1
|
Alves PT, de Souza AG, Bastos VAF, Miguel EL, Ramos ACS, Cameron LC, Goulart LR, Cunha TM. The Modulation of Septic Shock: A Proteomic Approach. Int J Mol Sci 2024; 25:10641. [PMID: 39408970 PMCID: PMC11476436 DOI: 10.3390/ijms251910641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Sepsis poses a significant challenge due its lethality, involving multiple organ dysfunction and impaired immune responses. Among several factors affecting sepsis, monocytes play a crucial role; however, their phenotype, proteomic profile, and function in septic shock remain unclear. Our aim was to fully characterize the subpopulations and proteomic profiles of monocytes seen in septic shock cases and discuss their possible impact on the disease. Peripheral blood monocyte subpopulations were phenotype based on CD14/CD16 expression by flow cytometry, and proteins were extracted from the monocytes of individuals with septic shock and healthy controls to identify changes in the global protein expression in these cells. Analysis using 2D-nanoUPLC-UDMSE identified 67 differentially expressed proteins in shock patients compared to controls, in which 44 were upregulated and 23 downregulated. These proteins are involved in monocyte reprogramming, immune dysfunction, severe hypotension, hypo-responsiveness to vasoconstrictors, vasodilation, endothelial dysfunction, vascular injury, and blood clotting, elucidating the disease severity and therapeutic challenges of septic shock. This study identified critical biological targets in monocytes that could serve as potential biomarkers for the diagnosis, prognosis, and treatment of septic shock, providing new insights into the pathophysiology of the disease.
Collapse
Affiliation(s)
- Patrícia Terra Alves
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Aline Gomes de Souza
- Department of Medical Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14040-900, SP, Brazil;
| | - Victor Alexandre F. Bastos
- Laboratory of Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Eduarda L. Miguel
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - Augusto César S. Ramos
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - L. C. Cameron
- Arthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada;
- Lorraine Protein Biochemistry Group, Graduate Program in Neurology, Gaffrée e Guinle University Hospital, Rio de Janeiro 20270-004, RJ, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Thúlio M. Cunha
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| |
Collapse
|
2
|
Wang X, Liu X, Xu L, Li Y, Zheng B, Xia C, Wang J, Liu H. Targeted delivery of type I TGF-β receptor-mimicking peptide to fibrotic kidney for improving kidney fibrosis therapy via enhancing the inhibition of TGF-β1/Smad and p38 MAPK pathways. Int Immunopharmacol 2024; 137:112483. [PMID: 38880023 DOI: 10.1016/j.intimp.2024.112483] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Renal fibrosis is a representative pathological feature of various chronic kidney diseases, and efficient treatment is needed. Interstitial myofibroblasts are a key driver of kidney fibrosis, which is dependent on the binding of TGF-β1 to type I TGF-β receptor (TβRI) and TGF-β1-related signaling pathways. Therefore, attenuating TGF-β1 activity by competing with TGF-β1 in myofibroblasts is an ideal strategy for treating kidney fibrosis. Recently, a novel TβRI-mimicking peptide RIPΔ demonstrated a high affinity for TGF-β1. Thus, it could be speculated that RIPΔ may be used for anti-fibrosis therapy. Platelet-derived growth factor β receptor (PDGFβR) is highly expressed in fibrotic kidney. In this study, we found that target peptide Z-RIPΔ, which is RIPΔ modified with PDGFβR-specific affibody ZPDGFβR, was specifically and highly taken up by TGF-β1-activated NIH3T3 fibroblasts. Moreover, Z-RIPΔ effectively inhibited the myofibroblast proliferation, migration and fibrosis response in vitro. In vivo and ex vivo experiments showed that Z-RIPΔ specifically targeted fibrotic kidney, improved the damaged renal function, and ameliorated kidney histopathology and renal fibrosis in UUO mice. Mechanistic studies showed that Z-RIPΔ hold the stronger inhibition of the TGF-β1/Smad and TGF-β1/p38 pathways than unmodified RIPΔ in vitro and in vivo. Furthermore, systemic administration of Z-RIPΔ to UUO mice led to minimal toxicity to major organs. Taken together, RIPΔ modified with ZPDGFβR increased its therapeutic efficacy and reduced its systemic toxicity, making it a potential candidate for targeted therapy for kidney fibrosis.
Collapse
Affiliation(s)
- Xiaohua Wang
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China; Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, Mudanjiang 157011, PR China; Department of Cell Biology, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Xiaohui Liu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Liming Xu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Yuting Li
- Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Bowen Zheng
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Caiyun Xia
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Jingru Wang
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China
| | - Haifeng Liu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang 157011, PR China; Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, Mudanjiang 157011, PR China.
| |
Collapse
|
3
|
Poole K, Iyer KS, Schmidtke DW, Petroll WM, Varner VD. Corneal keratocytes, fibroblasts, and myofibroblasts exhibit distinct transcriptional profiles in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582620. [PMID: 38464034 PMCID: PMC10925317 DOI: 10.1101/2024.02.28.582620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Purpose After stromal injury to the cornea, the release of growth factors and pro-inflammatory cytokines promotes the activation of quiescent keratocytes into a migratory fibroblast and/or fibrotic myofibroblast phenotype. Persistence of the myofibroblast phenotype can lead to corneal fibrosis and scarring, which are leading causes of blindness worldwide. This study aims to establish comprehensive transcriptional profiles for cultured corneal keratocytes, fibroblasts, and myofibroblasts to gain insights into the mechanisms through which these phenotypic changes occur. Methods Primary rabbit corneal keratocytes were cultured in either defined serum-free media (SF), fetal bovine serum (FBS) containing media, or in the presence of TGF-β1 to induce keratocyte, fibroblast, or myofibroblast phenotypes, respectively. Bulk RNA sequencing followed by bioinformatic analyses was performed to identify significant differentially expressed genes (DEGs) and enriched biological pathways for each phenotype. Results Genes commonly associated with keratocytes, fibroblasts, or myofibroblasts showed high relative expression in SF, FBS, or TGF-β1 culture conditions, respectively. Differential expression and functional analyses revealed novel DEGs for each cell type, as well as enriched pathways indicative of differences in proliferation, apoptosis, extracellular matrix (ECM) synthesis, cell-ECM interactions, cytokine signaling, and cell mechanics. Conclusions Overall, these data demonstrate distinct transcriptional differences among cultured corneal keratocytes, fibroblasts, and myofibroblasts. We have identified genes and signaling pathways that may play important roles in keratocyte differentiation, including many related to mechanotransduction and ECM biology. Our findings have revealed novel molecular markers for each cell type, as well as possible targets for modulating cell behavior and promoting physiological corneal wound healing.
Collapse
|
4
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
5
|
Lapthorn AR, Ilg MM, Dziewulski P, Cellek S. Hydroxypyridone anti-fungals selectively induce myofibroblast apoptosis in an in vitro model of hypertrophic scars. Eur J Pharmacol 2024; 967:176369. [PMID: 38325796 DOI: 10.1016/j.ejphar.2024.176369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
Hypertrophic scars are a common complication of burn injuries, yet there are no medications to prevent their formation. During scar formation, resident fibroblasts are transformed to myofibroblasts which become resistant to apoptosis. Previously, we have shown that hydroxypyridone anti-fungals can inhibit transformation of fibroblasts, isolated from hypertrophic scars, to myofibroblasts. This study aimed to investigate if these drugs can also target myofibroblast persistence. Primary human dermal fibroblasts, derived from burn scar tissue, were exposed to transforming growth factor beta-1 (TGF-β1) for 72 h to induce myofibroblast transformation. The cells were then incubated with three hydroxypyridone anti-fungals (ciclopirox, ciclopirox ethanolamine and piroctone olamine; 0.03-300 μM) for a further 72 h. The In-Cell ELISA method was utilised to quantify myofibroblast transformation by measuring alpha-smooth muscle actin (α-SMA) expression and DRAQ5 staining, to measure cell viability. TUNEL staining was utilised to assess if the drugs could induce apoptosis. When given to established myofibroblasts, the three hydroxypyridones did not reverse myofibroblast transformation, but instead elicited a concentration-dependent decrease in cell viability. TUNEL staining confirmed that the hydroxypyridone anti-fungals induced apoptosis in established myofibroblasts. This is the first study to show that hydroxypyridone anti-fungals are capable of inducing apoptosis in established myofibroblasts. Together with our previous results, we suggest that hydroxypyridone anti-fungals can prevent scar formation by preventing the formation of new myofibroblasts and by reducing the number of existing myofibroblasts.
Collapse
Affiliation(s)
- Alice Ruth Lapthorn
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, UK.
| | - Marcus Maximillian Ilg
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, UK
| | - Peter Dziewulski
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, UK; St. Andrew's Centre for Plastic Surgery and Burns, Broomfield Hospital, Chelmsford, UK; St Andrew's Anglia Ruskin Research Group (StAAR), Chelmsford, UK
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, School of Allied Health, Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Chelmsford, UK
| |
Collapse
|
6
|
Basalova N, Alexandrushkina N, Grigorieva O, Kulebyakina M, Efimenko A. Fibroblast Activation Protein Alpha (FAPα) in Fibrosis: Beyond a Perspective Marker for Activated Stromal Cells? Biomolecules 2023; 13:1718. [PMID: 38136590 PMCID: PMC10742035 DOI: 10.3390/biom13121718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
The development of tissue fibrosis is a complex process involving the interaction of multiple cell types, which makes the search for antifibrotic agents rather challenging. So far, myofibroblasts have been considered the key cell type that mediated the development of fibrosis and thus was the main target for therapy. However, current strategies aimed at inhibiting myofibroblast function or eliminating them fail to demonstrate sufficient effectiveness in clinical practice. Therefore, today, there is an unmet need to search for more reliable cellular targets to contribute to fibrosis resolution or the inhibition of its progression. Activated stromal cells, capable of active proliferation and invasive growth into healthy tissue, appear to be such a target population due to their more accessible localization in the tissue and their high susceptibility to various regulatory signals. This subpopulation is marked by fibroblast activation protein alpha (FAPα). For a long time, FAPα was considered exclusively a marker of cancer-associated fibroblasts. However, accumulating data are emerging on the diverse functions of FAPα, which suggests that this protein is not only a marker but also plays an important role in fibrosis development and progression. This review aims to summarize the current data on the expression, regulation, and function of FAPα regarding fibrosis development and identify promising advances in the area.
Collapse
Affiliation(s)
- Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Natalya Alexandrushkina
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| |
Collapse
|
7
|
Salminen A. The plasticity of fibroblasts: A forgotten player in the aging process. Ageing Res Rev 2023; 89:101995. [PMID: 37391015 DOI: 10.1016/j.arr.2023.101995] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Tissue-resident fibroblasts are mesenchymal cells which possess an impressive plasticity in their ability to modify their properties according to the requirements of the microenvironment. There are diverse subgroups of fibroblast phenotypes associated with different tissue pathological conditions, e.g., cancers, wound healing, and many fibrotic and inflammatory conditions. The heterogeneous phenotypes can be subdivided into fibrogenic and non-fibrogenic, inflammatory and immunosuppressive subtypes as well as cellular senescent subsets. A major hallmark of activated fibroblasts is that they contain different amounts of stress fibers combined with α-smooth muscle actin (α-SMA) protein, i.e., commonly this phenotype has been called the myofibroblast. Interestingly, several stresses associated with the aging process are potent inducers of myofibroblast differentiation, e.g., oxidative and endoplasmic reticulum stresses, extracellular matrix (ECM) disorders, inflammatory mediators, and telomere shortening. Accordingly, anti-aging treatments with metformin and rapamycin inhibited the differentiation of myofibroblasts in tissues. There is evidence that the senescent phenotype induced in cultured fibroblasts does not represent the phenotype of fibroblasts in aged tissues. Considering the versatile plasticity of fibroblasts as well as their frequency and structural importance in tissues, it does seem that fibroblasts are overlooked players in the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
8
|
Turishcheva EP, Vildanova MS, Vishnyakova PA, Matveeva DK, Saidova AA, Onishchenko GE, Smirnova EA. Phytohormones Affect Differentiation Status of Human Skin Fibroblasts via UPR Activation. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:810-822. [PMID: 37748877 DOI: 10.1134/s0006297923060093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 09/27/2023]
Abstract
Normalization of secretory activity and differentiation status of mesenchymal cells, including fibroblasts, is an important biomedical problem. One of the possible solutions is modulation of unfolded protein response (UPR) activated during fibroblast differentiation. Here, we investigated the effect of phytohormones on the secretory activity and differentiation of cultured human skin fibroblasts. Based on the analysis of expression of genes encoding UPR markers, abscisic acid (ABA) upregulated expression of the GRP78 and ATF4 genes, while gibberellic acid (GA) upregulated expression of CHOP. Evaluation of the biosynthetic activity of fibroblasts showed that ABA promoted secretion and synthesis of procollagen I and synthesis of fibronectin, as well as the total production of collagen and non-collagen proteins of the extracellular matrix (ECM). ABA also stimulated the synthesis of smooth muscle actin α (α-SMA), which is the marker of myofibroblasts, and increased the number of myofibroblasts in the cell population. On the contrary, GA increased the level of fibronectin secretion, but reduced procollagen I synthesis and the total production of the ECM collagen proteins. GA downregulated the synthesis of α-SMA and decreased the number of myofibroblasts in the cell population. Our results suggest that phytohormones modulate the biosynthetic activity of fibroblasts and affect their differentiation status.
Collapse
Affiliation(s)
| | - Mariya S Vildanova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Polina A Vishnyakova
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Healthcare of the Russian Federation, Moscow, 117997, Russia
- Peoples' Friendship University of Russia, Moscow, 117198, Russia
| | - Diana K Matveeva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia
| | - Aleena A Saidova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | | | - Elena A Smirnova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
9
|
Kalgudde Gopal S, Dai R, Stefanska AM, Ansari M, Zhao J, Ramesh P, Bagnoli JW, Correa-Gallegos D, Lin Y, Christ S, Angelidis I, Lupperger V, Marr C, Davies LC, Enard W, Machens HG, Schiller HB, Jiang D, Rinkevich Y. Wound infiltrating adipocytes are not myofibroblasts. Nat Commun 2023; 14:3020. [PMID: 37230982 DOI: 10.1038/s41467-023-38591-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
The origins of wound myofibroblasts and scar tissue remains unclear, but it is assumed to involve conversion of adipocytes into myofibroblasts. Here, we directly explore the potential plasticity of adipocytes and fibroblasts after skin injury. Using genetic lineage tracing and live imaging in explants and in wounded animals, we observe that injury induces a transient migratory state in adipocytes with vastly distinct cell migration patterns and behaviours from fibroblasts. Furthermore, migratory adipocytes, do not contribute to scar formation and remain non-fibrogenic in vitro, in vivo and upon transplantation into wounds in animals. Using single-cell and bulk transcriptomics we confirm that wound adipocytes do not convert into fibrogenic myofibroblasts. In summary, the injury-induced migratory adipocytes remain lineage-restricted and do not converge or reprogram into a fibrosing phenotype. These findings broadly impact basic and translational strategies in the regenerative medicine field, including clinical interventions for wound repair, diabetes, and fibrotic pathologies.
Collapse
Affiliation(s)
- Shruthi Kalgudde Gopal
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
- Institute of Lung Health and Immunity, Helmholtz Center Munich, Munich, Germany
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Ania Maria Stefanska
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Meshal Ansari
- Institute of Lung Health and Immunity, Helmholtz Center Munich, Munich, Germany
- Institute of AI for Health, Helmholtz Center Munich, Munich, Germany
| | - Jiakuan Zhao
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Pushkar Ramesh
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Johannes W Bagnoli
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilian University Munich, Munich, Germany
| | | | - Yue Lin
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Simon Christ
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany
| | - Ilias Angelidis
- Institute of Lung Health and Immunity, Helmholtz Center Munich, Munich, Germany
| | - Valerio Lupperger
- Institute of AI for Health, Helmholtz Center Munich, Munich, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Center Munich, Munich, Germany
| | - Lindsay C Davies
- Department of Microbiology, Tumour and Cell Biology (MTC), Karolinska Institute, Stockholm, Sweden
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilian University Munich, Munich, Germany
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Herbert B Schiller
- Institute of Lung Health and Immunity, Helmholtz Center Munich, Munich, Germany.
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany.
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
10
|
Al-Jazrawe M, Xu S, Poon R, Wei Q, Przybyl J, Varma S, van de Rijn M, Alman BA. CD142 Identifies Neoplastic Desmoid Tumor Cells, Uncovering Interactions Between Neoplastic and Stromal Cells That Drive Proliferation. CANCER RESEARCH COMMUNICATIONS 2023; 3:697-708. [PMID: 37377751 PMCID: PMC10128091 DOI: 10.1158/2767-9764.crc-22-0403] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/03/2023] [Accepted: 03/27/2023] [Indexed: 06/29/2023]
Abstract
The interaction between neoplastic and stromal cells within a tumor mass plays an important role in cancer biology. However, it is challenging to distinguish between tumor and stromal cells in mesenchymal tumors because lineage-specific cell surface markers typically used in other cancers do not distinguish between the different cell subpopulations. Desmoid tumors consist of mesenchymal fibroblast-like cells driven by mutations stabilizing beta-catenin. Here we aimed to identify surface markers that can distinguish mutant cells from stromal cells to study tumor-stroma interactions. We analyzed colonies derived from single cells from human desmoid tumors using a high-throughput surface antigen screen, to characterize the mutant and nonmutant cells. We found that CD142 is highly expressed by the mutant cell populations and correlates with beta-catenin activity. CD142-based cell sorting isolated the mutant population from heterogeneous samples, including one where no mutation was previously detected by traditional Sanger sequencing. We then studied the secretome of mutant and nonmutant fibroblastic cells. PTX3 is one stroma-derived secreted factor that increases mutant cell proliferation via STAT6 activation. These data demonstrate a sensitive method to quantify and distinguish neoplastic from stromal cells in mesenchymal tumors. It identifies proteins secreted by nonmutant cells that regulate mutant cell proliferation that could be therapeutically. Significance Distinguishing between neoplastic (tumor) and non-neoplastic (stromal) cells within mesenchymal tumors is particularly challenging, because lineage-specific cell surface markers typically used in other cancers do not differentiate between the different cell subpopulations. Here, we developed a strategy combining clonal expansion with surface proteome profiling to identify markers for quantifying and isolating mutant and nonmutant cell subpopulations in desmoid tumors, and to study their interactions via soluble factors.
Collapse
Affiliation(s)
- Mushriq Al-Jazrawe
- Hospital for Sick Children, Program in Developmental & Stem Cell Biology, Toronto, Ontario, Canada
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Steven Xu
- Hospital for Sick Children, Program in Developmental & Stem Cell Biology, Toronto, Ontario, Canada
| | - Raymond Poon
- Hospital for Sick Children, Program in Developmental & Stem Cell Biology, Toronto, Ontario, Canada
| | - Qingxia Wei
- Hospital for Sick Children, Program in Developmental & Stem Cell Biology, Toronto, Ontario, Canada
| | - Joanna Przybyl
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Benjamin A. Alman
- Hospital for Sick Children, Program in Developmental & Stem Cell Biology, Toronto, Ontario, Canada
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina
- Regeneration Next Initiative, Duke University, Durham, North Carolina
| |
Collapse
|
11
|
Bova RA, Lamont AC, Picou TJ, Ho VB, Gilchrist KH, Melton-Celsa AR. Shiga Toxin (Stx) Type 1a and Stx2a Translocate through a Three-Layer Intestinal Model. Toxins (Basel) 2023; 15:toxins15030207. [PMID: 36977098 PMCID: PMC10054274 DOI: 10.3390/toxins15030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Shiga toxins (Stxs) produced by ingested E. coli can induce hemolytic uremic syndrome after crossing the intact intestinal barrier, entering the bloodstream, and targeting endothelial cells in the kidney. The method(s) by which the toxins reach the bloodstream are not fully defined. Here, we used two polarized cell models to evaluate Stx translocation: (i) a single-layer primary colonic epithelial cell model and (ii) a three-cell-layer model with colonic epithelial cells, myofibroblasts, and colonic endothelial cells. We traced the movement of Stx types 1a and 2a across the barrier models by measuring the toxicity of apical and basolateral media on Vero cells. We found that Stx1a and Stx2a crossed both models in either direction. However, approximately 10-fold more Stx translocated in the three-layer model as compared to the single-layer model. Overall, the percentage of toxin that translocated was about 0.01% in the epithelial-cell-only model but up to 0.09% in the three-cell-layer model. In both models, approximately 3- to 4-fold more Stx2a translocated than Stx1a. Infection of the three-cell-layer model with Stx-producing Escherichia coli (STEC) strains showed that serotype O157:H7 STEC reduced barrier function in the model and that the damage was not dependent on the presence of the eae gene. Infection of the three-layer model with O26:H11 STEC strain TW08571 (Stx1a+ and Stx2a+), however, allowed translocation of modest amounts of Stx without reducing barrier function. Deletion of stx2a from TW08571 or the use of anti-Stx1 antibody prevented translocation of toxin. Our results suggest that single-cell models may underestimate the amount of Stx translocation and that the more biomimetic three-layer model is suited for Stx translocation inhibitor studies.
Collapse
Affiliation(s)
- Rebecca A. Bova
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Center for Biotechnology (4DBio3), Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD 20814, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Andrew C. Lamont
- Center for Biotechnology (4DBio3), Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD 20814, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Theodore J. Picou
- Center for Biotechnology (4DBio3), Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD 20814, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Vincent B. Ho
- Center for Biotechnology (4DBio3), Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD 20814, USA
| | - Kristin H. Gilchrist
- Center for Biotechnology (4DBio3), Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, MD 20814, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Angela R. Melton-Celsa
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Correspondence:
| |
Collapse
|
12
|
Hsiao Y, Wang I, Yang T. Fibrotic remodeling and tissue regeneration mechanisms define the therapeutic potential of human muscular progenitors. Bioeng Transl Med 2023; 8:e10439. [PMID: 36925693 PMCID: PMC10013817 DOI: 10.1002/btm2.10439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022] Open
Abstract
Fibrosis is an intrinsic biological reaction toward the challenges of tissue injury that is implicated in the wound-healing process. Although it is useful to efficiently mitigate the damage, progression of fibrosis is responsible for the morbidity and mortality occurring in a variety of diseases. Because of lacking effective treatments, there is an emerging need for exploring antifibrotic strategies. Cell therapy based on stem/progenitor cells is regarded as a promising approach for treating fibrotic diseases. Appropriate selection of cellular sources is required for beneficial results. Muscle precursor cells (MPCs) are specialized progenitors harvested from skeletal muscle for conducting muscle regeneration. Whether they are also effective in regulating fibrosis has seldom been explored and merits further investigation. MPCs were successfully harvested from all human samples regardless of demographic backgrounds. The extracellular matrices remodeling was enhanced through the paracrine effects mediated by MPCs. The suppression effects on fibrosis were confirmed in vivo when MPCs were transplanted into the diseased animals with oral submucous fibrosis. The data shown here revealed the potential of MPCs to be employed to simultaneously regulate both processes of fibrosis and tissue regeneration, supporting them as the promising cell candidates for development of the cell therapy for antifibrosis and tissue regeneration.
Collapse
Affiliation(s)
- Ya‐Chuan Hsiao
- Department of OphthalmologyTaipei City Hospital, Zhongxing BranchTaipeiTaiwan
- Department of OphthalmologyCollege of Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - I‐Han Wang
- Department of OtolaryngologyNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Tsung‐Lin Yang
- Department of OtolaryngologyNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan UniversityTaipeiTaiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
13
|
Saito S, Ohno SI, Harada Y, Kanno Y, Kuroda M. MiR-34a induces myofibroblast differentiation from renal fibroblasts. Clin Exp Nephrol 2023; 27:411-418. [PMID: 36808381 DOI: 10.1007/s10157-023-02329-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/07/2023] [Indexed: 02/21/2023]
Abstract
BACKGROUND Renal fibrosis is the common outcome of progressive kidney diseases. To avoid dialysis, the molecular mechanism of renal fibrosis must be explored further. MicroRNAs play key roles in renal fibrosis. MiR-34a is a transcriptional target of p53, which regulates the cell cycle and apoptosis. Previous studies demonstrated that miR-34a promotes renal fibrosis. However, the distinct roles of miR-34a in renal fibrosis have not been fully elucidated. Here, we identified the roles of miR-34a in renal fibrosis. METHOD We first analyzed p53 and miR-34a expression in kidney tissues in s UUO (unilateral ureteral obstruction) mouse model. Then, to confirm the effects of miR-34a in vitro, we transfected a miR-34a mimic into a kidney fibroblast cell line (NRK-49F) and analyzed. RESULTS We found that the expression of p53 and miR-34a was upregulated after UUO. Furthermore, after transfection of the miR-34a mimic into kidney fibroblasts, the expression of α-SMA was upregulated dramatically. In addition, α-SMA upregulation was greater upon transfection of the miR-34a mimic than upon treatment with TGF-β1. Moreover, high expression of Acta2 was maintained despite sufficient removal of the miR-34a mimic by changing the medium 4 times during the 9-day culture. After transfection of the miR-34a mimic into kidney fibroblasts, we did not detect phospho-SMAD2/3 by immunoblotting analysis. CONCLUSION Our study revealed that miR-34a induces myofibroblast differentiation from renal fibroblasts. Moreover, the miR-34a-induced upregulation of α-SMA was independent of the TGF-β/SMAD signaling pathway. In conclusion, our study indicated that the p53/miR-34a axis promotes the development of renal fibrosis.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Nephrology, Tokyo Medical University, Tokyo, Japan
| | - Shin-Ichiro Ohno
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| | - Yuichirou Harada
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Yoshihiko Kanno
- Department of Nephrology, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| |
Collapse
|
14
|
Rubino M, Travers JG, Headrick AL, Enyart BT, Lemieux ME, Cavasin MA, Schwisow JA, Hardy EJ, Kaltenbacher KJ, Felisbino MB, Jonas E, Ambardekar AV, Bristow MR, Koch KA, McKinsey TA. Inhibition of Eicosanoid Degradation Mitigates Fibrosis of the Heart. Circ Res 2023; 132:10-29. [PMID: 36475698 DOI: 10.1161/circresaha.122.321475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Organ fibrosis due to excessive production of extracellular matrix by resident fibroblasts is estimated to contribute to >45% of deaths in the Western world, including those due to cardiovascular diseases such as heart failure. Here, we screened for small molecule inhibitors with a common ability to suppress activation of fibroblasts across organ systems. METHODS High-content imaging of cultured cardiac, pulmonary, and renal fibroblasts was used to identify nontoxic compounds that blocked induction of markers of activation in response to the profibrotic stimulus, transforming growth factor-β1. SW033291, which inhibits the eicosanoid-degrading enzyme, 15-hydroxyprostaglandin dehydrogenase, was chosen for follow-up studies with cultured adult rat ventricular fibroblasts and human cardiac fibroblasts (CF), and for evaluation in mouse models of cardiac fibrosis and diastolic dysfunction. Additional mechanistic studies were performed with CFs treated with exogenous eicosanoids. RESULTS Nine compounds, including SW033291, shared a common ability to suppress transforming growth factor-β1-mediated activation of cardiac, pulmonary, and renal fibroblasts. SW033291 dose-dependently inhibited transforming growth factor-β1-induced expression of activation markers (eg, α-smooth muscle actin and periostin) in adult rat ventricular fibroblasts and normal human CFs, and reduced contractile capacity of the cells. Remarkably, the 15-hydroxyprostaglandin dehydrogenase inhibitor also reversed constitutive activation of fibroblasts obtained from explanted hearts from patients with heart failure. SW033291 blocked cardiac fibrosis induced by angiotensin II infusion and ameliorated diastolic dysfunction in an alternative model of systemic hypertension driven by combined uninephrectomy and deoxycorticosterone acetate administration. Mechanistically, SW033291-mediated stimulation of extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase signaling was required for the compound to block CF activation. Of the 12 exogenous eicosanoids that were tested, only 12(S)-hydroxyeicosatetraenoic acid, which signals through the G protein-coupled receptor, GPR31, recapitulated the suppressive effects of SW033291 on CF activation. CONCLUSIONS Inhibition of degradation of eicosanoids, arachidonic acid-derived fatty acids that signal through G protein-coupled receptors, is a potential therapeutic strategy for suppression of pathological organ fibrosis. In the heart, we propose that 15-hydroxyprostaglandin dehydrogenase inhibition triggers CF-derived autocrine/paracrine signaling by eicosanoids, including 12(S)-hydroxyeicosatetraenoic acid, to stimulate extracellular signal-regulated kinase 1/2 and block conversion of fibroblasts into activated cells that secrete excessive amounts of extracellular matrix and contribute to heart failure pathogenesis.
Collapse
Affiliation(s)
- Marcello Rubino
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Joshua G Travers
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Alaina L Headrick
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Blake T Enyart
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | | | - Maria A Cavasin
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Jessica A Schwisow
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Elizabeth J Hardy
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Keenan J Kaltenbacher
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Marina B Felisbino
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Eric Jonas
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Amrut V Ambardekar
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Michael R Bristow
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Keith A Koch
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| | - Timothy A McKinsey
- From the Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., J.A.S., E.J.H., K.J.K., M.B.F., E.J., A.V.A., M.R.B., K.A.K., T.A.M.).,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora (M.R., J.G.T., A.L.H., B.T.E., M.A.C., E.J.H., K.J.K., M.B.F., A.V.A., M.R.B., K.A.K., T.A.M.)
| |
Collapse
|
15
|
Bellei B, Migliano E, Picardo M. Therapeutic potential of adipose tissue-derivatives in modern dermatology. Exp Dermatol 2022; 31:1837-1852. [PMID: 35102608 DOI: 10.1111/exd.14532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022]
Abstract
Stem cell-mediated therapies in combination with biomaterial and growth factor-based approaches in regenerative medicine are rapidly evolving with increasing application beyond the dermatologic field. Adipose-derived stem cells (ADSCs) are the more frequently used adult stem cells due to their abundance and easy access. In the case of volumetric defects, adipose tissue can take the shape of defects, restoring the volume and enhancing the regeneration of receiving tissue. When regenerative purposes prevail on volume restoration, the stromal vascular fraction (SVF) rich in staminal cells, purified mesenchymal stem cells (MSCs) or their cell-free derivatives grafting are favoured. The therapeutic efficacy of acellular approaches is explained by the fact that a significant part of the natural propensity of stem cells to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines and extracellular matrix components. Therefore, the secretome's ability to modulate multiple targets simultaneously demonstrated preclinical and clinical efficacy in reversing pathological mechanisms of complex conditions such atopic dermatitis (AD), vitiligo, psoriasis, acne and Lichen sclerosus (LS), non-resolving wounds and alopecia. This review analysing both in vivo and in vitro models gives an overview of the clinical relevance of adipose tissue-derivatives such as autologous fat graft, stromal vascular fraction, purified stem cells and secretome for skin disorders application. Finally, we highlighted the major disease-specific limitations and the future perspective in this field.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Emilia Migliano
- Department of Plastic and Regenerative Surgery, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| |
Collapse
|
16
|
Turishcheva E, Vildanova M, Onishchenko G, Smirnova E. The Role of Endoplasmic Reticulum Stress in Differentiation of Cells of Mesenchymal Origin. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:916-931. [PMID: 36180988 PMCID: PMC9483250 DOI: 10.1134/s000629792209005x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 05/23/2023]
Abstract
Endoplasmic reticulum (ER) is a multifunctional membrane-enclosed organelle. One of the major ER functions is cotranslational transport and processing of secretory, lysosomal, and transmembrane proteins. Impaired protein processing caused by disturbances in the ER homeostasis results in the ER stress. Restoration of normal ER functioning requires activation of an adaptive mechanism involving cell response to misfolded proteins, the so-called unfolded protein response (UPR). Besides controlling protein folding, UPR plays a key role in other physiological processes, in particular, differentiation of cells of connective, muscle, epithelial, and neural tissues. Cell differentiation is induced by the physiological levels of ER stress, while excessive ER stress suppresses differentiation and can result in cell death. So far, it remains unknown whether UPR activation induces cell differentiation or if UPR is initiated by the upregulated synthesis of secretory proteins during cell differentiation. Cell differentiation is an important stage in the development of multicellular organisms and is tightly controlled. Suppression or excessive activation of this process can lead to the development of various pathologies in an organism. In particular, impairments in the differentiation of connective tissue cells can result in the development of fibrosis, obesity, and osteoporosis. Recently, special attention has been paid to fibrosis as one of the major complications of COVID-19. Therefore, studying the role of UPR in the activation of cell differentiation is of both theoretical and practical interest, as it might result in the identification of molecular targets for selective regulation of cell differentiation stages and as well as the potential to modulate the mechanisms involved in the development of various pathological states.
Collapse
Affiliation(s)
| | - Mariya Vildanova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Galina Onishchenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elena Smirnova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
17
|
Hou J, Yun Y, Cui C, Kim S. Ginsenoside Rh2 mitigates doxorubicin-induced cardiotoxicity by inhibiting apoptotic and inflammatory damage and weakening pathological remodelling in breast cancer-bearing mice. Cell Prolif 2022; 55:e13246. [PMID: 35534947 PMCID: PMC9201376 DOI: 10.1111/cpr.13246] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/10/2022] [Accepted: 04/18/2022] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVES There are presently a few viable ways to reduce cardiotoxicity of doxorubicin (Dox). The combination of chemotherapy agents with natural compounds delivers greater efficacy and reduces adverse effects in recent researches for cancer treatment. Here, we examined the potential effect of ginsenoside Rh2 on a Dox-based regimen in chemotherapy treatment. MATERIALS AND METHODS Human breast tumour (MDA-MB-231) xenograft nude mice, human cardiac ventricle fibroblasts, and human umbilical vein endothelial cells (HUVEC) were employed in the present study. Histology, immunohistochemistry, immunofluorescence, western blot, antibody array, and RNA-sequencing analyses were utilized to assess the protective effect of Rh2 on cardiotoxicity induced by Dox and the underlying mechanisms. RESULTS Rh2-reduced cardiotoxicity by inhibiting the cardiac histopathological changes, apoptosis and necrosis, and consequent inflammation. Pathological remodelling was attenuated by reducing fibroblast to myofibroblast transition (FMT) and endothelial-mesenchymal transition (EndMT) in hearts. RNA-sequencing analysis showed that Dox treatment predominantly targets cell cycle and attachment of microtubules and boosted tumour necrosis, chemokine and interferon-gamma production, response to cytokine and chemokine, and T cell activation, whereas Rh2 regulated these effects. Intriguingly, Rh2 also attenuated fibrosis via promoting senescence in myofibroblasts and reversing established myofibroblast differentiation in EndMT. CONCLUSIONS Rh2 regulates multiple pathways in the Dox-provoked heart, proposing a potential candidate for cancer supplement and therapy-associated cardiotoxicity.
Collapse
Affiliation(s)
- Jingang Hou
- Intelligent Synthetic Biology Center, Daejeon, Republic of Korea
| | - Yeejin Yun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Changhao Cui
- Research and Development Team 4, Sempio Foods Company, Cheongju, South Korea
| | - Sunchang Kim
- Intelligent Synthetic Biology Center, Daejeon, Republic of Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
18
|
Bellei B, Migliano E, Picardo M. Research update of adipose tissue-based therapies in regenerative dermatology. Stem Cell Rev Rep 2022; 18:1956-1973. [PMID: 35230644 DOI: 10.1007/s12015-022-10328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 12/09/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) have a spontaneous propensity to support tissue homeostasis and regeneration. Among the several sources of MSCs, adipose-derived tissue stem cells (ADSCs) have received major interest due to the higher mesenchymal stem cells concentration, ease, and safety of access. However, since a significant part of the natural capacity of ADSCs to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines, lipids, and extracellular matrix components, several studies focused on cell-free strategies. Furthermore, adipose cell-free derivatives are becoming more attractive especially for non-volumizing purposes, such as most dermatological conditions. However, when keratinocytes, fibroblasts, melanocytes, adipocytes, and hair follicle cells might not be locally sourced, graft of materials containing concentrated ADSCs is preferred. The usage of extracellular elements of adipose tissue aims to promote a self-autonomous regenerative microenvironment in the receiving area restoring physiological homeostasis. Hence, ADSCs or their paracrine activity are currently being studied in several dermatological settings including wound healing, skin fibrosis, burn, and aging.The present work analyzing both preclinical and clinical experiences gives an overview of the efficacy of adipose tissue-derivatives like autologous fat, the stromal vascular fraction (SVF), purified ADSCs, secretome and extracellular matrix graft in the field of regenerative medicine for the skin.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Emilia Migliano
- Department of Plastic and Reconstructive Surgery, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
19
|
Bi X, Li Y, Dong Z, Zhao J, Wu W, Zou J, Guo L, Lu F, Gao J. Recent Developments in Extracellular Matrix Remodeling for Fat Grafting. Front Cell Dev Biol 2021; 9:767362. [PMID: 34977018 PMCID: PMC8716396 DOI: 10.3389/fcell.2021.767362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Remodeling of the extracellular matrix (ECM), which provides structural and biochemical support for surrounding cells, is vital for adipose tissue regeneration after autologous fat grafting. Rapid and high-quality ECM remodeling can improve the retention rate after fat grafting by promoting neovascularization, regulating stem cells differentiation, and suppressing chronic inflammation. The degradation and deposition of ECM are regulated by various factors, including hypoxia, blood supply, inflammation, and stem cells. By contrast, ECM remodeling alters these regulatory factors, resulting in a dynamic relationship between them. Although researchers have attempted to identify the cellular sources of factors associated with tissue regeneration and regulation of the microenvironment, the factors and mechanisms that affect adipose tissue ECM remodeling remain incompletely understood. This review describes the process of adipose ECM remodeling after grafting and summarizes the factors that affect ECM reconstruction. Also, this review provides an overview of the clinical methods to avoid poor ECM remodeling. These findings may provide new ideas for improving the retention of adipose tissue after fat transplantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Possible Treatment of Myocardial Infarct Based on Tissue Engineering Using a Cellularized Solid Collagen Scaffold Functionalized with Arg-Glyc-Asp (RGD) Peptide. Int J Mol Sci 2021; 22:ijms222212563. [PMID: 34830447 PMCID: PMC8620820 DOI: 10.3390/ijms222212563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.
Collapse
|
21
|
Walters B, Turner PA, Rolauffs B, Hart ML, Stegemann JP. Controlled Growth Factor Delivery and Cyclic Stretch Induces a Smooth Muscle Cell-like Phenotype in Adipose-Derived Stem Cells. Cells 2021; 10:cells10113123. [PMID: 34831345 PMCID: PMC8624888 DOI: 10.3390/cells10113123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are an abundant and easily accessible multipotent stem cell source with potential application in smooth muscle regeneration strategies. In 3D collagen hydrogels, we investigated whether sustained release of growth factors (GF) PDGF-AB and TGF-β1 from GF-loaded microspheres could induce a smooth muscle cell (SMC) phenotype in ASCs, and if the addition of uniaxial cyclic stretch could enhance the differentiation level. This study demonstrated that the combination of cyclic stretch and GF release over time from loaded microspheres potentiated the differentiation of ASCs, as quantified by protein expression of early to late SMC differentiation markers (SMA, TGLN and smooth muscle MHC). The delivery of GFs via microspheres produced large ASCs with a spindle-shaped, elongated SMC-like morphology. Cyclic strain produced the largest, longest, and most spindle-shaped cells regardless of the presence or absence of growth factors or the growth factor delivery method. Protein expression and cell morphology data confirmed that the sustained release of GFs from GF-loaded microspheres can be used to promote the differentiation of ASCs into SMCs and that the addition of uniaxial cyclic stretch significantly enhances the differentiation level, as quantified by intermediate and late SMC markers and a SMC-like elongated cell morphology.
Collapse
Affiliation(s)
- Brandan Walters
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Paul A. Turner
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
| | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstraße 4, 79108 Freiburg, Germany;
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, 1107 Carl A. Gerstacker Building, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; (B.W.); (P.A.T.)
- Correspondence: (M.L.H.); (J.P.S.); Tel.: +49-(761)-270-26102 (M.L.H.); +001-(734)-764-8313 (J.P.S.)
| |
Collapse
|
22
|
Sun W, Byon CH, Kim DH, Choi HI, Park JS, Joo SY, Kim IJ, Jung I, Bae EH, Ma SK, Kim SW. Renoprotective Effects of Maslinic Acid on Experimental Renal Fibrosis in Unilateral Ureteral Obstruction Model via Targeting MyD88. Front Pharmacol 2021; 12:708575. [PMID: 34588982 PMCID: PMC8475766 DOI: 10.3389/fphar.2021.708575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Maslinic acid (MA), also named crategolic acid, is a pentacyclic triterpene extracted from fruits and vegetables. Although various beneficial pharmacological effects of MA have been revealed, its effect on renal fibrosis remains unclear. This study was designed to clarify whether MA could attenuate renal fibrosis and determine the putative underlying molecular mechanisms. We demonstrated that MA-treated mice with unilateral ureteral obstruction (UUO) developed a histological injury of low severity and exhibited downregulated expression of fibrotic markers, including α-smooth muscle actin (α-SMA), vimentin, and fibronectin by 38, 44 and 40%, and upregulated expression of E-cadherin by 70% as compared with untreated UUO mice. Moreover, MA treatment restored the expression levels of α-SMA, connective tissue growth factor, and vimentin to 10, 7.8 and 38% of those induced by transforming growth factor (TGF)-β in NRK49F cells. MA decreased expression of Smad2/3 phosphorylation and Smad4 in UUO kidneys and TGF-β treated NRK49F cells (p < 0.05, respectively). Notably, MA specifically interferes with MyD88, an adaptor protein, thereby mitigating Smad4 nuclear expression (p < 0.01 compared to TGF-β treated group) and ameliorating renal fibrotic changes (p < 0.01 for each fibrotic markers compared to TGF-β induced cells). In addition, in the UUO model and lipopolysaccharide-induced NRK49F cells, MA treatment decreased the expression of IL-1β, TGF-α and MCP-1, ICAM-1, associated with the suppression of NF-κB signaling. These findings suggest that MA is a potential agent that can reduce renal interstitial fibrosis, to some extent, via targeting TGF-β/Smad and MyD88 signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
23
|
Wang Y, Feng Q, Li Z, Bai X, Wu Y, Liu Y. Evaluating the Effect of Integra Seeded with Adipose Tissue-Derived Stem Cells or Fibroblasts in Wound Healing. Curr Drug Deliv 2021; 17:629-635. [PMID: 32394832 DOI: 10.2174/1567201817666200512104004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/27/2020] [Accepted: 04/18/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Extensive loss of skin in burn patients can have devastating consequences, both physically and mentally. Adipose-Derived Stem Cells (ADSCs) and fibroblasts are known to play significant roles in the process of wound healing. Recently, bioengineered skin has been considered for wound healing purposes. METHODS Investigate the effect of Integra seeded with ADSCs, fibroblasts, or both on wound healing. RESULTS We found that when Integra is seeded with ADSCs and fibroblasts, both types of cells incorporate and proliferate, the phenomenon becoming more robust when the cells are co-cultured on Integra, both in vitro; and in vivo;. In addition, when these cells are seeded on Integra, they stimulate epithelization with no signs of inflammation and skin necrosis being observed when transplanted on animals for 7 days. CONCLUSION ADSCs and fibroblasts seeded on Integra could decrease the number of α-SMA positive myofibroblasts, leading to scarless wound healing. The evidence from this study is strongly supportive that Integra seeded with ADSCs and fibroblasts is an appropriate and effective bioengineered skin for wound healing.
Collapse
Affiliation(s)
- Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Quanrui Feng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiping Wu
- Department of Plastic and Aesthetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yukun Liu
- Department of Plastic and Aesthetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
24
|
Ikeda S, Tsuji S, Ohama T, Sato K. Involvement of PP2A methylation in the adipogenic differentiation of bone marrow-derived mesenchymal stem cell. J Biochem 2021; 168:643-650. [PMID: 32663263 DOI: 10.1093/jb/mvaa077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/20/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent stem cells with ability to self-replicate and differentiate into mesodermal derivatives, such as adipocytes and osteoblasts. BM-MSCs are a critical component of the tumour microenvironment. They support tumour progression by recruiting additional BM-MSCs and by differentiating into myofibroblasts (also called cancer-associated fibroblasts). Protein phosphatase 2A (PP2A) is an essential serine/threonine protein phosphatase that regulates a broad range of cellular signalling. PP2A forms a heterotrimer to dephosphorylate specific substrates. The reversible methylesterification (methylation) of Leu309 in the catalytic subunit of PP2A (PP2Ac) regulates biogenesis of the PP2A holoenzyme. It is unknown whether the methylation of PP2Ac plays a role in BM-MSC differentiation. Our experiments determined that protein levels of PP2A subunits and PP2A methyltransferase (LCMT-1) are significantly altered during differentiation. PP2Ac methylation levels in BM-MSCs decrease over time in response to an adipogenic differentiation stimulus. However, blockage of PP2A demethylation using the PP2A dimethyl-esterase inhibitors enhanced adipocyte differentiation. This suggests that PP2Ac demethylation is involved in adipocyte differentiation resistance. The results of our study provide a greater understanding of the regulation of BM-MSCs differentiation by PP2A holoenzyme.
Collapse
Affiliation(s)
- Shunta Ikeda
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Shunya Tsuji
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| |
Collapse
|
25
|
A formalism for modelling traction forces and cell shape evolution during cell migration in various biomedical processes. Biomech Model Mechanobiol 2021; 20:1459-1475. [PMID: 33893558 PMCID: PMC8298374 DOI: 10.1007/s10237-021-01456-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 01/17/2023]
Abstract
The phenomenological model for cell shape deformation and cell migration Chen (BMM 17:1429–1450, 2018), Vermolen and Gefen (BMM 12:301–323, 2012), is extended with the incorporation of cell traction forces and the evolution of cell equilibrium shapes as a result of cell differentiation. Plastic deformations of the extracellular matrix are modelled using morphoelasticity theory. The resulting partial differential differential equations are solved by the use of the finite element method. The paper treats various biological scenarios that entail cell migration and cell shape evolution. The experimental observations in Mak et al. (LC 13:340–348, 2013), where transmigration of cancer cells through narrow apertures is studied, are reproduced using a Monte Carlo framework.
Collapse
|
26
|
Garrison CM, Schwarzbauer JE. Fibronectin fibril alignment is established upon initiation of extracellular matrix assembly. Mol Biol Cell 2021; 32:739-752. [PMID: 33625865 PMCID: PMC8108514 DOI: 10.1091/mbc.e20-08-0533] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The physical structure of the extracellular matrix (ECM) is tissue-specific and fundamental to normal tissue function. Proper alignment of ECM fibers is essential for the functioning of a variety of tissues. While matrix assembly in general has been intensively investigated, little is known about the mechanisms required for formation of aligned ECM fibrils. We investigated the initiation of fibronectin (FN) matrix assembly using fibroblasts that assemble parallel ECM fibrils and found that matrix assembly sites, where FN fibrillogenesis is initiated, were oriented in parallel at the cell poles. We show that these polarized matrix assembly sites progress into fibrillar adhesions and ultimately into aligned FN fibrils. Cells that assemble an unaligned meshwork matrix form matrix assembly sites around the cell periphery, but the distribution of matrix assembly sites in these cells could be modulated through micropatterning or mechanical stretch. While an elongated cell shape corresponds with a polarized matrix assembly site distribution, these two features are not absolutely linked, since we discovered that transforming growth factor beta (TGF-β1) enhances matrix assembly site polarity and assembly of aligned fibrils independent of cell elongation. We conclude that the ultimate orientation of FN fibrils is determined by the alignment and distribution of matrix assembly sites that form during the initial stages of cell–FN interactions.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
27
|
Venter C, Myburgh KH, Niesler CU. Co-culture of pro-inflammatory macrophages and myofibroblasts: Evaluating morphological phenotypes and screening the effects of signaling pathway inhibitors. Physiol Rep 2021; 9:e14704. [PMID: 33463904 PMCID: PMC7814483 DOI: 10.14814/phy2.14704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle regeneration is a complex process influenced by non-myogenic macrophages and fibroblasts, which acquire different phenotypes in response to changes in the injury milieu or changes in experimental conditions. In vitro, serum stimulates the differentiation of fibroblasts into myofibroblasts, while lipopolysaccharide (LPS) stimulates the polarization of unstimulated (M0) macrophages to acquire an M1 pro-inflammatory phenotype. We characterized these phenotypes using morphology (with circularity as shape descriptor; perfect circularity = 1.0) and phenotype-specific markers. Myofibroblasts (high α-smooth muscle actin [SMA] expression) had high circularity (mean 0.60 ± 0.03). Their de-differentiation to fibroblasts (low α-SMA expression) significantly lessened circularity (0.47 ± 0.01 and 0.35 ± 0.02 in 2% or 0% serum culture media respectively (p < 0.05). Unstimulated (M0) macrophages (no CD86 expression) had high circularity (0.72 ± 0.02) which decreased when stimulated to M1 macrophages (CD86 expression) (LPS; 0.61 ± 0.02; p < 0.05). Utilizing these established conditions, we then co-cultured M1 macrophages with myofibroblasts or myoblasts. M1 macrophages significantly decreased relative myofibroblast numbers (from 223 ± 22% to 64 ± 7%), but not myoblast numbers. This pro-inflammatory co-culture model was used to rapidly screen the following four compounds for ability to prevent M1 macrophage-mediated decrease in myofibroblast numbers: L-NAME (inducible nitric oxide synthase inhibitor), SB203580 (p38 mitogen-activated protein kinase inhibitor), SP600125 (c-Jun N-terminal kinase inhibitor) and LY294002 (phosphoinositide 3-kinase [PI3K] inhibitor). We found that LY294002 rescued myofibroblasts and decreased macrophage numbers. Myofibroblast rescue did not occur with L-NAME, SB203580 or SP600125 incubation. In conclusion, these data suggest a PI3K-associated mechanism whereby myofibroblasts can be rescued, despite simulated pro-inflammatory conditions.
Collapse
Affiliation(s)
- Colin Venter
- Discipline of BiochemistrySchool of Life SciencesUniversity of KwaZulu‐NatalScottsvilleSouth Africa
| | - Kathryn H. Myburgh
- Department Physiological SciencesStellenbosch UniversityMatielandSouth Africa
| | - Carola U. Niesler
- Discipline of BiochemistrySchool of Life SciencesUniversity of KwaZulu‐NatalScottsvilleSouth Africa
| |
Collapse
|
28
|
Karic V, Chandran R, Abrahamse H. Laser-Induced Differentiation of Human Adipose-Derived Stem Cells to Temporomandibular Joint Disc Cells. Lasers Surg Med 2020; 53:567-577. [PMID: 33030751 DOI: 10.1002/lsm.23332] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/16/2020] [Accepted: 09/27/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Temporomandibular disorder (TMD) is an incapacitating disease with temporomandibular joint (TMJ) disc degenerative changes in patients. Despite several research attempts to find a definitive treatment, there is no evidence of a permanent solution. The objective of the current study was to observe the role of 660 nm diode laser in the differentiation of human adipose-derived stem cells (ADSCs) to fibroblasts and chondrocytes. STUDY DESIGN/MATERIALS AND METHODS After irradiation, the morphology, viability, and adenosine triphosphate (ATP) proliferation of the ADSCs were analyzed at different time intervals. The differentiation of ADSCs toward fibroblastic and chondrogenic phenotypes was supported using flow cytometry and immunofluorescence at 1- and 2-week post-irradiation. RESULTS More than 90% of viable cells were observed in all experimental groups, with an increase in ATP proliferation. Flow cytometry analyses and immunofluorescence demonstrated the presence of chondrogenic and fibroblastic cell surface markers at 1- and 2-week post-irradiation. CONCLUSION This study has demonstrated methods to induce the differentiation of ADSCs toward fibroblastic and chondrogenic phenotypes with a 660 nm diode laser. The study also proposes a future alternative method of treatment for patients with degenerative TMJ disc disorders and presents a positive prospect in the application of photobiomodulation and ADSCs in the treatment of degenerative TMJ disc. Lasers Surg. Med. © 2020 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Vesna Karic
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO, Box 17011, Doornfontein, Johannesburg, 2028, South Africa.,Department of Prosthodontic and Oral Rehabilitation, and Laser Therapy in Dentistry, Division, School of Oral Sciences, Health Sciences Faculty, WITS University, PO Box, 2010, 7 York Street, Johannesburg, 2193, South Africa
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO, Box 17011, Doornfontein, Johannesburg, 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO, Box 17011, Doornfontein, Johannesburg, 2028, South Africa
| |
Collapse
|
29
|
Takeuchi S, Yamanouchi K, Sugihara H, Matsuwaki T, Nishihara M. Differentiation of skeletal muscle Mesenchymal progenitor cells to myofibroblasts is reversible. Anim Sci J 2020; 91:e13368. [PMID: 32285501 PMCID: PMC7216888 DOI: 10.1111/asj.13368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022]
Abstract
Accumulation of intramuscular adipose tissue (IMAT) and development of fibrous tissues due to accumulation of collagen both affect meat quality such as tenderness, texture, and flavor. Thus, it is important for the production of high‐quality meat to regulate the amount of adipose and fibrous tissues in skeletal muscle. IMAT is comprised of adipocytes, while collagens included in fibrous tissues are mainly produced by activated fibroblasts. Both adipocytes and fibroblasts are differentiated from their common ancestors, called mesenchymal progenitor cells (MPC). We previously established rat MPC clone, 2G11 cells. As several reports implicated the plasticity of fibroblast differentiation, in the present study, using 2G11 cells, we asked whether myofibroblasts differentiated from MPC are capable of re‐gaining adipogenic potential in vitro. By treating with bFGF, their αSMA expression was reduced and adipogenic potential was restored partially. Furthermore, by lowering cell density together with bFGF treatment, 2G11 cell‐derived myofibroblasts lost αSMA expression and showed the highest adipogenic potential, and this was along with their morphological change from flattened‐ to spindle‐like shape, which is typically observed with MPC. These results indicated that MPC‐derived myofibroblasts could re‐acquire adipogenic potential, possibly mediated through returning to an undifferentiated MPC‐like state.
Collapse
Affiliation(s)
- Shiho Takeuchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hidetoshi Sugihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takashi Matsuwaki
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
30
|
Sagaradze GD, Basalova NA, Efimenko AY, Tkachuk VA. Mesenchymal Stromal Cells as Critical Contributors to Tissue Regeneration. Front Cell Dev Biol 2020; 8:576176. [PMID: 33102483 PMCID: PMC7546871 DOI: 10.3389/fcell.2020.576176] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/18/2020] [Indexed: 12/27/2022] Open
Abstract
Adult stem cells that are tightly regulated by the specific microenvironment, or the stem cell niche, function to maintain tissue homeostasis and regeneration after damage. This demands the existence of specific niche components that can preserve the stem cell pool in injured tissues and restore the microenvironment for their subsequent appropriate functioning. This role may belong to mesenchymal stromal cells (MSCs) due to their resistance to damage signals and potency to be specifically activated in response to tissue injury and promote regeneration by different mechanisms. Increased amount of data indicate that activated MSCs are able to produce factors such as extracellular matrix components, growth factors, extracellular vesicles and organelles, which transiently substitute the regulatory signals from missing niche cells and restrict the injury-induced responses of them. MSCs may recruit functional cells into a niche or differentiate into missing cell components to endow a niche with ability to regulate stem cell fates. They may also promote the dedifferentiation of committed cells to re-establish a pool of functional stem cells after injury. Accumulated evidence indicates the therapeutic promise of MSCs for stimulating tissue regeneration, but the benefits of administered MSCs demonstrated in many injury models are less than expected in clinical studies. This emphasizes the importance of considering the mechanisms of endogenous MSC functioning for the development of effective approaches to their pharmacological activation or mimicking their effects. To achieve this goal, we integrate the current ideas on the contribution of MSCs in restoring the stem cell niches after damage and thereby tissue regeneration.
Collapse
Affiliation(s)
- Georgy D Sagaradze
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya A Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Anastasia Yu Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vsevolod A Tkachuk
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
31
|
Human Cardiac Fibroblast Number and Activation State Modulate Electromechanical Function of hiPSC-Cardiomyocytes in Engineered Myocardium. Stem Cells Int 2020; 2020:9363809. [PMID: 32724316 PMCID: PMC7381987 DOI: 10.1155/2020/9363809] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/11/2020] [Indexed: 11/27/2022] Open
Abstract
Cardiac tissue engineering using hiPSC-derived cardiomyocytes is a promising avenue for cardiovascular regeneration, pharmaceutical drug development, cardiotoxicity evaluation, and disease modeling. Limitations to these applications still exist due in part to the need for more robust structural support, organization, and electromechanical function of engineered cardiac tissues. It is well accepted that heterotypic cellular interactions impact the phenotype of cardiomyocytes. The current study evaluates the functional effects of coculturing adult human cardiac fibroblasts (hCFs) in 3D engineered tissues on excitation and contraction with the goal of recapitulating healthy, nonarrhythmogenic myocardium in vitro. A small population (5% of total cell number) of hCFs in tissues improves tissue formation, material properties, and contractile function. However, two perturbations to the hCF population create disease-like phenotypes in engineered cardiac tissues. First, increasing the percentage of hCFs to 15% resulted in tissues with increased ectopic activity and spontaneous excitation rate. Second, hCFs undergo myofibroblast activation in traditional two-dimensional culture, and this altered phenotype ablated the functional benefits of hCFs when incorporated into engineered cardiac tissues. Taken together, the results of this study demonstrate that human cardiac fibroblast number and activation state modulate electromechanical function of hiPSC-cardiomyocytes and that a low percentage of quiescent hCFs are a valuable cell source to advance a healthy electromechanical response of engineered cardiac tissue for regenerative medicine applications.
Collapse
|
32
|
Basalova N, Sagaradze G, Arbatskiy M, Evtushenko E, Kulebyakin K, Grigorieva O, Akopyan Z, Kalinina N, Efimenko A. Secretome of Mesenchymal Stromal Cells Prevents Myofibroblasts Differentiation by Transferring Fibrosis-Associated microRNAs within Extracellular Vesicles. Cells 2020; 9:cells9051272. [PMID: 32443855 PMCID: PMC7290371 DOI: 10.3390/cells9051272] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
Fibroblasts differentiation into myofibroblasts is a central event of tissue fibrosis. Multipotent mesenchymal stromal cells (MSCs) secretome can interfere with fibrosis development; despite precise underlying mechanisms remain unclear. In this study, we tested the hypothesis that MSC secretome can affect fibroblast’ differentiation into myofibroblasts by delivering regulatory RNAs, including microRNAs to these cells. Using the model of transforming growth factor-beta (TGFbeta)-induced fibroblast differentiation into myofibroblasts, we tested the activity of human MSC secretome components, specifically extracellular vesicles (MSC-EV). We showed that MSC-EV down-regulated secretion of extracellular matrix proteins by fibroblasts as well as suppressed their contractility resulting in prevention as well as reversion of fibroblasts differentiation to myofibroblasts. High-throughput sequencing of RNAs extracted from MSC-EV has revealed many fibrosis-associated microRNAs. Fibroblast treatment with MSC-EV led to direct transfer of microRNAs, which resulted in the elevation of most prominent fibrosis-associated microRNAs, including microRNA-21 and microRNA-29c. Using MSC-EV transfection by antagomirs to these microRNAs we demonstrated their involvement in the suppression of fibroblast differentiation in our model. Taken together, MSC secretome can suppress fibrosis by prevention of fibroblast differentiation into myofibroblasts as well as induce de-differentiation of the latter by direct transfer of specific microRNAs.
Collapse
Affiliation(s)
- Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia; (N.B.); (G.S.); (O.G.); (Z.A.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
| | - Georgy Sagaradze
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia; (N.B.); (G.S.); (O.G.); (Z.A.)
| | - Mikhail Arbatskiy
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
| | - Evgeniy Evtushenko
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia; (N.B.); (G.S.); (O.G.); (Z.A.)
| | - Zhanna Akopyan
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia; (N.B.); (G.S.); (O.G.); (Z.A.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
| | - Natalia Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia; (N.B.); (G.S.); (O.G.); (Z.A.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.); (K.K.); (N.K.)
- Correspondence:
| |
Collapse
|
33
|
Chamberlin T, Thompson V, Hillers-Ziemer LE, Walton BN, Arendt LM. Obesity reduces mammary epithelial cell TGFβ1 activity through macrophage-mediated extracellular matrix remodeling. FASEB J 2020; 34:8611-8624. [PMID: 32359100 PMCID: PMC7317547 DOI: 10.1096/fj.202000228rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Obesity is a risk factor for breast cancer in postmenopausal and high‐risk premenopausal women. Changes within the obese breast microenvironment may increase breast cancer risk. Transforming growth factor beta‐1 (TGFβ1) is a major regulator of mammary epithelial stem/progenitor cells, and its activity is dysregulated under conditions of obesity. Using a high‐fat diet model of obesity in mice and breast tissue from women, we observed that TGFβ1 activity is reduced in breast epithelial cells in obesity. Breast ducts and lobules demonstrated increased decorin in the extracellular matrix (ECM) surrounding epithelial cells, and we observed that decorin and latent TGFβ1 complexed together. Under conditions of obesity, macrophages expressed higher levels of decorin and were significantly increased in number surrounding breast epithelial cells. To investigate the relationship between macrophages and decorin expression, we treated obese mice with either IgG control or anti‐F4/80 antibodies to deplete macrophages. Mice treated with anti‐F4/80 antibodies demonstrated reduced decorin surrounding mammary ducts and enhanced TGFβ1 activity within mammary epithelial cells. Given the role of TGFβ1 as a tumor suppressor, reduced epithelial TGFβ1 activity and enhanced TGFβ1 within the ECM of obese mammary tissue may enhance breast cancer risk.
Collapse
Affiliation(s)
- Tamara Chamberlin
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Victoria Thompson
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Brenna N Walton
- Program in Molecular and Environmental Toxicology, University of Wisconsin-Madison, Madison, WI, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Program in Molecular and Environmental Toxicology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
34
|
Gremlich S, Roth-Kleiner M, Equey L, Fytianos K, Schittny JC, Cremona TP. Tenascin-C inactivation impacts lung structure and function beyond lung development. Sci Rep 2020; 10:5118. [PMID: 32198404 PMCID: PMC7083919 DOI: 10.1038/s41598-020-61919-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Tenascin-C (TNC) is an extracellular matrix protein expressed at high levels during lung organogenesis. Later, TNC is only transiently de novo expressed to orchestrate tissue repair in pathological situations. We previously showed that TNC inactivation affects lung development and thus evaluated here the implications on lung function in newborn/adult mice. Respiratory function parameters were measured in anesthetized and mechanically ventilated wild-type (WT) and TNC-deficient mice at 5 (P5) and 90 (P90) days of age under basal conditions, as well as following high tidal volume (HTV) ventilation. At P5, TNC-deficient mice showed an increased static compliance (Cst) and inspiratory capacity (IC) relative to WT at baseline and throughout HTV. At P90, however, Cst and IC were only elevated at baseline. Control non-ventilated newborn and adult TNC-deficient mice showed similar lung morphology, but less alpha smooth muscle actin (α-SMA) around small airways. SMA + cells were decreased by 50% in adult TNC-deficient lungs and collagen layer thickened around small airways. Increased surfactant protein C (SP-C) and altered TGFβ and TLR4 signaling pathways were also detected. Thus, TNC inactivation-related defects during organogenesis led to persisting functional impairment in adulthood. This might be of interest in the context of pulmonary diseases with thickened airway smooth muscle layer or ventilation heterogeneity, like asthma and COPD.
Collapse
Affiliation(s)
- Sandrine Gremlich
- Clinic of Neonatology, Department woman-mother-child, University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Matthias Roth-Kleiner
- Clinic of Neonatology, Department woman-mother-child, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lucile Equey
- Clinic of Neonatology, Department woman-mother-child, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kleanthis Fytianos
- Department of Bio-medical Research, University of Bern, Bern, Switzerland.,Division of Pulmonary Medicine, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
35
|
Kafarnik C, McClellan A, Dziasko M, Daniels JT, Guest DJ. Canine Corneal Stromal Cells Have Multipotent Mesenchymal Stromal Cell Properties In Vitro. Stem Cells Dev 2020; 29:425-439. [PMID: 31973649 DOI: 10.1089/scd.2019.0163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The objective of this study was to determine whether corneal stromal cells (CSCs) from the limbal and central corneal stroma in dogs have multipotent mesenchymal stem/stromal cell (MSC) properties, and whether this cell population can be differentiated into keratocyte-like cells (KDCs). Normal, donated, mesocephalic dog corneas were used to isolate CSC in vitro. Immunohistochemistry demonstrated a distinct population of CD90 expressing cells in the anterior stroma throughout the limbal and central cornea. CSC could be cultured from both the limbal and central cornea and the culture kinetics showed a progenitor cell profile. The CSC expressed stem cell markers CD90, CD73, CD105, N-cadherin, and Pax6, while CD34 was negative. Limbal and central CSC differentiated into osteoblasts, chondrocytes, and adipocytes confirming their multipotency. Coculturing allogeneic peripheral blood mononuclear cells (PBMCs) with limbal CSCs did not affect baseline PBMC proliferation indicating a degree of innate immune privilege. Limbal CSC could be differentiated into KDCs that expressed Keratocan, Lumican, and ALDH1A3 and downregulated Pax6 and N-cadherin. In conclusion, canine CSCs have multipotent MSC properties similarly described in humans and could serve as a source of cells for cell therapy and studying corneal diseases.
Collapse
Affiliation(s)
- Christiane Kafarnik
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom.,Rescue, Repair and Regeneration Theme, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Alyce McClellan
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| | - Marc Dziasko
- Rescue, Repair and Regeneration Theme, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Julie T Daniels
- Rescue, Repair and Regeneration Theme, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Deborah J Guest
- Centre for Preventive Medicine, Animal Health Trust, Newmarket, United Kingdom
| |
Collapse
|
36
|
Hu C, La H, Wei X, Zhou Y, Ou Q, Chen Z, Zhu X, Xu JY, Jin C, Gao F, Wang J, Zhang J, Zhang J, Lu L, Xu GT, Tian H. Transplantation Site Affects the Outcomes of Adipose-Derived Stem Cell-Based Therapy for Retinal Degeneration. Stem Cells Int 2020; 2020:9625798. [PMID: 32377204 PMCID: PMC7199575 DOI: 10.1155/2020/9625798] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/22/2019] [Accepted: 12/03/2019] [Indexed: 01/20/2023] Open
Abstract
Adipose-derived stem cells (ASCs) have shown a strong protective effect on retinal degenerative diseases (RDD) after being transplanted into the subretinal space in an animal model. Recently, several clinical trials have been conducted to treat RDD with intravitreal transplantation of stem cells, including ASCs. However, the outcomes of the clinical trials were not satisfactory. To investigate if the transplantation site alters the outcome of stem cell-based therapy for RDD, we isolated rat ASCs (rASCs) and labeled them with green fluorescent protein. Autologous rASCs were grafted into the vitreous chamber or subretinal space in a rat RDD model induced by sodium iodate (SI). The electric response was recorded by ERG. The anatomic structure of the retina was observed in cryosections of rat eyes at posttransplantation weeks 1, 2, and 4. Neural retina apoptosis and epiretinal membrane- (ERM-) like structure formation were investigated by immunostaining. The intravitreal transplantation of rASCs resulted in an extinguished electric response, although the rosette formation and apoptosis of neural retina were reduced. However, the rASCs that grafted in the subretinal space protected the retina from the damage caused by SI, including a partial recovering of the electric response and a reduction in rosette formation. Intravitreally grafted rASCs formed a membrane, resulting in retina folding at the injection site. Müller cells, retinal pigment epithelial cells, and microglial cells migrated from the retina to the rASC-formed membrane and subsequently formed an ERM-like structure. Furthermore, vitreous fluid promoted rASC migration, and rASC-conditioned medium enhanced Müller cell migration as indicated by in vitro studies. These data suggested that the vitreous chamber is not a good transplantation site for ASC-based therapy for RDD and that a deliberate decision should be made before transplantation of stem cells into the vitreous chamber to treat RDD in clinical trials.
Collapse
Affiliation(s)
- Chengyu Hu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Huanzhi La
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Xuancheng Wei
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Yue Zhou
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Zhiyang Chen
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Xiaoman Zhu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Caixia Jin
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Juan Wang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jingfa Zhang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| | - Jieping Zhang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, TUSM, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
- Department of Physiology and Pharmacology, TUSM, Shanghai, China
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology of Shanghai Tenth People's Hospital, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Laboratory of Clinical Visual Science, Department of Regenerative Medicine, Stem Cell Research Center, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Bachmann S, Jennewein M, Bubel M, Guthörl S, Pohlemann T, Oberringer M. Interacting adipose-derived stem cells and microvascular endothelial cells provide a beneficial milieu for soft tissue healing. Mol Biol Rep 2019; 47:111-122. [PMID: 31583562 DOI: 10.1007/s11033-019-05112-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
There is growing evidence suggesting that healing of chronic soft tissue wounds profits from the presence of adipose-derived stem cells (ADSC). Among the large spectrum of mechanisms by which ADSC might act, especially the interaction with the microvascular endothelial cell, a main player during angiogenesis, is of special interest. In the present 2D model on the basis of endothelial cell ADSC co-cultures, we focused on the identification of characteristics of both cell types in response to a typical condition in acute and chronic wounds: hypoxia. Parameters like proliferation capacity, migration, myofibroblastoid differentiation of ADSC and the quantification of important paracrine factors related to angiogenesis and inflammation were used to correlate our experimental model with the in vivo situation of soft tissue healing. ADSC were not negatively affected by hypoxia in terms of proliferation, referring to their excellent hypoxia tolerance. Myofibroblastoid differentiation among ADSC was enhanced by hypoxia in mono- but not in co-culture. Furthermore, co-cultures were able to migrate under hypoxia. These effects might be caused to some extent by the distinct milieu created by interacting ADSC and endothelial cells, which was characterized by modulated levels of interleukin-6, interleukin-8, monocyte chemoattractant protein-1 and vascular endothelial growth factor. The identification of these cell characteristics in the present 2D in vitro model provide new insights into the process of human soft tissue healing, and underpin a beneficial role of ADSC by regulating inflammation and angiogenesis.
Collapse
Affiliation(s)
- Sophie Bachmann
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany
| | - Martina Jennewein
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany
| | - Monika Bubel
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany
| | - Silke Guthörl
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany
| | - Martin Oberringer
- Department of Trauma-, Hand- and Reconstructive Surgery, Saarland University, Kirrberger Straße, Bldng. 57, 66421, Homburg, Germany.
| |
Collapse
|
38
|
Wu H, Tang WH, Zhao LM, Liu DF, Yang YZ, Zhang HT, Zhang Z, Hong K, Lin HC, Jiang H. Nanotechnology-assisted adipose-derived stem cell (ADSC) therapy for erectile dysfunction of cavernous nerve injury: In vivo cell tracking, optimized injection dosage, and functional evaluation. Asian J Androl 2019; 20:442-447. [PMID: 30004040 PMCID: PMC6116694 DOI: 10.4103/aja.aja_48_18] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cell therapy is a potentially promising option for erectile dysfunction; however, its risk of tumorigenicity is a clinical hurdle and the risk is positively related to the number of injected cells. Our previous study showed that nanotechnology improved adipose-derived stem cell (ADSC) therapy for erectile dysfunction of cavernous nerve injury (CNI) by attracting cells in the corpus cavernosum. These results indicated the possibility of using a reduced dosage of ADSCs for intracavernous injection. In this exploratory study, we used lower dosage (2 × 105 cells) of ADSCs for intracavernous injection (ICI) and the nanotechnology approach. Intracavernous pressure and mean arterial pressure were measured at day 28 to assess erectile function. The low-dose ADSC therapy group showed favorable treatment effects, and nanotechnology further improved these effects. In vivo imaging of ICI cells revealed that the fluorescein signals of NanoShuttle-bound ADSCs (NanoADSCs) were much stronger than those of ADSCs at days 0, 1, and 3. Both immunofluorescence and Western blot analysis showed a significant increase in smooth muscle, endothelium, and nerve tissue in the ADSC group compared to that in the CNI group; further improvement was achieved with assisted nanotechnology. These findings demonstrate that nanotechnology can be used to further improve the effect of small dosage of ADSCs to improve erectile function. Abundant NanoADSCs remain in the corpus cavernosum in vivo for at least 3 days. The mechanism of erectile function improvement may be related to the regeneration of the smooth muscle, endothelium, and nerve tissues.
Collapse
Affiliation(s)
- Han Wu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Wen-Hao Tang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China.,Department of Human Sperm Bank, Peking University Third Hospital, Beijing 100191, China
| | - Lian-Ming Zhao
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - De-Feng Liu
- Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Yu-Zhuo Yang
- Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China.,Department of Human Sperm Bank, Peking University Third Hospital, Beijing 100191, China
| | - Hai-Tao Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Kai Hong
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Hao-Cheng Lin
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing 100191, China.,Department of Andrology, Peking University Third Hospital, Beijing 100191, China.,Department of Human Sperm Bank, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
39
|
Zarei F, Abbaszadeh A. Application of Cell Therapy for Anti-Aging Facial Skin. Curr Stem Cell Res Ther 2019; 14:244-248. [PMID: 30421684 DOI: 10.2174/1574888x13666181113113415] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 08/30/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
The human skin undergoes the complex process of aging which is prompted by the interplay of intrinsic mechanisms and extrinsic influences. Aging is unavoidable but can be somewhat delayed. Numerous approaches have been developed to slow down facial skin aging process as it is of interest to stake holders in the beauty and fashion world as well as to plastic surgeons. Adipose-derived stem cell [ADSC] and mesenchymal stem cell [MSC] as potential anti-aging agents to some extent have provided a promising and effective alternative in managing skin and facial skin aging. Furthermore, bone marrow-derived mesenchymal stem cells [BMMSC] have exhibited similar ability to rejuvenate aged skin. This review is aimed at giving a comprehensive account of the application of stem cells especially ADSCs and MSCs to reduce or slow down the rate of facial skin aging process.
Collapse
Affiliation(s)
- Farshad Zarei
- Faculty of Medicine, Department of Surgery, Lorestan University of Medical Science, Khoramabad, Iran
| | - Abolfazl Abbaszadeh
- Faculty of Medicine, Department of Surgery, Lorestan University of Medical Science, Khoramabad, Iran
| |
Collapse
|
40
|
Adipose-Derived Tissue in the Treatment of Dermal Fibrosis: Antifibrotic Effects of Adipose-Derived Stem Cells. Ann Plast Surg 2019; 80:297-307. [PMID: 29309331 DOI: 10.1097/sap.0000000000001278] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment of hypertrophic scars and other fibrotic skin conditions with autologous fat injections shows promising clinical results; however, the underlying mechanisms of its antifibrotic action have not been comprehensively studied. Adipose-derived stem cells, or stromal cell-derived factors, inherent components of the transplanted fat tissue, seem to be responsible for its therapeutic effects on difficult scars. The mechanisms by which this therapeutic effect takes place are diverse and are mostly mediated by paracrine signaling, which switches on various antifibrotic molecular pathways, modulates the activity of the central profibrotic transforming growth factor β/Smad pathway, and normalizes functioning of fibroblasts and keratinocytes in the recipient site. Direct cell-to-cell communications and differentiation of cell types may also play a positive role in scar treatment, even though they have not been extensively studied in this context. A more thorough understanding of the fat tissue antifibrotic mechanisms of action will turn this treatment from an anecdotal remedy to a more controlled, timely administered technology.
Collapse
|
41
|
Chen H, Lui YS, Tan ZW, Lee JYH, Tan NS, Tan LP. Migration and Phenotype Control of Human Dermal Fibroblasts by Electrospun Fibrous Substrates. Adv Healthc Mater 2019; 8:e1801378. [PMID: 30901162 DOI: 10.1002/adhm.201801378] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/26/2019] [Indexed: 12/26/2022]
Abstract
Electrospun fibrous matrices, mimicking extracellular matrix (ECM) hierarchical structures, are potential scaffolds for wound healing. To design functional scaffolds, it is important to explore the interactions between scaffold topographic features and cellular responses, especially directional migration and phenotypic changes, which are critical functional aspects during wound healing. Here, accelerated and persistent migration of human dermal fibroblasts (HDFs) is observed on fibers with aligned orientation. Furthermore, aligned fibers can induce fibroblast-to-myofibroblast differentiation of HDFs. During wound healing, the presence of myofibroblasts advances wound repair by rendering contractile force and ECM deposition within the early and middle courses, but its continuous persistence in the later event may not be desired due to the contribution in pathological scarring. To tune the balance, it is noted in this work that the introduction of matricellular protein angiopoietin-like 4 (ANGPTL4) is capable of reversing the phenotypic alteration induced by aligned fibers, in a time-dependent manner. These results indicate fibrous matrices with oriented configuration are functional in mediating directional cell migration and phenotypic change. The discoveries further suggest that tissue-engineered fibrous grafts with precise alignment modulation and ANGPTL4 releasing properties may thus be promising to promote wound repair with minimizing scar formation.
Collapse
Affiliation(s)
- Huizhi Chen
- School of Materials Science and EngineeringNanyang Technological University 50 Nanyang Avenue Singapore 639798 Singapore
- Interdisciplinary Graduate SchoolNanyang Technological University 50 Nanyang Avenue Singapore 639798 Singapore
| | - Yuan Siang Lui
- School of Materials Science and EngineeringNanyang Technological University 50 Nanyang Avenue Singapore 639798 Singapore
| | - Zhen Wei Tan
- School of Biological SciencesNanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Justin Yin Hao Lee
- School of Biological SciencesNanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Nguan Soon Tan
- School of Biological SciencesNanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
- Lee Kong Chian School of MedicineNanyang Technological University 59 Nanyang Drive Singapore 636921 Singapore
| | - Lay Poh Tan
- School of Materials Science and EngineeringNanyang Technological University 50 Nanyang Avenue Singapore 639798 Singapore
| |
Collapse
|
42
|
Impact of HIV/simian immunodeficiency virus infection and viral proteins on adipose tissue fibrosis and adipogenesis. AIDS 2019; 33:953-964. [PMID: 30946149 DOI: 10.1097/qad.0000000000002168] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE HIV-infected patients receiving antiretroviral treatment (ART) often present adipose tissue accumulation and/or redistribution. adipose tissue has been shown to be an HIV/SIV reservoir and viral proteins as Tat or Nef can be released by infected immune cells and exert a bystander effect on adipocytes or precursors. Our aim was to demonstrate that SIV/HIV infection per se could alter adipose tissue structure and/or function. DESIGN Morphological and functional alterations of subcutaneous (SCAT) and visceral adipose tissue (VAT) were studied in SIV-infected macaques and HIV-infected ART-controlled patients. To analyze the effect of Tat or Nef, we used human adipose stem cells (ASCs) issued from healthy donors, and analyzed adipogenesis and extracellular matrix component production using two dimensional (2D) and three-dimensional (3D) culture models. METHODS Adipocyte size and index of fibrosis were determined on Sirius red-stained adipose tissue samples. Proliferating and adipocyte 2D-differentiating or 3D-differentiating ASCs were treated chronically with Tat or Nef. mRNA, protein expression and secretion were examined by RT-PCR, western-blot and ELISA. RESULTS SCAT and VAT from SIV-infected macaques displayed small adipocytes, decreased adipogenesis and severe fibrosis with collagen deposition. SCAT and VAT from HIV-infected ART-controlled patients presented similar alterations. In vitro, Tat and/or Nef induced a profibrotic phenotype in undifferentiated ASCs and altered adipogenesis and collagen production in adipocyte-differentiating ASCs. CONCLUSION We demonstrate here a specific role for HIV/SIV infection per se on adipose tissue fibrosis and adipogenesis, probably through the release of viral proteins, which could be involved in adipose tissue dysfunction contributing to cardiometabolic alterations of HIV-infected individuals.
Collapse
|
43
|
Lamaro-Cardoso A, Bachion MM, Morais JM, Fantinati MS, Milhomem AC, Almeida VL, Vinaud MC, Lino-Júnior RS. Photobiomodulation associated to cellular therapy improve wound healing of experimental full thickness burn wounds in rats. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 194:174-182. [PMID: 30999165 DOI: 10.1016/j.jphotobiol.2019.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 01/05/2023]
Abstract
Adipose derived stromal vascular fraction (SVF) is a method of cell therapy potentially applicable for treatment of full thickness burns. Here we investigated if the association of photobiomodulation (PBM) with SVF therapy could improve wound healing in experimentally induced full thickness burn wounds in rats compared to the topical agent 2% silver sulfadiazine in a dose-dependent manner. Sixty-six male Wistar rats were divided in 4 groups containing 5 animals each which received the following treatments: 2% sulfadiazine (SD), SVF, SVF plus PBM at 30 mW (SVFL30), and SVF plus PBM at 100 mW (SVFL100). Two donor animals were used for each experimental series with 3, 7 and 30 days. Digital photography, microscopic analysis with Hematoxilin and Eosin (H&E), quantification of collagen type I by picrosirius red staining analysis and wound contraction evaluation were performed in order to quantify the results. At day 3 SVF alone or combined with PBM promoted increased early inflammatory response compared to SD. At day 7 SVFL30 and SVFL100 enhanced inflammatory cells infiltration, angiogenesis and fibroblast content compared to SVF and SD groups. At day 30 collagen concentration and wound contraction were higher in SVFL30 when compared to the other groups. In conclusion PBM promotes a synergistic outcome with SVF therapy with a dose dependent effect potentializing wound healing of experimental full thickness burns in rats through amplification of early inflammatory response, enhanced angiogenesis, fibroblast content, accentuated wound contraction and collagen concentration.
Collapse
Affiliation(s)
| | - Maria M Bachion
- Faculty of Nursing, Federal University of Goiás, Goiânia, Brazil
| | - Júlia M Morais
- Faculty of Medicine, Federal University of Goiás, Jataí, Brazil
| | | | - Anália C Milhomem
- Laboratory of Experimental Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Vera L Almeida
- Laboratory of Experimental Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Marina C Vinaud
- Laboratory of Experimental Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Ruy S Lino-Júnior
- Laboratory of Experimental Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil.
| |
Collapse
|
44
|
Abrogation of transforming growth factor-β-induced tissue fibrosis in mice with a global genetic deletion of Nox4. J Transl Med 2019; 99:470-482. [PMID: 30470772 PMCID: PMC6530913 DOI: 10.1038/s41374-018-0161-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/12/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Excessive connective tissue deposition in skin and various internal organs is characteristic of systemic sclerosis (SSc). The profibrotic growth factor TGF-β plays a crucial role in SSc pathogenesis. The expression of NADPH oxidase 4 (NOX4), a critical mediator of oxidative stress, is potently stimulated by TGF-β. Here, we evaluated the effect of NOX4 on the development of TGF-β-induced tissue fibrosis. C57BL6/J control mice and Nox4 knockout mice were implanted subcutaneously with osmotic pumps containing either saline or 2.5 µg TGF-β1. After 28 days, skin and lung samples were isolated for histopathologic analysis, measurement of hydroxyproline content and gene expression analysis. Histopathology of skin and lungs from normal C57BL6/J mice treated with TGF-β1 showed profound dermal fibrosis and peribronchial and diffuse interstitial lung fibrosis. In contrast, TGF-β-treated Nox4 knockout mice showed normal skin and lung histology. Hydroxyproline levels in TGF-β-treated C57BL6/J mice skin and lungs demonstrated significant increases, however, hydroxyproline content of TGF-β-treated Nox4 knockout mice tissues was not changed. Expression of various profibrotic and fibrosis-associated genes was upregulated in skin and lungs of TGF-β1-treated C57BL6/J mice but was not significantly changed in TGF-β1-treated Nox4 knockout mice. The induction of skin and lung tissue fibrosis by TGF-β1 parenteral administration in mice was abrogated by the genetic deletion of Nox4 confirming that NOX4 is an essential mediator of the profibrotic effects of TGF-β. These results suggest Nox4 inhibition as a potential therapeutic target for SSc and other fibroproliferative disorders.
Collapse
|
45
|
Macrin D, Alghadeer A, Zhao YT, Miklas JW, Hussein AM, Detraux D, Robitaille AM, Madan A, Moon RT, Wang Y, Devi A, Mathieu J, Ruohola-Baker H. Metabolism as an early predictor of DPSCs aging. Sci Rep 2019; 9:2195. [PMID: 30778087 PMCID: PMC6379364 DOI: 10.1038/s41598-018-37489-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023] Open
Abstract
Tissue resident adult stem cells are known to participate in tissue regeneration and repair that follows cell turnover, or injury. It has been well established that aging impedes the regeneration capabilities at the cellular level, but it is not clear if the different onset of stem cell aging between individuals can be predicted or prevented at an earlier stage. Here we studied the dental pulp stem cells (DPSCs), a population of adult stem cells that is known to participate in the repair of an injured tooth, and its properties can be affected by aging. The dental pulp from third molars of a diverse patient group were surgically extracted, generating cells that had a high percentage of mesenchymal stem cell markers CD29, CD44, CD146 and Stro1 and had the ability to differentiate into osteo/odontogenic and adipogenic lineages. Through RNA seq and qPCR analysis we identified homeobox protein, Barx1, as a marker for DPSCs. Furthermore, using high throughput transcriptomic and proteomic analysis we identified markers for DPSC populations with accelerated replicative senescence. In particular, we show that the transforming growth factor-beta (TGF-β) pathway and the cytoskeletal proteins are upregulated in rapid aging DPSCs, indicating a loss of stem cell characteristics and spontaneous initiation of terminal differentiation. Importantly, using metabolic flux analysis, we identified a metabolic signature for the rapid aging DPSCs, prior to manifestation of senescence phenotypes. This metabolic signature therefore can be used to predict the onset of replicative senescence. Hence, the present study identifies Barx1 as a DPSCs marker and dissects the first predictive metabolic signature for DPSCs aging.
Collapse
Affiliation(s)
- Dannie Macrin
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, 603203, India
| | - Ammar Alghadeer
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, 98109, USA.,Department of Biomedical Dental Sciences, Imam Abdulrahman bin Faisal University, College of Dentistry, Dammam, 31441, Saudi Arabia
| | - Yan Ting Zhao
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, 98109, USA
| | - Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Abdiasis M Hussein
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
| | - Damien Detraux
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
| | - Aaron M Robitaille
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Pharmacology, University of Washington, Seattle, WA, 98109, USA
| | - Anup Madan
- Covance Genomics Laboratory, Redmond, WA, 98052, USA
| | - Randall T Moon
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Pharmacology, University of Washington, Seattle, WA, 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Arikketh Devi
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai, 603203, India
| | - Julie Mathieu
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.,Department of Comparative Medicine, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA. .,Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA. .,Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, 98109, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
46
|
Liu X, Zheng L, Zhou Y, Chen Y, Chen P, Xiao W. BMSC Transplantation Aggravates Inflammation, Oxidative Stress, and Fibrosis and Impairs Skeletal Muscle Regeneration. Front Physiol 2019; 10:87. [PMID: 30814953 PMCID: PMC6382023 DOI: 10.3389/fphys.2019.00087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/24/2019] [Indexed: 12/26/2022] Open
Abstract
Skeletal muscle contusion is one of the most common muscle injuries in sports medicine and traumatology. Bone marrow mesenchymal stem cell (BMSC) transplantation has been proposed as a promising strategy to promote skeletal muscle regeneration. However, the roles and underlying mechanisms of BMSCs in the regulation of skeletal muscle regeneration are still not completely clear. Here, we investigated the role of BMSC transplantation after muscle contusion. BMSCs were immediately transplanted into gastrocnemius muscles (GMs) following direct contusion. Comprehensive morphological and genetic analyses were performed after BMSC transplantation. BMSC transplantation exacerbated muscle fibrosis and inflammation, as evidenced by increased leukocyte and macrophage infiltration, increased inflammatory cytokines and chemokines, and increased matrix metalloproteinases. BMSC transplantation also increased muscle oxidative stress. Overall, BMSC transplantation aggravated inflammation, oxidative stress and fibrosis and impaired skeletal muscle regeneration. These results, shed new light on the role of BMSCs in regenerative medicine and indicate that caution is needed in the application of BMSCs for muscle injury.
Collapse
Affiliation(s)
- Xiaoguang Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lifang Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yongzhan Zhou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yingjie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
47
|
Zhou ZQ, Chen Y, Chai M, Tao R, Lei YH, Jia YQ, Shu J, Ren J, Li G, Wei WX, Han YD, Han Y. Adipose extracellular matrix promotes skin wound healing by inducing the differentiation of adipose‑derived stem cells into fibroblasts. Int J Mol Med 2019; 43:890-900. [PMID: 30535488 PMCID: PMC6317660 DOI: 10.3892/ijmm.2018.4006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Fibroblasts are the major effector cells of skin wound healing. Adipose‑derived stem cells can differentiate into fibroblasts under certain conditions. In the present study, it was hypothesized that adipose‑derived stem cells (ADSCs) could be induced by the adipose extracellular matrix (ECM) to differentiate into fibroblasts in order to promote skin wound healing. First, flow cytometry was used to detect the ratio of fibroblasts and relative expression of the fibroblast markers cytokeratin 19 (CK19) and vimentin in ADSCs. Then, the effect of the adipose ECM during the differentiation of ADSCs into fibroblasts was investigated by detecting the total amount of collagen fibers and degree of fibrosis, and the proliferation and cell cycle of differentiated fibroblasts, using the MTT assay and flow cytometry analysis respectively. Finally, a mouse skin wound model was established and treated with PBS, ADSC suspension or ECM + ADSCs to compare wound healing rate and expression of collagen I and collagen III by immunohistochemistry. Following induction of ADSCs with the adipose ECM, more fibroblasts were found, expression of CK19 and vimentin increased, and a greater degree of fibrosis occurred, which revealed the positive effect of the adipose ECM on the differentiation of ADSCs into fibroblasts. In addition, the induced fibroblasts had enhanced proliferation activity, with more cells in the S phase and fewer in the G2/M phase. The in vivo experiment indicated that the ECM produced by the ADSCs had a faster wound healing rate and increased expression of collagen I and collagen III compared with mice injected with PBS or ADSCs alone, which verified that ADSCs induced by the adipose ECM had a positive effect on skin wound healing. The present study demonstrated that the adipose ECM in combination with ADSCs may be a novel therapeutic target for the repair of skin injury, due to the ability of the adipose ECM to induce the differentiation of ADSCs into fibroblasts and to facilitate the wound healing process.
Collapse
Affiliation(s)
- Zhi-Qiang Zhou
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi Chen
- Institute of Bioengineering, Academy of Military Medical Research, Academy of Military Science of Chinese PLA, Beijing 100071, P.R. China
| | - Mi Chai
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yong-Hong Lei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi-Qing Jia
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jun Shu
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Guo Li
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Wen-Xin Wei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yu-Di Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| |
Collapse
|
48
|
Jia YY, Zhou JY, Chang Y, An F, Li XW, Xu XY, Sun XL, Xiong CY, Wang JL. Effect of Optimized Concentrations of Basic Fibroblast Growth Factor and Epidermal Growth Factor on Proliferation of Fibroblasts and Expression of Collagen: Related to Pelvic Floor Tissue Regeneration. Chin Med J (Engl) 2018; 131:2089-2096. [PMID: 30127219 PMCID: PMC6111681 DOI: 10.4103/0366-6999.239301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Fibroblasts were the main seed cells in the studies of tissue engineering of the pelvic floor ligament. Basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) were widely studied but at various concentrations. This study aimed to optimize the concentrations of combined bFGF and EGF by evaluating their effects on proliferation and collagen secretion of fibroblasts. Methods: Fibroblasts were differentiated from rat adipose mesenchymal stem cells (ADSCs). Flow cytometry and immunohistochemistry were used for cell identification. The growth factors were applied at concentrations of 0, 1, 10, and 100 ng/ml as three groups: (1) bFGF alone, (2) EGF alone, and (3) bFGF mixed with EGF. Cell proliferation was evaluated by Cell Counting Kit-8 assays. Expression of Type I and III collagen (Col-I and Col-III) mRNAs was evaluated by real-time quantitative reverse transcription-polymerase chain reaction. Statistical analysis was performed with SPSS software and GraphPad Prism using one-way analysis of variance and multiple t-test. Results: ADSCs were successfully isolated from rat adipose tissue as identified by expression of typical surface markers CD29, CD44, CD90, and CD45 in flow cytometry. Fibroblasts induced from ADSC, compared with ADSCs, were with higher mRNA expression levels of Col I and Col III (F = 1.29, P = 0.0390). bFGF, EGF, and the mixture of bFGF with EGF can enhanced fibroblasts proliferation, and the concentration of 10 ng/ml of the mixture of bFGF with EGF displayed most effectively (all P < 0.05). The expression levels of Col-I and Col-III mRNAs in fibroblasts displayed significant increases in the 10 ng/ml bFGF combined with EGF group (all P < 0.05). Conclusions: The optimal concentration of both bFGF and EGF to promote cell proliferation and collagen expression in fibroblasts was 10 ng/ml at which fibroblasts grew faster and secreted more Type I and III collagens into the extracellular matrix, which might contribute to the stability of the pelvic floor microenvironment.
Collapse
Affiliation(s)
- Yuan-Yuan Jia
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Jing-Yi Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Yue Chang
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Fang An
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Xiao-Wei Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Xiao-Yue Xu
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Xiu-Li Sun
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| | - Chun-Yang Xiong
- Department of Mechanics and Bioengineering, College of Engineering, Peking University, Beijing 100871, China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital; Beijing Key Laboratory of Female Pelvic Floor Disorders, Beijing 100044, China
| |
Collapse
|
49
|
Impact of Acellular Dermal Matrix on Postsurgical Wound Fluid Biomarkers in Prosthetic Breast Reconstruction. Ann Plast Surg 2018; 81:S89-S96. [PMID: 29851721 DOI: 10.1097/sap.0000000000001495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite the widespread practice of using biologic scaffolds for soft tissue reinforcement over prosthetic implants, the impact of acellular dermal matrix (ADM) on surgical wound fluid biomarkers over the initial postoperative period after prosthetic breast reconstruction remains poorly understood. METHODS Patients undergoing prosthetic breast reconstruction surgery where ADM was likely to be used were consented to have fluid samples collected from surgical drains after surgery. Sample collections occurred at an "Early" time point at 24 to 48 hours after surgery and then a "Late" time point approximately 1 to 2 weeks after surgery. All procedures were performed by a single surgeon. Acellular dermal matrix was placed when prosthetic coverage with autologous tissue could not be achieved. Laboratory analyses were performed in blinded fashion without the knowledge of whether the samples came from the ADM "Present" or "Not Present" group. RESULTS Twenty-one patients were in the ADM Present group and 18 patients were in the Not Present group. Both groups showed similar demographics based on age and body mass index. Analyses for cell concentration, protein concentration, extracellular matrix protein levels, cell proliferation activity, and matrix metalloproteinase activity showed no significant differences between wound fluid samples from the 2 groups. CONCLUSIONS The presence of ADM does not appear to significantly impact wound biomarkers in prosthetic breast reconstruction. The current study provides useful data regarding the impact of ADM on surgical wound fluid during the initial postoperative period, laying important groundwork for more extensive future studies on the impact of biologic scaffolds on wound biology.
Collapse
|
50
|
Liu X, Long X, Liu W, Zhao Y, Hayashi T, Yamato M, Mizuno K, Fujisaki H, Hattori S, Tashiro SI, Ogura T, Atsuzawa Y, Ikejima T. Type I collagen induces mesenchymal cell differentiation into myofibroblasts through YAP-induced TGF-β1 activation. Biochimie 2018; 150:110-130. [PMID: 29777737 DOI: 10.1016/j.biochi.2018.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022]
Abstract
In organ fibrosis, mechanical stress and transforming growth factor beta-1 (TGF-β1) promote differentiation into myofibroblast from mesenchymal cells, leading to extracellular matrix (ECM) remodeling or active synthesis, deposition or degradation of ECM components. A major component of ECM, type I collagen (col I) triple helical molecules assemble into fibrils or are denatured to gelatin without triple-helicity in remodeling. However, whether changes of ECM components in remodeling have influence on mesenchymal cell differentiation remains elusive. This study adopted three states of collagen I existing in ECM remodeling: molecular collagen, fibrillar collagen and gelatin to see what are characteristics in the effects on two cell lines of mesenchymal origin, murine 3T3-L1 embryonic fibroblast and murine C2C12 myoblasts. The results showed that all three forms of collagen I were capable of inducing these two cells to differentiate into myofibroblasts characterized by increased expression of alpha-smooth muscle actin (α-SMA) mRNA. The expression of α-SMA is positively regulated by TGF-β1. Nuclear translocation of Yes-associated protein (YAP) is involved in this process. Focal adhesion kinase (FAK) is activated in the cells cultured on molecular collagen-coated plates, contributing to YAP activation. On the other hand, in the cells cultured on fibrillar collagen gel or gelatin-coated plates, oxidative stress but not FAK induce YAP activation. In conclusion, the three physicochemically distinct forms of col I induce the differentiation of mesenchymal cells into myofibroblasts through different pathways.
Collapse
Affiliation(s)
- Xiaoling Liu
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xinyu Long
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Weiwei Liu
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yeli Zhao
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Toshihiko Hayashi
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, 162-8666, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Shin-Ichi Tashiro
- Department of Medical Education and Primary Care, Kyoto Prefectural University of Medicine, Kyoto, 603-8072, Japan
| | - Takaaki Ogura
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Yuji Atsuzawa
- Nippi Research Institute of Biomatrix, Ibaraki, 302-0017, Japan
| | - Takashi Ikejima
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|