1
|
Allen RS, Seifert AW. Spiny mice (Acomys) have evolved cellular features to support regenerative healing. Ann N Y Acad Sci 2025; 1544:5-26. [PMID: 39805008 PMCID: PMC11830558 DOI: 10.1111/nyas.15281] [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] [Indexed: 01/16/2025]
Abstract
Spiny mice (Acomys spp.) are warm-blooded (homeothermic) vertebrates whose ability to restore missing tissue through regenerative healing has coincided with the evolution of unique cellular and physiological adaptations across different tissue types. This review seeks to explore how these bizarre rodents deploy unique or altered injury response mechanisms to either enhance tissue repair or fully regenerate excised tissue compared to closely related, scar-forming mammals. First, we examine overall trends in healing Acomys tissues, including the cellular stress response, the ability to activate and maintain cell cycle progression, and the expression of certain features in reproductive adults that are normally associated with embryos. Second, we focus on specific cell types that exhibit precisely regulated proliferation to restore missing tissue. While Acomys utilize many of the same cell types involved in scar formation, these cells exhibit divergent activation profiles during regenerative healing. Considered together, current lines of evidence support sustained deployment of proregenerative pathways in conjunction with transient activation of fibrotic pathways to facilitate regeneration and improve tissue repair in Acomys.
Collapse
Affiliation(s)
- Robyn S. Allen
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
- The Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, USA
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
2
|
Weiss D, Yeung N, Ramachandra AB, Humphrey JD. Transcriptional regulation of postnatal aortic development. Cells Dev 2024; 180:203971. [PMID: 39426523 PMCID: PMC11634634 DOI: 10.1016/j.cdev.2024.203971] [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: 06/24/2024] [Revised: 08/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
The aorta exhibits tremendous changes in geometry, composition, and mechanical properties during postnatal development. These changes are necessarily driven by transcriptional changes, both genetically programmed and mechano-responsive, but there has not been a careful comparison of time-course changes in the transcriptional profile and biomechanical phenotype. Here, we show that the greatest period of differential gene expression in the normal postnatal mouse aorta occurs prior to weaning at three weeks of age though with important evolution of many transcripts thereafter. We identify six general temporal patterns, including transcripts that monotonically decrease to lower or increase to higher steady state values as well as those that either peak or dip prior to or near weaning. We show that diverse transcripts within individual groupings correlate well over time, and that sub-sets of these groups correlate well with the developmental progression of different biomechanical metrics that are expected to be involved in mechano-sensing. In particular, expression of genes for elastin and elastin-associated glycoproteins tend to correlate well with the ratio of systolic-to-diastolic stress whereas genes for collagen fibers correlate well with the daily rate of change of systolic stress and genes for mechano-sensing proteins tend to correlate well with the systolic stress itself. We conclude that different groupings of genes having different temporal expression patterns correlate well with different measures of the wall mechanics, hence emphasizing a need for age-dependent, gene-specific computational modeling of postnatal development.
Collapse
Affiliation(s)
- D Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical & Materials Engineering, University of Denver, Denver, CO, USA
| | - N Yeung
- School of the Biological Sciences, University of Cambridge, Cambridge, UK
| | - A B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Department of Mechanical Engineering, Iowa State University, Ames, IA, USA
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Eun K, Kim AY, Ryu S. Matricellular proteins in immunometabolism and tissue homeostasis. BMB Rep 2024; 57:400-416. [PMID: 38919018 PMCID: PMC11444987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/11/2023] [Accepted: 04/25/2024] [Indexed: 06/27/2024] Open
Abstract
Matricellular proteins are integral non-structural components of the extracellular matrix. They serve as essential modulators of immunometabolism and tissue homeostasis, playing critical roles in physiological and pathological conditions. These extracellular matrix proteins including thrombospondins, osteopontin, tenascins, the secreted protein acidic and rich in cysteine (SPARC) family, the Cyr61, CTGF, NOV (CCN) family, and fibulins have multi-faceted functions in regulating immune cell functions, metabolic pathways, and tissue homeostasis. They are involved in immune-metabolic regulation and influence processes such as insulin signaling, adipogenesis, lipid metabolism, and immune cell function, playing significant roles in metabolic disorders such as obesity and diabetes. Furthermore, their modulation of tissue homeostasis processes including cellular adhesion, differentiation, migration, repair, and regeneration is instrumental for maintaining tissue integrity and function. The importance of these proteins in maintaining physiological equilibrium is underscored by the fact that alterations in their expression or function often coincide with disease manifestation. This review contributes to our growing understanding of these proteins, their mechanisms, and their potential therapeutic applications. [BMB Reports 2024; 57(9): 400-416].
Collapse
Affiliation(s)
- Kyoungjun Eun
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 06974, Korea
| | - Ah Young Kim
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 06974, Korea
| | - Seungjin Ryu
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Institute of Natural Medicine, College of Medicine, Hallym Unviersity, Chuncheon 24252, Korea
- Department of Biochemistry, Chung-Ang University College of Medicine, Seoul 06974, Korea
| |
Collapse
|
4
|
Bariani MV, Grimm SL, Coarfa C, Velez Edwards DR, Yang Q, Walker CL, Ali M, Al-Hendy A. Altered extracellular matrix-related pathways accelerate the transition from normal to prefibroid myometrium in Black women. Am J Obstet Gynecol 2024; 231:324.e1-324.e12. [PMID: 38825029 PMCID: PMC11344675 DOI: 10.1016/j.ajog.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Black women experience a disproportionate impact of uterine fibroids compared to White women, including earlier diagnosis, higher frequency, and more severe symptoms. The etiology underlying this racial disparity remains elusive. OBJECTIVE The aim of this study was to evaluate the molecular differences in normal myometrium (fibroid-free uteri) and at-risk myometrium (fibroid-containing uteri) tissues in Black and White women. STUDY DESIGN We conducted whole-genome RNA-seq on normal and at-risk myometrium tissues obtained from both self-identified Black and White women (not Hispanic or Latino) to determine global gene expression profiles and to conduct enriched pathway analyses (n=3 per group). We initially assessed the differences within the same type of tissue (normal or at-risk myometrium) between races. Subsequently, we analyzed the transcriptome of normal myometrium compared to at-risk myometrium in each race and determined the differences between them. We validated our findings through real-time PCR (sample size range=5-12), western blot (sample size range=5-6), and immunohistochemistry techniques (sample size range=9-16). RESULTS The transcriptomic analysis revealed distinct profiles between Black and White women in normal and at-risk myometrium tissues. Interestingly, genes and pathways related to extracellular matrix and mechanosensing were more enriched in normal myometrium from Black than White women. Transcription factor enrichment analysis detected greater activity of the serum response transcription factor positional motif in normal myometrium from Black compared to White women. Furthermore, we observed increased expression levels of myocardin-related transcription factor-serum response factor and the serum response factor in the same comparison. In addition, we noted increased expression of both mRNA and protein levels of vinculin, a target gene of the serum response factor, in normal myometrium tissues from Black women as compared to White women. Importantly, the transcriptomic profile of normal to at-risk myometrium conversion differs between Black and White women. Specifically, we observed that extracellular matrix-related pathways are involved in the transition from normal to at-risk myometrium and that these processes are exacerbated in Black women. We found increased levels of Tenascin C, type I collagen alpha 1 chain, fibronectin, and phospho-p38 MAPK (Thr180/Tyr182, active) protein levels in at-risk over normal myometrium tissues from Black women, whereas such differences were not observed in samples from White women. CONCLUSION These findings indicate that the racial disparities in uterine fibroids may be attributed to heightened production of extracellular matrix in the myometrium in Black women, even before the tumors appear. Future research is needed to understand early life determinants of the observed racial differences.
Collapse
Affiliation(s)
| | - Sandra L Grimm
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX; Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX
| | - Cristian Coarfa
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, TX; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX; Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN; Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN; Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL
| | - Cheryl L Walker
- Center for Precision and Environmental Health, Baylor College of Medicine, Houston, TX
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL.
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL.
| |
Collapse
|
5
|
Wu H, Yang M, Yan C, Liu M, Wang H, Zhang W. Tenascin C activates the toll‑like receptor 4/NF‑κB signaling pathway to promote the development of polycystic ovary syndrome. Mol Med Rep 2024; 29:106. [PMID: 38666538 PMCID: PMC11082635 DOI: 10.3892/mmr.2024.13230] [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: 11/23/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a globally prevalent gynecological disorder among women of childbearing age. The present study aimed to investigate the role of tenascin C (TNC) in PCOS and its potential mechanisms. Fasting blood glucose and serum insulin, the homeostasis model assessment of insulin resistance and the serum hormone levels were determined in PCOS rats. In addition, H&E staining was used for assessing pathology. In addition, the effects of TNC on oxidative stress and inflammation response in PCOS rat and cell models was assessed. Furthermore, the roles of TNC on KGN cell proliferation and apoptosis were determined employing EdU assay and flow cytometry. TLR4/NF‑κB pathway‑related proteins were measured using western blotting, immunofluorescence and immunohistochemistry. It was found that the mRNA and protein expression was upregulated in PCOS rats and in KGN cells induced by dihydrotestosterone (DHT). Knockdown of TNC relieved the pathological characteristics and the endocrine abnormalities of PCOS rats. Knockdown of TNC inhibited ovarian cell apoptosis, oxidative stress and inflammation in PCOS rats. Knockdown of TNC reversed the DHT‑induced reduction in cell proliferation and increase in apoptosis in KGN cells. Furthermore, knockdown of TNC alleviated oxidative stress and inflammatory responses induced by DHT in KGN cells. Additionally, knockdown of TNC inhibited the toll‑like receptor 4 (TLR4)/NF‑κB signaling pathway in PCOS rats and DHT‑treated KGN cells. In conclusion, knockdown of TNC could ameliorate PCOS in both rats and a cell model by inhibiting cell apoptosis, oxidative stress and inflammation via the suppression of the TLR4/NF‑κB signaling pathway.
Collapse
Affiliation(s)
- Han Wu
- Center for Reproductive Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| | - Mo Yang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| | - Cuiping Yan
- Department of Women's Health Care, Taian Daiyue District Maternal and Child Health Care, Taian, Shandong 271021, P.R. China
| | - Mengchen Liu
- Center for Reproductive Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| | - Haoran Wang
- Center for Reproductive Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| | - Wenjuan Zhang
- Center for Reproductive Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong 271000, P.R. China
| |
Collapse
|
6
|
Kimura Y, Nakai Y, Ino Y, Akiyama T, Moriyama K, Ohira T, Saito T, Inaba Y, Kumagai K, Ryo A, Hirano H. Identification of gravity-responsive serum proteins in spaceflight mice using a quantitative proteomic approach with data-independent acquisition mass spectrometry. Proteomics 2024; 24:e2300214. [PMID: 38475964 DOI: 10.1002/pmic.202300214] [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: 05/09/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Physical inactivity associated with gravity unloading, such as microgravity during spaceflight and hindlimb unloading (HU), can cause various physiological changes. In this study, we attempted to identify serum proteins whose levels fluctuated in response to gravity unloading. First, we quantitatively assessed changes in the serum proteome profiles of spaceflight mice using mass spectrometry with data-independent acquisition. The serum levels of several proteins involved in the responses to estrogen and glucocorticoid, blood vessel maturation, osteoblast differentiation, and ossification were changed by microgravity exposure. Furthermore, a collective evaluation of serum proteomic data from spaceflight and HU mice identified 30 serum proteins, including Mmp2, Igfbp2, Tnc, Cdh5, and Pmel, whose levels varied to a similar extent in both gravity unloading models. These changes in serum levels could be involved in the physiological changes induced by gravity unloading. A collective evaluation of serum, femur, and soleus muscle proteome data of spaceflight mice also showed 24 serum proteins, including Igfbp5, Igfbp3, and Postn, whose levels could be associated with biological changes induced by microgravity. This study examined serum proteome profiles in response to gravity unloading, and may help deepen our understanding of microgravity adaptation mechanisms during prolonged spaceflight missions.
Collapse
Affiliation(s)
- Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yusuke Nakai
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yoko Ino
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Kayano Moriyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Takashi Ohira
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Yutaka Inaba
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Ken Kumagai
- Department of Orthopaedic Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Akihide Ryo
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Hisashi Hirano
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| |
Collapse
|
7
|
MA M, CHEN W, CAO HL, PAN J, ZHOU Q, TANG XL, WANG DJ. The diagnostic value of tenascin-C in acute aortic syndrome. J Geriatr Cardiol 2024; 21:359-368. [PMID: 38665282 PMCID: PMC11040054 DOI: 10.26599/1671-5411.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVES Misdiagnosis of acute aortic syndrome (AAS) significantly increases mortality. Tenascin-C (TN-C) is an extracellular matrix glycoprotein related to cardiovascular injury. The elevation of TN-C in AAS and whether it can discriminate sudden-onset of acute chest pain in Chinese remains unclear. METHODS We measured the plasma concentration of TN-C by ELISA in a cohort of 376 patients with chest or back pain. Measures to discriminate AAS from acute coronary syndrome (ACS) were compared and calculated. RESULTS From October 2016 to September 2021, 376 undiagnosed patients with chest or back pain were enrolled. 166 of them were finally diagnosed as AAS, 100 were ACS and 110 without cardiovascular diseases (NCV). TN-C was significantly elevated in AAS at 18.18 ng/mL (IQR: 13.10-27.68) compared with 7.51 ng/mL (IQR: 5.67-11.38) in ACS (P < 0.001) and 3.68 ng/mL (IQR: 2.50-5.29) in NCV (P < 0.001). There was no significant difference in TN-C level among the subtypes of AAS. Of the 166 AAS patients, the peaked level of TN-C was at acute stage (P = 0.012), then a slight of decrease was observed at subacute stage. The area under receiver operating characteristic curve for AAS patients versus NCV was 0.979 (95% CI: 0.964-0.994) for TN-C. At a cutoff level of 11.474 ng/mL, TN-C has a sensitivity of 76.0%, specificity of 85.5%, accuracy of 82.0%, positive predictive value (PPV) of 76.0%, negative predictive value (NPV) of 85.5%. Diagnostic performance of TN-C was superior to D-dimer and hs-cTnT. CONCLUSIONS The concentration of serum TN-C in AAS patients was significantly higher than that in ACS patients and NCV. TN-C could be a new biomarker to distinguish AAS patients in the early stage after symptoms onset from other pain diseases.
Collapse
Affiliation(s)
- Ming MA
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wei CHEN
- Department of Thoracic surgery, Kunshan Hospital of Traditional Chinese Medicine, Jiangsu, China
| | - Hai-Long CAO
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jun PAN
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing ZHOU
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin-Long TANG
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Dong-Jin WANG
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
- Department of Thoracic and Cardiovascular Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
8
|
De Magalhães CG, Cvekl A, Jaeger RG, Yan CYI. Lens Placode Modulates Extracellular Matrix Formation During Early Eye Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.30.569417. [PMID: 38076974 PMCID: PMC10705410 DOI: 10.1101/2023.11.30.569417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
The role extracellular matrix (ECM) in multiple events of morphogenesis has been well described, little is known about its specific role in early eye development. One of the first morphogenic events in lens development is placodal thickening, which converts the presumptive lens ectoderm from cuboidal to pseudostratified epithelium. This process occurs in the anterior pre-placodal ectoderm when the optic vesicle approaches the cephalic ectoderm. Since cells and ECM have a dynamic relationship of interdependence and modulation, we hypothesized that the ECM evolves with cell shape changes during lens placode formation. This study investigates changes in optic ECM including both protein distribution deposition, extracellular gelatinase activity and gene expression patterns during early optic development using chicken and mouse models. In particular, the expression of Timp2 , a metalloprotease inhibitor, corresponds with a decrease in gelatinase activity within the optic ECM. Furthermore, we demonstrate that optic ECM remodeling depends on BMP signaling in the placode. Together, our findings suggest that the lens placode plays an active role in remodeling the optic ECM during early eye development.
Collapse
|
9
|
Wagner R, Lieckfeldt P, Piyadasa H, Markel M, Riedel J, Stefanovici C, Peukert N, Patel D, Derraugh G, Min SAL, Gosemann JH, Deprest J, Pascoe CD, Tse WH, Lacher M, Mookherjee N, Keijzer R. Proteomic Profiling of Hypoplastic Lungs Suggests an Underlying Inflammatory Response in the Pathogenesis of Abnormal Lung Development in Congenital Diaphragmatic Hernia. Ann Surg 2023; 278:e411-e421. [PMID: 35920560 DOI: 10.1097/sla.0000000000005656] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The pathogenesis of lung hypoplasia in congenital diaphragmatic hernia (CDH), a common birth defect, is poorly understood. The diaphragmatic defect can be repaired surgically, but the abnormal lung development contributes to a high mortality in these patients. To understand the underlying pathobiology, we compared the proteomic profiles of fetal rat lungs at the alveolar stage (E21) that were either exposed to nitrofen in utero (CDH lungs, n=5) or exposed to vehicle only (non-CDH control lungs, n=5). Pathway analysis of proteomic datasets showed significant enrichment in inflammatory response proteins associated with cytokine signaling and Epstein Barr Virus in nitrofen CDH lungs. Among the 218 significantly altered proteins between CDH and non-CDH control lungs were Tenascin C, CREBBP, LYN, and STAT3. We showed that Tenascin C was decreased around the distal airway branches in nitrofen rat lungs and human CDH lungs, obtained from stillborn fetuses that did not receive pre- or postnatal treatment. In contrast, STAT3 was significantly increased in the airway epithelium of nitrofen lungs at E21. STAT3 inhibition after direct nitrofen exposure to fetal rat lung explants (E14.5) partially rescued the hypoplastic lung phenotype ex vivo by increasing peripheral lung budding. Moreover, we demonstrated that several STAT3-associated cytokines (IL-15, IL-9, andIL-2) are increased in fetal tracheal aspirates of CDH survivors compared with nonsurvivors after fetoscopic endoluminal tracheal occlusion. With our unbiased proteomics approach, we showed for the first time that downstream inflammatory processes are likely involved in the pathogenesis of abnormal lung development in CDH.
Collapse
Affiliation(s)
- Richard Wagner
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Paula Lieckfeldt
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Hadeesha Piyadasa
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Manitoba Center for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Moritz Markel
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Jan Riedel
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Camelia Stefanovici
- Department of Pathology, University of Manitoba, and Shared Services Manitoba, Winnipeg, MB, Canada
| | - Nicole Peukert
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Daywin Patel
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Gabrielle Derraugh
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Suyin A Lum Min
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
| | | | - Jan Deprest
- University Hospitals KU Leuven, Department of Development and Regeneration, Bio- medical Sciences, KU Leuven, Leuven, Belgium
| | - Christopher D Pascoe
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Wai Hei Tse
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Martin Lacher
- Department of Pediatric Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Neeloffer Mookherjee
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Manitoba Center for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health, University of Manitoba, Winnipeg, MB, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
10
|
Benn MC, Pot SA, Moeller J, Yamashita T, Fonta CM, Orend G, Kollmannsberger P, Vogel V. How the mechanobiology orchestrates the iterative and reciprocal ECM-cell cross-talk that drives microtissue growth. SCIENCE ADVANCES 2023; 9:eadd9275. [PMID: 36989370 PMCID: PMC10058249 DOI: 10.1126/sciadv.add9275] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Controlled tissue growth is essential for multicellular life and requires tight spatiotemporal control over cell proliferation and differentiation until reaching homeostasis. As cells synthesize and remodel extracellular matrix, tissue growth processes can only be understood if the reciprocal feedback between cells and their environment is revealed. Using de novo-grown microtissues, we identified crucial actors of the mechanoregulated events, which iteratively orchestrate a sharp transition from tissue growth to maturation, requiring a myofibroblast-to-fibroblast transition. Cellular decision-making occurs when fibronectin fiber tension switches from highly stretched to relaxed, and it requires the transiently up-regulated appearance of tenascin-C and tissue transglutaminase, matrix metalloprotease activity, as well as a switch from α5β1 to α2β1 integrin engagement and epidermal growth factor receptor signaling. As myofibroblasts are associated with wound healing and inflammatory or fibrotic diseases, crucial knowledge needed to advance regenerative strategies or to counter fibrosis and cancer progression has been gained.
Collapse
Affiliation(s)
- Mario C. Benn
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Simon A. Pot
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Tadahiro Yamashita
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Charlotte M. Fonta
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France
- Université Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Philip Kollmannsberger
- Biomedical Physics, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| |
Collapse
|
11
|
Humeres C, Venugopal H, Frangogiannis NG. The Role of Mechanosensitive Signaling Cascades in Repair and Fibrotic Remodeling of the Infarcted Heart. CARDIAC AND VASCULAR BIOLOGY 2023:61-100. [DOI: 10.1007/978-3-031-23965-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Song W, Qiu N. MiR-495-3p depletion contributes to myocardial ischemia/reperfusion injury in cardiomyocytes by targeting TNC. Regen Ther 2022; 21:380-388. [PMID: 36161101 PMCID: PMC9478495 DOI: 10.1016/j.reth.2022.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/08/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
Background Tenascin-C (TNC) has been found to abnormally express in myocardial ischemia/reperfusion injury (MI/RI), but its effect on cardiomyocytes apoptosis is unknown and is worthy of investigation. Methods H9C2 cells were given hypoxia/reoxygenation (H/R) treatment to obtain the replica of MI/RI in vitro. The effect of H/R on viability, apoptosis and inflammation was studied by CCK-8 assay, flow cytometry, mitochondrial membrane potential (MMP) and Ca2+ measurements as well as enzyme linked immunosorbent assay. We applied bioinformatics analysis and luciferase reporter assay to screened and validated TNC-targeting miR-495-3p which was then mechanistically investigated by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. With the assistance of cell transfection, rescue assays were conducted. Results H9C2 cells showed diminished viability, accelerated apoptosis, elevated tumour necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β), and TNC overexpression in response to H/R induction, while silencing of TNC partially reversed the effect of H/R treatment on the H9C2 cells. TNC silencing reduced Ca2+ level and enhanced MMP level in the H/R-stimulated cells. MiR-495-3p targeted TNC and showed a low expression in the H/R-stimulated cells. The expression of TNC was negatively regulated by miR-495-3p. Inhibition of miR-495-3p repressed viability and MMP level, and facilitated apoptosis and levels of Ca2+, TNF-α and IL-1β in the H/R-stimulated cells. The effect of TNC silencing and miR-495-3p depletion on H/R-induced cardiomyocyte injury was mutually reversed in vitro. Conclusion MiR-495-3p targeted TNC to regulate the apoptosis and inflammation of cardiomyocytes in H/R induction, which was associated with Ca2+ overload.
Collapse
Affiliation(s)
- Wei Song
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), 999 Donghai Avenue, Jiaojiang District, Taizhou, Zhejiang Province, China
| | - Naiyan Qiu
- Department of Cardiology, The Fifth People's Hospital of Jinan, No. 24297 Jingshi Road, Huaiyin District, Jinan, Shandong Province, 250021, China
| |
Collapse
|
13
|
Coutinho de Almeida R, Tuerlings M, Ramos Y, Den Hollander W, Suchiman E, Lakenberg N, Nelissen RGHH, Mei H, Meulenbelt I. Allelic expression imbalance in articular cartilage and subchondral bone refined genome-wide association signals in osteoarthritis. Rheumatology (Oxford) 2022; 62:1669-1676. [PMID: 36040165 PMCID: PMC10070069 DOI: 10.1093/rheumatology/keac498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES To present an unbiased approach to identify positional transcript single nucleotide polymorphisms (SNPs) of osteoarthritis (OA) risk loci by allelic expression imbalance (AEI) analyses using RNA sequencing of articular cartilage and subchondral bone from OA patients. METHODS RNA sequencing from 65 articular cartilage and 24 subchondral bone from OA patients was used for AEI analysis. AEI was determined for all genes present in the 100 regions reported by the GWAS catalog that were also expressed in cartilage or bone. The count fraction of the alternative allele (φ) was calculated for each heterozygous individual with the risk-SNP or with the SNP in linkage disequilibrium (LD) with it (r2 > 0.6). Furthermore, a meta-analysis was performed to generate a meta-φ (null hypothesis median φ = 0.49) and P-value for each SNP. RESULTS We identified 30 transcript SNPs (28 in cartilage and 2 in subchondral bone) subject to AEI in 29 genes. Notably, 10 transcript SNPs were located in genes not previously reported in the GWAS catalog, including two long intergenic non-coding RNAs (lincRNAs), MALAT1 (meta-φ = 0.54, FDR = 1.7x10-4) and ILF3-DT (meta-φ = 0.6, FDR = 1.75x10-5). Moreover, 12 drugs were interacting with 7 genes displaying AEI, of which 7 drugs have been already approved. CONCLUSIONS By prioritizing proxy transcript SNPs that mark AEI in cartilage and/or subchondral bone at loci harboring GWAS signals, we present an unbiased approach to identify the most likely functional OA risk-SNP and gene. We identified 10 new potential OA risk genes ready for further, translation towards underlying biological mechanisms.
Collapse
Affiliation(s)
- Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yolande Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter Den Hollander
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Eka Suchiman
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nico Lakenberg
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Dept. of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Mwase C, Phung TKN, O’Sullivan MJ, Mitchel JA, De Marzio M, Kılıç A, Weiss ST, Fredberg JJ, Park JA. Mechanical Compression of Human Airway Epithelial Cells Induces Release of Extracellular Vesicles Containing Tenascin C. Cells 2022; 11:cells11020256. [PMID: 35053372 PMCID: PMC8774246 DOI: 10.3390/cells11020256] [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: 12/14/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant remodeling of the asthmatic airway is not well understood but is thought to be attributable in part to mechanical compression of airway epithelial cells. Here, we examine compression-induced expression and secretion of the extracellular matrix protein tenascin C (TNC) from well-differentiated primary human bronchial epithelial (HBE) cells grown in an air-liquid interface culture. We measured TNC mRNA expression using RT-qPCR and secreted TNC protein using Western blotting and ELISA. To determine intracellular signaling pathways, we used specific inhibitors for either ERK or TGF-β receptor, and to assess the release of extracellular vesicles (EVs) we used a commercially available kit and Western blotting. At baseline, secreted TNC protein was significantly higher in asthmatic compared to non-asthmatic cells. In response to mechanical compression, both TNC mRNA expression and secreted TNC protein was significantly increased in both non-asthmatic and asthmatic cells. TNC production depended on both the ERK and TGF-β receptor pathways. Moreover, mechanically compressed HBE cells released EVs that contain TNC. These data reveal a novel mechanism by which mechanical compression, as is caused by bronchospasm, is sufficient to induce the production of ECM protein in the airway and potentially contribute to airway remodeling.
Collapse
Affiliation(s)
- Chimwemwe Mwase
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
| | - Thien-Khoi N. Phung
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
| | - Michael J. O’Sullivan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
| | - Jennifer A. Mitchel
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
| | - Margherita De Marzio
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Ayşe Kılıç
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Scott T. Weiss
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Jeffrey J. Fredberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
| | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.M.); (T.-K.N.P.); (M.J.O.); (J.A.M.); (M.D.M.); (S.T.W.); (J.J.F.)
- Correspondence: ; Tel.: +1-617-432-2726
| |
Collapse
|
15
|
Martins-Marques T. Connecting different heart diseases through intercellular communication. Biol Open 2021; 10:bio058777. [PMID: 34494646 PMCID: PMC8443862 DOI: 10.1242/bio.058777] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Well-orchestrated intercellular communication networks are pivotal to maintaining cardiac homeostasis and to ensuring adaptative responses and repair after injury. Intracardiac communication is sustained by cell-cell crosstalk, directly via gap junctions (GJ) and tunneling nanotubes (TNT), indirectly through the exchange of soluble factors and extracellular vesicles (EV), and by cell-extracellular matrix (ECM) interactions. GJ-mediated communication between cardiomyocytes and with other cardiac cell types enables electrical impulse propagation, required to sustain synchronized heart beating. In addition, TNT-mediated organelle transfer has been associated with cardioprotection, whilst communication via EV plays diverse pathophysiological roles, being implicated in angiogenesis, inflammation and fibrosis. Connecting various cell populations, the ECM plays important functions not only in maintaining the heart structure, but also acting as a signal transducer for intercellular crosstalk. Although with distinct etiologies and clinical manifestations, intercellular communication derailment has been implicated in several cardiac disorders, including myocardial infarction and hypertrophy, highlighting the importance of a comprehensive and integrated view of complex cell communication networks. In this review, I intend to provide a critical perspective about the main mechanisms contributing to regulate cellular crosstalk in the heart, which may be considered in the development of future therapeutic strategies, using cell-based therapies as a paradigmatic example. This Review has an associated Future Leader to Watch interview with the author.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
16
|
The bright side of fibroblasts: molecular signature and regenerative cues in major organs. NPJ Regen Med 2021; 6:43. [PMID: 34376677 PMCID: PMC8355260 DOI: 10.1038/s41536-021-00153-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a pathologic process characterized by the replacement of parenchymal tissue by large amounts of extracellular matrix, which may lead to organ dysfunction and even death. Fibroblasts are classically associated to fibrosis and tissue repair, and seldom to regeneration. However, accumulating evidence supports a pro-regenerative role of fibroblasts in different organs. While some organs rely on fibroblasts for maintaining stem cell niches, others depend on fibroblast activity, particularly on secreted molecules that promote cell adhesion, migration, and proliferation, to guide the regenerative process. Herein we provide an up-to-date overview of fibroblast-derived regenerative signaling across different organs and discuss how this capacity may become compromised with aging. We further introduce a new paradigm for regenerative therapies based on reverting adult fibroblasts to a fetal/neonatal-like phenotype.
Collapse
|
17
|
The role of physical cues in the development of stem cell-derived organoids. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 51:105-117. [PMID: 34120215 PMCID: PMC8964551 DOI: 10.1007/s00249-021-01551-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023]
Abstract
Organoids are a novel three-dimensional stem cells’ culture system that allows the in vitro recapitulation of organs/tissues structure complexity. Pluripotent and adult stem cells are included in a peculiar microenvironment consisting of a supporting structure (an extracellular matrix (ECM)-like component) and a cocktail of soluble bioactive molecules that, together, mimic the stem cell niche organization. It is noteworthy that the balance of all microenvironmental components is the most critical step for obtaining the successful development of an accurate organoid instead of an organoid with heterogeneous morphology, size, and cellular composition. Within this system, mechanical forces exerted on stem cells are collected by cellular proteins and transduced via mechanosensing—mechanotransduction mechanisms in biochemical signaling that dictate the stem cell specification process toward the formation of organoids. This review discusses the role of the environment in organoids formation and focuses on the effect of physical components on the developmental system. The work starts with a biological description of organoids and continues with the relevance of physical forces in the organoid environment formation. In this context, the methods used to generate organoids and some relevant published reports are discussed as examples showing the key role of mechanosensing–mechanotransduction mechanisms in stem cell-derived organoids.
Collapse
|
18
|
Albacete-Albacete L, Sánchez-Álvarez M, Del Pozo MA. Extracellular Vesicles: An Emerging Mechanism Governing the Secretion and Biological Roles of Tenascin-C. Front Immunol 2021; 12:671485. [PMID: 33981316 PMCID: PMC8107694 DOI: 10.3389/fimmu.2021.671485] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
ECM composition and architecture are tightly regulated for tissue homeostasis. Different disorders have been associated to alterations in the levels of proteins such as collagens, fibronectin (FN) or tenascin-C (TnC). TnC emerges as a key regulator of multiple inflammatory processes, both during physiological tissue repair as well as pathological conditions ranging from tumor progression to cardiovascular disease. Importantly, our current understanding as to how TnC and other non-collagen ECM components are secreted has remained elusive. Extracellular vesicles (EVs) are small membrane-bound particles released to the extracellular space by most cell types, playing a key role in cell-cell communication. A broad range of cellular components can be transported by EVs (e.g. nucleic acids, lipids, signalling molecules and proteins). These cargoes can be transferred to target cells, potentially modulating their function. Recently, several extracellular matrix (ECM) proteins have been characterized as bona fide EV cargoes, exosomal secretion being particularly critical for TnC. EV-dependent ECM secretion might underpin diseases where ECM integrity is altered, establishing novel concepts in the field such as ECM nucleation over long distances, and highlighting novel opportunities for diagnostics and therapeutic intervention. Here, we review recent findings and standing questions on the molecular mechanisms governing EV–dependent ECM secretion and its potential relevance for disease, with a focus on TnC.
Collapse
Affiliation(s)
- Lucas Albacete-Albacete
- Mechanoadaptation and Caveolae Biology Lab, Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Angel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
19
|
Park MY, Krishna Vasamsetti BM, Kim WS, Kang HJ, Kim DY, Lim B, Cho K, Kim JS, Chee HK, Park JH, Yang HS, Rallabandi HR, Ock SA, Park MR, Lee H, Hwang IS, Kim JM, Oh KB, Yun IJ. Comprehensive Analysis of Cardiac Xeno-Graft Unveils Rejection Mechanisms. Int J Mol Sci 2021; 22:ijms22020751. [PMID: 33451076 PMCID: PMC7828557 DOI: 10.3390/ijms22020751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Porcine heart xenotransplantation is a potential treatment for patients with end-stage heart failure. To understand molecular mechanisms of graft rejection after heart transplantation, we transplanted a 31-day-old alpha-1,3-galactosyltransferase knockout (GTKO) porcine heart to a five-year-old cynomolgus monkey. Histological and transcriptome analyses were conducted on xenografted cardiac tissue at rejection (nine days after transplantation). The recipient monkey's blood parameters were analyzed on days -7, -3, 1, 4, and 7. Validation was conducted by quantitative real-time PCR (qPCR) with selected genes. A non-transplanted GTKO porcine heart from an age-matched litter was used as a control. The recipient monkey showed systemic inflammatory responses, and the rejected cardiac graft indicated myocardial infarction and cardiac fibrosis. The transplanted heart exhibited a total of 3748 differentially expressed genes compared to the non-transplanted heart transcriptome, with 2443 upregulated and 1305 downregulated genes. Key biological pathways involved at the terminal stage of graft rejection were cardiomyopathies, extracellular interactions, and ion channel activities. The results of qPCR evaluation were in agreement with the transcriptome data. Transcriptome analysis of porcine cardiac tissue at graft rejection reveals dysregulation of the key molecules and signaling pathways, which play relevant roles on structural and functional integrities of the heart.
Collapse
Affiliation(s)
- Min Young Park
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Bala Murali Krishna Vasamsetti
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Wan Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Dongan-gu, Anyang 14068, Korea;
| | - Do-Young Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Byeonghwi Lim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Kahee Cho
- Primate Organ Transplantation Centre, Genia Inc., Sungnam 13201, Korea;
| | - Jun Seok Kim
- Department of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea; (J.S.K.); (H.K.C.)
| | - Hyun Keun Chee
- Department of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea; (J.S.K.); (H.K.C.)
| | - Jung Hwan Park
- Department of Nephrology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Hyun Suk Yang
- Department of Cardiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Harikrishna Reddy Rallabandi
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Sun A. Ock
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Mi-Ryung Park
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Heasun Lee
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - In-Sul Hwang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Jun-Mo Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| | - Ik Jin Yun
- Department of Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| |
Collapse
|
20
|
Matsumoto KI, Aoki H. The Roles of Tenascins in Cardiovascular, Inflammatory, and Heritable Connective Tissue Diseases. Front Immunol 2020; 11:609752. [PMID: 33335533 PMCID: PMC7736112 DOI: 10.3389/fimmu.2020.609752] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Tenascins are a family of multifunctional extracellular matrix (ECM) glycoproteins with time- and tissue specific expression patterns during development, tissue homeostasis, and diseases. There are four family members (tenascin-C, -R, -X, -W) in vertebrates. Among them, tenascin-X (TNX) and tenascin-C (TNC) play important roles in human pathologies. TNX is expressed widely in loose connective tissues. TNX contributes to the stability and maintenance of the collagen network, and its absence causes classical-like Ehlers-Danlos syndrome (clEDS), a heritable connective tissue disorder. In contrast, TNC is specifically and transiently expressed upon pathological conditions such as inflammation, fibrosis, and cancer. There is growing evidence that TNC is involved in inflammatory processes with proinflammatory or anti-inflammatory activity in a context-dependent manner. In this review, we summarize the roles of these two tenascins, TNX and TNC, in cardiovascular and inflammatory diseases and in clEDS, and we discuss the functional consequences of the expression of these tenascins for tissue homeostasis.
Collapse
Affiliation(s)
- Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| |
Collapse
|
21
|
Tenascin-C Function in Glioma: Immunomodulation and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:149-172. [PMID: 32845507 DOI: 10.1007/978-3-030-48457-6_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
First identified in the 1980s, tenascin-C (TNC) is a multi-domain extracellular matrix glycoprotein abundantly expressed during the development of multicellular organisms. TNC level is undetectable in most adult tissues but rapidly and transiently induced by a handful of pro-inflammatory cytokines in a variety of pathological conditions including infection, inflammation, fibrosis, and wound healing. Persistent TNC expression is associated with chronic inflammation and many malignancies, including glioma. By interacting with its receptor integrin and a myriad of other binding partners, TNC elicits context- and cell type-dependent function to regulate cell adhesion, migration, proliferation, and angiogenesis. TNC operates as an endogenous activator of toll-like receptor 4 and promotes inflammatory response by inducing the expression of multiple pro-inflammatory factors in innate immune cells such as microglia and macrophages. In addition, TNC drives macrophage differentiation and polarization predominantly towards an M1-like phenotype. In contrast, TNC shows immunosuppressive function in T cells. In glioma, TNC is expressed by tumor cells and stromal cells; high expression of TNC is correlated with tumor progression and poor prognosis. Besides promoting glioma invasion and angiogenesis, TNC has been found to affect the morphology and function of tumor-associated microglia/macrophages in glioma. Clinically, TNC can serve as a biomarker for tumor progression; and TNC antibodies have been utilized as an adjuvant agent to deliver anti-tumor drugs to target glioma. A better mechanistic understanding of how TNC impacts innate and adaptive immunity during tumorigenesis and tumor progression will open new therapeutic avenues to treat brain tumors and other malignancies.
Collapse
|
22
|
Hasegawa M, Yoshida T, Sudo A. Tenascin-C in Osteoarthritis and Rheumatoid Arthritis. Front Immunol 2020; 11:577015. [PMID: 33101302 PMCID: PMC7554343 DOI: 10.3389/fimmu.2020.577015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Tenascin-C (TNC) is a large multimodular glycoprotein of the extracellular matrix that consists of four distinct domains. Emerging evidence suggests that TNC may be involved in the pathogenesis of osteoarthritis (OA) and rheumatoid arthritis (RA). In this review, we summarize the current understanding of the role of TNC in cartilage and in synovial biology, across both OA and RA. TNC is expressed in association with the development of articular cartilage; the expression decreases during maturation of chondrocytes and disappears almost completely in adult articular cartilage. TNC expression is increased in diseased cartilage, synovium, and synovial fluid in OA and RA. In addition, elevated circulating TNC levels have been detected in the blood of RA patients. Thus, TNC could be used as a novel biochemical marker for OA and RA, although it has no specificity as a biochemical marker for these joint disorders. In a post-traumatic OA model of aged joints, TNC deficiency was shown to enhance cartilage degeneration. Treatment with TNC domains results in different, domain-specific effects, which are also dose-dependent. For instance, some TNC fragments including the fibrinogen-like globe domain might function as endogenous inducers of synovitis and cartilage matrix degradation through binding with toll-like receptor-4, while full-length TNC promotes cartilage repair and prevents the development of OA without exacerbating synovitis. The TNC peptide TNIIIA2 also prevents cartilage degeneration without causing synovial inflammation. The clinical significance of TNC effects on cartilage and synovium is unclear and understanding the clinical significance of TNC is not straightforward.
Collapse
Affiliation(s)
- Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshimichi Yoshida
- Department of Pathology & Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
23
|
Imanaka-Yoshida K, Tawara I, Yoshida T. Tenascin-C in cardiac disease: a sophisticated controller of inflammation, repair, and fibrosis. Am J Physiol Cell Physiol 2020; 319:C781-C796. [PMID: 32845719 DOI: 10.1152/ajpcell.00353.2020] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Toshimichi Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| |
Collapse
|
24
|
Spenlé C, Loustau T, Murdamoothoo D, Erne W, Beghelli-de la Forest Divonne S, Veber R, Petti L, Bourdely P, Mörgelin M, Brauchle EM, Cremel G, Randrianarisoa V, Camara A, Rekima S, Schaub S, Nouhen K, Imhof T, Hansen U, Paul N, Carapito R, Pythoud N, Hirschler A, Carapito C, Dumortier H, Mueller CG, Koch M, Schenke-Layland K, Kon S, Sudaka A, Anjuère F, Van Obberghen-Schilling E, Orend G. Tenascin-C Orchestrates an Immune-Suppressive Tumor Microenvironment in Oral Squamous Cell Carcinoma. Cancer Immunol Res 2020; 8:1122-1138. [PMID: 32665262 DOI: 10.1158/2326-6066.cir-20-0074] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/19/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Inherent immune suppression represents a major challenge in the treatment of human cancer. The extracellular matrix molecule tenascin-C promotes cancer by multiple mechanisms, yet the roles of tenascin-C in tumor immunity are incompletely understood. Using a 4NQO-induced oral squamous cell carcinoma (OSCC) model with abundant and absent tenascin-C, we demonstrated that tenascin-C enforced an immune-suppressive lymphoid stroma via CCL21/CCR7 signaling, leading to increased metastatic tumors. Through TLR4, tenascin-C increased expression of CCR7 in CD11c+ myeloid cells. By inducing CCL21 in lymphatic endothelial cells via integrin α9β1 and binding to CCL21, tenascin-C immobilized CD11c+ cells in the stroma. Inversion of the lymph node-to-tumor CCL21 gradient, recruitment of T regulatory cells, high expression of anti-inflammatory cytokines, and matrisomal components were hallmarks of the tenascin-C-instructed lymphoid stroma. Ablation of tenascin-C or CCR7 blockade inhibited the lymphoid immune-suppressive stromal properties, reducing tumor growth, progression, and metastasis. Thus, targeting CCR7 could be relevant in human head and neck tumors, as high tenascin-C expression and an immune-suppressive stroma correlate to poor patient survival.
Collapse
Affiliation(s)
- Caroline Spenlé
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Thomas Loustau
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Devadarssen Murdamoothoo
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - William Erne
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | | | - Romain Veber
- Institut de Biologie Moléculaire et Cellulaire, CNRS, UPR3572 Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Luciana Petti
- Université Côte d'Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | - Pierre Bourdely
- Université Côte d'Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | | | - Eva-Maria Brauchle
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany
| | - Gérard Cremel
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Vony Randrianarisoa
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Abdouramane Camara
- Institut de Biologie Moléculaire et Cellulaire, CNRS, UPR3572 Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Samah Rekima
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.,Centre Antoine Lacassagne, Nice, France
| | - Sebastian Schaub
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.,Centre Antoine Lacassagne, Nice, France
| | - Kelly Nouhen
- Université Côte d'Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | - Thomas Imhof
- Institute for Dental Research and Oral, Musculoskeletal Research, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine (IMM), University Hospital Muenster, Muenster, Germany
| | | | | | | | | | | | - Hélène Dumortier
- Institut de Biologie Moléculaire et Cellulaire, CNRS, UPR3572 Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Christopher G Mueller
- Institut de Biologie Moléculaire et Cellulaire, CNRS, UPR3572 Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Manuel Koch
- Institute for Dental Research and Oral, Musculoskeletal Research, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute of Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany.,The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," Eberhard Karls University Tübingen, Tübingen, Germany
| | - Shigeyuki Kon
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Anne Sudaka
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.,Centre Antoine Lacassagne, Nice, France
| | - Fabienne Anjuère
- Université Côte d'Azur, CNRS, IPMC, Valbonne-Sophia Antipolis, France
| | | | - Gertraud Orend
- Université Strasbourg, INSERM U1109-MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, and The Tumor Microenvironment Laboratory, Hopital Civil, Institut d'Hématologie et d'Immunologie, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
25
|
Giantsis IA, Diakakis NE, Avdi M. High Frequencies of TNC and COL5A1 Genotypes Associated With Low Risk for Superficial Digital Flexor Tendinopathy in Greek Indigenous Horse Breeds Compared With Warmblood Horses. J Equine Vet Sci 2020; 92:103173. [PMID: 32797795 DOI: 10.1016/j.jevs.2020.103173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
Tendinopathies constitute a very common category of musculoskeletal disorders, causing economic losses in the equine industry and animal welfare issues in horse populations. Sport and racehorses are in general sensitive to tendinopathies, whereas local indigenous horse populations are often more tolerant to various disorders. Particularly, indigenous Greek horse breeds have evolved and adapted in the rough topographic features of mountainous and semimountainous Greek terrain and are less prone to develop tendinopathies. Susceptibility to tendinopathy has been proposed to be associated with three specific variants in the tenascin-C and collagen type 5 α-1 genes. The present study was designed to analyze these genes and estimate the frequency of the aforementioned variants in three indigenous Greek horse populations, namely the pinia (peneia) breed, the Messara breed, and the nondescript Macedonian pacer, in comparison with a population of warmbloods. Results revealed high frequencies of genotypes with high risk for tendinopathy in the warmblood population and high frequency of low risk genotypes in the Greek breeds. Apart from the three variants, two novel polymorphisms were detected, one of which may also be associated with risk for tendinopathy. Our findings (a) are in agreement with previous ones demonstrating that risk for tendinopathies is associated with particular gene variants and also (b) highlight the value of Greek indigenous horse breeds as a genetic resource that (c) may be used in selective breeding schemes for decreasing the risk for tendinopathies in genetically improved breeds.
Collapse
Affiliation(s)
- Ioannis A Giantsis
- Department of Animal Science, Faculty of Agricultural Sciences, University of Western Macedonia, Florina, Greece
| | - Nikolaos E Diakakis
- Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melpomeni Avdi
- Department of Animal Production, Faculty of Agriculture, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
26
|
Belviso I, Romano V, Sacco AM, Ricci G, Massai D, Cammarota M, Catizone A, Schiraldi C, Nurzynska D, Terzini M, Aldieri A, Serino G, Schonauer F, Sirico F, D’Andrea F, Montagnani S, Di Meglio F, Castaldo C. Decellularized Human Dermal Matrix as a Biological Scaffold for Cardiac Repair and Regeneration. Front Bioeng Biotechnol 2020; 8:229. [PMID: 32266249 PMCID: PMC7099865 DOI: 10.3389/fbioe.2020.00229] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
The complex and highly organized environment in which cells reside consists primarily of the extracellular matrix (ECM) that delivers biological signals and physical stimuli to resident cells. In the native myocardium, the ECM contributes to both heart compliance and cardiomyocyte maturation and function. Thus, myocardium regeneration cannot be accomplished if cardiac ECM is not restored. We hypothesize that decellularized human skin might make an easily accessible and viable alternate biological scaffold for cardiac tissue engineering (CTE). To test our hypothesis, we decellularized specimens of both human skin and human myocardium and analyzed and compared their composition by histological methods and quantitative assays. Decellularized dermal matrix was then cut into 600-μm-thick sections and either tested by uniaxial tensile stretching to characterize its mechanical behavior or used as three-dimensional scaffold to assess its capability to support regeneration by resident cardiac progenitor cells (hCPCs) in vitro. Histological and quantitative analyses of the dermal matrix provided evidence of both effective decellularization with preserved tissue architecture and retention of ECM proteins and growth factors typical of cardiac matrix. Further, the elastic modulus of the dermal matrix resulted comparable with that reported in literature for the human myocardium and, when tested in vitro, dermal matrix resulted a comfortable and protective substrate promoting and supporting hCPC engraftment, survival and cardiomyogenic potential. Our study provides compelling evidence that dermal matrix holds promise as a fully autologous and cost-effective biological scaffold for CTE.
Collapse
Affiliation(s)
- Immacolata Belviso
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Veronica Romano
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Anna Maria Sacco
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Diana Massai
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Marcella Cammarota
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Angiolina Catizone
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Chiara Schiraldi
- Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Daria Nurzynska
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mara Terzini
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Alessandra Aldieri
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Gianpaolo Serino
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Fabrizio Schonauer
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Felice Sirico
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco D’Andrea
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Franca Di Meglio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
27
|
Integrin Signaling in Glioma Pathogenesis: From Biology to Therapy. Int J Mol Sci 2020; 21:ijms21030888. [PMID: 32019108 PMCID: PMC7037280 DOI: 10.3390/ijms21030888] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 11/17/2022] Open
Abstract
Integrins are a large family of transmembrane adhesion receptors, which play a key role in interactions of a cell with the surrounding stroma. Integrins are comprised of non-covalently linked α and β chains, which form heterodimeric receptor complexes. The signals from integrin receptors are combined with those originating from growth factor receptors and participate in orchestrating morphological changes of cells, organization of the cytoskeleton, stimulation of cell proliferation and rescuing cells from programmed cell death induced by extracellular matrix (ECM) detachment. Upon binding to specific ligands or ECM components, integrin dimers activate downstream signaling pathways, including focal adhesion kinase, phosphoinositide-3-kinase (PI3K) and AKT kinases, which regulate migration, invasion, proliferation and survival. Expression of specific integrins is upregulated in both tumor cells and stromal cells in a tumor microenvironment. Therefore, integrins became an attractive therapeutic target for many cancers, including the most common primary brain tumors-gliomas. In this review we provide an overview of the involvement of integrin signaling in glioma pathogenesis, formation of the tumor niche and brain tissue infiltration. We will summarize up-to-date therapeutic strategies for gliomas focused on interference with integrin ligand-receptor signaling.
Collapse
|
28
|
Serum tenascin-C levels in atrium predict atrial structural remodeling processes in patients with atrial fibrillation. J Interv Card Electrophysiol 2019; 59:401-406. [PMID: 31820271 DOI: 10.1007/s10840-019-00670-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 11/19/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Fibro-inflammatory processes in the extracellular matrix are closely associated with progressive structural remodeling in atrial fibrillation (AF). Serum concentrations of tenascin-C (TNC), an extracellular matrix glycoprotein, and of high-sensitivity C-reactive protein (CRP) might serve as a marker of remodeling and progressive inflammation of the aorta and in myocardial diseases. This study aimed to clarify relationships between TNC and CRP in patients with AF. METHODS This study included 38 patients with AF and five controls without left ventricular dysfunction who underwent catheter ablation. Blood was collected immediately before ablation from the left atrium (LA), right atrium (RA), and femoral artery (FA), and left and right atrial pressure was measured. Levels of TNC in the LA (TNC-LA), RA (TNC-RA), and FA (TNC-FA) and high-sensitivity C-reactive protein (CRP) were measured. Atrial size was also determined by echocardiography. RESULTS Levels of TNC corrected by atrial size were maximal in the LA, followed by the RA (3.69 ± 0.32 and 2.87 ± 0.38 ng/mL/cm, respectively). Mean transverse diameter corrected by body surface area was larger and mean atrial pressure was greater in the LA than the RA. A relationship was found between CRP from the femoral vein and TNC-LA and TNC-RA, but not TNC-FA. None of TNC-LA, TNC-RA, or TNC-FA correlated with ANP or BNP in the femoral vein. CONCLUSIONS Intracardiac (atrial) TNC expression plays an important role in the development of remodeling processes in the atrium with AF. Tenascin-C from the LA and RA (but not TNC, ANP, and BNP from FA) might serve as novel markers of these processes.
Collapse
|
29
|
Haage V, Elmadany N, Roll L, Faissner A, Gutmann DH, Semtner M, Kettenmann H. Tenascin C regulates multiple microglial functions involving TLR4 signaling and HDAC1. Brain Behav Immun 2019; 81:470-483. [PMID: 31271872 DOI: 10.1016/j.bbi.2019.06.047] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 01/15/2023] Open
Abstract
Tenascin C (Tnc) is an extracellular matrix glycoprotein, expressed in the CNS during development, as well as in the setting of inflammation, fibrosis and cancer, which operates as an activator of Toll-like receptor 4 (TLR4). Although TLR4 is highly expressed in microglia, the effect of Tnc on microglia has not been elucidated to date. Herein, we demonstrate that Tnc regulates microglial phagocytic activity at an early postnatal age (P4), and that this process is partially dependent on microglial TLR4 expression. We further show that Tnc regulates proinflammatory cytokine/chemokine production, chemotaxis and phagocytosis in primary microglia in a TLR4-dependent fashion. Moreover, Tnc induces histone-deacetylase 1 (HDAC1) expression in microglia, such that HDAC1 inhibition by MS-275 decreases Tnc-induced microglial IL-6 and TNF-α production. Finally, Tnc-/- cortical microglia have reduced HDAC1 expression levels at P4. Taken together, these findings establish Tnc as a regulator of microglia function during early postnatal development.
Collapse
Affiliation(s)
- Verena Haage
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Lars Roll
- Zellmorphologie und Molekulare Neurobiologie, Fakultät für Biologie und Biotechnologie, Ruhr-Universität Bochum, Bochum, Nordrhein-Wastfalen 44801, Germany
| | - Andreas Faissner
- Zellmorphologie und Molekulare Neurobiologie, Fakultät für Biologie und Biotechnologie, Ruhr-Universität Bochum, Bochum, Nordrhein-Wastfalen 44801, Germany
| | - David H Gutmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.
| |
Collapse
|
30
|
Li H, Ge X, Pan K, Sui M, Cai H, Cui C, Li C, Lu S. The Predictive Role of Tenascin-C and Cellular Communication Network Factor 3 (CCN3) in Post Hepatectomy Liver Failure in a Rat Model and 50 Patients Following Partial Hepatectomy. Med Sci Monit 2019; 25:6755-6766. [PMID: 31494663 PMCID: PMC6752097 DOI: 10.12659/msm.917331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Matricellular proteins of the extracellular matrix (ECM) include tenascin-C (TNC) and cellular communication network factor 3 (CCN3). This study aimed to investigate the role of TNC and CCN3 as prognostic factors for post hepatectomy liver failure (PHLF) in a rat model of partial hepatectomy and 50 patients following partial hepatectomy. Material/Methods Sprague-Dawley rats underwent 85% (n=53) or 90% hepatectomy (n=53) in the partial hepatectomy (PHx) model. TNC and CCN3 mRNA expression in residual liver tissue was evaluated using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and enzyme-linked immunoassay (ELISA) determined the serum levels of TNC and CCN3. In 50 patients who underwent partial hepatectomy, TNC and CCN3 serum levels were measured on postoperative day 1 and day 3. Results In the rat partial hepatectomy model, mRNA and serum levels of TNC and CCN3 were significantly increased within the first 24 h, and were higher in the 90% PHx group compared with the 85% PHx group. Fifty patients who underwent partial hepatectomy, included patients with PHLF (n=12) and patients without PHLF (n=38). Multivariate analysis confirmed that serum levels on postoperative day 3 TNChigh+CCN3high was a significant predictor of PHLF, which was associated with more than twice the risk of severe morbidity when compared with the low-risk patients (80% vs. 30%) and a significantly longer hospital stay (17 days vs. 8 days). Conclusions Further studies are needed to evaluate the potential role of the matricellular proteins, TNC and CCN3 as early clinical predictors for PHLF.
Collapse
Affiliation(s)
- Hao Li
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Xinlan Ge
- Institute of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Ke Pan
- Institute of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Minghao Sui
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Huayong Cai
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Chao Cui
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Chonghui Li
- Institute of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Shichun Lu
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| |
Collapse
|
31
|
Tenascin-C promotes chronic pressure overload-induced cardiac dysfunction, hypertrophy and myocardial fibrosis. J Hypertens 2019; 36:847-856. [PMID: 29283973 DOI: 10.1097/hjh.0000000000001628] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS Left ventricular (LV) hypertrophy is characterized by cardiomyocyte hypertrophy and interstitial fibrosis ultimately leading to increased myocardial stiffness and reduced contractility. There is substantial evidence that the altered expression of matrix metalloproteinases (MMP) and Tenascin-C (TN-C) are associated with the progression of adverse LV remodeling. However, the role of TN-C in the development of LV hypertrophy because of chronic pressure overload as well as the regulatory role of TN-C on MMPs remains unknown. METHODS AND RESULTS In a knockout mouse model of TN-C, we investigated the effect of 10 weeks of pressure overload using transverse aortic constriction (TAC). Cardiac function was determined by magnetic resonance imaging. The expression of MMP-2 and MMP-9, CD147 as well as myocardial fibrosis were assessed by immunohistochemistry. The expression of TN-C was assessed by RT-qPCR and ELISA. TN-C knockout mice showed marked reduction in fibrosis (P < 0.001) and individual cardiomyocytes size (P < 0.01), in expression of MMP-2 (P < 0.05) and MMP-9 (P < 0.001) as well as preserved cardiac function (P < 0.01) in comparison with wild-type mice after 10 weeks of TAC. In addition, CD147 expression was markedly increased under pressure overload (P < 0.01), irrespectively of genotype. TN-C significantly increased the expression of the markers of hypertrophy such as ANP and BNP as well as MMP-2 in H9c2 cells (P < 0.05, respectively). CONCLUSION Our results are pointed toward a novel signaling mechanism that contributes to LV remodeling via MMPs upregulation, cardiomyocyte hypertrophy as well as myocardial fibrosis by TN-C under chronic pressure overload.
Collapse
|
32
|
Lopez AL, Larina IV. Second harmonic generation microscopy of early embryonic mouse hearts. BIOMEDICAL OPTICS EXPRESS 2019; 10:2898-2908. [PMID: 31259060 PMCID: PMC6583332 DOI: 10.1364/boe.10.002898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 05/15/2023]
Abstract
The understanding of biomechanical regulation of early heart development in genetic mouse models can contribute to improved management of congenital cardiovascular defects and embryonic cardiac failures in humans. The extracellular matrix (ECM), and particularly fibrillar collagen, are central to heart biomechanics, regulating tissue strength, elasticity and contractility. Here, we explore second harmonic generation (SHG) microscopy for visualization of establishing cardiac fibers such as collagen in mouse embryos through the earliest stages of development. We detected significant increase in SHG positive fibrillar content and organization over the first 24 hours after initiation of contractions. SHG microscopy revealed regions of higher fibrillar organization in regions of higher contractility and reduced fibrillar content and organization in mouse Mlc2a model with cardiac contractility defect, suggesting regulatory role of mechanical load in production and organization of structural fibers from the earliest stages. Simultaneous volumetric SHG and two-photon excitation microscopy of vital fluorescent reporter EGFP in the heart was demonstrated. In summary, these data set SHG microscopy as a valuable non-bias imaging tool to investigate mouse embryonic cardiogenesis and biomechanics.
Collapse
Affiliation(s)
- Andrew L. Lopez
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Irina V. Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
33
|
Zappe M, Feldner A, Arnold C, Sticht C, Hecker M, Korff T. NFAT5 Isoform C Controls Biomechanical Stress Responses of Vascular Smooth Muscle Cells. Front Physiol 2018; 9:1190. [PMID: 30190682 PMCID: PMC6115610 DOI: 10.3389/fphys.2018.01190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/07/2018] [Indexed: 01/10/2023] Open
Abstract
Vascular cells are continuously exposed to mechanical stress that may wreak havoc if exceeding physiological levels. Consequently, mechanisms facing such a challenge are indispensable and contribute to the adaptation of the cellular phenotype. To this end, vascular smooth muscle cells (VSMCs) activate mechanoresponsive transcription factors promoting their proliferation and migration to initiate remodeling the arterial wall. In mechanostimulated VSMCs, we identified nuclear factor of activated T-cells 5 (NFAT5) as transcriptional regulator protein and intended to unravel mechanisms controlling its expression and nuclear translocation. In cultured human VSMCs, blocking RNA synthesis diminished both baseline and stretch-induced NFAT5 mRNA expression while inhibition of the proteasome promoted accumulation of the NFAT5 protein. Detailed PCR analyses indicated a decrease in expression of NFAT5 isoform A and an increase in isoform C in mechanoactivated VSMCs. Upon overexpression, only NFAT5c was capable to enter the nucleus in control- and stretch-stimulated VSMCs. As evidenced by analyses of NFAT5c mutants, nuclear translocation required palmitoylation, phosphorylation at Y143 and was inhibited by phosphorylation at S1197. On the functional level, overexpression of NFAT5c forces its accumulation in the nucleus as well as transcriptional activity and stimulated VSMC proliferation and migration. These findings suggest that NFAT5 is continuously expressed and degraded in resting VSMCs while expression and accumulation of isoform C in the nucleus is facilitated during biomechanical stress to promote an activated VSMC phenotype.
Collapse
Affiliation(s)
- Maren Zappe
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Anja Feldner
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Caroline Arnold
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Markus Hecker
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
34
|
Imanaka-Yoshida K, Matsumoto KI. Multiple Roles of Tenascins in Homeostasis and Pathophysiology of Aorta. Ann Vasc Dis 2018; 11:169-180. [PMID: 30116408 PMCID: PMC6094038 DOI: 10.3400/avd.ra.17-00118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tenascins are a family of large extracellular matrix (ECM) glycoproteins. Four family members (tenascin-C, -R, -X, and -W) have been identified to date. Each member consists of the same types of structural domains and exhibits time- and tissue-specific expression patterns, suggesting their specific roles in embryonic development and tissue remodeling. Among them, the significant involvement of tenascin-C (TNC) and tenascin-X (TNX) in the progression of vascular diseases has been examined in detail. TNC is strongly up-regulated under pathological conditions, induced by a number of inflammatory mediators and mechanical stress. TNC has diverse functions, particularly in the regulation of inflammatory responses. Recent studies suggest that TNC is involved in the pathophysiology of aneurysmal and dissecting lesions, in part by protecting the vascular wall from destructive mechanical stress. TNX is strongly expressed in vascular walls, and its distribution is often reciprocal to that of TNC. TNX is involved in the stability and maintenance of the collagen network and elastin fibers. A deficiency in TNX results in a form of Ehlers–Danlos syndrome (EDS). Although their exact roles in vascular diseases have not yet been elucidated, TNC and TNX are now being recognized as promising biomarkers for diagnosis and risk stratification of vascular diseases.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie, Japan.,Mie University Research Center for Matrix Biology, Tsu, Mie, Japan
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Shimane, Japan
| |
Collapse
|
35
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
36
|
Mwila-Kazimbaya K, Garcia MP, Bosomprah S, Laban NM, Chisenga CC, Permar SR, Simuyandi M, Munsaka S, Chilengi R. Effect of innate antiviral glycoproteins in breast milk on seroconversion to rotavirus vaccine (Rotarix) in children in Lusaka, Zambia. PLoS One 2017; 12:e0189351. [PMID: 29284036 PMCID: PMC5746212 DOI: 10.1371/journal.pone.0189351] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/24/2017] [Indexed: 12/20/2022] Open
Abstract
Introduction Rotavirus vaccines have been introduced into national immunization programmes to mitigate morbidity and mortality associated rotavirus diarrhoea. Lower vaccine effectiveness has however been noted in low-middle income countries, but little is known about the role of maternal components found in breast milk. This study assessed the effect of lactoferrin, lactadherin, and tenascin-c on rotavirus vaccine seroconversion. Methods This was a retrospective cohort study of 128 infants who had been fully immunized with Rotarix™. Serum samples were collected from the infant at baseline and one month after second rotavirus vaccine dose. Breast milk samples were collected from mothers at baseline. Standard ELISA was used to determine titres of rotavirus-specific immunologlobulin G and A in breast milk and serum as well as concentrations of lactoferrin, lactadherin, and tenascin-c. Poisson regression model with robust standard error was used to estimate the effect of breast milk components on seroconversion. The components were modelled on log base 2 so that the effect would be interpreted as a doubling of the concentration. Results In a multivariable analysis adjusting for maternal age, maternal HIV status, seropositivity at baseline, sex, age of child at vaccination as well as breast milk IgA and IgG, we found evidence of independent effect of LA (Adjusted IRR = 0.95; 95% CI = 0.91–0.99; P = 0.019) on seroconversion while there was no evidence for TNC (Adjusted IRR = 1.00; 95% CI = 0.85–1.17; P = 0.967) and LF (Adjusted RR = 1.01; 95% CI = 0.96–1.05); P = 0.802). We explored the joint effects of the three components but we found no evidence (Adjusted RR = 0.95; 95% CI = 0.81; P = 0.535). Conclusion High breast milk concentrations of lactadherin might play a role in infant’s failure to seroconvert to rotavirus vaccines. Further research to understand this observed association is an important consideration.
Collapse
Affiliation(s)
- Katayi Mwila-Kazimbaya
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of biomedical sciences, School of Health sciences, University of Zambia, Lusaka, Zambia
| | | | - Samuel Bosomprah
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of Biostatistics, School of Public Health, University of Ghana, Legon, Accra, Ghana
| | | | | | - Sallie Robey Permar
- Department of Paediatrics, Human Vaccine Institute, Duke University, Durham, North Carolina
| | | | - Sody Munsaka
- Department of biomedical sciences, School of Health sciences, University of Zambia, Lusaka, Zambia
| | - Roma Chilengi
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
37
|
Sun Z, Schwenzer A, Rupp T, Murdamoothoo D, Vegliante R, Lefebvre O, Klein A, Hussenet T, Orend G. Tenascin-C Promotes Tumor Cell Migration and Metastasis through Integrin α9β1-Mediated YAP Inhibition. Cancer Res 2017; 78:950-961. [PMID: 29259017 DOI: 10.1158/0008-5472.can-17-1597] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
Tenascin-C is an extracellular matrix molecule that drives progression of many types of human cancer, but the basis for its actions remains obscure. In this study, we describe a cell-autonomous signaling mechanism explaining how tenascin-C promotes cancer cell migration in the tumor microenvironment. In a murine xenograft model of advanced human osteosarcoma, tenascin-C and its receptor integrin α9β1 were determined to be essential for lung metastasis of tumor cells. We determined that activation of this pathway also reduced tumor cell-autonomous expression of target genes for the transcription factor YAP. In clinical specimens, a genetic signature comprising four YAP target genes represents prognostic impact. Taken together, our results illuminate how tumor cell deposition of tenascin-C in the tumor microenvironment promotes invasive migration and metastatic progression.Significance: These results illuminate how the extracellular matrix glycoprotein tenascin-C in the tumor microenvironment promotes invasive migration and metastatic progression by employing integrin α9β1, abolishing actin stress fiber formation, inhibiting YAP and its target gene expression, with potential implications for cancer prognosis and therapy. Cancer Res; 78(4); 950-61. ©2017 AACR.
Collapse
Affiliation(s)
- Zhen Sun
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Anja Schwenzer
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Tristan Rupp
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Devadarssen Murdamoothoo
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Rolando Vegliante
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Olivier Lefebvre
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annick Klein
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Thomas Hussenet
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Gertraud Orend
- INSERM U1109 - MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hôpital Civil, Institut d'Hématologie et d'Immunologie, Strasbourg, France. .,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
38
|
Landry NM, Cohen S, Dixon IMC. Periostin in cardiovascular disease and development: a tale of two distinct roles. Basic Res Cardiol 2017; 113:1. [PMID: 29101484 DOI: 10.1007/s00395-017-0659-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022]
Abstract
Tissue development and homeostasis are dependent upon the concerted synthesis, maintenance, and degradation of extracellular matrix (ECM) molecules. Cardiac fibrosis is now recognized as a primary contributor to incidence of heart failure, particularly heart failure with preserved ejection fraction, wherein cardiac filling in diastole is compromised. Periostin is a cell-associated protein involved in cell fate determination, proliferation, tumorigenesis, and inflammatory responses. As a non-structural component of the ECM, secreted 90 kDa periostin is emerging as an important matricellular factor in cardiac mesenchymal tissue development. In addition, periostin's role as a mediator in cell-matrix crosstalk has also garnered attention for its association with fibroproliferative diseases in the myocardium, and for its association with TGF-β/BMP signaling. This review summarizes the phylogenetic history of periostin, its role in cardiac development, and the major signaling pathways influencing its expression in cardiovascular pathology. Further, we provide a synthesis of the current literature to distinguish the multiple roles of periostin in cardiac health, development and disease. As periostin may be targeted for therapeutic treatment of cardiac fibrosis, these insights may shed light on the putative timing for application of periostin-specific therapies.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada
| | - Smadar Cohen
- Regenerative Medicine and Stem Cell Research Center, Ilse Katz Institute for Nanoscale Science and Technology, Beersheba, Israel.,Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, Institute of Cardiovascular Sciences, University of Manitoba, Winnipeg, Canada. .,Laboratory of Molecular Cardiology, St. Boniface Hospital Albrechtsen Research Centre, R3010-351 Taché Avenue, Winnipeg, MB R2H 2A6, Canada.
| |
Collapse
|
39
|
The Roles of Matricellular Proteins in Oncogenic Virus-Induced Cancers and Their Potential Utilities as Therapeutic Targets. Int J Mol Sci 2017; 18:ijms18102198. [PMID: 29065446 PMCID: PMC5666879 DOI: 10.3390/ijms18102198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
Matricellular proteins differ from other classical extracellular matrix proteins; for instance, they are transiently expressed as soluble proteins rather than being constitutively expressed in pathological conditions, such as acute viral infections. Accumulating studies have revealed that matricellular proteins, including osteopontin and tenascin-C, both of which interact with integrin heterodimers, are involved in inflammatory diseases, autoimmune disorders, and cancers. The concentrations of these matricellular proteins are elevated in the plasma of patients with certain types of cancers, indicating that they play important roles in oncogenesis. Chronic viral infections are associated with certain cancers, which are distinct from non-viral cancers. Viral oncogenes play critical roles in the development and progression of such cancers. It is vital to investigate the mechanisms of tumorigenesis and, particularly, the mechanism by which viral proteins induce tumor progression. Viral proteins have been shown to influence not only the viral-infected cancer cells, but also the stromal cells and matricellular proteins that constitute the extracellular matrix that surrounds tumor tissues. In this review, we summarize the recent progress on the involvement of matricellular proteins in oncogenic virus-induced cancers to elucidate the mechanism of oncogenesis and consider the possible role of matricellular proteins as therapeutic targets in virus-induced cancers.
Collapse
|
40
|
Goldstein NB, Koster MI, Jones KL, Gao B, Hoaglin LG, Robinson SE, Wright MJ, Birlea SI, Luman A, Lambert KA, Shellman YG, Fujita M, Robinson WA, Roop DR, Norris DA, Birlea SA. Repigmentation of Human Vitiligo Skin by NBUVB Is Controlled by Transcription of GLI1 and Activation of the β-Catenin Pathway in the Hair Follicle Bulge Stem Cells. J Invest Dermatol 2017; 138:657-668. [PMID: 29054607 DOI: 10.1016/j.jid.2017.09.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/17/2017] [Accepted: 09/21/2017] [Indexed: 12/22/2022]
Abstract
Vitiligo repigmentation is a complex process in which the melanocyte-depleted interfollicular epidermis is repopulated by melanocyte precursors from hair follicle bulge that proliferate, migrate, and differentiate into mature melanocytes on their way to the epidermis. The strongest stimulus for vitiligo repigmentation is narrow-band UVB (NBUVB), but how the hair follicle melanocyte precursors are activated by UV light has not been extensively studied. To better understand this process, we developed an application that combined laser capture microdissection and subsequent whole transcriptome RNA sequencing of hair follicle bulge melanocyte precursors and compared their gene signatures to that of regenerated mature epidermal melanocytes from NBUVB-treated vitiligo skin. Using this strategy, we found up-regulation of TNC, GJB6, and THBS1 in the hair follicle bulge melanocytes and of TYR in the epidermal melanocytes of the NBUVB-treated vitiligo skin. We validated these results by quantitative real-time-PCR using NBUVB-treated vitiligo skin and untreated normal skin. We also identified that GLI1, a candidate stem cell-associated gene, is significantly up-regulated in the melanocytes captured from NBUVB-treated vitiligo bulge compared with untreated vitiligo bulge. These signals are potential key players in the activation of bulge melanocyte precursors during vitiligo repigmentation.
Collapse
Affiliation(s)
| | - Maranke I Koster
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA
| | - Kenneth L Jones
- Department of Hematology, University of Colorado, Aurora, Colorado, USA; Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Bifeng Gao
- Sequencing and Microarray Core, University of Colorado, Aurora, Colorado, USA
| | - Laura G Hoaglin
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA
| | | | - Michael J Wright
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Smaranda I Birlea
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Abigail Luman
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Karoline A Lambert
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Yiqun G Shellman
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | | | - Dennis R Roop
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA
| | - David A Norris
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Stanca A Birlea
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado Aurora, Colorado, USA.
| |
Collapse
|
41
|
Wang Z, Wei Q, Han L, Cao K, Lan T, Xu Z, Wang Y, Gao Y, Xue J, Shan F, Feng J, Xie X. Tenascin-c renders a proangiogenic phenotype in macrophage via annexin II. J Cell Mol Med 2017; 22:429-438. [PMID: 28857429 PMCID: PMC5742692 DOI: 10.1111/jcmm.13332] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 06/21/2017] [Indexed: 12/31/2022] Open
Abstract
Tenascin-c is an extracellular matrix glycoprotein, the expression of which relates to the progression of atherosclerosis, myocardial infarction and heart failure. Annexin II acts as a cell surface receptor of tenascin-c. This study aimed to delineate the role of tenascin-c and annexin II in macrophages presented in atherosclerotic plaque. Animal models with atherosclerotic lesions were established using ApoE-KO mice fed with high-cholesterol diet. The expression of tenascin-c and annexin II in atherosclerotic lesions was determined by qRT-PCR, Western blot and immunohistochemistry analysis. Raw 264.7 macrophages and human primary macrophages were exposed to 5, 10 and 15 μg/ml tenascin-c for 12 hrs. Cell migration as well as the proangiogenic ability of macrophages was examined. Additionally, annexin II expression was delineated in raw 264.7 macrophages under normal condition (20% O2 ) for 12 hrs or hypoxic condition (1% O2 ) for 6-12 hrs. The expression of tenascin-c and annexin II was markedly augmented in lesion aorta. Tenascin-c positively regulated macrophage migration, which was dependent on the expression of annexin II in macrophages. VEGF release from macrophages and endothelial tube induction by macrophage were boosted by tenascin-c and attenuated by annexin II blocking. Furthermore, tenascin-c activated Akt/NF-κB and ERK signalling through annexin II. Lastly, hypoxia conditioning remarkably facilitates annexin II expression in macrophages through hypoxia-inducible factor (HIF)-1α but not HIF-2α. In conclusion, tenascin-c promoted macrophage migration and VEGF expression through annexin II, the expression of which was modulated by HIF-1α.
Collapse
Affiliation(s)
- Zhiyang Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Qi Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Liang Han
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Keqing Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Tianfeng Lan
- Institute of Integrated Medical Information, Xi'an, China
| | - Zhenjie Xu
- Institute of Integrated Medical Information, Xi'an, China
| | - Yingjuan Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China
| | - Yuan Gao
- Department of Traditional Chinese Medicine, College of Life Science, Northwest University, Xi'an, China
| | - Jing Xue
- Department of Traditional Chinese Medicine, College of Life Science, Northwest University, Xi'an, China
| | - Fei Shan
- Department of Cardiovascular Surgery, Affiliated Hospital of Yan'an University, Yan'an, China
| | - Jun Feng
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Xin Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, China.,Institute of Integrated Medical Information, Xi'an, China
| |
Collapse
|
42
|
Zhao B, Hu M, Wu H, Ren C, Wang J, Cui S. Tenascin-C expression and its associated pathway in BMSCs following co-culture with mechanically stretched ligament fibroblasts. Mol Med Rep 2017; 15:2465-2472. [PMID: 28447748 PMCID: PMC5428738 DOI: 10.3892/mmr.2017.6329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/09/2017] [Indexed: 01/09/2023] Open
Abstract
The occurrence of pelvic organ prolapse (POP) is closely associated with alterations in the extracellular matrix proteins of the supporting ligament. Bone marrow mesenchymal stem cells (BMSCs) have the potential to differentiate into a variety of cell types, including osteoblasts, chondroblasts and adipocytes. Therefore, BMSCs have the potential to improve the clinical outcomes of POP. Tenascin-C is a large glycoprotein that is present in the ECM and is involved in morphogenetic movements, and tissue patterning and repair. The aim of the present study was to investigate the effect of mechanical stretching on tenascin-C expression during the differentiation of BMSCs induced by pelvic ligament fibroblasts. BMSCs were isolated from 7-day-old Sprague Dawley rats. Fibroblasts were obtained from rat pelvic ligaments and, at the fourth passage, were subjected to 10% deformation with 1 Hz, periodic one-way mechanical stretch stimulation, followed by co-culture with BMSCs. The co-culture with stretched fibroblasts increased tenascin-C and transforming growth factor (TGF)-β expression levels, compared with groups without mechanical stimulation. Neutralizing anti-TGF-β1 antibodies, and inhibitors of TGF-β receptor, mitogen-activated protein kinase (MAPK) kinase and MAPK, decreased tenascin-C expression levels induced by TGF-β and mechanical stretching. The results of the present study suggested that the regulation of tenascin-C expression levels in BMSCs co-cultured with mechanically stretched pelvic ligament fibroblasts is mediated via the soluble growth factor TGF-β and the MAPK signaling pathway. In addition, these results indicated that in an indirect co-culture system, pelvic ligament fibroblasts with mechanical stretch stimulation may promote the synthesis of tenascin-C and BMSC differentiation into pelvic ligament fibroblasts.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mengcai Hu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huiyan Wu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chenchen Ren
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jianshe Wang
- Department of Clinical Medicine, Hebi Polytechnic College, Hebi, Henan 458030, P.R. China
| | - Shihong Cui
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
43
|
Effects of Tenascin-C Knockout on Cerebral Vasospasm After Experimental Subarachnoid Hemorrhage in Mice. Mol Neurobiol 2017; 55:1951-1958. [DOI: 10.1007/s12035-017-0466-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022]
|
44
|
Midgett M, López CS, David L, Maloyan A, Rugonyi S. Increased Hemodynamic Load in Early Embryonic Stages Alters Endocardial to Mesenchymal Transition. Front Physiol 2017; 8:56. [PMID: 28228731 PMCID: PMC5296359 DOI: 10.3389/fphys.2017.00056] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/23/2017] [Indexed: 12/30/2022] Open
Abstract
Normal blood flow is essential for proper heart formation during embryonic development, as abnormal hemodynamic load (blood pressure and shear stress) results in cardiac defects seen in congenital heart disease. However, the progressive detrimental remodeling processes that relate altered blood flow to cardiac defects remain unclear. Endothelial-mesenchymal cell transition is one of the many complex developmental events involved in transforming the early embryonic outflow tract into the aorta, pulmonary trunk, interventricular septum, and semilunar valves. This study elucidated the effects of increased hemodynamic load on endothelial-mesenchymal transition remodeling of the outflow tract cushions in vivo. Outflow tract banding was used to increase hemodynamic load in the chicken embryo heart between Hamburger and Hamilton stages 18 and 24. Increased hemodynamic load induced increased cell density in outflow tract cushions, fewer cells along the endocardial lining, endocardium junction disruption, and altered periostin expression as measured by confocal microscopy analysis. In addition, 3D focused ion beam scanning electron microscopy analysis determined that a portion of endocardial cells adopted a migratory shape after outflow tract banding that is more irregular, elongated, and with extensive cellular projections compared to normal cells. Proteomic mass-spectrometry analysis quantified altered protein composition after banding that is consistent with a more active stage of endothelial-mesenchymal transition. Outflow tract banding enhances the endothelial-mesenchymal transition phenotype during formation of the outflow tract cushions, suggesting that endothelial-mesenchymal transition is a critical developmental process that when disturbed by altered blood flow gives rise to cardiac malformation and defects.
Collapse
Affiliation(s)
- Madeline Midgett
- Biomedical Engineering, Oregon Health and Science University Portland, OR, USA
| | - Claudia S López
- Biomedical Engineering, Oregon Health and Science UniversityPortland, OR, USA; Multiscale Microscopy Core, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science UniversityPortland, OR, USA
| | - Larry David
- Proteomics Core, Oregon Health and Science University Portland, OR, USA
| | - Alina Maloyan
- Knight Cardiovascular Institute, Oregon Health and Science University Portland, OR, USA
| | - Sandra Rugonyi
- Biomedical Engineering, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
45
|
Di Liddo R, Aguiari P, Barbon S, Bertalot T, Mandoli A, Tasso A, Schrenk S, Iop L, Gandaglia A, Parnigotto PP, Conconi MT, Gerosa G. Nanopatterned acellular valve conduits drive the commitment of blood-derived multipotent cells. Int J Nanomedicine 2016; 11:5041-5055. [PMID: 27789941 PMCID: PMC5068475 DOI: 10.2147/ijn.s115999] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Considerable progress has been made in recent years toward elucidating the correlation among nanoscale topography, mechanical properties, and biological behavior of cardiac valve substitutes. Porcine TriCol scaffolds are promising valve tissue engineering matrices with demonstrated self-repopulation potentiality. In order to define an in vitro model for investigating the influence of extracellular matrix signaling on the growth pattern of colonizing blood-derived cells, we cultured circulating multipotent cells (CMC) on acellular aortic (AVL) and pulmonary (PVL) valve conduits prepared with TriCol method and under no-flow condition. Isolated by our group from Vietnamese pigs before heart valve prosthetic implantation, porcine CMC revealed high proliferative abilities, three-lineage differentiative potential, and distinct hematopoietic/endothelial and mesenchymal properties. Their interaction with valve extracellular matrix nanostructures boosted differential messenger RNA expression pattern and morphologic features on AVL compared to PVL, while promoting on both matrices the commitment to valvular and endothelial cell-like phenotypes. Based on their origin from peripheral blood, porcine CMC are hypothesized in vivo to exert a pivotal role to homeostatically replenish valve cells and contribute to hetero- or allograft colonization. Furthermore, due to their high responsivity to extracellular matrix nanostructure signaling, porcine CMC could be useful for a preliminary evaluation of heart valve prosthetic functionality.
Collapse
Affiliation(s)
- Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova; Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS
| | - Paola Aguiari
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Silvia Barbon
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova; Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS
| | - Thomas Bertalot
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova
| | - Amit Mandoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova
| | - Alessia Tasso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova
| | - Sandra Schrenk
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova
| | - Laura Iop
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Alessandro Gandaglia
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova; Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS
| | - Gino Gerosa
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| |
Collapse
|
46
|
Fujimoto M, Shiba M, Kawakita F, Liu L, Nakasaki A, Shimojo N, Imanaka-Yoshida K, Yoshida T, Suzuki H. Epidermal growth factor-like repeats of tenascin-C-induced constriction of cerebral arteries via activation of epidermal growth factor receptors in rats. Brain Res 2016; 1642:436-444. [DOI: 10.1016/j.brainres.2016.04.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 01/01/2023]
|
47
|
Andrés-Delgado L, Mercader N. Interplay between cardiac function and heart development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1707-16. [PMID: 26952935 PMCID: PMC4906158 DOI: 10.1016/j.bbamcr.2016.03.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 12/24/2022]
Abstract
Mechanotransduction refers to the conversion of mechanical forces into biochemical or electrical signals that initiate structural and functional remodeling in cells and tissues. The heart is a kinetic organ whose form changes considerably during development and disease. This requires cardiomyocytes to be mechanically durable and able to mount coordinated responses to a variety of environmental signals on different time scales, including cardiac pressure loading and electrical and hemodynamic forces. During physiological growth, myocytes, endocardial and epicardial cells have to adaptively remodel to these mechanical forces. Here we review some of the recent advances in the understanding of how mechanical forces influence cardiac development, with a focus on fluid flow forces. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Laura Andrés-Delgado
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Nadia Mercader
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain; Institute of Anatomy, University of Bern, Bern, Switzerland.
| |
Collapse
|
48
|
Waite C, Mejia R, Ascoli M. Gq/11-Dependent Changes in the Murine Ovarian Transcriptome at the End of Gestation. Biol Reprod 2016; 94:62. [PMID: 26843449 PMCID: PMC4829089 DOI: 10.1095/biolreprod.115.136952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/01/2016] [Indexed: 11/26/2022] Open
Abstract
Parturition in rodents is highly dependent on the engagement of the luteal prostaglandin F2 alpha receptor, which, through activation of the Gq/11 family of G proteins, increases the expression of Akr1c18, leading to an increase in progesterone catabolism. To further understand the involvement of Gq/11 on luteolysis and parturition, we used microarray analysis to compare the ovarian transcriptome of mice with a granulosa/luteal cell-specific deletion of Galphaq/11 with their control littermates on Day 18 of pregnancy, when mice from both genotypes are pregnant, and on Day 22, when mice with a granulosa/luteal cell-specific deletion of Galphaq/11 are still pregnant but their control littermates are 1–2 days postpartum. Ovarian genes up-regulated at the end of gestation in a Galphaq/11 -dependent fashion include genes involved in focal adhesion and extracellular matrix interactions. Genes down-regulated at the end of gestation in a Galphaq/11-dependent manner include Serpina6 (which encodes corticosteroid-binding globulin); Enpp2 (which encodes autotaxin, the enzyme responsible for the synthesis of lysophosphatidic acid); genes involved in protein processing and export; reproductive genes, such as Lhcgr; the three genes needed to convert progesterone to estradiol (Cyp17a1, Hsd17b7, and Cyp19a1); and Inha. Activation of ovarian Gq/11 by engagement of the prostaglandin F2 alpha receptor on Day 18 of pregnancy recapitulated the regulation of many but not all of these genes. Thus, although the ovarian transcriptome at the end of gestation is highly dependent on the activation of Gq/11, not all of these changes are dependent on the actions of prostaglandin F2 alpha.
Collapse
Affiliation(s)
- Courtney Waite
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rachel Mejia
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Mario Ascoli
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
49
|
Yokokawa T, Sugano Y, Nakayama T, Nagai T, Matsuyama TA, Ohta-Ogo K, Ikeda Y, Ishibashi-Ueda H, Nakatani T, Yasuda S, Takeishi Y, Ogawa H, Anzai T. Significance of myocardial tenascin-C expression in left ventricular remodelling and long-term outcome in patients with dilated cardiomyopathy. Eur J Heart Fail 2016; 18:375-85. [PMID: 26763891 PMCID: PMC5066704 DOI: 10.1002/ejhf.464] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 09/12/2015] [Accepted: 09/22/2015] [Indexed: 12/23/2022] Open
Abstract
Aim Dilated cardiomyopathy (DCM) has a variety of causes, and no useful approach to predict left ventricular (LV) remodelling and long‐term outcome has yet been established. Myocardial tenascin‐C (TNC) is known to appear under pathological conditions, possibly to regulate cardiac remodelling. The aim of this study was to clarify the significance of myocardial TNC expression in LV remodelling and the long‐term outcome in DCM. Methods and results One hundred and twenty‐three consecutive DCM patients who underwent endomyocardial biopsy for initial diagnosis were studied. Expression of TNC in biopsy sections was analysed immunohistochemically to quantify the ratio of the TNC‐positive area to the whole myocardial tissue area (TNC area). Clinical parameters associated with TNC area were investigated. The patients were divided into two groups based on receiver operating characteristic analysis of TNC area to predict death: high TNC group with TNC area ≥2.3% (22 patients) and low TNC group with TNC area <2.3% (101 patients). High TNC was associated with diabetes mellitus. Comparing echocardiographic findings between before and 9 months after endomyocardial biopsy, the low TNC group was associated with decreased LV end‐diastolic diameter and increased LV ejection fraction, whereas the high TNC group was not. Survival analysis revealed a worse outcome in the high TNC group than in the low TNC group (P < 0.001). Multivariable Cox regression analysis revealed that TNC area was independently associated with poor outcome (HR = 1.347, P = 0.032). Conclusions Increased myocardial TNC expression was associated with worse LV remodeling and long‐term outcome in DCM.
Collapse
Affiliation(s)
- Tetsuro Yokokawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Yasuo Sugano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Takafumi Nakayama
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Toshiyuki Nagai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Taka-Aki Matsuyama
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Keiko Ohta-Ogo
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | | | - Takeshi Nakatani
- Department of Transplantation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, Fukushima, Japan
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| |
Collapse
|
50
|
Kasprzycka M, Hammarström C, Haraldsen G. Tenascins in fibrotic disorders-from bench to bedside. Cell Adh Migr 2015; 9:83-9. [PMID: 25793575 DOI: 10.4161/19336918.2014.994901] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although fibrosis is becoming increasingly recognized as a major cause of morbidity and mortality in chronic inflammatory diseases, available treatment strategies are limited. Tenascins constitute a family of matricellular proteins, primarily modulating interactions of cells with other matrix components and growth factors. Data obtained from tenascin C deficient mice show important roles of this molecule in several models of fibrosis. Moreover there is growing evidence that tenascin C has a strong impact on chronic inflammation, myofibroblast differentiation and recruitment. Tenascin C as well as tenascin X has furthermore been shown to affect TGF-β activation and signaling. Taken together these data suggest that these proteins might be important factors in fibrosis development and make them attractive both as biological markers and as targets for therapeutical intervention. So far most clinical research in fibrosis has been focused on tenascin C. This review aims at summarizing our up-to-date knowledge on the involvement of tenascin C in the pathogenesis of fibrotic disorders.
Collapse
|