod1. The findings of the current study offer some insight into the potential mechanism for the delayed wound healing in diabetes and provide a potential therapeutic target for diabetic foot.
Collapse
Affiliation(s)
- Mengdie Hu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxi Wu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuan Yang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liyan Zhou
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tingting Zeng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhou
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuan Wang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guojuan Lao
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meng Ren
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
288
|
Adipocytic Progenitor Cells Give Rise to Pathogenic Myofibroblasts: Adipocyte-to-Mesenchymal Transition and Its Emerging Role in Fibrosis in Multiple Organs. Curr Rheumatol Rep 2020; 22:79. [PMID: 32978695 PMCID: PMC7518402 DOI: 10.1007/s11926-020-00957-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose of Review Adipocytes have recently been shown to be able to reprogram to a myofibroblastic phenotype in a process termed adipocyte mesenchymal transition (AMT). This review seeks to discuss the relevance of this process to disease and explore its mechanisms. Recent Findings AMT occurs in multiple organs and diseases, transdifferentiation goes through a precursor cell and there is a reversible process that can be influenced by metabolic stress, myeloid cells, immune dysregulation, and pharmacological intervention. Summary AMT is a newly appreciated and highly relevant process in multiple forms of fibrosis. Targeting AMT may serve as a novel method of treating fibrosis.
Collapse
|
289
|
Grim JC, Aguado BA, Vogt BJ, Batan D, Andrichik CL, Schroeder ME, Gonzalez-Rodriguez A, Yavitt FM, Weiss RM, Anseth KS. Secreted Factors From Proinflammatory Macrophages Promote an Osteoblast-Like Phenotype in Valvular Interstitial Cells. Arterioscler Thromb Vasc Biol 2020; 40:e296-e308. [PMID: 32938214 DOI: 10.1161/atvbaha.120.315261] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Resident valvular interstitial cells (VICs) activate to myofibroblasts during aortic valve stenosis progression, which further promotes fibrosis or even differentiate into osteoblast-like cells that can lead to calcification of valve tissue. Inflammation is a hallmark of aortic valve stenosis, so we aimed to determine proinflammatory cytokines secreted from M1 macrophages that give rise to a transient VIC phenotype that leads to calcification of valve tissue. Approach and Results: We designed hydrogel biomaterials as valve extracellular matrix mimics enabling the culture of VICs in either their quiescent fibroblast or activated myofibroblast phenotype in response to the local matrix stiffness. When VIC fibroblasts and myofibroblasts were treated with conditioned media from THP-1-derived M1 macrophages, we observed robust reduction of αSMA (alpha smooth muscle actin) expression, reduced stress fiber formation, and increased proliferation, suggesting a potent antifibrotic effect. We further identified TNF (tumor necrosis factor)-α and IL (interleukin)-1β as 2 cytokines in M1 media that cause the observed antifibrotic effect. After 7 days of culture in M1 conditioned media, VICs began differentiating into osteoblast-like cells, as measured by increased expression of RUNX2 (runt-related transcription factor 2) and osteopontin. We also identified and validated IL-6 as a critical mediator of the observed pro-osteogenic effect. CONCLUSIONS Proinflammatory cytokines in M1 conditioned media inhibit myofibroblast activation in VICs (eg, TNF-α and IL-1β) and promote their osteogenic differentiation (eg, IL-6). Together, our work suggests inflammatory M1 macrophages may drive a myofibroblast-to-osteogenic intermediate VIC phenotype, which may mediate the switch from fibrosis to calcification during aortic valve stenosis progression.
Collapse
Affiliation(s)
- Joseph C Grim
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Brian A Aguado
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Brandon J Vogt
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Dilara Batan
- BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Division of Biochemistry (D.B.), University of Colorado Boulder, Boulder
| | - Cassidy L Andrichik
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Megan E Schroeder
- BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Materials Science and Engineering Program (M.E.S., K.S.A.), University of Colorado Boulder, Boulder
| | - Andrea Gonzalez-Rodriguez
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - F Max Yavitt
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa, Iowa City (R.M.W.)
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering (J.C.G., B.A.A., B.J.V., C.L.A., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,BioFrontiers Institute (J.C.G., B.A.A., D.B., M.E.S., A.G.-R., F.M.Y., K.S.A.), University of Colorado Boulder, Boulder.,Materials Science and Engineering Program (M.E.S., K.S.A.), University of Colorado Boulder, Boulder
| |
Collapse
|
290
|
Zhang Z, Kruglikov I, Zhao S, Zi Z, Gliniak CM, Li N, Wang M, Zhu Q, Kusminski CM, Scherer PE. Dermal adipocytes contribute to the metabolic regulation of dermal fibroblasts. Exp Dermatol 2020; 30:102-111. [DOI: 10.1111/exd.14181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Zhuzhen Zhang
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | | | - Shangang Zhao
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Zhenzhen Zi
- Department of Biochemistry University of Texas Southwestern Medical Center Dallas TX USA
| | - Christy M. Gliniak
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Na Li
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - May‐yun Wang
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Qingzhang Zhu
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Christine M. Kusminski
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center University of Texas Southwestern Medical Center Dallas TX USA
| |
Collapse
|
291
|
Wang Y, Guerrero-Juarez CF, Qiu Y, Du H, Chen W, Figueroa S, Plikus MV, Nie Q. A multiscale hybrid mathematical model of epidermal-dermal interactions during skin wound healing. Exp Dermatol 2020; 28:493-502. [PMID: 30801791 DOI: 10.1111/exd.13909] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/13/2019] [Indexed: 12/18/2022]
Abstract
Following injury, skin activates a complex wound healing programme. While cellular and signalling mechanisms of wound repair have been extensively studied, the principles of epidermal-dermal interactions and their effects on wound healing outcomes are only partially understood. To gain new insight into the effects of epidermal-dermal interactions, we developed a multiscale, hybrid mathematical model of skin wound healing. The model takes into consideration interactions between epidermis and dermis across the basement membrane via diffusible signals, defined as activator and inhibitor. Simulations revealed that epidermal-dermal interactions are critical for proper extracellular matrix deposition in the dermis, suggesting these signals may influence how wound scars form. Our model makes several theoretical predictions. First, basal levels of epidermal activator and inhibitor help to maintain dermis in a steady state, whereas their absence results in a raised, scar-like dermal phenotype. Second, wound-triggered increase in activator and inhibitor production by basal epidermal cells, coupled with fast re-epithelialization kinetics, reduces dermal scar size. Third, high-density fibrin clot leads to a raised, hypertrophic scar phenotype, whereas low-density fibrin clot leads to a hypotrophic phenotype. Fourth, shallow wounds, compared to deep wounds, result in overall reduced scarring. Taken together, our model predicts the important role of signalling across dermal-epidermal interface and the effect of fibrin clot density and wound geometry on scar formation. This hybrid modelling approach may be also applicable to other complex tissue systems, enabling the simulation of dynamic processes, otherwise computationally prohibitive with fully discrete models due to a large number of variables.
Collapse
Affiliation(s)
- Yangyang Wang
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Department of Mathematics, University of California, Irvine, Irvine, California
| | - Christian F Guerrero-Juarez
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Department of Mathematics, University of California, Irvine, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California
| | - Yuchi Qiu
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Department of Mathematics, University of California, Irvine, Irvine, California
| | - Huijing Du
- Department of Mathematics, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Weitao Chen
- Department of Mathematics, University of California, Riverside, Riverside, California
| | - Seth Figueroa
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Department of Mathematics, University of California, Irvine, Irvine, California
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California
| | - Qing Nie
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,Department of Mathematics, University of California, Irvine, Irvine, California.,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California
| |
Collapse
|
292
|
Januszyk M, Chen K, Henn D, Foster DS, Borrelli MR, Bonham CA, Sivaraj D, Wagh D, Longaker MT, Wan DC, Gurtner GC. Characterization of Diabetic and Non-Diabetic Foot Ulcers Using Single-Cell RNA-Sequencing. MICROMACHINES 2020; 11:mi11090815. [PMID: 32872278 PMCID: PMC7570277 DOI: 10.3390/mi11090815] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022]
Abstract
Background: Recent advances in high-throughput single-cell sequencing technologies have led to their increasingly widespread adoption for clinical applications. However, challenges associated with tissue viability, cell yield, and delayed time-to-capture have created unique obstacles for data processing. Chronic wounds, in particular, represent some of the most difficult target specimens, due to the significant amount of fibrinous debris, extracellular matrix components, and non-viable cells inherent in tissue routinely obtained from debridement. Methods: Here, we examined the feasibility of single cell RNA sequencing (scRNA-seq) analysis to evaluate human chronic wound samples acquired in the clinic, subjected to prolonged cold ischemia time, and processed without FACS sorting. Wound tissue from human diabetic and non-diabetic plantar foot ulcers were evaluated using an optimized 10X Genomics scRNA-seq platform and analyzed using a modified data pipeline designed for low-yield specimens. Cell subtypes were identified informatically and their distributions and transcriptional programs were compared between diabetic and non-diabetic tissue. Results: 139,000 diabetic and non-diabetic wound cells were delivered for 10X capture after either 90 or 180 min of cold ischemia time. cDNA library concentrations were 858.7 and 364.7 pg/µL, respectively, prior to sequencing. Among all barcoded fragments, we found that 83.5% successfully aligned to the human transcriptome and 68% met the minimum cell viability threshold. The average mitochondrial mRNA fraction was 8.5% for diabetic cells and 6.6% for non-diabetic cells, correlating with differences in cold ischemia time. A total of 384 individual cells were of sufficient quality for subsequent analyses; from this cell pool, we identified transcriptionally-distinct cell clusters whose gene expression profiles corresponded to fibroblasts, keratinocytes, neutrophils, monocytes, and endothelial cells. Fibroblast subpopulations with differing fibrotic potentials were identified, and their distributions were found to be altered in diabetic vs. non-diabetic cells. Conclusions: scRNA-seq of clinical wound samples can be achieved using minor modifications to standard processing protocols and data analysis methods. This simple approach can capture widespread transcriptional differences between diabetic and non-diabetic tissue obtained from matched wound locations.
Collapse
Affiliation(s)
- Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Dominic Henn
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Deshka S. Foster
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Mimi R. Borrelli
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Clark A. Bonham
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Dharshan Sivaraj
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Dhananjay Wagh
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Michael T. Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
| | - Geoffrey C. Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.J.); (K.C.); (D.H.); (D.S.F.); (M.R.B.); (C.A.B.); (D.S.); (M.T.L.); (D.C.W.)
- Correspondence: ; Tel.: +1-650-736-2776
| |
Collapse
|
293
|
Distinct fibroblast subsets regulate lacteal integrity through YAP/TAZ-induced VEGF-C in intestinal villi. Nat Commun 2020; 11:4102. [PMID: 32796823 PMCID: PMC7428020 DOI: 10.1038/s41467-020-17886-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
Emerging evidence suggests that intestinal stromal cells (IntSCs) play essential roles in maintaining intestinal homeostasis. However, the extent of heterogeneity within the villi stromal compartment and how IntSCs regulate the structure and function of specialized intestinal lymphatic capillary called lacteal remain elusive. Here we show that selective hyperactivation or depletion of YAP/TAZ in PDGFRβ+ IntSCs leads to lacteal sprouting or regression with junctional disintegration and impaired dietary fat uptake. Indeed, mechanical or osmotic stress regulates IntSC secretion of VEGF-C mediated by YAP/TAZ. Single-cell RNA sequencing delineated novel subtypes of villi fibroblasts that upregulate Vegfc upon YAP/TAZ activation. These populations of fibroblasts were distributed in proximity to lacteal, suggesting that they constitute a peri-lacteal microenvironment. Our findings demonstrate the heterogeneity of IntSCs and reveal that distinct subsets of villi fibroblasts regulate lacteal integrity through YAP/TAZ-induced VEGF-C secretion, providing new insights into the dynamic regulatory mechanisms behind lymphangiogenesis and lymphatic remodeling. Intestinal stromal cells (IntSCs) play essential roles in maintaining intestinal homeostasis. Here the authors show that VEGF-C expression in specialized IntSCs is regulated by YAP/TAZ, and VEGF-C is responsible for maintaining lacteal integrity, thus influencing dietary fat drainage into lacteals.
Collapse
|
294
|
Mascharak S, desJardins-Park HE, Longaker MT. Fibroblast Heterogeneity in Wound Healing: Hurdles to Clinical Translation. Trends Mol Med 2020; 26:1101-1106. [PMID: 32800679 DOI: 10.1016/j.molmed.2020.07.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
Recent work has revealed that fibroblasts are remarkably heterogeneous cells, but the appropriate lens through which to study this variation (lineage, phenotype, and plasticity) and its relevance to human biology remain unclear. In this opinion article, we comment on recent breakthroughs in our understanding of fibroblast heterogeneity during skin wound healing, and on open questions that must be addressed to clinically translate these findings in order to minimize scarring in patients. We emphasize the need for experimental models of wound healing that better approximate human biology, as well as comparison of scarring and regenerative phenotypes to uncover master regulators of fibrosis.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heather E desJardins-Park
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
295
|
Wang M, Lessard SG, Singh P, Pannellini T, Chen T, Rourke BJ, Chowdhury L, Craveiro V, Sculco PK, Meulen MCH, Otero M. Knee fibrosis is associated with the development of osteoarthritis in a murine model of tibial compression. J Orthop Res 2020. [DOI: 10.1002/jor.24815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- Mengying Wang
- HSS Research Institute Hospital for Special Surgery New York New York
- School of Public Health, Xi'an Jiaotong University Health Science Center Xi'an China
| | | | - Purva Singh
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Tania Pannellini
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Tony Chen
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Brennan J. Rourke
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Luvana Chowdhury
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Vinicius Craveiro
- HSS Research Institute Hospital for Special Surgery New York New York
| | - Peter K. Sculco
- The Stavros Niarchos Foundation Complex Joint Reconstruction Center Hospital for Special Surgery New York New York
| | - Marjolein C. H. Meulen
- HSS Research Institute Hospital for Special Surgery New York New York
- Sibley School of Mechanical and Aerospace Engineering Cornell University Ithaca New York
- Meinig School of Biomedical Engineering Cornell University Ithaca New York
| | - Miguel Otero
- HSS Research Institute Hospital for Special Surgery New York New York
| |
Collapse
|
296
|
Abbasi S, Sinha S, Labit E, Rosin NL, Yoon G, Rahmani W, Jaffer A, Sharma N, Hagner A, Shah P, Arora R, Yoon J, Islam A, Uchida A, Chang CK, Stratton JA, Scott RW, Rossi FMV, Underhill TM, Biernaskie J. Distinct Regulatory Programs Control the Latent Regenerative Potential of Dermal Fibroblasts during Wound Healing. Cell Stem Cell 2020; 27:396-412.e6. [PMID: 32755548 DOI: 10.1016/j.stem.2020.07.008] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/25/2020] [Accepted: 07/09/2020] [Indexed: 01/12/2023]
Abstract
Dermal fibroblasts exhibit considerable heterogeneity during homeostasis and in response to injury. Defining lineage origins of reparative fibroblasts and regulatory programs that drive fibrosis or, conversely, promote regeneration will be essential for improving healing outcomes. Using complementary fate-mapping approaches, we show that hair follicle mesenchymal progenitors make limited contributions to wound repair. In contrast, extrafollicular progenitors marked by the quiescence-associated factor Hic1 generated the bulk of reparative fibroblasts and exhibited functional divergence, mediating regeneration in the center of the wound neodermis and scar formation in the periphery. Single-cell RNA-seq revealed unique transcriptional, regulatory, and epithelial-mesenchymal crosstalk signatures that enabled mesenchymal competence for regeneration. Integration with scATAC-seq highlighted changes in chromatin accessibility within regeneration-associated loci. Finally, pharmacological modulation of RUNX1 and retinoic acid signaling or genetic deletion of Hic1 within wound-activated fibroblasts was sufficient to modulate healing outcomes, suggesting that reparative fibroblasts have latent but modifiable regenerative capacity.
Collapse
Affiliation(s)
- Sepideh Abbasi
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elodie Labit
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Nicole L Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Grace Yoon
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Waleed Rahmani
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Arzina Jaffer
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Nilesh Sharma
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Andrew Hagner
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Prajay Shah
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Rohit Arora
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jessica Yoon
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Anowara Islam
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Aya Uchida
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Chih Kai Chang
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Jo Anne Stratton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - R Wilder Scott
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - T Michael Underhill
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
297
|
Koliaraki V, Prados A, Armaka M, Kollias G. The mesenchymal context in inflammation, immunity and cancer. Nat Immunol 2020; 21:974-982. [PMID: 32747813 DOI: 10.1038/s41590-020-0741-2] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022]
Abstract
Mesenchymal cells are mesoderm-derived stromal cells that are best known for providing structural support to organs, synthesizing and remodeling the extracellular matrix (ECM) and regulating development, homeostasis and repair of tissues. Recent detailed mechanistic insights into the biology of fibroblastic mesenchymal cells have revealed they are also significantly involved in immune regulation, stem cell maintenance and blood vessel function. It is now becoming evident that these functions, when defective, drive the development of complex diseases, such as various immunopathologies, chronic inflammatory disease, tissue fibrosis and cancer. Here, we provide a concise overview of the contextual contribution of fibroblastic mesenchymal cells in physiology and disease and bring into focus emerging evidence for both their heterogeneity at the single-cell level and their tissue-specific, spatiotemporal functional diversity.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
298
|
Mateus Gonçalves L, Pereira E, Werneck de Castro JP, Bernal-Mizrachi E, Almaça J. Islet pericytes convert into profibrotic myofibroblasts in a mouse model of islet vascular fibrosis. Diabetologia 2020; 63:1564-1575. [PMID: 32424539 PMCID: PMC7354906 DOI: 10.1007/s00125-020-05168-7] [Citation(s) in RCA: 26] [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: 01/02/2020] [Accepted: 04/02/2020] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Islet vascular fibrosis may play an important role in the progression of type 2 diabetes, but there are no mouse models allowing detailed mechanistic studies to understand how a dysfunctional islet microvasculature contributes to diabetes pathogenesis. Here we report that the transgenic AktTg mouse, unlike other mouse strains, shows an increased deposition of extracellular matrix (ECM) proteins in perivascular regions, allowing us to study the cellular mechanisms that lead to islet vascular fibrosis. METHODS Using immunohistochemistry, we labelled the islet microvasculature and ECM in pancreas sections of AktTg mice and human donors and performed lineage tracing to follow the fate of islet pericytes. We compared islet microvascular responses in living pancreas slices from wild-type and AktTg mice. RESULTS We found that vascular pericytes proliferate extensively, convert into profibrotic myofibroblasts and substantially contribute to vascular fibrosis in the AktTg mouse model. The increased deposition of collagen I, fibronectin and periostin within the islet is associated with diminished islet perfusion as well as impaired capillary responses to noradrenaline (norepinephrine) and to high glucose in living pancreas slices. CONCLUSIONS/INTERPRETATION Our study thus illustrates how the AktTg mouse serves to elucidate a cellular mechanism in the development of islet vascular fibrosis, namely a change in pericyte phenotype that leads to vascular dysfunction. Because beta cells in the AktTg mouse are more numerous and larger, and secrete more insulin, in future studies we will test the role beta cell secretory products play in determining the phenotype of pericytes and other cells residing in the islet microenvironment under physiological and pathophysiological conditions. Graphical abstract.
Collapse
Affiliation(s)
- Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Obesity and Comorbidities Research Center, Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - João Pedro Werneck de Castro
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Miami VA Health Care System, Miami, FL, 33136, USA
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
299
|
Wu C, Zhang Z, Zhou K, Chen W, Tao J, Li C, Xin H, Song Y, Ai F. Preparation and characterization of borosilicate-bioglass-incorporated sodium alginate composite wound dressing for accelerated full-thickness skin wound healing. Biomed Mater 2020; 15:055009. [PMID: 32422624 DOI: 10.1088/1748-605x/ab9421] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Full-thickness skin injury is a serious and intractable clinical problem. Wound dressing is urgently needed to treat serious skin defects or induce skin reconstruction. For the first time, we demonstrated a borosilicate bioglass (BBG)-incorporated sodium alginate (SA) wound dressing by a simple and effective technique for accelerated wound healing. The physical and chemical properties, in vitro and in vivo properties of SA-BBG composite wound dressing have been investigated. The results show that the SA-BBG composite dressing possesses good water absorption performance. The boron and silicon ions in BBG can maintain stable and sustained release. Most importantly, the SA-BBG composite wound dressing shows outstanding wound healing ability in full-thickness skin defects in rats. The wounds treated with SA-BBG composite dressing groups had almost closed at day 15. When the ratio of sodium alginate to bioglass in the sponge is 3:1, the wound healing effect is the best. In conclusion, the SA-BBG composite dressing shows great potential for application in skin wound healing and SA3BBG works best.
Collapse
Affiliation(s)
- Chunxuan Wu
- The Second Clinical Medical School, Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
- These authors contributed equally to this work
| | - Zhongjie Zhang
- Xiaogan Central Hospital, Xiaogan, Hubei 432000, People's Republic of China
- These authors contributed equally to this work
| | - Kui Zhou
- School of Mechanic & Electronic Engineering, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Weigao Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jun Tao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Chen Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Hongbo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Yulin Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Fanrong Ai
- School of Mechanic & Electronic Engineering, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| |
Collapse
|
300
|
El-Hattab MY, Nagumo Y, Gourronc FA, Klingelhutz AJ, Ankrum JA, Sander EA. Human Adipocyte Conditioned Medium Promotes In Vitro Fibroblast Conversion to Myofibroblasts. Sci Rep 2020; 10:10286. [PMID: 32581231 PMCID: PMC7314785 DOI: 10.1038/s41598-020-67175-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Adipocytes and adipose tissue derived cells have been investigated for their potential to contribute to the wound healing process. However, the details of how these cells interact with other essential cell types, such as myofibroblasts/fibroblasts, remain unclear. Using a novel in-vitro 3D human adipocyte/pre-adipocyte spheroid model, we investigated whether adipocytes and their precursors (pre-adipocytes) secrete factors that affect human dermal fibroblast behavior. We found that both adipocyte and pre-adipocyte conditioned medium induced the migration of fibroblasts, but only adipocyte conditioned medium induced fibroblast differentiation into a highly contractile, collagen producing myofibroblast phenotype. Furthermore, adipocyte mediated myofibroblast induction occurred through a TGF-β independent mechanism. Our findings contribute to a better understanding on the involvement of adipose tissue in wound healing, and may help to uncover and develop fat-related wound healing treatments.
Collapse
Affiliation(s)
- Mariam Y El-Hattab
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA
| | - Yoshiaki Nagumo
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA
- Department of Plastic Surgery, Kindai University, Faculty of Medicine, Higashiosaka, Osaka, Japan
| | - Francoise A Gourronc
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.
| | - Edward A Sander
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, IA, USA.
- Department of Orthopedics and Rehabilitation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|