1
|
Reed EB, Sitikov A, Shin KWD, Hamanaka RB, Cetin-Atalay R, Mutlu GM, Mongin AA, Dulin NO. Gα12 and Gα13 proteins are required for transforming growth factor-β-induced myofibroblast differentiation. Biochem J 2024; 481:1937-1948. [PMID: 39621448 DOI: 10.1042/bcj20240317] [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: 06/26/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
Myofibroblast differentiation, characterized by accumulation of cytoskeletal and extracellular matrix proteins by fibroblasts, is a key process in wound healing and pathogenesis of tissue fibrosis. Transforming growth factor-β (TGF-β) is the most powerful known driver of myofibroblast differentiation. TGF-β signals through transmembrane receptor serine/threonine kinases that phosphorylate Smad transcription factors (Smad2/3) leading to activation of transcription of target genes. Heterotrimeric G proteins mediate distinct signaling from seven-transmembrane G protein coupled receptors, which are not known to be linked to Smad activation. We tested whether G protein signaling plays any role in TGF-β-induced myofibroblast differentiation, using primary cultured human lung fibroblasts. Activation of Gαs by cholera toxin blocked TGF-β-induced myofibroblast differentiation without affecting Smad2/3 phosphorylation. Neither inhibition of Gαi by pertussis toxin nor siRNA-mediated combined knockdown of Gαq and Gα11 had a significant effect on TGF-β-induced myofibroblast differentiation. In contrast, combined knockdown of Gα12 and Gα13 significantly inhibited TGF-β-stimulated expression of myofibroblast marker proteins (collagen-1, fibronectin, smooth-muscle α-actin), with siGα12 being significantly more potent than siGα13. Mechanistically, combined knockdown of Gα12 and Gα13 resulted in substantially reduced phosphorylation of Smad2 and Smad3 in response to TGF-β, which was accompanied by a significant decrease in the expression of TGF-β receptors (TGFBR1, TGFBR2) and of Smad3. Thus, our study uncovers a novel role of Gα12/13 proteins in the control of TGF-β signaling and myofibroblast differentiation.
Collapse
Affiliation(s)
- Eleanor B Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Kun Woo D Shin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Rengül Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, U.S.A
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, U.S.A
| |
Collapse
|
2
|
Li L, Ma M, Zuo G, Xiao J, Chen J, He X, Song Z. Effect of manganese amino acid complexes on growth performance, meat quality, breast muscle and bone development in broilers. Br Poult Sci 2024; 65:582-594. [PMID: 38994893 DOI: 10.1080/00071668.2024.2346640] [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: 11/26/2023] [Accepted: 02/14/2024] [Indexed: 07/13/2024]
Abstract
1. This study was conducted to investigate the effects of dietary supplementation of manganese (Mn) amino acid complexes on growth performance, Mn deposition, meat quality, breast muscle and bone development of broilers.2. A total of 504, one-day-old male Arbor Acres broilers were randomly divided into seven treatments; control diet (CON; basal diet, no extra Mn addition), manganese diet (MnN as Numine®-Mn; CON + 40, 80, 120 or 160 mg Mn/kg), manganese-S group (MnS; CON + 120 mg Mn/kg as MnSO4·H2O), manganese-A diet (MnA as Mn from hydrolysed feather meal; CON + 40 mg Mn/kg as MnA).3. There were no significant differences for average daily gain (ADG) or feed intake (ADFI) among diets during the feed phases (p > 0.05). The FCR in the starter and over the whole period were quadratically affected by dietary MnN dosage and gave the lowest FCR at 80 mg/kg (p < 0.05). The Mn content of thigh muscle, jejunum, heart, pancreas, liver and tibia increased linearly with MnN addition (p < 0.05).4. For meat quality, MnN significantly increased colour (a*), pH45 min and pH24 h, reduced shear force, drip loss and pressure loss of breast muscle (p < 0.05).5. Moreover, MnN significantly upregulated MYOD expression at d 21 and SOD expression at d 42, decreased MuRF1 and Atrogin-1 mRNA level at d 42 in breast muscle. Transcriptome analysis revealed that the regulating effect of MnN on muscle development significantly enriched signalling pathways such as adhesion, ECM-receptor, MAPK, mTOR and AMPK. Furthermore, dietary MnN significantly affected tibia length and growth plate development (p < 0.05) and promoted growth plate chondrocytes by increasing SOX-9, Runx-2, Mef2c, TGF-β, Ihh, Bcl-2 and Beclin1 and decreasing Bax and Caspase-3 (p < 0.05) expression which affect longitudinal tibial development.6. In conclusion, Mn amino acid complexes could improve growth performance, tissue Mn deposition, breast muscle development, meat quality and bone development.
Collapse
Affiliation(s)
- L Li
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, China
- R&D Department, Hunan Engineering Research Center of Poultry Production Safety, Hunan, China
| | - M Ma
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, China
- R&D Department, Hunan Engineering Research Center of Poultry Production Safety, Hunan, China
| | - G Zuo
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, China
- R&D Department, Hunan Engineering Research Center of Poultry Production Safety, Hunan, China
- Technical R&D Department, Beijing Deyuanshun Biotechnology Co, Ltd, Beijing, China
| | - J Xiao
- Technical R&D Department, Hunan Xiang Jia Husbandry Limited by Share Ltd, Changde, Hunan, China
| | - J Chen
- Technical R&D Department, Hunan Xiang Jia Husbandry Limited by Share Ltd, Changde, Hunan, China
| | - X He
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, China
- R&D Department, Hunan Engineering Research Center of Poultry Production Safety, Hunan, China
| | - Z Song
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, China
- R&D Department, Hunan Engineering Research Center of Poultry Production Safety, Hunan, China
| |
Collapse
|
3
|
Usman K, Fouadi M, Nwozor KO, Aminazadeh F, Nair P, Luo H, Sin DD, Osei ET, Hackett TL. Interleukin-1α inhibits transforming growth factor-β1 and β2-induced extracellular matrix production, remodeling and signaling in human lung fibroblasts: Master regulator in lung mucosal repair. Matrix Biol 2024; 132:47-58. [PMID: 39147560 DOI: 10.1016/j.matbio.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-β (TGF-β) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS Stimulation of lung fibroblasts with TGF-β1 and TGF-β2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-β1 and TGF-β2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-β1 or TGF-β2. Mechanistically, IL-1α administration led to the suppression of TGF-β1 and TGF-β2 signaling, through downregulation of mRNA and protein for TGF-β receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION IL-1α acts as a master regulator, modulating TGF-β1 and TGF-β2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-β signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-β1 or TGF-β2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.
Collapse
Affiliation(s)
- Kauna Usman
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| | - May Fouadi
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kingsley Okechukwu Nwozor
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Fatemeh Aminazadeh
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Parameswaran Nair
- Division of Respirology, St Joseph's Healthcare Hamilton & McMaster University, ON L8N 4A6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Emmanuel Twumasi Osei
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Biology, University of British Columbia, Okanagan, BC V1V 1V7, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
4
|
Kent RN, Jewett ME, Buck TP, Said M, Hold LA, Crawford EA, Killian ML, Abraham AC, Huang AH, Baker BM. Engineered Microenvironmental Cues from Fiber-Reinforced Hydrogel Composites Drive Tenogenesis and Aligned Collagen Deposition. Adv Healthc Mater 2024; 13:e2400529. [PMID: 38441411 PMCID: PMC11281874 DOI: 10.1002/adhm.202400529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Indexed: 03/25/2024]
Abstract
Effective tendon regeneration following injury is contingent on appropriate differentiation of recruited cells and deposition of mature, aligned, collagenous extracellular matrix that can withstand the extreme mechanical demands placed on the tissue. As such, myriad biomaterial approaches have been explored to provide biochemical and physical cues that encourage tenogenesis and template aligned matrix deposition in lieu of dysfunctional scar tissue formation. Fiber-reinforced hydrogels present an ideal biomaterial system toward this end given their transdermal injectability, tunable stiffness over a range amenable to tenogenic differentiation of progenitors, and capacity for modular inclusion of biochemical cues. Here, tunable and modular, fiber-reinforced, synthetic hydrogels are employed to elucidate salient microenvironmental determinants of tenogenesis and aligned collagen deposition by tendon progenitor cells. Transforming growth factor β3 drives a cell fate switch toward pro-regenerative or pro-fibrotic phenotypes, which can be biased toward the former by culture in softer microenvironments or inhibition of the RhoA/ROCK activity. Furthermore, studies demonstrate that topographical anisotropy in fiber-reinforced hydrogels critically mediates the alignment of de novo collagen fibrils, reflecting native tendon architecture. These findings inform the design of cell-free, injectable, synthetic hydrogels for tendon tissue regeneration and, likely, that of a range of load-bearing connective tissues.
Collapse
Affiliation(s)
- Robert N. Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Maggie E. Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Trevor P. Buck
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed Said
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - LeeAnn A. Hold
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Eileen A. Crawford
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Megan L. Killian
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Adam C. Abraham
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Alice H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10027
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
5
|
Cheng KC, Chong PCT, Hsieh CC, Lin YT, Ye CH, Khumsupan D, Lu JJ, Yu WC, Cheng KW, Yap KY, Kou WS, Cheng MT, Hsu CC, Sheen LY, Lin SP, Wei AC, Yu SH. Identification of anti-fibrotic and pro-apoptotic bioactive compounds from Ganoderma formosanum and their possible mechanisms in modulating TGF-β1-induced lung fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118008. [PMID: 38458343 DOI: 10.1016/j.jep.2024.118008] [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: 12/15/2023] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Compendium of Materia Medica and the Classic of Materia Medica, the two most prominent records of traditional Chinese medicine, documented the therapeutic benefits of Ganoderma sinense particularly in addressing pulmonary-related ailments. Ganoderma formosanum, an indigenous subspecies of G. sinense from Taiwan, has demonstrated the same therapeutic properties. AIM OF THE STUDY The aim of this study is to identify bioactive compounds and evaluate the potential of G. formosanum extracts as a novel treatment to alleviate pulmonary fibrosis (PF). Using an in-house drug screening platform, two-stage screening was performed to determine their anti-fibrotic efficacy. METHODS AND MATERIALS G. formosanum was fractionated into four partitions by solvents of different polarities. To determine their antifibrotic and pro-apoptotic properties, the fractions were analyzed using two TGF-β1-induced pulmonary fibrosis cell models (NIH-3T3) and human pulmonary fibroblast cell lines, immunoblot, qRT-PCR, and annexin V assays. Subsequently, transcriptomic analysis was conducted to validate the findings and explore possible molecular pathways. The identification of potential bioactive compounds was achieved through UHPLC-MS/MS analysis, while molecular interaction study was investigated by multiple ligands docking and molecular dynamic simulations. RESULTS The ethyl acetate fraction (EAF) extracted from G. formosanum demonstrated substantial anti-fibrotic and pro-apoptotic effects on TGF-β1-induced fibrotic models. Moreover, the EAF exhibited no discernible cytotoxicity. Untargeted UHPLC-MS/MS analysis identified potential bioactive compounds in EAF, including stearic acid, palmitic acid, and pentadecanoic acid. Multiple ligands docking and molecular dynamic simulations further confirmed that those bioactive compounds possess the ability to inhibit TGF-β receptor 1. CONCLUSION Potential bioactive compounds in G. formosanum were successfully extracted and identified in the EAF, whose anti-fibrotic and pro-apoptotic properties could potentially modulate pulmonary fibrosis. This finding not only highlights the EAF's potential as a promising therapeutic candidate to treat pulmonary fibrosis, but it also elucidates how Ganoderma confers pulmonary health benefits as described in the ancient texts.
Collapse
Affiliation(s)
- Kuan-Chen Cheng
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C; Institute of Food Science Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C; Department of Optometry, Asia University, No. 500, Lioufeng Rd., Wufeng, Taichung, Taiwan. R.O.C; Department of Medical Research, China Medical University Hospital, China Medical University, No. 91, Hsueh-Shih Rd., Taichung, Taiwan. R.O.C
| | - Patrick Chun Theng Chong
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Chen-Che Hsieh
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Yu-Te Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan. R.O.C
| | - Chih-Hung Ye
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Darin Khumsupan
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Jheng-Jhe Lu
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Wei-Chieh Yu
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Kai-Wen Cheng
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Kah Yi Yap
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Weng Si Kou
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Meng-Tsung Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, No.33, Linsen S. Rd., Taipei, 100025, Taiwan. R.O.C
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C; Leeuwenhoek Laboratories Co. Ltd., No. 71, Fanglan Rd, Taipei, 106038, Taiwan. R.O.C
| | - Lee-Yan Sheen
- Institute of Food Science Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C
| | - Shin-Ping Lin
- School of Food Safety, Taipei Medical University, No. 250, Wu-Hsing Street, Taipei, Taiwan. R.O.C
| | - An-Chi Wei
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan. R.O.C
| | - Shu-Han Yu
- Institute of Biotechnology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, Taiwan. R.O.C.
| |
Collapse
|
6
|
Reed EB, Sitikov A, Hamanaka RB, Cetin-Atalay R, Mutlu GM, Mongin AA, Dulin NO. Critical role of Gα12 and Gα13 proteins in TGF-β-induced myofibroblast differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596473. [PMID: 38854083 PMCID: PMC11160726 DOI: 10.1101/2024.05.29.596473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Myofibroblast differentiation, characterized by accumulation of cytoskeletal and extracellular matrix proteins by fibroblasts, is a key process in wound healing and pathogenesis of tissue fibrosis. Transforming growth factor-β (TGF-β) is the most powerful known driver of myofibroblast differentiation. TGF-β signals through transmembrane receptor serine/threonine kinases that phosphorylate Smad transcription factors (Smad2/3) leading to activation of transcription of target genes. Heterotrimeric G proteins mediate a distinct signaling from seven-transmembrane G protein coupled receptors, not commonly linked to Smad activation. We asked if G protein signaling plays any role in TGF-β-induced myofibroblast differentiation, using primary cultured human lung fibroblasts. Activation of Gαs by cholera toxin blocked TGF-β-induced myofibroblast differentiation without affecting Smad2/3 phosphorylation. Inhibition of Gαi by pertussis toxin, or siRNA-mediated combined knockdown of Gαq and Gα11 had no significant effect on TGF-β-induced myofibroblast differentiation. A combined knockdown of Gα12 and Gα13 resulted in a drastic inhibition of TGF-β-stimulated expression of myofibroblast marker proteins (collagen-1, fibronectin, smooth-muscle α-actin), with siGα12 being significantly more potent than siGα13. Mechanistically, a combined knockdown of Gα12 and Gα13 resulted in a substantially reduced phosphorylation of Smad2 and Smad3 in response to TGF-β, which was accompanied by a significant decrease in the expression of TGFβ receptors (TGFBR1, TGFBR2) and of Smad3 under siGα12/13 conditions. In conclusion, our study uncovers a novel role of Gα12/13 proteins in the control of TGF-β signaling and myofibroblast differentiation.
Collapse
Affiliation(s)
- Eleanor B. Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Robert B. Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Rengül Cetin-Atalay
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Alexander A. Mongin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY
| | - Nickolai O. Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
7
|
Kumari J, Paul O, Verdellen L, Berking B, Chen W, Gerrits L, Postma J, Wagener FADTG, Kouwer PHJ. Conductive Polyisocyanide Hydrogels Inhibit Fibrosis and Promote Myogenesis. ACS APPLIED BIO MATERIALS 2024; 7:3258-3270. [PMID: 38593039 PMCID: PMC11110048 DOI: 10.1021/acsabm.4c00210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Reliable in vitro models closely resembling native tissue are urgently needed for disease modeling and drug screening applications. Recently, conductive biomaterials have received increasing attention in the development of in vitro models as they permit exogenous electrical signals to guide cells toward a desired cellular response. Interestingly, they have demonstrated that they promote cellular proliferation and adhesion even without external electrical stimulation. This paper describes the development of a conductive, fully synthetic hydrogel based on hybrids of the peptide-modified polyisocyanide (PIC-RGD) and the relatively conductive poly(aniline-co-N-(4-sulfophenyl)aniline) (PASA) and its suitability as the in vitro matrix. We demonstrate that incorporating PASA enhances the PIC-RGD hydrogel's electroactive nature without significantly altering the fibrous architecture and nonlinear mechanics of the PIC-RGD network. The biocompatibility of our model was assessed through phenotyping cultured human foreskin fibroblasts (HFF) and murine C2C12 myoblasts. Immunofluorescence analysis revealed that PIC-PASA hydrogels inhibit the fibrotic behavior of HFFs while promoting myogenesis in C2C12 cells without electrical stimulation. The composite PIC-PASA hydrogel can actively change the cell fate of different cell types, providing an attractive tool to improve skin and muscle repair.
Collapse
Affiliation(s)
- Jyoti Kumari
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Department
of Dentistry—Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, The Netherlands
| | - Odile Paul
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Lisa Verdellen
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Bela Berking
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Wen Chen
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Lotte Gerrits
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Jelle Postma
- Department
of General Instrumentation, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Frank A. D. T. G. Wagener
- Department
of Dentistry—Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, The Netherlands
| | - Paul H. J. Kouwer
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| |
Collapse
|
8
|
Zhang X, Han P, Qiu J, Huang F, Luo Q, Cheng J, Shan K, Yang Y, Zhang C. Single-cell RNA sequencing reveals the complex cellular niche of pterygium. Ocul Surf 2024; 32:91-103. [PMID: 38290663 DOI: 10.1016/j.jtos.2024.01.013] [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: 10/12/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE Pterygium is a vision-threatening conjunctival fibrovascular degenerated disease with a high global prevalence up to 12 %, while no absolute pharmacotherapy has been applied in clinics. In virtue of single-cell RNA sequencing (scRNA-seq) technique, our study investigated underlying pathogeneses and potential therapeutic targets of pterygium from the cellular transcriptional level. METHODS A total of 45605 cells from pterygium of patients and conjunctiva of normal controls (NC) were conducted with scRNA-seq, and then analyzed via integrated analysis, pathway enrichment, pseudotime trajectory, and cell-cell communications. Besides, immunofluorescence and western blot were performed in vivo and in vitro to verify our findings. RESULTS In brief, 9 major cellular types were defined, according to canonical markers. Subsequently, we further determined the subgroups of each major cell lineages. Several newly identified cell sub-clusters could promote pterygium, including immuno-fibroblasts, epithelial mesenchymal transition (EMT)-epithelial cells, and activated vascular endothelial cells (activated-vEndo). Besides, we also probed the enrichment of immune cells in pterygium. Particularly, macrophages, recruited by ACKR1+activated-vEndo, might play an important role in the development of pterygium by promoting angiogenesis, immune suppression, and inflammation. CONCLUSION An intricate cellular niche was revealed in pterygium via scRNA-seq analysis and the interactions between macrophages and ACKR1+ activated-vEndo might be the key part in the development of pterygia.
Collapse
Affiliation(s)
- Xueling Zhang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Peizhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jini Qiu
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Feifei Huang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Qiting Luo
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Jingyi Cheng
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China
| | - Kun Shan
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| | - Yujing Yang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| | - Chaoran Zhang
- Department of Ophthalmology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China; Department of Ophthalmology, Shanghai Medical College, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
9
|
Leong E, Al-Bitar H, Marshall JS, Bezuhly M. Ketotifen directly modifies the fibrotic response of human skin fibroblasts. Sci Rep 2024; 14:7076. [PMID: 38528089 DOI: 10.1038/s41598-024-57776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
Fibrosis is a destructive, end-stage disease process. In the skin, it is associated with systemic sclerosis and scarring with considerable health burden. Ketotifen is a clinical antihistamine and mast cell stabilizer. Studies have demonstrated mast cell-dependent anti-fibrotic effects of ketotifen but direct effects on fibroblasts have not been determined. Human dermal fibroblasts were treated with pro-fibrotic transforming growth factor-β1 (TGFβ) followed by ketotifen or control treatments to determine direct effects on fibrotic fibroblasts. Ketotifen impaired TGFβ-induced α-smooth muscle actin gene and protein responses and decreased cytoskeletal- and contractility-associated gene responses associated with fibrosis. Ketotifen reduced Yes-associated protein phosphorylation, transcriptional coactivator with PDZ binding motif transcript and protein levels, and phosphorylation of protein kinase B. In a fibroblast-populated collagen gel contraction assay, ketotifen reduced the contractile activity of TGFβ-activated fibroblasts. In a murine model of bleomycin-induced skin fibrosis, collagen density and dermal thickness were significantly decreased in ketotifen-treated mice supporting in vitro findings. These results support a novel, direct anti-fibrotic activity of ketotifen, reducing pro-fibrotic phenotypic changes in fibroblasts and reducing collagen fibres in fibrotic mouse skin. Together, these findings suggest novel therapeutic potential and a novel mechanism of action for ketotifen in the context of fibrosis.
Collapse
Affiliation(s)
- Edwin Leong
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada
| | - Haya Al-Bitar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada.
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
| | - Michael Bezuhly
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
- Division of Plastic Surgery, Izaak Walton Killam Health Centre, 5850/5980 University Avenue, PO Box 9700, Halifax, NS, B3K 6R8, Canada.
- Department of Surgery, Dalhousie University, Halifax, Canada.
| |
Collapse
|
10
|
Nelson AR, Christiansen SL, Naegle KM, Saucerman JJ. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts. Proc Natl Acad Sci U S A 2024; 121:e2303513121. [PMID: 38266046 PMCID: PMC10835125 DOI: 10.1073/pnas.2303513121] [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: 03/01/2023] [Accepted: 11/30/2023] [Indexed: 01/26/2024] Open
Abstract
Fibroblasts are essential regulators of extracellular matrix deposition following cardiac injury. These cells exhibit highly plastic responses in phenotype during fibrosis in response to environmental stimuli. Here, we test whether and how candidate anti-fibrotic drugs differentially regulate measures of cardiac fibroblast phenotype, which may help identify treatments for cardiac fibrosis. We conducted a high-content microscopy screen of human cardiac fibroblasts treated with 13 clinically relevant drugs in the context of TGFβ and/or IL-1β, measuring phenotype across 137 single-cell features. We used the phenotypic data from our high-content imaging to train a logic-based mechanistic machine learning model (LogiMML) for fibroblast signaling. The model predicted how pirfenidone and Src inhibitor WH-4-023 reduce actin filament assembly and actin-myosin stress fiber formation, respectively. Validating the LogiMML model prediction that PI3K partially mediates the effects of Src inhibition, we found that PI3K inhibition reduces actin-myosin stress fiber formation and procollagen I production in human cardiac fibroblasts. In this study, we establish a modeling approach combining the strengths of logic-based network models and regularized regression models. We apply this approach to predict mechanisms that mediate the differential effects of drugs on fibroblasts, revealing Src inhibition acting via PI3K as a potential therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Anders R. Nelson
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| | - Steven L. Christiansen
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
- Department of Biochemistry, Brigham Young University, Provo, UT84602
| | - Kristen M. Naegle
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA22903
| |
Collapse
|
11
|
Hung CC, Chen KH, Hsu HH, Chang MY, Ko YC, Yang HY, Yang CW. Noscapine alleviates unilateral ureteral obstruction-induced inflammation and fibrosis by regulating the TGFβ1/Smads signaling pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119594. [PMID: 37730129 DOI: 10.1016/j.bbamcr.2023.119594] [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: 02/25/2023] [Revised: 09/02/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023]
Abstract
Renal fibrosis is a common pathway leading to progressive renal function loss in various forms of chronic kidney disease. Many fibrogenic factors regulate renal fibrosis; two key players are post-injury inflammation and transforming growth factor-β1 (TGF-β1)-induced myofibroblast differentiation. Myofibroblast differentiation is tightly regulated by the microtubule polymerization. Noscapine, an antitussive plant alkaloid, is a potent microtubule-interfering agent previously identified as a potential anticancer compound. Here, we examined how noscapine affects renal fibrogenesis in an in vitro renal fibroblast model and an in vivo unilateral ureteral obstruction (UUO) model. UUO mice were intraperitoneally treated with noscapine at 1 day before UUO surgery and daily thereafter. At 7 days post-surgery, kidneys were collected for further analysis. To analyze whether noscapine inhibits downstream TGF-β1-related signaling, we pre-incubated NRK-49F fibroblasts with noscapine and then performed TGF-β1 stimulation. In UUO mice, noscapine attenuated extracellular matrix protein deposition and the expression levels of type I collagen, type IV collagen, α-smooth muscle actin, and fibronectin. In addition, noscapine decreased tubulointerstitial inflammation in UUO kidneys by reducing TLR2 expression, modulating NLRP3 inflammasome activation, reducing macrophage infiltration, and antagonizing the M2 macrophage phenotype. Furthermore, noscapine pre-incubation suppressed the TGF-β1-induced fibroblast-myofibroblast transformation by downregulating the TGF-β/Smads signaling pathways in NRK-49F cells. These results suggest that noscapine reduces tubulointerstitial inflammation and fibrosis in the kidneys of UUO mice and inhibits the fibroblast-myofibroblast transformation induced by TGF-β1. Noscapine is an over-the-counter antitussive that has been used safely for several decades. Therefore, noscapine is an attractive therapeutic agent for inhibiting renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Cheng-Chieh Hung
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan.
| | - Kuan-Hsing Chen
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Hsiang-Hao Hsu
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Ming-Yang Chang
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Yi-Ching Ko
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Huang-Yu Yang
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Chih-Wei Yang
- Department of Nephrology and Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| |
Collapse
|
12
|
Ma M, Li L, Zuo G, Xiao J, Chen J, He X, Song Z. Effect of Zinc Amino Acid Complexes on Growth Performance, Tissue Zinc Concentration, and Muscle Development of Broilers. Biol Trace Elem Res 2024; 202:291-306. [PMID: 37086354 DOI: 10.1007/s12011-023-03661-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023]
Abstract
The present study aimed to evaluate the effects of zinc amino acid complexes on growth performance, tissue zinc concentration, and muscle development in broilers. A total of 504 day-old male arbor acres broilers were randomly divided into seven treatments (fed with a basal diet or a basal diet supplemented with 120 mg kg-1 Zn as ZnSO4, 30, 60, 90 or 120 mg kg-1 Zn as ZnN, or 30 mg kg-1 Zn as ZnA separately). Each group had six replicates, with 12 birds per replicate. The results showed that the addition of 60 mg kg-1 ZnN significantly increased (P < 0.05) the average daily gain (ADG) and breast muscle percentage of broilers. Zinc concentration of ZnN and ZnA added groups were higher than (P < 0.05) that in the Zn sulfate group under the same addition dose. Except for the 30 mg kg-1 ZnN group, the muscle fiber diameter and cross-sectional area (CSA) were significantly increased (P < 0.05) in the ZnN addition groups. Compared with the basal diet group, adding ZnN significantly increased (P < 0.05) the expression of MTOR, MYOD, and MYOG at day 21 and decreased (P < 0.05) the expression of Atrogin-1. The expression levels of AKT, MTOR, P70S6K, and MYOD were increased at day 42, while the expression levels of MuRF1 and Atrogin-1 were decreased. Adhesion, backbone regulation of actin, MAPK, mTOR, and AMPK were significantly enriched as indicated by KEGG pathway enrichment analysis. In conclusion, zinc amino acid complexes could improve growth performance, tissue zinc concentration, and regulate breast muscle development.
Collapse
Affiliation(s)
- Mengmeng Ma
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, 410128, China
| | - Liwei Li
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, 410128, China
| | - Gang Zuo
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, 410128, China
- Beijing Deyuanshun Biotechnology Co., Ltd., Beijing, 102206, China
| | - Jian Xiao
- Hunan Xiang Jia Husbandry Limited By Share Ltd., Changde, 415000, Hunan, China
| | - Junlie Chen
- Hunan Xiang Jia Husbandry Limited By Share Ltd., Changde, 415000, Hunan, China
| | - Xi He
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, 410128, China
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, 410128, China
| | - Zehe Song
- College of Animal Science and Technology, Hunan Agricultural University, Hunan, 410128, China.
- Hunan Engineering Research Center of Poultry Production Safety, Changsha, 410128, China.
| |
Collapse
|
13
|
Kumari J, Hammink R, Baaij J, Wagener FADTG, Kouwer PHJ. Antifibrotic properties of hyaluronic acid crosslinked polyisocyanide hydrogels. BIOMATERIALS ADVANCES 2024; 156:213705. [PMID: 38006784 DOI: 10.1016/j.bioadv.2023.213705] [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: 07/22/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
Fibrosis is characterized by the formation of fibrous connective tissue in response to primary injury. As a result, an affected organ may lose part of its functionality due to chronic, organ-specific tissue damage. Since fibrosis is a leading cause of death worldwide, targeting fibrotic diseases with antifibrotic hydrogels can be a lifesaving therapeutic strategy. This study developed a novel hybrid antifibrotic hydrogel by combining the synthetic polyisocyanide (PIC) with hyaluronic acid (HA). Gels of PIC are highly tailorable, thermosensitive, and strongly biomimetic in architecture and mechanical properties, whereas HA is known to promote non-fibrotic fetal wound healing and inhibits inflammatory signaling. The developed HA-PIC hybrids were biocompatible with physical properties comparable to those of the PIC gels. The antifibrotic nature of the gels was assessed by 3D cultures of human foreskin fibroblasts in the presence (or absence as control) of TGFβ1 that promotes differentiation into myofibroblasts, a critical step in fibrosis. Proliferation and macroscopic contraction assays and studies on the formation of stress fibers and characteristic fibrosis markers all indicate a strong antifibrotic nature of HA-PIC hydrogel. We showed that these effects originate from both the lightly crosslinked architecture and the presence of HA itself. The hybrid displaying both these effects shows the strongest antifibrotic nature and is a promising candidate for use as in vivo treatment for skin fibrosis.
Collapse
Affiliation(s)
- Jyoti Kumari
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands; Department of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, the Netherlands
| | - Roel Hammink
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Jochem Baaij
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Frank A D T G Wagener
- Department of Dentistry - Orthodontics and Craniofacial Biology, Radboud University Medical Centre, 6525 EX Nijmegen, the Netherlands.
| | - Paul H J Kouwer
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands.
| |
Collapse
|
14
|
Reed EB, Orbeta S, Miao BA, Sitikov A, Chen B, Levitan I, Solway J, Mutlu GM, Fang Y, Mongin AA, Dulin NO. Anoctamin-1 is induced by TGF-β and contributes to lung myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol 2024; 326:L111-L123. [PMID: 38084409 PMCID: PMC11279757 DOI: 10.1152/ajplung.00155.2023] [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: 05/16/2023] [Revised: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 12/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by progressive scarring of the lungs and resulting in deterioration in lung function. Transforming growth factor-β (TGF-β) is one of the most established drivers of fibrotic processes. TGF-β promotes the transformation of tissue fibroblasts to myofibroblasts, a key finding in the pathogenesis of pulmonary fibrosis. We report here that TGF-β robustly upregulates the expression of the calcium-activated chloride channel anoctamin-1 (ANO1) in human lung fibroblasts (HLFs) at mRNA and protein levels. ANO1 is readily detected in fibrotic areas of IPF lungs in the same area with smooth muscle α-actin (SMA)-positive myofibroblasts. TGF-β-induced myofibroblast differentiation (determined by the expression of SMA, collagen-1, and fibronectin) is significantly inhibited by a specific ANO1 inhibitor, T16Ainh-A01, or by siRNA-mediated ANO1 knockdown. T16Ainh-A01 and ANO1 siRNA attenuate profibrotic TGF-β signaling, including activation of RhoA pathway and AKT, without affecting initial Smad2 phosphorylation. Mechanistically, TGF-β treatment of HLFs results in a significant increase in intracellular chloride levels, which is prevented by T16Ainh-A01 or by ANO1 knockdown. The downstream mechanism involves the chloride-sensing "with-no-lysine (K)" kinase (WNK1). WNK1 siRNA significantly attenuates TGF-β-induced myofibroblast differentiation and signaling (RhoA pathway and AKT), whereas the WNK1 kinase inhibitor WNK463 is largely ineffective. Together, these data demonstrate that 1) ANO1 is a TGF-β-inducible chloride channel that contributes to increased intracellular chloride concentration in response to TGF-β; and 2) ANO1 mediates TGF-β-induced myofibroblast differentiation and fibrotic signaling in a manner dependent on WNK1 protein but independent of WNK1 kinase activity.NEW & NOTEWORTHY This study describes a novel mechanism of differentiation of human lung fibroblasts (HLFs) to myofibroblasts: the key process in the pathogenesis of pulmonary fibrosis. Transforming growth factor-β (TGF-β) drives the expression of calcium-activated chloride channel anoctmin-1 (ANO1) leading to an increase in intracellular levels of chloride. The latter recruits chloride-sensitive with-no-lysine (K) kinase (WNK1) to activate profibrotic RhoA and AKT signaling pathways, possibly through activation of mammalian target of rapamycin complex-2 (mTORC2), altogether promoting myofibroblast differentiation.
Collapse
Affiliation(s)
- Eleanor B Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Shaina Orbeta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Bernadette A Miao
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Irena Levitan
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Julian Solway
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Yun Fang
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, United States
| | - Nickolai O Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, Illinois, United States
| |
Collapse
|
15
|
Dong C, Yu Z, Du J, Zhang Y, Liu W, Huang Z, Xiong S, Wang T, Song Y, Ma X. Montelukast Attenuates Retraction of Expanded Flap by Inhibiting Capsule Formation around Silicone Expander through TGF-β1 Signaling. Plast Reconstr Surg 2023; 152:1044e-1052e. [PMID: 36988445 DOI: 10.1097/prs.0000000000010459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
BACKGROUND Tissue expansion has tremendous applications in plastic surgery, but flap retraction provides insufficient tissue for use. Inspired by the use of montelukast to suppress capsular contracture, the authors investigated the effects of montelukast on capsule formation around the expander and retraction of the expanded scalp of the rat. METHODS Thirty-six male Sprague-Dawley rats were randomly divided into control and montelukast groups. In each group, 12 expanded flaps with or without capsules were harvested for histologic and molecular analysis; the six remaining expanded flaps were transferred to repair defects. Myofibroblast and transforming growth factor-β1 expression in the capsule was determined using immunofluorescence. Capsule ultrastructure was observed using transmission electron microscopy. Related protein expression in the capsules was detected using Western blot analysis. RESULTS A comparison of control and montelukast groups revealed that areas of the harvested expanded flaps with capsules were greater (2.04 ± 0.11 cm 2 versus 2.42 ± 0.12 cm 2 , respectively; P = 0.04); the retraction rate decreased (41.3% ± 2.16% versus 28.13% ± 2.17%, respectively; P < 0.01). However, the increased areas and decreased retraction disappeared after capsule removal. The number of myofibroblasts declined. Thin, sparse collagen fibers were observed in the capsules. The expression of COL1, COL3, TGF-β1, EGR1, and phosphorylated ERK1/2 in the capsules decreased. Furthermore, the recipient area repaired by the transferred expanded flap was increased from 4.25 ± 0.39 cm 2 to 6.58 ± 0.31 cm 2 ( P < 0.01). CONCLUSION Montelukast attenuates retraction of the expanded flap by inhibiting capsule formation through suppressing transforming growth factor-β1 signaling. CLINICAL RELEVANCE STATEMENT This study provides novel insights into a method for increasing the area of the expanded flap.
Collapse
Affiliation(s)
- Chen Dong
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Zhou Yu
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Jing Du
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Yu Zhang
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Wei Liu
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Zhaosong Huang
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Shaoheng Xiong
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Tong Wang
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Yajuan Song
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| | - Xianjie Ma
- From the Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University
| |
Collapse
|
16
|
Phillips AT, Boumil EF, Venkatesan A, Tilstra-Smith C, Castro N, Knox BE, Henty-Ridilla JL, Bernstein AM. The formin DAAM1 regulates the deubiquitinase activity of USP10 and integrin homeostasis. Eur J Cell Biol 2023; 102:151347. [PMID: 37562219 PMCID: PMC10839120 DOI: 10.1016/j.ejcb.2023.151347] [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: 07/12/2022] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
The differentiation of fibroblasts into pathological myofibroblasts during wound healing is characterized by increased cell surface expression of αv-integrins. Our previous studies found that the deubiquitinase (DUB) USP10 removes ubiquitin from αv-integrins, leading to cell surface integrin accumulation, subsequent TGFβ1 activation, and pathological myofibroblast differentiation. In this study, a yeast two-hybrid screen revealed a novel binding partner for USP10, the formin, DAAM1. We found that DAAM1 binds to and inhibits USP10's DUB activity through the FH2 domain of DAAM1 independent of its actin functions. The USP10/DAAM1 interaction was also supported by proximity ligation assay (PLA) in primary human corneal fibroblasts. Treatment with TGFβ1 significantly increased USP10 and DAAM1 protein expression, PLA signal, and co-localization to actin stress fibers. DAAM1 siRNA knockdown significantly reduced co-precipitation of USP10 and DAAM1 on purified actin stress fibers, and β1- and β5-integrin ubiquitination. This resulted in increased αv-, β1-, and β5-integrin total protein levels, αv-integrin recycling, and extracellular fibronectin (FN) deposition. Together, our data demonstrate that DAAM1 inhibits USP10's DUB activity on integrins subsequently regulating cell surface αv-integrin localization and FN accumulation.
Collapse
Affiliation(s)
- Andrew T Phillips
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Edward F Boumil
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Arunkumar Venkatesan
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Christine Tilstra-Smith
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Nileyma Castro
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA
| | - Barry E Knox
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Jessica L Henty-Ridilla
- SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Audrey M Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, 750 East Adams Street, Syracuse, NY 13210, USA; SUNY Upstate Medical University, Biochemistry and Molecular Biology, 750 East Adams Street, Syracuse, NY 13210, USA; New York VA Health Care, Syracuse VA Medical Center, 800 Irving Ave, Syracuse 13210, USA.
| |
Collapse
|
17
|
Piñero-Pérez R, López-Cabrera A, Álvarez-Córdoba M, Cilleros-Holgado P, Talaverón-Rey M, Suárez-Carrillo A, Munuera-Cabeza M, Gómez-Fernández D, Reche-López D, Romero-González A, Romero-Domínguez JM, de Pablos RM, Sánchez-Alcázar JA. Actin Polymerization Defects Induce Mitochondrial Dysfunction in Cellular Models of Nemaline Myopathies. Antioxidants (Basel) 2023; 12:2023. [PMID: 38136143 PMCID: PMC10740811 DOI: 10.3390/antiox12122023] [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: 10/24/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Nemaline myopathy (NM) is one of the most common forms of congenital myopathy and it is identified by the presence of "nemaline bodies" (rods) in muscle fibers by histopathological examination. The most common forms of NM are caused by mutations in the Actin Alpha 1 (ACTA1) and Nebulin (NEB) genes. Clinical features include hypotonia and muscle weakness. Unfortunately, there is no curative treatment and the pathogenetic mechanisms remain unclear. In this manuscript, we examined the pathophysiological alterations in NM using dermal fibroblasts derived from patients with mutations in ACTA1 and NEB genes. Patients' fibroblasts were stained with rhodamine-phalloidin to analyze the polymerization of actin filaments by fluorescence microscopy. We found that patients' fibroblasts showed incorrect actin filament polymerization compared to control fibroblasts. Actin filament polymerization defects were associated with mitochondrial dysfunction. Furthermore, we identified two mitochondrial-boosting compounds, linoleic acid (LA) and L-carnitine (LCAR), that improved the formation of actin filaments in mutant fibroblasts and corrected mitochondrial bioenergetics. Our results indicate that cellular models can be useful to study the pathophysiological mechanisms involved in NM and to find new potential therapies. Furthermore, targeting mitochondrial dysfunction with LA and LCAR can revert the pathological alterations in NM cellular models.
Collapse
Affiliation(s)
- Rocío Piñero-Pérez
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Alejandra López-Cabrera
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Mónica Álvarez-Córdoba
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Paula Cilleros-Holgado
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Marta Talaverón-Rey
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Alejandra Suárez-Carrillo
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Manuel Munuera-Cabeza
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - David Gómez-Fernández
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Diana Reche-López
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Ana Romero-González
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - José Manuel Romero-Domínguez
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| | - Rocío M. de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain;
- Instituto of Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío (HUVR)/CSIC/Universidad de Sevilla, 41012 Sevilla, Spain
| | - José A. Sánchez-Alcázar
- Departamento de Fisiología, Anatomía y Biología Celular, Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (R.P.-P.); (A.L.-C.); (M.Á.-C.); (P.C.-H.); (M.T.-R.); (A.S.-C.); (M.M.-C.); (D.G.-F.); (D.R.-L.); (A.R.-G.); (J.M.R.-D.)
| |
Collapse
|
18
|
Reed EB, Orbeta S, Miao BA, Sitikov A, Chen B, Levitan I, Solway J, Mutlu GM, Fang Y, Mongin AA, Dulin NO. Anoctamin-1 is induced by TGF-beta and contributes to lung myofibroblast differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544093. [PMID: 37333255 PMCID: PMC10274757 DOI: 10.1101/2023.06.07.544093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by progressive scarring of the lungs and resulting in deterioration in lung function. Transforming growth factor-beta (TGF-β) is one of the most established drivers of fibrotic processes. TGF-β promotes transformation of tissue fibroblasts to myofibroblasts, a key finding in the pathogenesis of pulmonary fibrosis. We report here that TGF-β robustly upregulates the expression of the calcium-activated chloride channel Anoctamin-1 (ANO1) in human lung fibroblasts (HLF) at mRNA and protein levels. ANO1 is readily detected in fibrotic areas of IPF lungs in the same area with smooth muscle alpha-actin (SMA)-positive myofibroblasts. TGF-β-induced myofibroblast differentiation (determined by the expression of SMA, collagen-1 and fibronectin) is significantly inhibited by a specific ANO1 inhibitor, T16Ainh-A01, or by siRNA-mediated ANO1 knockdown. T16Ainh-A01 and ANO1 siRNA attenuate pro-fibrotic TGF-β signaling, including activation of RhoA pathway and AKT, without affecting initial Smad2 phosphorylation. Mechanistically, TGF-β treatment of HLF results in a significant increase in intracellular chloride levels, which is prevented by T16Ainh-A01 or by ANO1 knockdown. The downstream mechanism involves the chloride-sensing "with-no-lysine (K)" kinase (WNK1). WNK1 siRNA significantly attenuates TGF-β-induced myofibroblast differentiation and signaling (RhoA pathway and AKT), whereas the WNK1 kinase inhibitor WNK463 is largely ineffective. Together, these data demonstrate that (i) ANO1 is a TGF-β-inducible chloride channel that contributes to increased intracellular chloride concentration in response to TGF-β; and (ii) ANO1 mediates TGF-β-induced myofibroblast differentiation and fibrotic signaling in a manner dependent on WNK1 protein, but independent of WNK1 kinase activity.
Collapse
Affiliation(s)
- Eleanor B. Reed
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Shaina Orbeta
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY
| | - Bernadette A. Miao
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Albert Sitikov
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Bohao Chen
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Irena Levitan
- Departments of Medicine, Pharmacology and Bioengineering, University of Illinois at Chicago, Chicago, IL
| | - Julian Solway
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Gökhan M. Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Yun Fang
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| | - Alexander A. Mongin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY
| | - Nickolai O. Dulin
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Warwar R, Zupan AM, Nietupski C, Manzanares M, Hurley EG, Schutte SC. Uterine fibroid cell cytoskeletal organization is affected by altered G protein-coupled estrogen receptor-1 and phosphatidylinositol 3-kinase signaling. F&S SCIENCE 2023; 4:327-338. [PMID: 37797815 DOI: 10.1016/j.xfss.2023.09.007] [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: 05/10/2023] [Revised: 09/05/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE To determine whether cyclic strain affects fibroid cell cytoskeletal organization, proliferation, and collagen synthesis differently than myometrial cells. DESIGN A basic science study using primary cultures of patient-matched myometrial and fibroid cells. SETTING Academic laboratory. PATIENT(S) Premenopausal women undergoing myomectomy or hysterectomy for the treatment of symptomatic uterine fibroids. INTERVENTION(S) Application of uniaxial strain patterns mimicking periovulation, menses, or dysmenorrhea using the Flexcell tension system or static control. Secondarily, inhibition of G protein-coupled estrogen receptor-1 and phosphatidylinositol 3-kinase. MAIN OUTCOME MEASURE(S) Cell alignment, cell number, and collagen content. RESULT(S) Menses-strained cells demonstrated the most variation in cell alignment, cell proliferation, and procollagen content between myometrial and fibroid cells. Procollagen content decreased in myometrial cells with increasing strain amplitude and decreasing frequency. G protein-coupled estrogen receptor-1 inhibition decreases cellular alignment in the presence of strain. CONCLUSION(S) Mechanotransduction affecting cytoskeletal arrangement through the G protein-coupled estrogen receptor-1-phosphatidylinositol 3-kinase pathway is altered in fibroid cells. These results highlight the importance of incorporating mechanical stimulation into the in vitro study of fibroid pathology.
Collapse
Affiliation(s)
- Rachel Warwar
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Andreja Moset Zupan
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Carolyn Nietupski
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Maricela Manzanares
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Emily G Hurley
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Stacey C Schutte
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
20
|
Rao WT, Jiang S, Shen YH, Wang YH, Liu SN, Tang JD, Xing JF. Myofibroblasts: A New Factor Affecting the Hyperlipidemia-Induced Elastic Abnormality of Corpus Cavernosum in Rabbits Detected by 2-D Shear Wave Elastography. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:2336-2345. [PMID: 37544829 DOI: 10.1016/j.ultrasmedbio.2023.07.010] [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: 02/14/2023] [Revised: 06/22/2023] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
OBJECTIVE Two-dimensional shear wave elastography (2-D SWE) has been proven to detect hyperlipidemia-induced elastic abnormality in the corpus cavernosum. This study investigated cytological factors affecting the elasticity of the corpus cavernosum in rabbits with hyperlipidemia using single-cell RNA sequencing (scRNA-seq). METHODS Male New Zealand white rabbits were randomly divided into a hyperlipidemia group (high-cholesterol diet) and a control group (standard diet). Penile 2-D SWE was performed to detect the elastic abnormality in the corpus cavernosum. ScRNA-seq was performed to observe cellular changes in the corpus cavernosum of rabbits with hyperlipidemia. Immunohistochemistry, immunofluorescence and histological examinations were conducted to verify the results of scRNA-seq. RESULTS Two-dimensional SWE revealed that the Young's modulus of the corpus cavernosum was significantly greater in the hyperlipidemia group than that in the control group (p < 0.001). Histological findings revealed extracellular matrix accumulation within the corpus cavernosum, with stronger staining of collagen types I and Ⅲ. ScRNA-seq revealed that fibroblasts, smooth muscle cells, and endothelial cells were the major cell types in the corpus cavernosum. A novel subtype of fibroblasts (myofibroblast) was discovered in the hyperlipidemia group, which was verified by immunofluorescence staining and gene ontology analysis. Fibroblasts, smooth muscle cells and endothelial cells were three cellular sources for myofibroblasts. CONCLUSION Myofibroblasts are activated and proliferate and secrete large amounts of collagen fibers in the corpus cavernosum during hyperlipidemia, leading to abnormal Young's modulus detected by 2-D SWE and their recognition as a new factor affecting the hyperlipidemia-induced elastic abnormality of the corpus cavernosum.
Collapse
Affiliation(s)
- Wan-Ting Rao
- Department of Medical Ultrasound, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuai Jiang
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yi-Hao Shen
- Department of Medical Ultrasound, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yan-He Wang
- Department of Medical Ultrasound, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Sen-Ning Liu
- Department of Medical Ultrasound, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jing-Dong Tang
- Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jin-Fang Xing
- Department of Medical Ultrasound, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China; Shanghai Key Laboratory of Vascular Lesions Regulation and Remodeling, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
21
|
Nelson AR, Christiansen SL, Naegle KM, Saucerman JJ. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530599. [PMID: 36909540 PMCID: PMC10002757 DOI: 10.1101/2023.03.01.530599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Fibroblasts are essential regulators of extracellular matrix deposition following cardiac injury. These cells exhibit highly plastic responses in phenotype during fibrosis in response to environmental stimuli. Here, we test whether and how candidate anti-fibrotic drugs differentially regulate measures of cardiac fibroblast phenotype, which may help identify treatments for cardiac fibrosis. We conducted a high content microscopy screen of human cardiac fibroblasts treated with 13 clinically relevant drugs in the context of TGFβ and/or IL-1β, measuring phenotype across 137 single-cell features. We used the phenotypic data from our high content imaging to train a logic-based mechanistic machine learning model (LogiMML) for fibroblast signaling. The model predicted how pirfenidone and Src inhibitor WH-4-023 reduce actin filament assembly and actin-myosin stress fiber formation, respectively. Validating the LogiMML model prediction that PI3K partially mediates the effects of Src inhibition, we found that PI3K inhibition reduces actin-myosin stress fiber formation and procollagen I production in human cardiac fibroblasts. In this study, we establish a modeling approach combining the strengths of logic-based network models and regularized regression models, apply this approach to predict mechanisms that mediate the differential effects of drugs on fibroblasts, revealing Src inhibition acting via PI3K as a potential therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Anders R. Nelson
- University of Virginia School of Medicine, Charlottesville, VA 22903
| | - Steven L. Christiansen
- University of Virginia School of Medicine, Charlottesville, VA 22903
- Brigham Young University Department of Biochemistry, Provo, UT 84602
| | - Kristen M. Naegle
- University of Virginia School of Medicine, Charlottesville, VA 22903
| | | |
Collapse
|
22
|
Atzemian N, Dovrolis N, Ragia G, Portokallidou K, Kolios G, Manolopoulos VG. Beyond the Rhythm: In Silico Identification of Key Genes and Therapeutic Targets in Atrial Fibrillation. Biomedicines 2023; 11:2632. [PMID: 37893006 PMCID: PMC10604372 DOI: 10.3390/biomedicines11102632] [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: 07/31/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is a prevalent cardiac arrhythmia worldwide and is characterized by a high risk of thromboembolism, ischemic stroke, and fatality. The precise molecular mechanisms of AF pathogenesis remain unclear. The purpose of this study was to use bioinformatics tools to identify novel key genes in AF, provide deeper insights into the molecular pathogenesis of AF, and uncover potential therapeutic targets. Four publicly available raw RNA-Seq datasets obtained through the ENA Browser, as well as proteomic analysis results, both derived from atrial tissues, were used in this analysis. Differential gene expression analysis was performed and cross-validated with proteomics results to identify common genes/proteins between them. A functional enrichment pathway analysis was performed. Cross-validation analysis revealed five differentially expressed genes, namely FGL2, IGFBP5, NNMT, PLA2G2A, and TNC, in patients with AF compared with those with sinus rhythm (SR). These genes play crucial roles in various cardiovascular functions and may be part of the molecular signature of AF. Furthermore, functional enrichment analysis revealed several pathways related to the extracellular matrix, inflammation, and structural remodeling. This study highlighted five key genes that constitute promising candidates for further experimental exploration as biomarkers as well as therapeutic targets for AF.
Collapse
Affiliation(s)
- Natalia Atzemian
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Konstantina Portokallidou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (N.A.); (G.R.); (K.P.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
| |
Collapse
|
23
|
Poe A, Martinez Yus M, Wang H, Santhanam L. Lysyl oxidase like-2 in fibrosis and cardiovascular disease. Am J Physiol Cell Physiol 2023; 325:C694-C707. [PMID: 37458436 PMCID: PMC10635644 DOI: 10.1152/ajpcell.00176.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 09/01/2023]
Abstract
Fibrosis is an important and essential reparative response to injury that, if left uncontrolled, results in the excessive synthesis, deposition, remodeling, and stiffening of the extracellular matrix, which is deleterious to organ function. Thus, the sustained activation of enzymes that catalyze matrix remodeling and cross linking is a fundamental step in the pathology of fibrotic diseases. Recent studies have implicated the amine oxidase lysyl oxidase like-2 (LOXL2) in this process and established significantly elevated expression of LOXL2 as a key component of profibrotic conditions in several organ systems. Understanding the relationship between LOXL2 and fibrosis as well as the mechanisms behind these relationships can offer significant insights for developing novel therapies. Here, we summarize the key findings that demonstrate the link between LOXL2 and fibrosis and inflammation, examine current therapeutics targeting LOXL2 for the treatment of fibrosis, and discuss future directions for experiments and biomedical engineering.
Collapse
Affiliation(s)
- Alan Poe
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Marta Martinez Yus
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| | - Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Anesthesiology and CCM, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
24
|
Angelini A, Trial J, Saltzman AB, Malovannaya A, Cieslik KA. A defective mechanosensing pathway affects fibroblast-to-myofibroblast transition in the old male mouse heart. iScience 2023; 26:107283. [PMID: 37520701 PMCID: PMC10372839 DOI: 10.1016/j.isci.2023.107283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/12/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
The cardiac fibroblast interacts with an extracellular matrix (ECM), enabling myofibroblast maturation via a process called mechanosensing. Although in the aging male heart, ECM is stiffer than in the young mouse, myofibroblast development is impaired, as demonstrated in 2-D and 3-D experiments. In old male cardiac fibroblasts, we found a decrease in actin polymerization, α-smooth muscle actin (α-SMA), and Kindlin-2 expressions, the latter an effector of the mechanosensing. When Kindlin-2 levels were manipulated via siRNA interference, young fibroblasts developed an old-like fibroblast phenotype, whereas Kindlin-2 overexpression in old fibroblasts reversed the defective phenotype. Finally, inhibition of overactivated extracellular regulated kinases 1 and 2 (ERK1/2) in the old male fibroblasts rescued actin polymerization and α-SMA expression. Pathological ERK1/2 overactivation was also attenuated by Kindlin-2 overexpression. In contrast, old female cardiac fibroblasts retained an operant mechanosensing pathway. In conclusion, we identified defective components of the Kindlin/ERK/actin/α-SMA mechanosensing axis in aged male fibroblasts.
Collapse
Affiliation(s)
- Aude Angelini
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - JoAnn Trial
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Alexander B. Saltzman
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Anna Malovannaya
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Katarzyna A. Cieslik
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
VanSlyke JK, Boswell BA, Musil LS. ErbBs in Lens Cell Fibrosis and Secondary Cataract. Invest Ophthalmol Vis Sci 2023; 64:6. [PMID: 37418274 PMCID: PMC10337807 DOI: 10.1167/iovs.64.10.6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/30/2023] [Indexed: 07/08/2023] Open
Abstract
Purpose TGFβ-induced epithelial-to-myofibroblast transition (EMyT) of lens cells has been linked to the most common vision-disrupting complication of cataract surgery-namely, posterior capsule opacification (PCO; secondary cataract). Although inhibitors of the ErbB family of receptor tyrosine kinases have been shown to block some PCO-associated processes in model systems, our knowledge of ErbB signaling in the lens is very limited. Here, we investigate the expression of ErbBs and their ligands in primary cultures of chick lens epithelial cells (dissociated cell-derived monolayer cultures [DCDMLs]) and how TGFβ affects ErbB function. Methods DCDMLs were analyzed by immunofluorescence microscopy and Western blotting under basal and profibrotic conditions. Results Small-molecule ErbB kinase blockers, including the human therapeutic lapatinib, selectively inhibit TGFβ-induced EMyT of DCDMLs. Lens cells constitutively express ErbB1 (EGFR), ErbB2, and ErbB4 protein on the plasma membrane and release into the medium ErbB-activating ligand. Culturing DCDMLs with TGFβ increases soluble bioactive ErbB ligand and markedly alters ErbBs, reducing total and cell surface ErbB2 and ErbB4 while increasing ErbB1 expression and homodimer formation. Similar, TGFβ-dependent changes in relative ErbB expression are induced when lens cells are exposed to the profibrotic substrate fibronectin. A single, 1-hour treatment with lapatinib inhibits EMyT in DCDMLs assessed 6 days later. Short-term exposure to lower doses of lapatinib is also capable of eliciting a durable response when combined with suboptimal levels of a mechanistically distinct multikinase inhibitor. Conclusions Our findings support ErbB1 as a therapeutic target for fibrotic PCO, which could be leveraged to pharmaceutically preserve the vision of millions of patients with cataracts.
Collapse
Affiliation(s)
- Judy K. VanSlyke
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Bruce A. Boswell
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| | - Linda S. Musil
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
26
|
Lam KH, Shihabeddin TZ, Awkal JA, Najjar AM, Miron-Mendoza M, Maruri DP, Varner VD, Petroll WM, Schmidtke DW. Effects of Topography and PDGF on the Response of Corneal Keratocytes to Fibronectin-Coated Surfaces. J Funct Biomater 2023; 14:217. [PMID: 37103307 PMCID: PMC10144166 DOI: 10.3390/jfb14040217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
During corneal wound healing, corneal keratocytes are exposed to both biophysical and soluble cues that cause them to transform from a quiescent state to a repair phenotype. How keratocytes integrate these multiple cues simultaneously is not well understood. To investigate this process, primary rabbit corneal keratocytes were cultured on substrates patterned with aligned collagen fibrils and coated with adsorbed fibronectin. After 2 or 5 days of culture, keratocytes were fixed and stained to assess changes in cell morphology and markers of myofibroblastic activation by fluorescence microscopy. Initially, adsorbed fibronectin had an activating effect on the keratocytes as evidenced by changes in cell shape, stress fiber formation, and expression of alpha-smooth muscle actin (α-SMA). The magnitude of these effects depended upon substrate topography (i.e., flat substrate vs aligned collagen fibrils) and decreased with culture time. When keratocytes were simultaneously exposed to adsorbed fibronectin and soluble platelet-derived growth factor-BB (PDGF-BB), the cells elongated and had reduced expression of stress fibers and α-SMA. In the presence of PDGF-BB, keratocytes plated on the aligned collagen fibrils elongated in the direction of the fibrils. These results provide new information on how keratocytes respond to multiple simultaneous cues and how the anisotropic topography of aligned collagen fibrils influences keratocyte behavior.
Collapse
Affiliation(s)
- Kevin H. Lam
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Tarik Z. Shihabeddin
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jacob A. Awkal
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Alex M. Najjar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Miguel Miron-Mendoza
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel P. Maruri
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Victor D. Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W. Matthew Petroll
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David W. Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Integration of Transcriptomics and Non-Targeted Metabolomics Reveals the Underlying Mechanism of Skeletal Muscle Development in Duck during Embryonic Stage. Int J Mol Sci 2023; 24:ijms24065214. [PMID: 36982289 PMCID: PMC10049352 DOI: 10.3390/ijms24065214] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Skeletal muscle is an important economic trait in duck breeding; however, little is known about the molecular mechanisms of its embryonic development. Here, the transcriptomes and metabolomes of breast muscle of Pekin duck from 15 (E15_BM), 21 (E21_BM), and 27 (E27_BM) days of incubation were compared and analyzed. The metabolome results showed that the differentially accumulated metabolites (DAMs), including the up-regulated metabolites, l-glutamic acid, n-acetyl-1-aspartylglutamic acid, l-2-aminoadipic acid, 3-hydroxybutyric acid, bilirubin, and the significantly down-regulated metabolites, palmitic acid, 4-guanidinobutanoate, myristic acid, 3-dehydroxycarnitine, and s-adenosylmethioninamine, were mainly enriched in metabolic pathways, biosynthesis of secondary metabolites, biosynthesis of cofactors, protein digestion and absorption, and histidine metabolism, suggesting that these pathways may play important roles in the muscle development of duck during the embryonic stage. Moreover, a total of 2142 (1552 up-regulated and 590 down-regulated), 4873 (3810 up-regulated and 1063 down-regulated), and 2401 (1606 up-regulated and 795 down-regulated) DEGs were identified from E15_BM vs. E21_BM, E15_BM vs. E27_BM and E21_BM vs. E27_BM in the transcriptome, respectively. The significantly enriched GO terms from biological processes were positive regulation of cell proliferation, regulation of cell cycle, actin filament organization, and regulation of actin cytoskeleton organization, which were associated with muscle or cell growth and development. Seven significant pathways, highly enriched by FYN, PTK2, PXN, CRK, CRKL, PAK, RHOA, ROCK, INSR, PDPK1, and ARHGEF, were focal adhesion, regulation of actin cytoskeleton, wnt signaling pathway, insulin signaling pathway, extracellular matrix (ECM)-receptor interaction, cell cycle, and adherens junction, which participated in regulating the development of skeletal muscle in Pekin duck during the embryonic stage. KEGG pathway analysis of the integrated transcriptome and metabolome indicated that the pathways, including arginine and proline metabolism, protein digestion and absorption, and histidine metabolism, were involved in regulating skeletal muscle development in embryonic Pekin duck. These findings suggested that the candidate genes and metabolites involved in crucial biological pathways may regulate muscle development in the Pekin duck at the embryonic stage, and increased our understanding of the molecular mechanisms underlying the avian muscle development.
Collapse
|
28
|
Horii Y, Matsuda S, Toyota C, Morinaga T, Nakaya T, Tsuchiya S, Ohmuraya M, Hironaka T, Yoshiki R, Kasai K, Yamauchi Y, Takizawa N, Nagasaka A, Tanaka A, Kosako H, Nakaya M. VGLL3 is a mechanosensitive protein that promotes cardiac fibrosis through liquid-liquid phase separation. Nat Commun 2023; 14:550. [PMID: 36754961 PMCID: PMC9908974 DOI: 10.1038/s41467-023-36189-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Myofibroblasts cause tissue fibrosis by producing extracellular matrix proteins, such as collagens. Humoral factors like TGF-β, and matrix stiffness are important for collagen production by myofibroblasts. However, the molecular mechanisms regulating their ability to produce collagen remain poorly characterised. Here, we show that vestigial-like family member 3 (VGLL3) is specifically expressed in myofibroblasts from mouse and human fibrotic hearts and promotes collagen production. Further, substrate stiffness triggers VGLL3 translocation into the nucleus through the integrin β1-Rho-actin pathway. In the nucleus, VGLL3 undergoes liquid-liquid phase separation via its low-complexity domain and is incorporated into non-paraspeckle NONO condensates containing EWS RNA-binding protein 1 (EWSR1). VGLL3 binds EWSR1 and suppresses miR-29b, which targets collagen mRNA. Consistently, cardiac fibrosis after myocardial infarction is significantly attenuated in Vgll3-deficient mice, with increased miR-29b expression. Overall, our results reveal an unrecognised VGLL3-mediated pathway that controls myofibroblasts' collagen production, representing a novel therapeutic target for tissue fibrosis.
Collapse
Affiliation(s)
- Yuma Horii
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichi Matsuda
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Chikashi Toyota
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takumi Morinaga
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeo Nakaya
- Department of Pathology, Jichi Medical University, Tochigi, Japan
| | - Soken Tsuchiya
- Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo College of Medicine, Hyogo, Japan
| | - Takanori Hironaka
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yoshiki
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kotaro Kasai
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuto Yamauchi
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Noburo Takizawa
- Department of Disease Control, Kyushu University, Fukuoka, Japan.,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiomi Nagasaka
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Tanaka
- Department of Pathology, Jichi Medical University, Tochigi, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Michio Nakaya
- Department of Disease Control, Kyushu University, Fukuoka, Japan. .,Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan. .,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
29
|
Hironaka T, Takizawa N, Yamauchi Y, Horii Y, Nakaya M. The well-developed actin cytoskeleton and Cthrc1 expression by actin-binding protein drebrin in myofibroblasts promote cardiac and hepatic fibrosis. J Biol Chem 2023; 299:102934. [PMID: 36690273 PMCID: PMC9988570 DOI: 10.1016/j.jbc.2023.102934] [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: 10/15/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Fibrosis is mainly triggered by inflammation in various tissues, such as heart and liver tissues, and eventually leads to their subsequent dysfunction. Fibrosis is characterized by the excessive accumulation of extracellular matrix proteins (e.g., collagens) produced by myofibroblasts. The well-developed actin cytoskeleton of myofibroblasts, one of the main features differentiating them from resident fibroblasts in tissues under inflammatory conditions, contributes to maintaining their ability to produce excessive extracellular matrix proteins. However, the molecular mechanisms via which the actin cytoskeleton promotes the production of fibrosis-related genes in myofibroblasts remain unclear. In this study, we found, via single-cell analysis, that developmentally regulated brain protein (drebrin), an actin-binding protein, was specifically expressed in cardiac myofibroblasts with a well-developed actin cytoskeleton in fibrotic hearts. Moreover, our immunocytochemistry analysis revealed that drebrin promoted actin cytoskeleton formation and myocardin-related transcription factor-serum response factor signaling. Comprehensive single-cell analysis and RNA-Seq revealed that the expression of collagen triple helix repeat containing 1 (Cthrc1), a fibrosis-promoting secreted protein, was regulated by drebrin in cardiac myofibroblasts via myocardin-related transcription factor-serum response factor signaling. Furthermore, we observed the profibrotic effects of drebrin exerted via actin cytoskeleton formation and the Cthrc1 expression regulation by drebrin in liver myofibroblasts (hepatic stellate cells). Importantly, RNA-Seq demonstrated that drebrin expression levels increased in human fibrotic heart and liver tissues. In summary, our results indicated that the well-developed actin cytoskeleton and Cthrc1 expression due to drebrin in myofibroblasts promoted cardiac and hepatic fibrosis, suggesting that drebrin is a therapeutic target molecule for fibrosis.
Collapse
Affiliation(s)
- Takanori Hironaka
- Department of Disease Control, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Noburo Takizawa
- Department of Disease Control, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuto Yamauchi
- Department of Disease Control, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuma Horii
- Department of Disease Control, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Michio Nakaya
- Department of Disease Control, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
30
|
Chitosan, chondroitin sulfate, and hyaluronic acid based in-situ forming scaffold for efficient cell grafting. Int J Biol Macromol 2023; 225:938-951. [PMID: 36410536 DOI: 10.1016/j.ijbiomac.2022.11.157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/26/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
Current cell grafting techniques are majorly dependent on seeding cells on a pre-formed scaffold. However, cells grow in a 2-dimensional (2D) space in such constructs, not mimicking the tissue's 3-dimensional (3D) architecture. The present study evaluated a unique poly-electrolyte complexation (PEC) based strategy for the 3D engraftment of cells in a porous polymeric scaffold. The scaffold was synthesized using a positively charged polysaccharide chitosan (CH) and negatively charged glycosaminoglycans chondroitin sulfate (CS) and hyaluronic acid (HA). Two different scaffolds were synthesized, one using CH and CS [CH-CS] and another using CH and CS + HA [CH-(CS-HA)]. The physicochemical characterization of both the PECs confirmed electrostatic interactions, leading to a porous and viscoelastic PEC formation. Fibroblast cells were grafted and seeded in both scaffolds to evaluate the effect of different scaffold compositions and the difference between seeded and grafted cells. Imaging studies confirmed that grafting of the fibroblast cells supports cellular proliferation. The qPCR studies demonstrated increased expression of functional markers TGF-β, α-SMA, collagen-I, and fibronectin in the CH-(CS-HA) grafted cells. In summary, it was demonstrated that an in-situ forming PEC of CH, CS, and HA had good physicochemical properties for cell grafting and supported grafted cells with improved function.
Collapse
|
31
|
Schröder SK, Tag CG, Weiskirchen S, Weiskirchen R. Phalloidin Staining for F-Actin in Hepatic Stellate Cells. Methods Mol Biol 2023; 2669:55-66. [PMID: 37247054 DOI: 10.1007/978-1-0716-3207-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
During the development of liver fibrosis, hepatic stellate cells undergo a transition from a quiescent phenotype into a proliferative, fibrogenic, and contractile, α-smooth muscle actin-positive myofibroblast. These cells acquire properties that are strongly associated with the reorganization of the actin cytoskeleton. Actin possesses a unique ability to polymerize into filamentous actin (F-actin) form its monomeric globular state (G-actin). F-actin can form robust actin bundles and cytoskeletal networks by interacting with a number of actin-binding proteins that provide important mechanical and structural support for a multitude of cellular processes including intracellular transport, cell motility, polarity, cell shape, gene regulation, and signal transduction. Therefore, stains with actin-specific antibodies and phalloidin conjugates for actin staining are widely used to visualize actin structures in myofibroblasts. Here we present an optimized protocol for F-actin staining for hepatic stellate cells using a fluorescent phalloidin.
Collapse
Affiliation(s)
- Sarah K Schröder
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany.
| | - Carmen G Tag
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institut für Molekulare Pathobiochemie, Experimentelle Gentherapie und Klinische Chemie (IFMPEGKC), Universitätsklinikum Aachen AöR, Aachen, Germany.
| |
Collapse
|
32
|
Song J, Gao H, Zhang H, George OJ, Hillman AS, Fox JM, Jia X. Matrix Adhesiveness Regulates Myofibroblast Differentiation from Vocal Fold Fibroblasts in a Bio-orthogonally Cross-linked Hydrogel. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51669-51682. [PMID: 36367478 PMCID: PMC10350853 DOI: 10.1021/acsami.2c13852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Repeated mechanical and chemical insults cause an irreversible alteration of extracellular matrix (ECM) composition and properties, giving rise to vocal fold scarring that is refractory to treatment. Although it is well known that fibroblast activation to myofibroblast is the key to the development of the pathology, the lack of a physiologically relevant in vitro model of vocal folds impedes mechanistic investigations on how ECM cues promote myofibroblast differentiation. Herein, we describe a bio-orthogonally cross-linked hydrogel platform that recapitulates the alteration of matrix adhesiveness due to enhanced fibronectin deposition when vocal fold wound healing is initiated. The synthetic ECM (sECM) was established via the cycloaddition reaction of tetrazine (Tz) with slow (norbornene, Nb)- and fast (trans-cyclooctene, TCO)-reacting dienophiles. The relatively slow Tz-Nb ligation allowed the establishment of the covalent hydrogel network for 3D cell encapsulation, while the rapid and efficient Tz-TCO reaction enabled precise conjugation of the cell-adhesive RGDSP peptide in the hydrogel network. To mimic the dynamic changes of ECM composition during wound healing, RGDSP was conjugated to cell-laden hydrogel constructs via a diffusion-controlled bioorthognal ligation method 3 days post encapsulation. At a low RGDSP concentration (0.2 mM), fibroblasts residing in the hydrogel remained quiescent when maintained in transforming growth factor beta 1 (TGF-β1)-conditioned media. However, at a high concentration (2 mM), RGDSP potentiated TGF-β1-induced myofibroblast differentiation, as evidenced by the formation of an actin cytoskeleton network, including F-actin and alpha-smooth muscle actin. The RGDSP-driven fibroblast activation to myofibroblast was accompanied with an increase in the expression of wound healing-related genes, the secretion of profibrotic cytokines, and matrix contraction required for tissue remodeling. This work represents the first step toward the establishment of a 3D hydrogel-based cellular model for studying myofibroblast differentiation in a defined niche associated with vocal fold scarring.
Collapse
Affiliation(s)
- Jiyeon Song
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - Hanyuan Gao
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - He Zhang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - Olivia J. George
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - Ashlyn S. Hillman
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, USA
| | - Joseph. M. Fox
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, Delaware, USA
| |
Collapse
|
33
|
Vijayan AN, Solaimuthu A, Murali P, Gopi J, Y MT, R AP, Korrapati PS. Decorin mediated biomimetic PCL-gelatin nano-framework to impede scarring. Int J Biol Macromol 2022; 219:907-918. [PMID: 35952816 DOI: 10.1016/j.ijbiomac.2022.08.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/05/2022]
Abstract
Scars occur as a result of fibrosis after tissue damage or surgery and reports suggest that excessive Transforming growth factor-β (TGF-β) activity during the process of wound healing leads to progressive fibrosis. Decorin is an extracellular matrix (ECM) protein which regulates collagen fibrillogenesis. However, targeted delivery and effective protein therapy remains a challenge owing to degradation byproteases. Hence, we aimed to deliver Decorin in a sustainable mode for the reduction of TGF-β levels and subsequent scar formation. Herein, we have fabricated PCL-Gelatin bio-mimetic scaffolds to optimize the bio-activity and provide localized delivery of recombinant Decorin. The degradation and drug release patterns reveals that this biomaterial is biodegradable and offers sustained release of the recombinant Decorin. Decorin loaded nanofiber displayed lower adhesion and proliferation rates in in-vitro conditions. Moreover, Decorin loaded scaffolds demonstrated morphological changes in cells, specifically targeting the myofibroblast. The expression of TGF-β was also scrutinized to understand the effect of Decorin loaded nanofibers. Besides, in the in-vitro fibrotic model, Decorin loaded nanofibers efficiently reduced the expression of ECM related proteins. Therefore, we report the sustained delivery of the recombinant Decorin from nanofiber dressing to potentially obstruct scar formation during the process of wound healing.
Collapse
Affiliation(s)
- Ane Nishitha Vijayan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anbuthiruselvan Solaimuthu
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Padmaja Murali
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Janani Gopi
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Madhan Teja Y
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Akshaya Priya R
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Purna Sai Korrapati
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
34
|
Zhang Z, Zhang W, Blakes R, Sundby LJ, Shi Z, Rockey DC, Ervasti JM, Nam YJ. Fibroblast fate determination during cardiac reprogramming by remodeling of actin filaments. Stem Cell Reports 2022; 17:1604-1619. [PMID: 35688153 PMCID: PMC9287671 DOI: 10.1016/j.stemcr.2022.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022] Open
Abstract
Fibroblasts can be reprogrammed into induced cardiomyocyte-like cells (iCMs) by forced expression of cardiogenic transcription factors. However, it remains unknown how fibroblasts adopt a cardiomyocyte (CM) fate during their spontaneous ongoing transdifferentiation toward myofibroblasts (MFs). By tracing fibroblast lineages following cardiac reprogramming in vitro, we found that most mature iCMs are derived directly from fibroblasts without transition through the MF state. This direct conversion is attributable to mutually exclusive induction of cardiac sarcomeres and MF cytoskeletal structures in the cytoplasm of fibroblasts during reprogramming. For direct fate switch from fibroblasts to iCMs, significant remodeling of actin isoforms occurs in fibroblasts, including induction of α-cardiac actin and decrease of the actin isoforms predominant in MFs. Accordingly, genetic or pharmacological ablation of MF-enriched actin isoforms significantly enhances cardiac reprogramming. Our results demonstrate that remodeling of actin isoforms is required for fibroblast to CM fate conversion by cardiac reprogramming.
Collapse
Affiliation(s)
- Zhentao Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Wenhui Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert Blakes
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Lauren J Sundby
- Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Zengdun Shi
- Department of Internal Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Don C Rockey
- Department of Internal Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Young-Jae Nam
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
35
|
Li LX, Agborbesong E, Zhang L, Zhang X, Zhou JX, Li X. Crosstalk between lysine methyltransferase Smyd2 and TGF-β-Smad3 signaling promotes renal fibrosis in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2022; 323:F227-F242. [PMID: 35759739 PMCID: PMC9359663 DOI: 10.1152/ajprenal.00452.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited genetic disorder, which is caused by mutations of PKD1 or PKD2 gene and is characterized by renal fluid-filled cyst formation and interstitial fibrosis. PKD1 gene mutation results in the upregulation of SET and MYND domain-containing lysine methyltransferase 2 (SMYD2) in Pkd1 mutant mouse and ADPKD patient kidneys. However, the role and mechanism of Smyd2 in the regulation of renal fibrosis in ADPKD remains elusive. In this study, we show that: 1) the expression of Smyd2 can be regulated by TGF-β-Smad3 in normal rat kidney 49F (NRK-49F) cells and mouse fibroblast NIH3T3 cells; 2) knockdown of Smyd2 and inhibition of Smyd2 with its specific inhibitor, AZ505, decreases TGF-β-induced expression of α-smooth muscle actin (α-SMA), fibronectin, collagens 1 and 3 and plasminogen activator inhibitor-1( PAI1) in NRK-49F cells; 3) Smyd2 regulates the transcription of fibrotic marker genes through binding on the promoters of those genes or through methylating histone H3 to indirectly regulate the expression of those genes; and 4) knockout and inhibition of Smyd2 significantly decreases renal fibrosis in Pkd1 knockout mice, supporting that targeting Smyd2 can not only delay cyst growth but also attenuate renal fibrosis in ADPKD. This study identifies a crosstalk between TGF-β signaling and Smyd2 in the regulation of fibrotic gene transcription and the activation of fibroblasts in cystic kidneys, suggesting that targeting Smyd2 with AZ505 is a potential therapeutic strategy for ADPKD treatment.
Collapse
Affiliation(s)
- Linda Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Lu Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Xiaoqin Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
36
|
Nho RS, Ballinger MN, Rojas MM, Ghadiali SN, Horowitz JC. Biomechanical Force and Cellular Stiffness in Lung Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:750-761. [PMID: 35183510 PMCID: PMC9088200 DOI: 10.1016/j.ajpath.2022.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/25/2022] [Accepted: 02/08/2022] [Indexed: 01/10/2023]
Abstract
Lung fibrosis is characterized by the continuous accumulation of extracellular matrix (ECM) proteins produced by apoptosis-resistant (myo)fibroblasts. Lung epithelial injury promotes the recruitment and activation of fibroblasts, which are necessary for tissue repair and restoration of homeostasis. However, under pathologic conditions, a vicious cycle generated by profibrotic growth factors/cytokines, multicellular interactions, and matrix-associated signaling propagates the wound repair response and promotes lung fibrosis characterized not only by increased quantities of ECM proteins but also by changes in the biomechanical properties of the matrix. Importantly, changes in the biochemical and biomechanical properties of the matrix itself can serve to perpetuate fibroblast activity and propagate fibrosis, even in the absence of the initial stimulus of injury. The development of novel experimental models and methods increasingly facilitates our ability to interrogate fibrotic processes at the cellular and molecular levels. The goal of this review is to discuss the impact of ECM conditions in the development of lung fibrosis and to introduce new approaches to more accurately model the in vivo fibrotic microenvironment. This article highlights the pathologic roles of ECM in terms of mechanical force and the cellular interactions while reviewing in vitro and ex vivo models of lung fibrosis. The improved understanding of the fundamental mechanisms that contribute to lung fibrosis holds promise for identification of new therapeutic targets and improved outcomes.
Collapse
Affiliation(s)
- Richard S Nho
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| | - Megan N Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Mauricio M Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Samir N Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Jeffrey C Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
37
|
Choo YY, Sakai T, Komatsu S, Ikebe R, Jeffers A, Singh KP, Idell S, Tucker TA, Ikebe M. Calponin 1 contributes to myofibroblast differentiation of human pleural mesothelial cells. Am J Physiol Lung Cell Mol Physiol 2022; 322:L348-L364. [PMID: 35018804 PMCID: PMC8858681 DOI: 10.1152/ajplung.00289.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/02/2022] [Accepted: 01/03/2022] [Indexed: 11/22/2022] Open
Abstract
Pleural mesothelial cells (PMCs) can become myofibroblasts via mesothelial-mesenchymal transition (MesoMT) and contribute to pleural organization, fibrosis, and rind formation. However, how these transformed mesothelial cells contribute to lung fibrosis remains unclear. Here, we investigated the mechanism of contractile myofibroblast differentiation of PMCs. Transforming growth factor-β (TGF-β) induced marked upregulation of calponin 1 expression, which was correlated with notable cytoskeletal rearrangement in human PMCs (HPMCs) to produce stress fibers. Downregulation of calponin 1 expression reduced stress fiber formation. Interestingly, induced stress fibers predominantly contain α-smooth muscle actin (αSMA) associated with calponin 1 but not β-actin. Calponin 1-associated stress fibers also contained myosin II and α-actinin. Furthermore, focal adhesions were aligned with the produced stress fibers. These results suggest that calponin 1 facilitates formation of stress fibers that resemble contractile myofibrils. Supporting this notion, TGF-β significantly increased the contractile activity of HPMCs, an effect that was abolished by downregulation of calponin 1 expression. We infer that differentiation of HPMCs to contractile myofibroblasts facilitates stiffness of scar tissue in pleura to promote pleural fibrosis (PF) and that upregulation of calponin 1 plays a central role in this process.
Collapse
Affiliation(s)
- Young-Yeon Choo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Satoshi Komatsu
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Karan P Singh
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| |
Collapse
|
38
|
Halim A, Narayanan G, Hato T, Ho L, Wan D, Siedlecki AM, Rhee EP, Allegretti AS, Nigwekar SU, Zehnder D, Hiemstra TF, Bonventre JV, Charytan DM, Kalim S, Thadhani R, Lu T, Lim K. Myocardial Cytoskeletal Adaptations in Advanced Kidney Disease. J Am Heart Assoc 2022; 11:e022991. [PMID: 35179046 PMCID: PMC9075094 DOI: 10.1161/jaha.121.022991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/24/2021] [Indexed: 11/18/2022]
Abstract
Background The myocardial cytoskeleton functions as the fundamental framework critical for organelle function, bioenergetics and myocardial remodeling. To date, impairment of the myocardial cytoskeleton occurring in the failing heart in patients with advanced chronic kidney disease has been largely undescribed. Methods and Results We conducted a 3-arm cross-sectional cohort study of explanted human heart tissues from patients who are dependent on hemodialysis (n=19), hypertension (n=10) with preserved renal function, and healthy controls (n=21). Left ventricular tissues were subjected to pathologic examination and next-generation RNA sequencing. Mechanistic and interference RNA studies utilizing in vitro human cardiac fibroblast models were performed. Left ventricular tissues from patients undergoing hemodialysis exhibited increased myocardial wall thickness and significantly greater fibrosis compared with hypertension patients (P<0.05) and control (P<0.01). Transcriptomic analysis revealed that the focal adhesion pathway was significantly enriched in hearts from patients undergoing hemodialysis. Hearts from patients undergoing hemodialysis exhibited dysregulated components of the focal adhesion pathway including reduced β-actin (P<0.01), β-tubulin (P<0.01), vimentin (P<0.05), and increased expression of vinculin (P<0.05) compared with controls. Cytoskeletal adaptations in hearts from the hemodialysis group were associated with impaired mitochondrial bioenergetics, including dysregulated mitochondrial dynamics and fusion, and loss of cell survival pathways. Mechanistic studies revealed that cytoskeletal changes can be driven by uremic and metabolic abnormalities of chronic kidney disease, in vitro. Furthermore, focal adhesion kinase silencing via interference RNA suppressed major cytoskeletal proteins synergistically with mineral stressors found in chronic kidney disease in vitro. Conclusions Myocardial failure in advanced chronic kidney disease is characterized by impairment of the cytoskeleton involving disruption of the focal adhesion pathway, mitochondrial failure, and loss of cell survival pathways.
Collapse
Affiliation(s)
- Arvin Halim
- Division of Nephrology and HypertensionIndiana University School of MedicineIndianapolisIN
| | - Gayatri Narayanan
- Division of Nephrology and HypertensionIndiana University School of MedicineIndianapolisIN
| | - Takashi Hato
- Division of Nephrology and HypertensionIndiana University School of MedicineIndianapolisIN
| | - Lilun Ho
- Department of Computer Science, Computer Science and Artificial Intelligence LaboratoryMassachusetts Institute of TechnologyCambridgeMA
| | - Douglas Wan
- Division of CardiologyUniversity of Toronto and Sunnybrook Health Sciences CentreTorontoCanada
| | | | - Eugene P. Rhee
- Division of Nephrology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Andrew S. Allegretti
- Division of Nephrology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Sagar U. Nigwekar
- Division of Nephrology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | - Daniel Zehnder
- Department of Nephrology and Department of Acute MedicineNorth Cumbria University Hospital NHS TrustCarlisleUnited Kingdom
| | - Thomas F. Hiemstra
- Cambridge Clinical Trials Unit and School of Clinical MedicineUniversity of CambridgeUnited Kingdom
| | | | - David M. Charytan
- Division of NephrologyNew York University School of MedicineNew YorkNY
| | - Sahir Kalim
- Division of Nephrology, Department of MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMA
| | | | - Tzongshi Lu
- Renal Division, Brigham and Women’s HospitalHarvard Medical SchoolBostonMA
| | - Kenneth Lim
- Division of Nephrology and HypertensionIndiana University School of MedicineIndianapolisIN
| |
Collapse
|
39
|
Wegner E, Slotina E, Mickan T, Truffel S, Arand C, Wagner D, Ritz U, Rommens PM, Gercek E, Drees P, Baranowski A. Pleiotropic Long-Term Effects of Atorvastatin on Posttraumatic Joint Contracture in a Rat Model. Pharmaceutics 2022; 14:pharmaceutics14030523. [PMID: 35335899 PMCID: PMC8950153 DOI: 10.3390/pharmaceutics14030523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/18/2022] [Accepted: 02/25/2022] [Indexed: 11/19/2022] Open
Abstract
The antifibrotic effect of atorvastatin has already been demonstrated in several organ systems. In the present study, a rat model was used to investigate the effect of atorvastatin on posttraumatic joint contracture. Forty-eight Sprague Dawley rats were equally randomized into an atorvastatin group and a control group. After initial joint trauma, knee joints were immobilized for intervals of 2 weeks (n = 16) or 4 weeks (n = 16) or immobilized for 4 weeks with subsequent remobilization for another 4 weeks (n = 16). Starting from the day of surgery, animals received either atorvastatin or placebo daily. After euthanasia at week 2, 4 or 8, joint contracture was determined, histological examinations were performed, and gene expression was assessed. The results suggest that the joint contracture was primarily arthrogenic. Atorvastatin failed to significantly affect contracture formation and showed a reduction in myofibroblast numbers to 98 ± 58 (control: 319 ± 113, p < 0.01) and a reduction in joint capsule collagen to 60 ± 8% (control: 73 ± 9%, p < 0.05) at week 2. Gene expression of α-smooth muscle actin (α-SMA), collagen type I, transforming growth factor β1 (TGF-β1) and interleukin-6 (IL-6) was not significantly affected by atorvastatin. Atorvastatin decreases myofibroblast number and collagen deposition but does not result in an improvement in joint mobility.
Collapse
|
40
|
Jia B, Yu S, Yu D, Liu N, Zhang S, Wu A. Mycotoxin deoxynivalenol affects myoblast differentiation via downregulating cytoskeleton and ECM-integrin-FAK-RAC-PAK signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112850. [PMID: 34607188 DOI: 10.1016/j.ecoenv.2021.112850] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 06/13/2023]
Abstract
As a common mycotoxin, deoxynivalenol (DON) contaminates cereal grains and feed in field or during processing and storage. DON elicits a spectrum of adverse effects in animals including anorexia and growth retardation. Especially, the presence of DON has also been detected in muscle, suggesting that DON may has the potential to affect the development of muscle. However, the relevant research is very rare and the molecular mechanism remains unclear. Myoblasts differentiation into multinucleated myotubes is one of the crucial steps of skeletal muscle development. In the present study, we investigated the effects of DON on differentiation of myoblasts using murine C2C12 cells model. The results indicated that DON dose-dependent inhibited the formation of myotubes in C2C12 cells. After performing omics techniques, a total of 149 differentially expressed genes were identified. The expression of cytoskeleton proteins and extracellular matrix (ECM) proteins were downregulated by DON. Furthermore, DON significantly downregulated the expression of integrin αv and integrin β5, leading to inhibition of the ECM-integrin receptor interaction. The focal adhesion kinase (FAK) and phosphorylated forms, ras-related C3 botulinum toxin substrate (RAC) and p21-activated kinases 1 (PAK1) were also downregulated by DON. Taken together, our findings suggest that DON has the potent to affect the differentiation of myoblasts via downregulating of cytoskeleton and ECM-integrin-FAK-RAC-PAK signaling pathway.
Collapse
Affiliation(s)
- Bingxuan Jia
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Song Yu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dianzhen Yu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Na Liu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuo Zhang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100021, China
| | - Aibo Wu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
41
|
Micus LC, Trautschold-Krause FS, Jelit AL, Schön MP, Lorenz VN. NF-кB c-Rel modulates pre-fibrotic changes in human fibroblasts. Arch Dermatol Res 2021; 314:943-951. [PMID: 34888734 PMCID: PMC9522690 DOI: 10.1007/s00403-021-02310-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/03/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022]
Abstract
Skin fibrosis is one central hallmark of the heterogeneous autoimmune disease systemic sclerosis. So far, there are hardly any standardized and effective treatment options. Pathogenic mechanisms underlying fibrosis comprise excessive and uncontrolled myofibroblast differentiation, increased extracellular matrix protein (ECM) synthesis and an intensification of the forces exerted by the cytoskeleton. A deeper understanding of fibroblast transformation could help to prevent or reverse fibrosis by specifically interfering with abnormally regulated signaling pathways. The transcription factor NF-κB has been implicated in the progression of fibrotic processes. However, the cellular processes regulated by NF-κB in fibrosis as well as the NF-κB isoforms preferentially involved are still completely unknown. In an in vitro model of fibrosis, we consistently observed the induction of the c-Rel subunit of NF-κB. Functional abrogation of c-Rel by siRNA resulted in diminished cell contractility of dermal fibroblasts in relaxed, but not in stressed 3D collagen matrices. Furthermore, directed migration was reduced after c-Rel silencing and total N-cadherin expression level was diminished, possibly mediating the observed cellular defects. Therefore, NF-кB c-Rel impacts central cellular adhesion markers and processes which negatively regulate fibrotic progression in SSc pathophysiology.
Collapse
Affiliation(s)
- Lara Carolina Micus
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen Lower Saxony, Robert Koch Str. 40, 37075, Göttingen, Germany
| | - Franziska Susanne Trautschold-Krause
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen Lower Saxony, Robert Koch Str. 40, 37075, Göttingen, Germany
| | - Anna Lena Jelit
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen Lower Saxony, Robert Koch Str. 40, 37075, Göttingen, Germany
| | - Michael Peter Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen Lower Saxony, Robert Koch Str. 40, 37075, Göttingen, Germany
| | - Verena Natalie Lorenz
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen Lower Saxony, Robert Koch Str. 40, 37075, Göttingen, Germany.
| |
Collapse
|
42
|
Martin M, Zhang J, Miao Y, He M, Kang J, Huang HY, Chou CH, Huang TS, Hong HC, Su SH, Wong SS, Harper RL, Wang L, Bhattacharjee R, Huang HD, Chen ZB, Malhotra A, Rabinovitch M, Hagood JS, Shyy JYJ. Role of endothelial cells in pulmonary fibrosis via SREBP2 activation. JCI Insight 2021; 6:125635. [PMID: 34806652 PMCID: PMC8663776 DOI: 10.1172/jci.insight.125635] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 10/06/2021] [Indexed: 01/22/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with limited treatment options. Despite endothelial cells (ECs) comprising 30% of the lung cellular composition, the role of EC dysfunction in pulmonary fibrosis (PF) remains unclear. We hypothesize that sterol regulatory element-binding protein 2 (SREBP2) plays a critical role in the pathogenesis of PF via EC phenotypic modifications. Transcriptome data demonstrate that SREBP2 overexpression in ECs led to the induction of the TGF, Wnt, and cytoskeleton remodeling gene ontology pathways and the increased expression of mesenchymal genes, such as snail family transcriptional repressor 1 (snai1), α-smooth muscle actin, vimentin, and neural cadherin. Furthermore, SREBP2 directly bound to the promoter regions and transactivated these mesenchymal genes. This transcriptomic change was associated with an epigenetic and phenotypic switch in ECs, leading to increased proliferation, stress fiber formation, and ECM deposition. Mice with endothelial-specific transgenic overexpression of SREBP2 (EC-SREBP2[N]-Tg mice) that were administered bleomycin to induce PF demonstrated exacerbated vascular remodeling and increased mesenchymal transition in the lung. SREBP2 was also found to be markedly increased in lung specimens from patients with IPF. These results suggest that SREBP2, induced by lung injury, can exacerbate PF in rodent models and in human patients with IPF.
Collapse
Affiliation(s)
- Marcy Martin
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA.,Vera Moulton Wall Center for Pulmonary Vascular Diseases.,Stanford Cardiovascular Institute, and.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Jiao Zhang
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Yifei Miao
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Ming He
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Jian Kang
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Hsi-Yuan Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China.,Warshel Institute for Computational Biology, and School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong Province, China
| | - Chih-Hung Chou
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Tse-Shun Huang
- Department of Bioengineering and Institute of Engineering in Medicine and
| | - Hsiao-Chin Hong
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Shu-Han Su
- Institute of Bioinformatics and Systems Biology, Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Simon S Wong
- Division of Respiratory Medicine, Department of Pediatrics, UCSD, La Jolla, California, USA
| | - Rebecca L Harper
- Vera Moulton Wall Center for Pulmonary Vascular Diseases.,Stanford Cardiovascular Institute, and.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Lingli Wang
- Vera Moulton Wall Center for Pulmonary Vascular Diseases.,Stanford Cardiovascular Institute, and.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Rakesh Bhattacharjee
- Division of Respiratory Medicine, Department of Pediatrics, UCSD, La Jolla, California, USA
| | - Hsien-Da Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Longgang District, Shenzhen, Guangdong Province, China.,Warshel Institute for Computational Biology, and School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong Province, China
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, Duarte, California, USA
| | - Atul Malhotra
- Division of Pulmonary and Critical Care Medicine, UCSD, La Jolla, California, USA
| | - Marlene Rabinovitch
- Vera Moulton Wall Center for Pulmonary Vascular Diseases.,Stanford Cardiovascular Institute, and.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - James S Hagood
- Division of Respiratory Medicine, Department of Pediatrics, UCSD, La Jolla, California, USA.,Division of Pulmonology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
43
|
Development and Characterization of Alkaline Phosphatase-Positive Human Umbilical Cord Perivascular Cells. Cells 2021; 10:cells10113011. [PMID: 34831233 PMCID: PMC8616437 DOI: 10.3390/cells10113011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/25/2022] Open
Abstract
Human umbilical cord perivascular cells (HUCPVCs), harvested from human umbilical cord perivascular tissue, show potential for future use as an alternative to mesenchymal stromal cells. Here, we present the results for the characterization of the properties alkaline phosphatase-positive HUCPVCs (ALP(+)-HUCPVCs). These ALP(+)-HUCPVCs were created from HUCPVCs in this study by culturing in the presence of activated vitamin D3, an inhibitor of bone morphogenetic protein signaling and transforming growth factor-beta1 (TGF-β1). The morphological characteristics, cell proliferation, gene expression, and mineralization-inducing ability of ALP(+)-HUCPVCs were investigated at the morphological, biological, and genetic levels. ALP(+)-HUCPVCs possess high ALP gene expression and activity in cells and a slow rate of cell growth. The morphology of ALP(+)-HUCPVCs is fibroblast-like, with an increase in actin filaments containing alpha-smooth muscle actin. In addition to ALP expression, the gene expression levels of type I collagen, osteopontin, elastin, fibrillin-1, and cluster of differentiation 90 are increased in ALP(+)-HUCPVCs. ALP(+)-HUCPVCs do not have the ability to induce mineralization nodules, which may be due to the restriction of phosphate uptake into matrix vesicles. Moreover, ALP(+)-HUCPVCs may produce anti-mineralization substances. We conclude that ALP(+)-HUCPVCs induced from HUCPVCs by a TGF-β1 stimulation possess myofibroblast-like properties that have little mineralization-inducing ability.
Collapse
|
44
|
Mendoza FA, Jimenez SA. Serine-Threonine Kinase inhibition as antifibrotic therapy: TGF-β and ROCK inhibitors. Rheumatology (Oxford) 2021; 61:1354-1365. [PMID: 34664623 DOI: 10.1093/rheumatology/keab762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/18/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Serine-threonine kinases mediate the phosphorylation of intracellular protein targets, transferring a phosphorus group from an ATP molecule to the specific amino acid residues within the target proteins. Serine-threonine kinases regulate multiple key cellular functions. From this large group of kinases, transforming growth factor beta (TGF-β) through the serine-threonine activity of its receptors and Rho kinase (ROCK) play an important role in the development and maintenance of fibrosis in various human diseases, including systemic sclerosis. In recent years, multiple drugs targeting and inhibiting these kinases, have been developed, opening the possibility of becoming potential antifibrotic agents of clinical value for treating fibrotic diseases. This review analyzes the contribution of TGF- β and ROCK-mediated serine-threonine kinase molecular pathways to the development and maintenance of pathological fibrosis and the potential clinical use of their inhibition.
Collapse
Affiliation(s)
- Fabian A Mendoza
- Division of Rheumatology, Department of Medicine. Thomas Jefferson University. Philadelphia, PA, USA 19107.,Jefferson Institute of Molecular Medicine and Scleroderma Center. Thomas Jefferson University. Philadelphia, PA, USA 19107
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center. Thomas Jefferson University. Philadelphia, PA, USA 19107
| |
Collapse
|
45
|
Yoodee S, Noonin C, Sueksakit K, Kanlaya R, Chaiyarit S, Peerapen P, Thongboonkerd V. Effects of secretome derived from macrophages exposed to calcium oxalate crystals on renal fibroblast activation. Commun Biol 2021; 4:959. [PMID: 34381146 PMCID: PMC8358035 DOI: 10.1038/s42003-021-02479-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
The association between kidney stone disease and renal fibrosis has been widely explored in recent years but its underlying mechanisms remain far from complete understanding. Using label-free quantitative proteomics (nanoLC-ESI-LTQ-Orbitrap MS/MS), this study identified 23 significantly altered secreted proteins from calcium oxalate monohydrate (COM)-exposed macrophages (COM-MP) compared with control macrophages (Ctrl-MP) secretome. Functional annotation and protein-protein interactions network analysis revealed that these altered secreted proteins were involved mainly in inflammatory response and fibroblast activation. BHK-21 renal fibroblasts treated with COM-MP secretome had more spindle-shaped morphology with greater spindle index. Immunofluorescence study and gelatin zymography revealed increased levels of fibroblast activation markers (α-smooth muscle actin and F-actin) and fibrotic factors (fibronectin and matrix metalloproteinase-9 and -2) in the COM-MP secretome-treated fibroblasts. Our findings indicate that proteins secreted from macrophages exposed to COM crystals induce renal fibroblast activation and may play important roles in renal fibrogenesis in kidney stone disease.
Collapse
Affiliation(s)
- Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chadanat Noonin
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanyarat Sueksakit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rattiyaporn Kanlaya
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sakdithep Chaiyarit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Paleerath Peerapen
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
46
|
Medication-Related Osteonecrosis of the Jaws Initiated by Zoledronic Acid and Potential Pathophysiology. Dent J (Basel) 2021; 9:dj9080085. [PMID: 34435997 PMCID: PMC8392270 DOI: 10.3390/dj9080085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
The aim of this systematic review is to present an up-to-date review of available publications investigating the cellular mechanisms initiating the development of medication-related osteonecrosis of the jaw caused by zoledronic acid. Electronic searches of MEDLINE/PubMed and Scopus were conducted on the 3 June 2019. A total of 804 publications were identified, of which 11 met the inclusion criteria and were, therefore, included in this study. All the included studies were in vitro studies investigating various human cells. The current review found that zoledronic acid in various concentrations increased apoptosis and decreased migration and proliferation of epithelial cells, fibroblasts, osteoblasts, endothelial cells and dental pulp stem cells, which can affect local tissue homeostasis. The consequences of zoledronic acid were found to be both time- and dose-dependent. The pathophysiology of medication-related osteonecrosis of the jaw is likely a multifactorial process involving prolonged wound healing, chronic inflammation and altered bone remodelling following the administration of zoledronic acid. Further research is needed to identify the exact pathophysiology to optimise management and treatment.
Collapse
|
47
|
Chavkin NW, Sano S, Wang Y, Oshima K, Ogawa H, Horitani K, Sano M, MacLauchlan S, Nelson A, Setia K, Vippa T, Watanabe Y, Saucerman JJ, Hirschi KK, Gokce N, Walsh K. The Cell Surface Receptors Ror1/2 Control Cardiac Myofibroblast Differentiation. J Am Heart Assoc 2021; 10:e019904. [PMID: 34155901 PMCID: PMC8403294 DOI: 10.1161/jaha.120.019904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/22/2021] [Indexed: 12/25/2022]
Abstract
Background A hallmark of heart failure is cardiac fibrosis, which results from the injury-induced differentiation response of resident fibroblasts to myofibroblasts that deposit extracellular matrix. During myofibroblast differentiation, fibroblasts progress through polarization stages of early proinflammation, intermediate proliferation, and late maturation, but the regulators of this progression are poorly understood. Planar cell polarity receptors, receptor tyrosine kinase-like orphan receptor 1 and 2 (Ror1/2), can function to promote cell differentiation and transformation. In this study, we investigated the role of the Ror1/2 in a model of heart failure with emphasis on myofibroblast differentiation. Methods and Results The role of Ror1/2 during cardiac myofibroblast differentiation was studied in cell culture models of primary murine cardiac fibroblast activation and in knockout mouse models that underwent transverse aortic constriction surgery to induce cardiac injury by pressure overload. Expression of Ror1 and Ror2 were robustly and exclusively induced in fibroblasts in hearts after transverse aortic constriction surgery, and both were rapidly upregulated after early activation of primary murine cardiac fibroblasts in culture. Cultured fibroblasts isolated from Ror1/2 knockout mice displayed a proinflammatory phenotype indicative of impaired myofibroblast differentiation. Although the combined ablation of Ror1/2 in mice did not result in a detectable baseline phenotype, transverse aortic constriction surgery led to the death of all mice by day 6 that was associated with myocardial hyperinflammation and vascular leakage. Conclusions Together, these results show that Ror1/2 are essential for the progression of myofibroblast differentiation and for the adaptive remodeling of the heart in response to pressure overload.
Collapse
Affiliation(s)
- Nicholas W. Chavkin
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Soichi Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Ying Wang
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Kosei Oshima
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Hayato Ogawa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Keita Horitani
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Miho Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Susan MacLauchlan
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Anders Nelson
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVA
| | - Karishma Setia
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Tanvi Vippa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Yosuke Watanabe
- Vascular Biology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Jeffrey J. Saucerman
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVA
| | - Karen K. Hirschi
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Cardiovascular Research CenterSchool of MedicineYale UniversityNew HavenCT
| | - Noyan Gokce
- Boston University School of MedicineBostonMA
| | - Kenneth Walsh
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| |
Collapse
|
48
|
Fernández Pérez ER, Harmacek LD, O'Connor BP, Danhorn T, Vestal B, Maier LA, Koelsch TL, Leach SM. Prognostic accuracy of a peripheral blood transcriptome signature in chronic hypersensitivity pneumonitis. Thorax 2021; 77:86-90. [PMID: 34183448 DOI: 10.1136/thoraxjnl-2020-214790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/14/2021] [Indexed: 11/04/2022]
Abstract
The prognostic value of peripheral blood mononuclear cell (PBMC) expression profiles, when used in patients with chronic hypersensitivity pneumonitis (CHP), as an adjunct to traditional clinical assessment is unknown. RNA-seq analysis on PBMC from 37 patients with CHP at initial presentation determined that (1) 74 differentially expressed transcripts at a 10% false discovery rate distinguished those with (n=10) and without (n=27) disease progression, defined as absolute FVC and/or diffusing capacity of the lungs for carbon monoxide (DLCO) decline of ≥10% and increased fibrosis on chest CT images within 24 months, and (2) classification models based on gene expression and clinical factors strongly outperform models based solely on clinical factors (baseline FVC%, DLCO% and chest CT fibrosis).
Collapse
Affiliation(s)
- Evans R Fernández Pérez
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Department of Medicine, Denver, Colorado, USA
| | - Laura D Harmacek
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Brian P O'Connor
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Thomas Danhorn
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, Auroa, Colorado, USA
| | - Brian Vestal
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Lisa A Maier
- Division of Occupational Health and Environmental Health Sciences, National Jewish Health Department of Medicine, Denver, Colorado, USA
| | - Tilman L Koelsch
- Thoracic Radiology, National Jewish Health, Denver, Colorado, USA
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
49
|
Dissecting the Involvement of Ras GTPases in Kidney Fibrosis. Genes (Basel) 2021; 12:genes12060800. [PMID: 34073961 PMCID: PMC8225075 DOI: 10.3390/genes12060800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
Many different regulatory mechanisms of renal fibrosis are known to date, and those related to transforming growth factor-β1 (TGF-β1)-induced signaling have been studied in greater depth. However, in recent years, other signaling pathways have been identified, which contribute to the regulation of these pathological processes. Several studies by our team and others have revealed the involvement of small Ras GTPases in the regulation of the cellular processes that occur in renal fibrosis, such as the activation and proliferation of myofibroblasts or the accumulation of extracellular matrix (ECM) proteins. Intracellular signaling mediated by TGF-β1 and Ras GTPases are closely related, and this interaction also occurs during the development of renal fibrosis. In this review, we update the available in vitro and in vivo knowledge on the role of Ras and its main effectors, such as Erk and Akt, in the cellular mechanisms that occur during the regulation of kidney fibrosis (ECM synthesis, accumulation and activation of myofibroblasts, apoptosis and survival of tubular epithelial cells), as well as the therapeutic strategies for targeting the Ras pathway to intervene on the development of renal fibrosis.
Collapse
|
50
|
González-Pérez M, Camasão DB, Mantovani D, Alonso M, Rodríguez-Cabello JC. Biocasting of an elastin-like recombinamer and collagen bi-layered model of the tunica adventitia and external elastic lamina of the vascular wall. Biomater Sci 2021; 9:3860-3874. [PMID: 33890956 DOI: 10.1039/d0bm02197k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of techniques for fabricating vascular wall models will foster the development of preventive and therapeutic therapies for treating cardiovascular diseases. However, the physical and biological complexity of vascular tissue represents a major challenge, especially for the design and the production of off-the-shelf biomimetic vascular replicas. Herein, we report the development of a biocasting technique that can be used to replicate the tunica adventitia and the external elastic lamina of the vascular wall. Type I collagen embedded with neonatal human dermal fibroblast (HDFn) and an elastic click cross-linkable, cell-adhesive and protease-sensitive elastin-like recombinamer (ELR) hydrogel were investigated as readily accessible and tunable layers to the envisaged model. Mechanical characterization confirmed that the viscous and elastic attributes predominated in the collagen and ELR layers, respectively. In vitro maturation confirmed that the collagen and ELR provided a favorable environment for the HDFn viability, while histology revealed the wavy and homogenous morphology of the ELR and collagen layer respectively, the cell polarization towards the cell-attachment sites encoded on the ELR, and the enhanced expression of glycosaminoglycan-rich extracellular matrix and differentiation of the embedded HDFn into myofibroblasts. As a complementary assay, 30% by weight of the collagen layer was substituted with the ELR. This model proved the possibility to tune the composition and confirm the versatile character of the technology developed, while revealing no significant differences with respect to the original construct. On-demand modification of the model dimensions, number and composition of the layers, as well as the type and density of the seeded cells, can be further envisioned, thus suggesting that this bi-layered model may be a promising platform for the fabrication of biomimetic vascular wall models.
Collapse
Affiliation(s)
- Miguel González-Pérez
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| | - Dimitria Bonizol Camasão
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC, Canada G1V 0A6
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Québec, Division of Regenerative Medicine, Laval University, Québec, QC, Canada G1V 0A6
| | - Matilde Alonso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| | - José Carlos Rodríguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, 47011 Valladolid, Spain.
| |
Collapse
|