1
|
Tsoneva Y, Velikova T, Nikolaev G. Circadian clock regulation of myofibroblast fate. Cell Signal 2025; 131:111774. [PMID: 40169063 DOI: 10.1016/j.cellsig.2025.111774] [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: 12/04/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Fibrosis-related disorders represent an increasing medical and economic burden on a worldwide scale, accounting for one-third of all disease-related deaths with limited therapeutic options. As central mediators in fibrosis development, myofibroblasts have been gaining increasing attention in the last 20 years as potential targets for fibrosis attenuation and reversal. While various aspects of myofibroblast physiology have been proposed as treatment targets, many of these approaches have shown limited long-term efficacy so far. However, ongoing research is uncovering new potential strategies for targeting myofibroblast activity, offering hope for more effective treatments in the future. The circadian molecular clock is a feature of almost every cell in the human body that dictates the rhythmic nature of various aspects of human physiology and behavior in response to changes in the surrounding environment. The dysregulation of these rhythms with aging is considered to be one of the underlying reasons behind the development of multiple aging-related chronic disorders, with fibrotic tissue scarring being a common pathological complication among the majority of them. Myofibroblast dysregulation due to skewed circadian clockwork might significantly contribute to fibrotic scar persistence. In the current review, we highlight the role of the circadian clock in the context of myofibroblast activation and deactivation and examine its dysregulation as a driver of fibrogenesis.
Collapse
Affiliation(s)
- Yoanna Tsoneva
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", Bulgaria.
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak str, 1407 Sofia, Bulgaria.
| | - Georgi Nikolaev
- Department of Cell and Developmental Biology, Faculty of Biology, Sofia University "St. Kliment Ohridski", Bulgaria.
| |
Collapse
|
2
|
Mueller MC, Blomberg R, Tanneberger AE, Davis-Hall D, Neeves KB, Magin CM. Female Fibroblast Activation Is Estrogen-Mediated in Sex-Specific 3D-Bioprinted Pulmonary Artery Adventitia Models. ACS Biomater Sci Eng 2025; 11:2935-2945. [PMID: 40285704 DOI: 10.1021/acsbiomaterials.5c00123] [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] [Indexed: 04/29/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a form of pulmonary vascular disease characterized by scarring of the small blood vessels that results in reduced blood flow and increased blood pressure in the lungs. Over time, this increase in blood pressure causes damage to the heart. Idiopathic (IPAH) impacts male and female patients differently, with female patients showing a higher disease susceptibility (4:1 female-to-male ratio) but experiencing longer survival rates postdiagnosis compared to male patients. This complex sex dimorphism is known as the estrogen paradox. Prior studies suggest that estrogen signaling may be pathologic in the pulmonary vasculature and protective in the heart, yet the mechanisms underlying these sex differences in IPAH remain unclear. Many previous studies of PAH relied on male cells or cells of undisclosed origin for in vitro modeling. Here, we present a dynamic, three-dimensional (3D)-bioprinted model incorporating cells and circulating sex hormones from female patients to specifically study how female patients respond to changes in microenvironmental stiffness and sex hormone signaling on the cellular level. Poly(ethylene glycol)-α methacrylate (PEGαMA)-based hydrogels containing female human pulmonary artery adventitia fibroblasts (hPAAFs) from IPAH or control donors were 3D bioprinted to mimic pulmonary artery adventitia. These biomaterials were initially soft, like healthy blood vessels, and then stiffened using light to mimic vessel scarring in PAH. These 3D-bioprinted models showed that stiffening the microenvironment around female IPAH hPAAFs led to hPAAF activation. On both the protein and gene-expression levels, cellular activation markers significantly increased in stiffened samples and were highest in IPAH patient-derived cells. Treatment with a selective estrogen receptor modulator, which is currently in clinical trials for IPAH treatment, reduced the expression of hPAAF activation markers, demonstrating that hPAAF activation is one pathologic response mediated by estrogen signaling in the vasculature. These results showed the utility of sex-specific, 3D-bioprinted pulmonary artery adventitia models for preclinical drug discovery and validation.
Collapse
Affiliation(s)
- Mikala C Mueller
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
| | - Rachel Blomberg
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
| | - Alicia E Tanneberger
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
| | - Duncan Davis-Hall
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
| | - Keith B Neeves
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora 80045, Colorado, United States
- Hemophilia and Thrombosis Center, University of Colorado, Anschutz Medical Campus, Aurora 80045, Colorado, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver|Anschutz Medical Campus, Aurora 80045, Colorado, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora 80045, Colorado, United States
- Division of Pulmonary Sciences & Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora 80045, Colorado, United States
| |
Collapse
|
3
|
Mueller MC, Blomberg R, Tanneberger AE, Davis-Hall D, Neeves KB, Magin CM. Female fibroblast activation is estrogen-mediated in sex-specific 3D-bioprinted pulmonary artery adventitia models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633670. [PMID: 39896610 PMCID: PMC11785021 DOI: 10.1101/2025.01.17.633670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Pulmonary arterial hypertension (PAH) impacts male and female patients in different ways. Female patients exhibit a greater susceptibility to disease (4:1 female-to-male ratio) but live longer after diagnosis than male patients. This complex sexual dimorphism is known as the estrogen paradox. Prior studies suggest that estrogen signaling may be pathologic in the pulmonary vasculature and protective in the heart, yet the mechanisms underlying these sex-differences in PAH remain unclear. PAH is a form of a pulmonary vascular disease that results in scarring of the small blood vessels, leading to impaired blood flow and increased blood pressure. Over time, this increase in blood pressure causes damage to the heart. Many previous studies of PAH relied on male cells or cells of undisclosed origin for in vitro modeling. Here we present a dynamic, 3D-bioprinted model that incorporates cells and circulating sex hormones from female patients to specifically study how female patients respond to changes in microenvironmental stiffness and sex hormone signaling. Poly(ethylene glycol)-alpha methacrylate (PEGαMA)-based hydrogels containing female human pulmonary artery adventitia fibroblasts (hPAAFs) from idiopathic PAH (IPAH) or control donors were 3D bioprinted to mimic pulmonary artery adventitia. These biomaterials were initially soft, like healthy blood vessels, and then stiffened using light to mimic vessel scarring in PAH. These 3D-bioprinted models showed that stiffening the microenvironment around female IPAH hPAAFs led to hPAAF activation. On both the protein and gene-expression levels, cellular activation markers significantly increased in stiffened samples and were highest in IPAH patient-derived cells. Treatment with a selective estrogen receptor modulator reduced expression hPAAF activation markers, demonstrating that hPAAF activation is a one pathologic response mediated by estrogen signaling in the vasculature, validating that drugs currently in clinical trials could be evaluated in sex-specific 3D-bioprinted pulmonary artery adventitia models.
Collapse
|
4
|
Şahin A, Babayev H, Cirigliano L, Preto M, Falcone M, Altıntas E, Gül M. Unveiling the molecular Hallmarks of Peyronie's disease: a comprehensive narrative review. Int J Impot Res 2024; 36:801-808. [PMID: 38454161 DOI: 10.1038/s41443-024-00845-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/26/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024]
Abstract
Peyronie's disease, a fibroinflammatory disorder, detrimentally impacts the sexual well-being of men and their partners. The manifestation of fibrotic plaques within penile tissue, attributed to dysregulated fibrogenesis, is pathognomonic for this condition. The onset of fibrosis hinges on the perturbation of the equilibrium between matrix metalloproteinases (MMPs), crucial enzymes governing the extracellular matrix, and tissue inhibitors of MMPs (TIMPs). In the context of Peyronie's disease, there is an elevation in TIMP levels coupled with a decline in MMP levels, culminating in fibrogenesis. Despite the scant molecular insights into fibrotic pathologies, particularly in the context of Peyronie's disease, a comprehensive literature search spanning 1995 to 2023, utilizing PubMed Library, was conducted to elucidate these mechanisms. The findings underscore the involvement of growth factors such as FGF and PDGF, and cytokines like IL-1 and IL-6, alongside PAI-1, PTX-3, HIF, and IgG4 in the fibrotic cascade. Given the tissue-specific modulation of fibrosis, comprehending the molecular underpinnings of penile fibrosis becomes imperative for the innovation of novel and efficacious therapies targeting Peyronie's disease. This review stands as a valuable resource for researchers and clinicians engaged in investigating the molecular basis of fibrotic diseases, offering guidance for advancements in understanding Peyronie's disease.
Collapse
Affiliation(s)
- Ali Şahin
- Selcuk University School of Medicine, 42250, Konya, Turkey
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265, Davos, Switzerland
| | - Lorenzo Cirigliano
- Department of Urology, Molinette Hospital, University of Torino, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Mirko Preto
- Department of Urology, Molinette Hospital, University of Torino, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Marco Falcone
- Department of Urology, Molinette Hospital, University of Torino, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Emre Altıntas
- Department of Urology, Selcuk University School of Medicine, 42250, Konya, Turkey
| | - Murat Gül
- Department of Urology, Molinette Hospital, University of Torino, AOU Città della Salute e della Scienza di Torino, Turin, Italy.
- Department of Urology, Selcuk University School of Medicine, 42250, Konya, Turkey.
| |
Collapse
|
5
|
Zheng S, Liu Y. Progress in the Study of Fra-2 in Respiratory Diseases. Int J Mol Sci 2024; 25:7143. [PMID: 39000247 PMCID: PMC11240912 DOI: 10.3390/ijms25137143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 07/16/2024] Open
Abstract
Fos-related antigen-2 (Fra-2) is a member of the activating protein-1 (AP-1) family of transcription factors. It is involved in controlling cell growth and differentiation by regulating the production of the extracellular matrix (ECM) and coordinating the balance of signals within and outside the cell. Fra-2 is not only closely related to bone development, metabolism, and immune system and eye development but also in the progression of respiratory conditions like lung tumors, asthma, pulmonary fibrosis, and chronic obstructive pulmonary disease (COPD). The increased expression and activation of Fra-2 in various lung diseases has been shown in several studies. However, the specific molecular mechanisms through which Fra-2 affects the development of respiratory diseases are not yet understood. The purpose of this research is to summarize and delineate advancements in the study of the involvement of transcription factor Fra-2 in disorders related to the respiratory system.
Collapse
Affiliation(s)
- Shuping Zheng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
6
|
Luna-Angulo A, Landa-Solís C, Escobar-Cedillo RE, Estrada-Mena FJ, Sánchez-Chapul L, Gómez-Díaz B, Carrillo-Mora P, Avilés-Arnaut H, Jiménez-Hernández L, Jiménez-Hernández DA, Miranda-Duarte A. Pharmacological Treatments and Therapeutic Targets in Muscle Dystrophies Generated by Alterations in Dystrophin-Associated Proteins. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1060. [PMID: 39064489 PMCID: PMC11279157 DOI: 10.3390/medicina60071060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases of genetic origin characterized by progressive skeletal muscle degeneration and weakness. There are several types of MDs, varying in terms of age of onset, severity, and pattern of the affected muscles. However, all of them worsen over time, and many patients will eventually lose their ability to walk. In addition to skeletal muscle effects, patients with MDs may present cardiac and respiratory disorders, generating complications that could lead to death. Interdisciplinary management is required to improve the surveillance and quality of life of patients with an MD. At present, pharmacological therapy is only available for Duchene muscular dystrophy (DMD)-the most common type of MD-and is mainly based on the use of corticosteroids. Other MDs caused by alterations in dystrophin-associated proteins (DAPs) are less frequent but represent an important group within these diseases. Pharmacological alternatives with clinical potential in patients with MDs and other proteins associated with dystrophin have been scarcely explored. This review focuses on drugs and molecules that have shown beneficial effects, mainly in experimental models involving alterations in DAPs. The mechanisms associated with the effects leading to promising results regarding the recovery or maintenance of muscle strength and reduction in fibrosis in the less-common MDs (i.e., with respect to DMD) are explored, and other therapeutic targets that could contribute to maintaining the homeostasis of muscle fibers, involving different pathways, such as calcium regulation, hypertrophy, and maintenance of satellite cell function, are also examined. It is possible that some of the drugs explored here could be used to affordably improve the muscular function of patients until a definitive treatment for MDs is developed.
Collapse
Affiliation(s)
- Alexandra Luna-Angulo
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Carlos Landa-Solís
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, División de Biotecnología, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Rosa Elena Escobar-Cedillo
- Departamento de Electromiografía y Distrofia Muscular, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Francisco Javier Estrada-Mena
- Laboratorio de Biología Molecular, Universidad Panamericana, Facultad de Ciencias de la Salud, Augusto Rodin 498, Ciudad de México 03920, Mexico
| | - Laura Sánchez-Chapul
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Benjamín Gómez-Díaz
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Paul Carrillo-Mora
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Hamlet Avilés-Arnaut
- Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo Leon, Av. Universidad s/n Ciudad Universitaria, San Nicolas de los Garza 66455, Mexico
| | | | | | - Antonio Miranda-Duarte
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| |
Collapse
|
7
|
Ilg MM, Harding S, Lapthorn AR, Kirvell S, Ralph DJ, Bustin SA, Ball G, Cellek S. Temporal gene signature of myofibroblast transformation in Peyronie's disease: first insights into the molecular mechanisms of irreversibility. J Sex Med 2024; 21:278-287. [PMID: 38383071 DOI: 10.1093/jsxmed/qdae006] [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: 08/31/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 02/23/2024]
Abstract
BACKGROUND Transformation of resident fibroblasts to profibrotic myofibroblasts in the tunica albuginea is a critical step in the pathophysiology of Peyronie's disease (PD). We have previously shown that myofibroblasts do not revert to the fibroblast phenotype and we suggested that there is a point of no return at 36 hours after induction of the transformation. However, the molecular mechanisms that drive this proposed irreversibility are not known. AIM Identify molecular pathways that drive the irreversibility of myofibroblast transformation by analyzing the expression of the genes involved in the process in a temporal fashion. METHODS Human primary fibroblasts obtained from tunica albuginea of patients with Peyronie's disease were transformed to myofibroblasts using transforming growth factor beta 1 (TGF-β1). The mRNA of the cells was collected at 0, 24, 36, 48, and 72 hours after stimulation with TGF-β1 and then analyzed using a Nanostring nCounter Fibrosis panel. The gene expression results were analyzed using Reactome pathway analysis database and ANNi, a deep learning-based inference algorithm based on a swarm approach. OUTCOMES The study outcome was the time course of changes in gene expression during transformation of PD-derived fibroblasts to myofibroblasts. RESULTS The temporal analysis of the gene expression revealed that the majority of the changes at the gene expression level happened within the first 24 hours and remained so throughout the 72-hour period. At 36 hours, significant changes were observed in genes involved in MAPK-Hedgehog signaling pathways. CLINICAL TRANSLATION This study highlights the importance of early intervention in clinical management of PD and the future potential of new drugs targeting the point of no return. STRENGTHS AND LIMITATIONS The use of human primary cells and confirmation of results with further RNA analysis are the strengths of this study. The study was limited to 760 genes rather than the whole transcriptome. CONCLUSION This study is to our knowledge the first analysis of temporal gene expression associated with the regulation of the transformation of resident fibroblasts to profibrotic myofibroblasts in PD. Further research is warranted to investigate the role of the MAPK-Hedgehog signaling pathways in reversibility of PD.
Collapse
Affiliation(s)
- Marcus M Ilg
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Sophie Harding
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Alice R Lapthorn
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Sara Kirvell
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - David J Ralph
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
- Urology Department, University College London, London, W1G 8PH, United Kingdom
| | - Stephen A Bustin
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Graham Ball
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| | - Selim Cellek
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, CM1 1SQ, United Kingdom
| |
Collapse
|
8
|
Rampioni Vinciguerra GL, Capece M, Scafetta G, Rentsch S, Vecchione A, Lovat F, Croce CM. Role of Fra-2 in cancer. Cell Death Differ 2024; 31:136-149. [PMID: 38104183 PMCID: PMC10850073 DOI: 10.1038/s41418-023-01248-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Abstract
Fos-related antigen-2 (Fra-2) is the most recently discovered member of the Fos family and, by dimerizing with Jun proteins, forms the activator protein 1 (AP-1) transcription factor. By inducing or repressing the transcription of several target genes, Fra-2 is critically involved in the modulation of cell response to a variety of extracellular stimuli, stressors and intracellular changes. In physiological conditions, Fra-2 has been found to be ubiquitously expressed in human cells, regulating differentiation and homeostasis of bone, muscle, nervous, lymphoid and other tissues. While other AP-1 members, like Jun and Fos, are well characterized, studies of Fra-2 functions in cancer are still at an early stage. Due to the lack of a trans-activating domain, which is present in other Fos proteins, it has been suggested that Fra-2 might inhibit cell transformation, eventually exerting an anti-tumor effect. In human malignancies, however, Fra-2 activity is enhanced (or induced) by dysregulation of microRNAs, oncogenes and extracellular signaling, suggesting a multifaceted role. Therefore, Fra-2 can promote or prevent transformation, proliferation, migration, epithelial-mesenchymal transition, drug resistance and metastasis formation in a tumor- and context-dependent manner. Intriguingly, recent data reports that Fra-2 is also expressed in cancer associated cells, contributing to the intricate crosstalk between neoplastic and non-neoplastic cells, that leads to the evolution and remodeling of the tumor microenvironment. In this review we summarize three decades of research on Fra-2, focusing on its oncogenic and anti-oncogenic effects in tumor progression and dissemination.
Collapse
Affiliation(s)
- Gian Luca Rampioni Vinciguerra
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Marina Capece
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Giorgia Scafetta
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Sydney Rentsch
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Francesca Lovat
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
9
|
Zhao Y, Ding W, Zhang P, Deng L, Long Y, Lu J, Shiri F, Heidari Majd M. Improving Tamoxifen Performance in Inducing Apoptosis and Hepatoprotection by Loading on a Dual Nanomagnetic Targeting System. Anticancer Agents Med Chem 2024; 24:1016-1028. [PMID: 38685808 DOI: 10.2174/0118715206289666240423091244] [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: 12/15/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Although tamoxifen (TMX) belongs to selective estrogen receptor modulators (SERMs) and selectively binds to estrogen receptors, it affects other estrogen-producing tissues due to passive diffusion and non-differentiation of normal and cancerous cells and leads to side effects. METHODS The problems expressed about tamoxifen (TMX) encouraged us to design a new drug delivery system based on magnetic nanoparticles (MNPs) to simultaneously target two receptors on cancer cells through folic acid (FA) and hyaluronic acid (HA) groups. The mediator of binding of two targeting agents to MNPs is a polymer linker, including dopamine, polyethylene glycol, and terminal amine (DPN). RESULTS Zeta potential, dynamic light scattering (DLS), and Field emission scanning electron microscopy (FESEM) methods confirmed that MNPs-DPN-HA-FA has a suitable size of ~105 nm and a surface charge of -41 mV, and therefore, it can be a suitable option for carrying TMX and increasing its solubility. The cytotoxic test showed that the highest concentration of MNPs-DPN-HA-FA-TMX decreased cell viability to about 11% after 72 h of exposure compared to the control. While the protective effect of modified MNPs on normal cells was evident, unlike tamoxifen, the survival rate of liver cells, even after 180 min of treatment, was not significantly different from the control group. The protective effect of MNPs was also confirmed by examining the amount of malondialdehyde, and no significant difference was observed in the amount of lipid peroxidation caused by modified MNPs compared to the control. Flow cytometry proved that TMX loaded onto modified MNPs can induce apoptosis by targeting the overexpressed receptors on cancer cells. Real-time PCR showed that the modified MNPs activated the intrinsic and extrinsic mitochondrial pathways of apoptosis, so the Bak1/Bclx ratio for MNPs-DPN-HAFA- TMX and free TMX was 70.82 and 0.38, respectively. Also, the expression of the caspase-3 gene increased 430 times compared to the control. On the other hand, only TNF gene expression, which is responsible for metastasis in some tumors, was decreased by both free TMX and MNPs-DPN-HA-FA-TMX. Finally, molecular docking proved that MNPs-DPN-HA-FA-TMX could provide a very stable interaction with both CD44 and folate receptors, induce apoptosis in cancer cells, and reduce hepatotoxicity. CONCLUSION All the results showed that MNPs-DPN-HA-FA-TMX can show good affinity to cancer cells using targeting agents and induce apoptosis in metastatic breast ductal carcinoma T-47D cell lines. Also, the protective effects of MNPs on hepatocytes are quite evident, and they can reduce the side effects of TMX.
Collapse
Affiliation(s)
- Yanfang Zhao
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Wanbao Ding
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Peixian Zhang
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Lei Deng
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Yi Long
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Jiuqin Lu
- Key Laboratory of Tumor Immunological Prevention and Treatment, Department of Oncology, Yan'An Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | | | - Mostafa Heidari Majd
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
10
|
Urban L, Čoma M, Lacina L, Szabo P, Sabová J, Urban T, Šuca H, Lukačín Š, Zajíček R, Smetana K, Gál P. Heterogeneous response to TGF-β1/3 isoforms in fibroblasts of different origins: implications for wound healing and tumorigenesis. Histochem Cell Biol 2023; 160:541-554. [PMID: 37707642 DOI: 10.1007/s00418-023-02221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 09/15/2023]
Abstract
Identification of therapeutic targets for treating fibrotic diseases and cancer remains challenging. Our study aimed to investigate the effects of TGF-β1 and TGF-β3 on myofibroblast differentiation and extracellular matrix deposition in different types of fibroblasts, including normal/dermal, cancer-associated, and scar-derived fibroblasts. When comparing the phenotype and signaling pathways activation we observed extreme heterogeneity of studied markers across different fibroblast populations, even within those isolated from the same tissue. Specifically, the presence of myofibroblast and deposition of extracellular matrix were dependent on the origin of the fibroblasts and the type of treatment they received (TGF-β1 vs. TGF-β3). In parallel, we detected activation of canonical signaling (pSMAD2/3) across all studied fibroblasts, albeit to various extents. Treatment with TGF-β1 and TGF-β3 resulted in the activation of canonical and several non-canonical pathways, including AKT, ERK, and ROCK. Among studied cells, cancer-associated fibroblasts displayed the most heterogenic response to TGF-β1/3 treatments. In general, TGF-β1 demonstrated a more potent activation of signaling pathways compared to TGF-β3, whereas TGF-β3 exhibited rather an inhibitory effect in keloid- and hypertrophic scar-derived fibroblasts suggesting its clinical potential for scar treatment. In summary, our study has implications for comprehending the role of TGF-β signaling in fibroblast biology, fibrotic diseases, and cancer. Future research should focus on unraveling the mechanisms beyond differential fibroblast responses to TGF-β isomers considering inherent fibroblast heterogeneity.
Collapse
Affiliation(s)
- Lukáš Urban
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11, Košice, Slovak Republic
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Ondavská, 040 11, Košice, Slovak Republic
| | - Matúš Čoma
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11, Košice, Slovak Republic
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Ondavská, 040 11, Košice, Slovak Republic
| | - Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 2, 128 00, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 128 08, Prague, Czech Republic
| | - Pavol Szabo
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 2, 128 00, Prague, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Jana Sabová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11, Košice, Slovak Republic
| | - Tomáš Urban
- Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, 100 00, Prague, Czech Republic
| | - Hubert Šuca
- Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, 100 00, Prague, Czech Republic
| | - Štefan Lukačín
- Department of Heart Surgery, East-Slovak Institute of Cardiovascular Diseases Inc, 040 11, Košice, Slovak Republic
| | - Robert Zajíček
- Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, 100 00, Prague, Czech Republic
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, U Nemocnice 2, 128 00, Prague, Czech Republic.
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic.
| | - Peter Gál
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 040 11, Košice, Slovak Republic.
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Ondavská, 040 11, Košice, Slovak Republic.
- Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, 100 00, Prague, Czech Republic.
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, 832 32, Bratislava, Slovak Republic.
- Institute of Neurobiology, Biomedical Research Center of the Slovak Academy of Sciences, 040 01, Košice, Slovak Republic.
| |
Collapse
|
11
|
Zomer HD, Cooke PS. Targeting estrogen signaling and biosynthesis for aged skin repair. Front Physiol 2023; 14:1281071. [PMID: 38028803 PMCID: PMC10645088 DOI: 10.3389/fphys.2023.1281071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Non-healing skin wounds are disproportionally prevalent in older adults. Current treatments do not account for the particularities of aged skin and result in inadequate outcomes. Overall, healing chronic wounds in the elderly remains a major unmet clinical need. Estrogens play a critical role in reproduction but also have important actions in non-reproductive organs. Estrogen biosynthesis and signaling pathways are locally activated during physiological wound healing, processes that are inhibited in elderly estrogen-deprived skin. Estrogen deprivation has been shown to be a critical mediator of impaired wound healing in both postmenopausal women and aged men, and topical estrogen application reverses age-associated delayed wound healing in both elderly men and women. These data indicate that adequate estrogen biosynthesis and properly regulated estrogen signaling pathways are essential for normal wound healing and can be targeted to optimize tissue repair in the elderly. However, due to fundamental questions regarding how to safely restore estrogen signaling locally in skin wounds, there are currently no therapeutic strategies addressing estrogen deficiency in elderly chronic wounds. This review discusses established and recent literature in this area and proposes the hypothesis that estrogen plays a pleiotropic role in skin aging and that targeting estrogen signaling and biosynthesis could promote skin repair in older adults.
Collapse
Affiliation(s)
- Helena D. Zomer
- Department of Physiological Sciences, University of Florida, Gainesville, FL, United States
| | | |
Collapse
|
12
|
Maleddu A, Zhu J, Clay MR, Wilky BA. Current therapies and future prospective for locally aggressive mesenchymal tumors. Front Oncol 2023; 13:1160239. [PMID: 37546427 PMCID: PMC10401592 DOI: 10.3389/fonc.2023.1160239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/11/2023] [Indexed: 08/08/2023] Open
Abstract
Locally aggressive mesenchymal tumors comprise a heterogeneous group of soft tissue and bone tumors with intermediate histology, incompletely understood biology, and highly variable natural history. Despite having a limited to absent ability to metastasize and excellent survival prognosis, locally aggressive mesenchymal tumors can be symptomatic, require prolonged and repeat treatments including surgery and chemotherapy, and can severely impact patients' quality of life. The management of locally aggressive tumors has evolved over the years with a focus on minimizing morbid treatments. Extensive oncologic surgeries and radiation are pillars of care for high grade sarcomas, however, play a more limited role in management of locally aggressive mesenchymal tumors, due to propensity for local recurrence despite resection, and the risk of transformation to a higher-grade entity following radiation. Patients should ideally be evaluated in specialized sarcoma centers that can coordinate complex multimodal decision-making, taking into consideration the individual patient's clinical presentation and history, as well as any available prognostic factors into customizing therapy. In this review, we aim to discuss the biology, clinical management, and future treatment frontiers for three representative locally aggressive mesenchymal tumors: desmoid-type fibromatosis (DF), tenosynovial giant cell tumor (TSGCT) and giant cell tumor of bone (GCTB). These entities challenge clinicians with their unpredictable behavior and responses to treatment, and still lack a well-defined standard of care despite recent progress with newly approved or promising experimental drugs.
Collapse
Affiliation(s)
- Alessandra Maleddu
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Jessica Zhu
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Michael Roy Clay
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Breelyn Ann Wilky
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
13
|
Khallouki F, Hajji L, Saber S, Bouddine T, Edderkaoui M, Bourhia M, Mir N, Lim A, El Midaoui A, Giesy JP, Aboul-Soud MAM, Silvente-Poirot S, Poirot M. An Update on Tamoxifen and the Chemo-Preventive Potential of Vitamin E in Breast Cancer Management. J Pers Med 2023; 13:jpm13050754. [PMID: 37240924 DOI: 10.3390/jpm13050754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Breast cancer (BC) is the most common female cancer in terms of incidence and mortality worldwide. Tamoxifen (Nolvadex) is a widely prescribed, oral anti-estrogen drug for the hormonal treatment of estrogen-receptor-positive BC, which represents 70% of all BC subtypes. This review assesses the current knowledge on the molecular pharmacology of tamoxifen in terms of its anticancer and chemo-preventive actions. Due to the importance of vitamin E compounds, which are widely taken as a supplementary dietary component, the review focuses only on the potential importance of vitamin E in BC chemo-prevention. The chemo-preventive and onco-protective effects of tamoxifen combined with the potential effects of vitamin E can alter the anticancer actions of tamoxifen. Therefore, methods involving an individually designed, nutritional intervention for patients with BC warrant further consideration. These data are of great importance for tamoxifen chemo-prevention strategies in future epidemiological studies.
Collapse
Affiliation(s)
- Farid Khallouki
- Biology Department, FSTE, Moulay Ismail University of Meknes, BP 609, Errachidia 52000, Morocco
- Biology Department, Faculty of Sciences, Moulay Ismail University of Meknes, BP. 11201 Zitoune, Meknes 50050, Morocco
| | - Lhoussain Hajji
- Biology Department, Faculty of Sciences, Moulay Ismail University of Meknes, BP. 11201 Zitoune, Meknes 50050, Morocco
| | - Somayya Saber
- Biology Department, FSTE, Moulay Ismail University of Meknes, BP 609, Errachidia 52000, Morocco
- Biology Department, Faculty of Sciences, Moulay Ismail University of Meknes, BP. 11201 Zitoune, Meknes 50050, Morocco
| | - Toufik Bouddine
- Biology Department, Faculty of Sciences, Moulay Ismail University of Meknes, BP. 11201 Zitoune, Meknes 50050, Morocco
| | - Mouad Edderkaoui
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center & University of California, Los Angeles, CA 90048, USA
| | - Mohammed Bourhia
- Higher Institute of Nursing Professions and Technical Health, Laayoune 70000, Morocco
| | - Nora Mir
- Biology Department, Faculty of Sciences, Moulay Ismail University of Meknes, BP. 11201 Zitoune, Meknes 50050, Morocco
| | - Adrian Lim
- Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center & University of California, Los Angeles, CA 90048, USA
| | - Adil El Midaoui
- Biology Department, FSTE, Moulay Ismail University of Meknes, BP 609, Errachidia 52000, Morocco
| | - John P Giesy
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA
- Department of Environmental Sciences, Baylor University, Waco, TX 76706, USA
| | - Mourad A M Aboul-Soud
- Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Sandrine Silvente-Poirot
- Cancer Research Center of Toulouse, UMR 1037 INSERM, UMR 5071 CNRS, University of Toulouse III, Equipe labellisée par la Ligue Nationale Contre le Cancer, 31037 Toulouse, France
- French Network for Nutrition And Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France
| | - Marc Poirot
- Cancer Research Center of Toulouse, UMR 1037 INSERM, UMR 5071 CNRS, University of Toulouse III, Equipe labellisée par la Ligue Nationale Contre le Cancer, 31037 Toulouse, France
- French Network for Nutrition And Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France
| |
Collapse
|
14
|
Cancer-Associated Fibroblast Diversity Shapes Tumor Metabolism in Pancreatic Cancer. Cancers (Basel) 2022; 15:cancers15010061. [PMID: 36612058 PMCID: PMC9817728 DOI: 10.3390/cancers15010061] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Despite extensive research, the 5-year survival rate of pancreatic cancer (PDAC) patients remains at only 9%. Patients often show poor treatment response, due partly to a highly complex tumor microenvironment (TME). Cancer-associated fibroblast (CAF) heterogeneity is characteristic of the pancreatic TME, where several CAF subpopulations have been identified, such as myofibroblastic CAFs (myCAFs), inflammatory CAFs (iCAFs), and antigen presenting CAFs (apCAFs). In PDAC, cancer cells continuously adapt their metabolism (metabolic switch) to environmental changes in pH, oxygenation, and nutrient availability. Recent advances show that these environmental alterations are all heavily driven by stromal CAFs. CAFs and cancer cells exchange cytokines and metabolites, engaging in a tight bidirectional crosstalk, which promotes tumor aggressiveness and allows constant adaptation to external stress, such as chemotherapy. In this review, we summarize CAF diversity and CAF-mediated metabolic rewiring, in a PDAC-specific context. First, we recapitulate the most recently identified CAF subtypes, focusing on the cell of origin, activation mechanism, species-dependent markers, and functions. Next, we describe in detail the metabolic crosstalk between CAFs and tumor cells. Additionally, we elucidate how CAF-driven paracrine signaling, desmoplasia, and acidosis orchestrate cancer cell metabolism. Finally, we highlight how the CAF/cancer cell crosstalk could pave the way for new therapeutic strategies.
Collapse
|
15
|
Tamoxifen Prevents Peritendinous Adhesions: A Preliminary Report. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4250771. [PMID: 36177054 PMCID: PMC9514950 DOI: 10.1155/2022/4250771] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Background Scarless healing comprises the ultimate goal after an injury. Since tendon healing results in a fibrotic scar, an injured tendon can never regain the mechanical potential and strength of its uninjured form. A wide variety of studies focus on the tendon healing with an absent or minimal peritendinous adhesions. However, no simple method has managed it at all. Possible complex actions and peritendinous environmental events take place during the tendon healing process. Tamoxifen (TAM), besides its breast cancer-related usage, is a potent antifibrotic drug. Here, we aimed to reduce the peritendinous adhesion with TAM administration. Methods Achilles tendons of 44 Wistar albino rats were randomly distributed in 4 groups. In group 1, bilateral lower extremities were used as control and sham. Groups 2 and 3 were comprised of low-dose (1 mg/kg) and high-dose (40 mg/kg) systemic administration of TAM, respectively. Group 4 included local administration (1 mg/kg) of TAM. Biomechanical, macroscopical, and histopathological analyses were done and compared statistically. Biomechanically, the maximum force that led to tendon rupture was determined, and tensile force data were recorded via tensile testing device. Macroscopical and histopathological analysis were composed of the quantity, quality, and grade of peritendinous adhesions. Results Macroscopic and histopathologic findings revealed that groups 2 and 3 had a variety of values ranging between slight to severe adhesions. In group 2, almost half of the animals had moderate adhesions, whereas in group 3, the majority of the animals had moderate adhesions. There were no animals with moderate or severe adhesions in group 4. Statistically significant values were calculated between sham and control groups. Biomechanically, group 2 showed the most significant result. The tendons in group 2 had the highest stiffness when maximal force was applied to rupture the tendons. Henceforth, all these consequences were proven statistically. Conclusion We achieved less peritendinous adhesion with the local administration of TAM when compared to systemic administration of TAM. A better understanding of the peritendinous environmental process will reveal to develop new therapies in the prevention of peritendinous adhesions.
Collapse
|
16
|
Chen B, Mu C, Zhang Z, He X, Liu X. The Love-Hate Relationship Between TGF-β Signaling and the Immune System During Development and Tumorigenesis. Front Immunol 2022; 13:891268. [PMID: 35720407 PMCID: PMC9204485 DOI: 10.3389/fimmu.2022.891268] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Since TGF-β was recognized as an essential secreted cytokine in embryogenesis and adult tissue homeostasis a decade ago, our knowledge of the role of TGF-β in mammalian development and disease, particularly cancer, has constantly been updated. Mounting evidence has confirmed that TGF-β is the principal regulator of the immune system, as deprivation of TGF-β signaling completely abrogates adaptive immunity. However, enhancing TGF-β signaling constrains the immune response through multiple mechanisms, including boosting Treg cell differentiation and inducing CD8+ T-cell apoptosis in the disease context. The love-hate relationship between TGF-β signaling and the immune system makes it challenging to develop effective monotherapies targeting TGF-β, especially for cancer treatment. Nonetheless, recent work on combination therapies of TGF-β inhibition and immunotherapy have provide insights into the development of TGF-β-targeted therapies, with favorable outcomes in patients with advanced cancer. Hence, we summarize the entanglement between TGF-β and the immune system in the developmental and tumor contexts and recent progress on hijacking crucial TGF-β signaling pathways as an emerging area of cancer therapy.
Collapse
Affiliation(s)
- Baode Chen
- Department of Laboratory Medicine, Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chenglin Mu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhiwei Zhang
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Xuelin He
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Institute for Intelligent Bio/Chem Manufacturing (iBCM), Zhejiang University (ZJU)-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| |
Collapse
|
17
|
Reszegi A, Tátrai P, Regős E, Kovalszky I, Baghy K. Syndecan-1 in liver pathophysiology. Am J Physiol Cell Physiol 2022; 323:C289-C294. [PMID: 35704700 DOI: 10.1152/ajpcell.00039.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Syndecan-1 is a heparan sulfate/chondroitin sulfate proteoglycan (PG) of the cell surface and the extracellular matrix, which regulates a broad spectrum of physiological and pathological processes such as cell proliferation, migration, inflammation, matrix remodeling, wound healing, or tumorigenesis. Syndecan-1 represents the major PG of the liver, expressed by hepatocytes and cholangiocytes, and its elevated expression is a characteristic feature of liver diseases. The highest syndecan-1 expression is found in liver cirrhosis and in hepatocellular carcinoma (HCC) developed in cirrhotic livers. In addition, as being a hepatitis C receptor, hepatitis C virus (HCV) infected livers produce extremely large amounts of syndecan-1. The serum levels of the cleaved (shedded) extracellular domain has clinical significance, as its increased concentration reflects on poor prognosis in cirrhosis as well as in cancer. In vivo experiments confirmed that syndecan-1 protects against early stages of fibrogenesis mainly by enhanced clearance of transforming growth factor beta (TGFβ1) and thrombospondin-1 via circulation, and against hepatocarcinogenesis by interfering with several signaling pathways and enhancing cell cycle blockade. In addition, syndecan-1 is capable to hinder lipid metabolism and ribosomal biogenesis in induced cancer models.. These observations together with its participation in the uptake of viruses (e.g. HCV, SARS-CoV-2) indicate that syndecan-1 is a central player in liver pathologies.
Collapse
Affiliation(s)
- Andrea Reszegi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Eszter Regős
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornelia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
18
|
Chung E, Yafi FA. Pharmacotherapy in Peyronie's disease: a state-of-the-art review on established contemporary and emerging drugs. Expert Opin Pharmacother 2022; 23:1035-1042. [PMID: 35209789 DOI: 10.1080/14656566.2022.2043274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Current clinical guidelines on Peyronie's disease (PD) advocate non-surgical treatment options as the first-line therapy despite inconsistent clinical outcomes when compared to definitive penile reconstructive surgery. AREAS COVERED This article examines the current understanding of established contemporary and emerging pharmacotherapies for PD. Emphasis has been placed on published clinical studies on drugs in the last 10 years. EXPERT OPINION Published studies have shown that combination therapy is likely more effective than monotherapy. Combined treatment modalities involving various oral and/or intralesional pharmacotherapies together with mechanical devices or clinical psychosexual therapy may provide additional or synergistic benefits for PD patients. A multidisciplinary approach coupled with more novel targets for pharmacological intervention could deliver a more effective treatment paradigm to prevent or at least delay the need for definitive penile reconstructive surgery. Drugs targeting the inhibition of TGF-β1 pathway and myofibroblast transformation are of great interest and studies into next-generation genetic sequencing and transcriptional biomarker regulatory pathways in PD will provide useful insights into the pathophysiology of PD, and assist the development of future regenerative technology including cellular-based therapies to target various anti-fibrotic molecular mechanisms and the potential to be integrated into existing treatment armamentarium for PD.
Collapse
Affiliation(s)
- Eric Chung
- AndroUrology Centre, Brisbane Qld and Sydney, NSW, Australia.,Department of Urology, University of Queensland, Princess Alexandra Hospital, Brisbane, QLD, Australia.,Department of Urology, Macquarie University Hospital, Sydney, NSW, Australia
| | - Faysal A Yafi
- Department of Urology, University of California Irvine, Orange, CA, USA
| |
Collapse
|
19
|
Plaut S. Scoping review and interpretation of myofascial pain/fibromyalgia syndrome: An attempt to assemble a medical puzzle. PLoS One 2022; 17:e0263087. [PMID: 35171940 PMCID: PMC8849503 DOI: 10.1371/journal.pone.0263087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Myofascial Pain Syndrome (MPS) is a common, overlooked, and underdiagnosed condition and has significant burden. MPS is often dismissed by clinicians while patients remain in pain for years. MPS can evolve into fibromyalgia, however, effective treatments for both are lacking due to absence of a clear mechanism. Many studies focus on central sensitization. Therefore, the purpose of this scoping review is to systematically search cross-disciplinary empirical studies of MPS, focusing on mechanical aspects, and suggest an organic mechanism explaining how it might evolve into fibromyalgia. Hopefully, it will advance our understanding of this disease. METHODS Systematically searched multiple phrases in MEDLINE, EMBASE, COCHRANE, PEDro, and medRxiv, majority with no time limit. Inclusion/exclusion based on title and abstract, then full text inspection. Additional literature added on relevant side topics. Review follows PRISMA-ScR guidelines. PROSPERO yet to adapt registration for scoping reviews. FINDINGS 799 records included. Fascia can adapt to various states by reversibly changing biomechanical and physical properties. Trigger points, tension, and pain are a hallmark of MPS. Myofibroblasts play a role in sustained myofascial tension. Tension can propagate in fascia, possibly supporting a tensegrity framework. Movement and mechanical interventions treat and prevent MPS, while living sedentarily predisposes to MPS and recurrence. CONCLUSIONS MPS can be seen as a pathological state of imbalance in a natural process; manifesting from the inherent properties of the fascia, triggered by a disrupted biomechanical interplay. MPS might evolve into fibromyalgia through deranged myofibroblasts in connective tissue ("fascial armoring"). Movement is an underemployed requisite in modern lifestyle. Lifestyle is linked to pain and suffering. The mechanism of needling is suggested to be more mechanical than currently thought. A "global percutaneous needle fasciotomy" that respects tensegrity principles may treat MPS/fibromyalgia more effectively. "Functional-somatic syndromes" can be seen as one entity (myofibroblast-generated-tensegrity-tension), sharing a common rheuma-psycho-neurological mechanism.
Collapse
Affiliation(s)
- Shiloh Plaut
- School of Medicine, St. George’s University of London, London, United Kingdom
| |
Collapse
|
20
|
Lewoniewska S, Oscilowska I, Forlino A, Palka J. Understanding the Role of Estrogen Receptor Status in PRODH/POX-Dependent Apoptosis/Survival in Breast Cancer Cells. BIOLOGY 2021; 10:biology10121314. [PMID: 34943229 PMCID: PMC8698543 DOI: 10.3390/biology10121314] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 04/15/2023]
Abstract
It has been suggested that activation of estrogen receptor α (ER α) stimulates cell proliferation. In contrast, estrogen receptor β (ER β) has anti-proliferative and pro-apoptotic activity. Although the role of estrogens in estrogen receptor-positive breast cancer progression has been well established, the mechanism of their effect on apoptosis is not fully understood. It has been considered that ER status of breast cancer cells and estrogen availability might determine proline dehydrogenase/proline oxidase (PRODH/POX)-dependent apoptosis. PRODH/POX is a mitochondrial enzyme that converts proline into pyrroline-5-carboxylate (P5C). During this process, ATP (adenosine triphosphate) or ROS (reactive oxygen species) are produced, facilitating cell survival or death, respectively. However, the critical factor in driving PRODH/POX-dependent functions is proline availability. The amount of this amino acid is regulated at the level of prolidase (proline releasing enzyme), collagen biosynthesis (proline utilizing process), and glutamine, glutamate, α-ketoglutarate, and ornithine metabolism. Estrogens were found to upregulate prolidase activity and collagen biosynthesis. It seems that in estrogen receptor-positive breast cancer cells, prolidase supports proline for collagen biosynthesis, limiting its availability for PRODH/POX-dependent apoptosis. Moreover, lack of free proline (known to upregulate the transcriptional activity of hypoxia-inducible factor 1, HIF-1) contributes to downregulation of HIF-1-dependent pro-survival activity. The complex regulatory mechanism also involves PRODH/POX expression and activity. It is induced transcriptionally by p53 and post-transcriptionally by AMPK (AMP-activated protein kinase), which is regulated by ERs. The review also discusses the role of interconversion of proline/glutamate/ornithine in supporting proline to PRODH/POX-dependent functions. The data suggest that PRODH/POX-induced apoptosis is dependent on ER status in breast cancer cells.
Collapse
Affiliation(s)
- Sylwia Lewoniewska
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Ilona Oscilowska
- Department of Analysis and Bioanalysis of Medicines, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Antonella Forlino
- Department of Molecular Medicine, University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy;
| | - Jerzy Palka
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-748-5706
| |
Collapse
|
21
|
Carver W, Fix E, Fix C, Fan D, Chakrabarti M, Azhar M. Effects of emodin, a plant-derived anthraquinone, on TGF-β1-induced cardiac fibroblast activation and function. J Cell Physiol 2021; 236:7440-7449. [PMID: 34041746 PMCID: PMC8530838 DOI: 10.1002/jcp.30416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022]
Abstract
Cardiac fibrosis accompanies a number of pathological conditions and results in altered myocardial structure, biomechanical properties and function. The signaling networks leading to fibrosis are complex, contributing to the general lack of progress in identifying effective therapeutic approaches to prevent or reverse this condition. Several studies have shown protective effects of emodin, a plant-derived anthraquinone, in animal models of fibrosis. A number of questions remain regarding the mechanisms whereby emodin impacts fibrosis. Transforming growth factor beta 1 (TGF-β1) is a potent stimulus of fibrosis and fibroblast activation. In the present study, experiments were performed to evaluate the effects of emodin on activation and function of cardiac fibroblasts following treatment with TGF-β1. We demonstrate that emodin attenuates TGF-β1-induced fibroblast activation and collagen accumulation in vitro. Emodin also inhibits activation of several canonical (SMAD2/3) and noncanonical (Erk1/2) TGF-β signaling pathways, while activating the p38 pathway. These results suggest that emodin may provide an effective therapeutic agent for fibrosis that functions via specific TGF-β signaling pathways.
Collapse
Affiliation(s)
- Wayne Carver
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Ethan Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Charity Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Mrinmay Chakrabarti
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| |
Collapse
|
22
|
Safety and clinical outcome of tamoxifen in Duchenne Muscular Dystrophy. Neuromuscul Disord 2021; 31:803-813. [PMID: 34304968 DOI: 10.1016/j.nmd.2021.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 01/25/2023]
Abstract
Patients having Duchenne Muscular Dystrophy (DMD) are currently being treated with corticosteroids, which slow down disease progression at the expense of serious adverse effects. Tamoxifen is a pro-drug some of whose metabolites interact with the nuclear estrogen receptor, leading to anti-fibrotic and muscle-protective effects as has been demonstrated in a murine model of DMD. Here we report the results from a monocentric single arm prospective study in 13 ambulant boys aged 6-14 years with genetically confirmed DMD, aimed to assess the safety of tamoxifen and its impact on disease progression. Boys were treated for up to 3 years with 20 mg/day of oral tamoxifen, in addition to their ongoing corticosteroid treatment. For 8 of these patients, outcome was compared to an age- and performance-matched 12-month natural history dataset. The primary end point was the 6-minute walk test. Secondary end points were the NorthStar assessment, timed function tests, pulmonary function, the biomarker creatine phosphokinase and adverse effects. No adverse effects were noticed other than mild gynecomastia in 4 boys. Tamoxifen-treated patients retained motor and respiratory function, compared with a significant deterioration of age-matched historical control patients receiving corticosteroids only. These encouraging findings warrant a larger clinical trial to substantiate the use of tamoxifen in Duchenne Muscular Dystrophy.
Collapse
|
23
|
Zhang F, Qin F, Yuan J. Molecular Mechanisms and Current Pharmacotherapy of Peyronie's Disease: A Review. Front Pharmacol 2021; 12:643641. [PMID: 34093182 PMCID: PMC8173627 DOI: 10.3389/fphar.2021.643641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/14/2021] [Indexed: 02/05/2023] Open
Abstract
Peyronie’s disease (PD) is a localized fibrotic lesion of the penis that has adverse effects on men’s health. In this review, we summarized the molecular mechanisms and pharmacotherapies of PD. A literature search was conducted using PubMed and Cochrane Library during 2001–2020. Although no oral or topical medication demonstrated efficacy in monotherapy of PD, several intralesional medications have yielded promising results. Currently, the effective strategy in management of PD should be combined modality therapy, including but not limited to pharmacotherapy, mechanical therapy, and psychotherapy. Meanwhile, basic research is still necessary to facilitate the development of novel and more reliable treatments. In future, more attention should be given simultaneously to epigenetic changes, inflammatory cytokines, the abnormal wound-healing process, and profibrotic and anti-fibrotic factors to provide more options for this refractory disease.
Collapse
Affiliation(s)
- Fuxun Zhang
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Jiuhong Yuan
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, China.,Department of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Blum KM, Roby LC, Zbinden JC, Chang YC, Mirhaidari GJM, Reinhardt JW, Yi T, Barker JC, Breuer CK. Sex and Tamoxifen confound murine experimental studies in cardiovascular tissue engineering. Sci Rep 2021; 11:8037. [PMID: 33850181 PMCID: PMC8044102 DOI: 10.1038/s41598-021-87006-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/25/2021] [Indexed: 02/01/2023] Open
Abstract
Tissue engineered vascular grafts hold promise for the creation of functional blood vessels from biodegradable scaffolds. Because the precise mechanisms regulating this process are still under investigation, inducible genetic mouse models are an important and widely used research tool. However, here we describe the importance of challenging the baseline assumption that tamoxifen is inert when used as a small molecule inducer in the context of cardiovascular tissue engineering. Employing a standard inferior vena cava vascular interposition graft model in C57BL/6 mice, we discovered differences in the immunologic response between control and tamoxifen-treated animals, including occlusion rate, macrophage infiltration and phenotype, the extent of foreign body giant cell development, and collagen deposition. Further, differences were noted between untreated males and females. Our findings demonstrate that the host-response to materials commonly used in cardiovascular tissue engineering is sex-specific and critically impacted by exposure to tamoxifen, necessitating careful model selection and interpretation of results.
Collapse
Affiliation(s)
- Kevin M Blum
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, USA
| | - Lauren C Roby
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- College of Medicine, The Ohio State University, Columbus, USA
| | - Jacob C Zbinden
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, USA
| | - Yu-Chun Chang
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, USA
| | - Gabriel J M Mirhaidari
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, USA
| | - James W Reinhardt
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
| | - Tai Yi
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
| | - Jenny C Barker
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, USA.
| |
Collapse
|
25
|
Tabeling C, Wienhold SM, Birnhuber A, Brack MC, Nouailles G, Kershaw O, Firsching TC, Gruber AD, Lienau J, Marsh LM, Olschewski A, Kwapiszewska G, Witzenrath M. Pulmonary fibrosis in Fra-2 transgenic mice is associated with decreased numbers of alveolar macrophages and increased susceptibility to pneumococcal pneumonia. Am J Physiol Lung Cell Mol Physiol 2021; 320:L916-L925. [PMID: 33655757 DOI: 10.1152/ajplung.00505.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a deadly condition characterized by progressive respiratory dysfunction. Exacerbations due to airway infections are believed to promote disease progression, and presence of Streptococcus in the lung microbiome has been associated with the progression of IPF and mortality. The aim of this study was to analyze the effect of lung fibrosis on susceptibility to pneumococcal pneumonia and bacteremia. The effects of subclinical (low dose) infection with Streptococcus pneumoniae were studied in a well characterized fos-related antigen-2 (Fra-2) transgenic (TG) mouse model of spontaneous, progressive pulmonary fibrosis. Forty-eight hours after transnasal infection with S. pneumoniae, bacterial load was assessed in lung tissue, bronchoalveolar lavage (BAL), blood, and spleen. Leukocyte subsets and cytokine levels were analyzed in BAL and blood. Lung compliance and arterial blood gases were assessed. In contrast to wildtype mice, low dose lung infection with S. pneumoniae in Fra-2 TG mice resulted in substantial pneumonia including weight loss, increased lung bacterial load, and bacteremia. BAL alveolar macrophages were reduced in Fra-2 TG mice compared to the corresponding WT mice. Proinflammatory cytokines and chemokines (IL-1β, IL-6, TNF-α, and CXCL1) were elevated upon infection in BAL supernatant and plasma of Fra-2 TG mice. Lung compliance was decreased in Fra-2 TG mice following low dose infection with S. pneumoniae. Pulmonary fibrosis increases susceptibility to pneumococcal pneumonia and bacteremia possibly via impaired alveolar bacterial clearance.
Collapse
Affiliation(s)
- Christoph Tabeling
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra-Maria Wienhold
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Markus C Brack
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Geraldine Nouailles
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olivia Kershaw
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Theresa C Firsching
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Jasmin Lienau
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Martin Witzenrath
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,German Center for Lung Research (DZL), Partner Site Charité, Berlin, Germany
| |
Collapse
|
26
|
Abarca-Buis RF, Mandujano-Tinoco EA, Cabrera-Wrooman A, Krötzsch E. The complexity of TGFβ/activin signaling in regeneration. J Cell Commun Signal 2021; 15:7-23. [PMID: 33481173 DOI: 10.1007/s12079-021-00605-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
The role of transforming growth factor β TGFβ/activin signaling in wound repair and regeneration is highly conserved in the animal kingdom. Various studies have shown that TGF-β/activin signaling can either promote or inhibit different aspects of the regeneration process (i.e., proliferation, differentiation, and re-epithelialization). It has been demonstrated in several biological systems that some of the different cellular responses promoted by TGFβ/activin signaling depend on the activation of Smad-dependent or Smad-independent signal transduction pathways. In the context of regeneration and wound healing, it has been shown that the type of R-Smad stimulated determines the different effects that can be obtained. However, neither the possible roles of Smad-independent pathways nor the interaction of the TGFβ/activin pathway with other complex signaling networks involved in the regenerative process has been studied extensively. Here, we review the important aspects concerning the TGFβ/activin signaling pathway in the regeneration process. We discuss data regarding the role of TGF-β/activin in the most common animal regenerative models to demonstrate how this signaling promotes or inhibits regeneration, depending on the cellular context.
Collapse
Affiliation(s)
- René Fernando Abarca-Buis
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico.
| | - Edna Ayerim Mandujano-Tinoco
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| |
Collapse
|
27
|
Rha EY, Kim JW, Kim JH, Yoo G. Angiotensin-Converting Enzyme Inhibitor, Captopril, Improves Scar Healing in Hypertensive Rats. Int J Med Sci 2021; 18:975-983. [PMID: 33456355 PMCID: PMC7807183 DOI: 10.7150/ijms.50197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/22/2020] [Indexed: 12/03/2022] Open
Abstract
Pathological cutaneous scars, with aberrant extracellular matrix accumulation, have multiple origins. Antihypertensive medications, such as calcium channel blockers, have been used to treat pathological scars. However, a relationship between angiotensin-converting enzyme (ACE) inhibitors, pathological scars, and blood pressure (BP) has never been reported. Here, we aimed to compare the differences in scar development and the effects of the administration of systemic ACE inhibitor on scar tissue in a normotensive rat, the Wistar Kyoto rat (WKY), a hypertensive rat, and the spontaneously hypertensive rat (SHR). Using an 8-mm punch, we created two full-thickness skin defects in a total of 32 rats (16 WKY and 16 SHR) to obtain a total of 64 wounds. We established control WKY (n = 16), captopril-treated WKY (n = 16), control SHR (n = 16), and captopril-treated SHR (n = 16) groups and started captopril (100 mg/g per day) treatment on day 21 in the appropriate groups. The BP of all groups was measured at 0, 3, and 5 weeks. The scar area was measured by histopathological examination, and scarring was expressed in terms of scar area and fibroblast and capillary counts. The expression of heat shock protein (HSP) 47, type I and III collagens, alpha-smooth muscle actin (α-SMA), Ki67, and vascular endothelial growth factor (VEGF) was investigated using immunohistochemistry. The scar area and fibroblast count were significantly higher in control SHR than in control WKY. The scar area, fibroblast count, and capillary count were significantly smaller in captopril-treated SHR than in control SHR. Immunostaining for α-SMA, Ki67, and VEGF also showed a noticeable decrease in scarring in the treated SHR compared with that in control SHR. Thus, BP affects scar development in a rat model, and an ACE inhibitor is more effective at reducing scars in hypertensive rats than in normotensive rats.
Collapse
Affiliation(s)
- Eun Young Rha
- Department of Plastic and Reconstructive Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Won Kim
- Department of Plastic and Reconstructive Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jun Hyeok Kim
- Department of Plastic and Reconstructive Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gyeol Yoo
- Department of Plastic and Reconstructive Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
28
|
Transforming Growth Factor-β Signaling in Fibrotic Diseases and Cancer-Associated Fibroblasts. Biomolecules 2020; 10:biom10121666. [PMID: 33322749 PMCID: PMC7763058 DOI: 10.3390/biom10121666] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling is essential in embryo development and maintaining normal homeostasis. Extensive evidence shows that TGF-β activation acts on several cell types, including epithelial cells, fibroblasts, and immune cells, to form a pro-fibrotic environment, ultimately leading to fibrotic diseases. TGF-β is stored in the matrix in a latent form; once activated, it promotes a fibroblast to myofibroblast transition and regulates extracellular matrix (ECM) formation and remodeling in fibrosis. TGF-β signaling can also promote cancer progression through its effects on the tumor microenvironment. In cancer, TGF-β contributes to the generation of cancer-associated fibroblasts (CAFs) that have different molecular and cellular properties from activated or fibrotic fibroblasts. CAFs promote tumor progression and chronic tumor fibrosis via TGF-β signaling. Fibrosis and CAF-mediated cancer progression share several common traits and are closely related. In this review, we consider how TGF-β promotes fibrosis and CAF-mediated cancer progression. We also discuss recent evidence suggesting TGF-β inhibition as a defense against fibrotic disorders or CAF-mediated cancer progression to highlight the potential implications of TGF-β-targeted therapies for fibrosis and cancer.
Collapse
|
29
|
Yamamoto A, Morioki H, Nakae T, Miyake Y, Harada T, Noda S, Mitsuoka S, Matsumoto K, Tomimatsu M, Kanemoto S, Tanaka S, Maeda M, Conway SJ, Imaizumi K, Fujio Y, Obana M. Transcription factor old astrocyte specifically induced substance is a novel regulator of kidney fibrosis. FASEB J 2020; 35:e21158. [PMID: 33150680 PMCID: PMC7821213 DOI: 10.1096/fj.202001820r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/07/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Prevention of kidney fibrosis is an essential requisite for effective therapy in preventing chronic kidney disease (CKD). Here, we identify Old astrocyte specifically induced substance (OASIS)/cAMP responsive element‐binding protein 3‐like 1 (CREB3l1), a CREB/ATF family transcription factor, as a candidate profibrotic gene that drives the final common pathological step along the fibrotic pathway in CKD. Although microarray data from diseased patient kidneys and fibrotic mouse model kidneys both exhibit OASIS/Creb3l1 upregulation, the pathophysiological roles of OASIS in CKD remains unknown. Immunohistochemistry revealed that OASIS protein was overexpressed in human fibrotic kidney compared with normal kidney. Moreover, OASIS was upregulated in murine fibrotic kidneys, following unilateral ureteral obstruction (UUO), resulting in an increase in the number of OASIS‐expressing pathological myofibroblasts. In vitro assays revealed exogenous TGF‐β1 increased OASIS expression coincident with fibroblast‐to‐myofibroblast transition and OASIS contributed to TGF‐β1–mediated myofibroblast migration and increased proliferation. Significantly, in vivo kidney fibrosis induced via UUO or ischemia/reperfusion injury was ameliorated by systemic genetic knockout of OASIS, accompanied by reduced myofibroblast proliferation. Microarrays revealed that the transmembrane glycoprotein Bone marrow stromal antigen 2 (Bst2) expression was reduced in OASIS knockout myofibroblasts. Interestingly, a systemic anti‐Bst2 blocking antibody approach attenuated kidney fibrosis in normal mice but not in OASIS knockout mice after UUO, signifying Bst2 functions downstream of OASIS. Finally, myofibroblast‐restricted OASIS conditional knockouts resulted in resistance to kidney fibrosis. Taken together, OASIS in myofibroblasts promotes kidney fibrosis, at least in part, via increased Bst2 expression. Thus, we have identified and demonstrated that OASIS signaling is a novel regulator of kidney fibrosis.
Collapse
Affiliation(s)
- Ayaha Yamamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Hitomi Morioki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Takafumi Nakae
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Yoshiaki Miyake
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Takeo Harada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Shunsuke Noda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Sayuri Mitsuoka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Kotaro Matsumoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Masashi Tomimatsu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Soshi Kanemoto
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Makiko Maeda
- Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.,Radioisotope Research Center, Institute for Radiation Sciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
30
|
Critical role of estrogen in the progression of chronic liver diseases. Hepatobiliary Pancreat Dis Int 2020; 19:429-434. [PMID: 32299655 DOI: 10.1016/j.hbpd.2020.03.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Estrogens regulate sexual function and also have a significant role in various pathophysiological processes. Estrogens have a non-reproductive role as the modulators of the immune system, growth, neuronal function, and metabolism. Estrogen receptors are expressed in the liver and their impaired expression and function are implicated with obesity and liver associated metabolic dysfunctions. The purpose of the current review is to discuss the disparity role of estrogens on several forms of liver diseases. DATA SOURCES A comprehensive search in PubMed and EMBASE was conducted using the keywords "estrogens and liver diseases", "estradiol and liver diseases", "hormones and liver diseases", "endocrine function in liver diseases", and "female hormones in liver diseases". Relevant papers published before September 30, 2019 were included. RESULTS The present review confirms the imperative role of estrogen in various forms of chronic liver diseases. Estrogens play a key role in maintaining homeostasis and make the liver less susceptible to several forms of chronic liver diseases in healthy premenopausal individuals. In contrast, clinical studies also showed increased estrogen levels with chronic liver diseases. CONCLUSIONS Several studies reported the protective role of estrogens in chronic liver diseases and this has been widely accepted and confirmed in experimental studies using ovariectomized rat models. However, in a few clinical studies, increased estrogen levels are also implicated in chronic liver diseases. Therefore, further studies are warranted at molecular level to explore the role of estrogen in various forms of chronic liver diseases.
Collapse
|
31
|
Ilg MM, Stafford SJ, Mateus M, Bustin SA, Carpenter MJ, Muneer A, Bivalacqua TJ, Ralph DJ, Cellek S. Phosphodiesterase Type 5 Inhibitors and Selective Estrogen Receptor Modulators Can Prevent But Not Reverse Myofibroblast Transformation in Peyronie's Disease. J Sex Med 2020; 17:1848-1864. [PMID: 32771352 DOI: 10.1016/j.jsxm.2020.06.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Myofibroblast transformation is a key step in the pathogenesis of Peyronie's disease (PD). Phosphodiesterase type 5 inhibitors (PDE5is) and selective estrogen receptor modulators (SERMs) can prevent the formation of fibrosis in in vitro and in vivo models of PD. However, it is unknown whether these drugs can also reverse established fibrosis. AIM To investigate whether PDE5is and SERMs can reverse transforming growth factor beta 1 (TGF-β1)-induced myofibroblast transformation and determine the point of no return. METHODS In-Cell enzyme-linked immunosorbent assay was used to quantify TGF-β1-induced myofibroblast transformation of human primary fibroblasts isolated from tunica albuginea (TA) of patients undergoing surgery for treatment of PD. Extracellular matrix production and collagen contraction assays were used as secondary assays. Reverse transcription-quantitative polymerase chain reaction and In-Cell enzyme-linked immunosorbent assay were used to measure drug target expression. PDE5i (vardenafil) and SERM (tamoxifen) were applied at various time points after TGF-β1. OUTCOMES Reversibility of myofibroblast transformation and drug target expression were investigated in a time-dependent manner in TA-derived fibroblasts. RESULTS Vardenafil or tamoxifen could not reverse the myofibroblast traits of alpha-smooth muscle actin expression and extracellular matrix production, whereas only tamoxifen affected collagen contraction after 72 hours of TGF-β1 treatment. Phosphodiesterase 5A and estrogen receptor (ER)-β were downregulated after 72 hours, and estrogen receptor -α protein could not be quantified. Tamoxifen could prevent myofibroblast transformation until 36 hours after TGF-β1 treatment, whereas vardenafil could prevent only 24 hours after TGF-β1 treatment. This was mirrored by downregulation of drug targets on mRNA and protein level. Furthermore, antifibrotic signaling pathways, peroxisome proliferator-activated receptor gamma and betaglycan (TGFB receptor III), were significantly downregulated after 36 hours of TGF-β1 exposure, as opposed to upregulation of profibrotic thrombospondin-1 at the same time point. CLINICAL TRANSLATION This study suggests that using PDE5is and SERMs might only help for early-phase PD and further highlights the need to test drugs at the appropriate stage of the disease based on their mechanism of action. STRENGTHS & LIMITATIONS The study uses primary human TA-derived fibroblasts that enhances translatability of the results. Limitations include that only 1 example of PDE5i- and SERM-type drug was tested. Time course experiments were only performed for marker expression experiments and not for functional assays. CONCLUSION This is the first study to demonstrate that timing for administration of drugs affecting myofibroblast transformation appears to be vital in in vitro models of PD, where 36 hours of TGF-β1 treatment can be suggested as a "point of no return" for myofibroblast transformation. Ilg MM, Stafford SJ, Mateus M, et al. Phosphodiesterase Type 5 Inhibitors and Selective Estrogen Receptor Modulators Can Prevent But Not Reverse Myofibroblast Transformation in Peyronie's Disease. J Sex Med 2020;17:1848-1864.
Collapse
Affiliation(s)
- Marcus M Ilg
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK.
| | - Simon J Stafford
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Marta Mateus
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Stephen A Bustin
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Michael J Carpenter
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| | - Asif Muneer
- Department of Urology, University College London, London, UK; NIHR Biomedical Research Centre, University College London, London, UK
| | - Trinity J Bivalacqua
- James Buchanan Brady Urologic Institute, John Hopkins University, Baltimore, MD, USA
| | - David J Ralph
- Department of Urology, University College London, London, UK
| | - Selim Cellek
- Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, Essex, UK
| |
Collapse
|
32
|
Napolitano A, Mazzocca A, Spalato Ceruso M, Minelli A, Baldo F, Badalamenti G, Silletta M, Santini D, Tonini G, Incorvaia L, Vincenzi B. Recent Advances in Desmoid Tumor Therapy. Cancers (Basel) 2020; 12:cancers12082135. [PMID: 32752153 PMCID: PMC7463981 DOI: 10.3390/cancers12082135] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
The desmoid tumor is a locally aggressive proliferative disease within the family of soft-tissue sarcomas. Despite its relatively good prognosis, the clinical management of desmoid tumors requires constant multidisciplinary evaluation due to its highly variable clinical behavior. Recently, active surveillance has being regarded as the appropriate strategy at diagnosis, as indolent persistence or spontaneous regressions are not uncommon. Here, we review the most recent advances in desmoid tumor therapy, including low-dose chemotherapy and treatment with tyrosine kinase inhibitors. We also explore the recent improvements in our knowledge of the molecular biology of this disease, which are leading to clinical trials with targeted agents.
Collapse
Affiliation(s)
- Andrea Napolitano
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Alessandro Mazzocca
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Mariella Spalato Ceruso
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Alessandro Minelli
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Francesca Baldo
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Giuseppe Badalamenti
- Department of Medical Oncology, Policlinico “Paolo Giaccone”, 90127 Palermo, Italy; (G.B.); (L.I.)
| | - Marianna Silletta
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Daniele Santini
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Giuseppe Tonini
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
| | - Lorena Incorvaia
- Department of Medical Oncology, Policlinico “Paolo Giaccone”, 90127 Palermo, Italy; (G.B.); (L.I.)
| | - Bruno Vincenzi
- Department of Medical Oncology, Università Campus Bio-Medico, 00128 Rome, Italy; (A.N.); (A.M.); (M.S.C.); (A.M.); (F.B.); (M.S.); (D.S.); (G.T.)
- Correspondence: ; Tel.: +39-06-225411227
| |
Collapse
|
33
|
Krakhotkin DV, Chernylovskyi VA, Mottrie A, Greco F, Bugaev RA. New insights into the pathogenesis of Peyronie's disease: A narrative review. Chronic Dis Transl Med 2020; 6:165-181. [PMID: 32885153 PMCID: PMC7451633 DOI: 10.1016/j.cdtm.2020.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Indexed: 12/11/2022] Open
Abstract
Peyronie's disease (PD) is a benign, progressive fibrotic disorder characterized by scar or plaques within the tunica albuginea (TA) of the penis. This study provides new insights into the pathogenesis of PD based on data from different studies regarding the roles of cytokines, cell signaling pathways, biochemical mechanisms, genetic factors responsible for fibrogenesis. A growing body of literature has shown that PD is a chronically impaired, localized, wound healing process within the TA and the Smith space. It is caused by the influence of different pathological stimuli, most often the effects of mechanical stress during sexual intercourse in genetically sensitive individuals with unusual anatomical TA features, imbalanced matrix metalloproteinase/tissue inhibitor of metalloproteinase (MMP/TIMP), and suppressed antioxidant systems during chronic inflammation. Other intracellular signal cascades are activated during fibrosis along with low expression levels of their negative regulators and transforming growth factor-β1 signaling. The development of multikinase agents with minimal side effects that can block several signal cell pathways would significantly improve fibrosis in PD tissues by acting on common downstream mediators.
Collapse
Affiliation(s)
- Denis V Krakhotkin
- Outpatient Department, Central District Hospital, Kamenolomni, Rostov Region, Russia
| | | | - Alexandre Mottrie
- Department of Urology, Onze Lieve Vrouw Hospital, Aalst, Belgium.,ORSI Academy, Melle, Belgium
| | | | - Ruslan A Bugaev
- Outpatient Department, Central District Hospital, Kamenolomni, Rostov Region, Russia
| |
Collapse
|
34
|
Reprogramming of Mesothelial-Mesenchymal Transition in Chronic Peritoneal Diseases by Estrogen Receptor Modulation and TGF-β1 Inhibition. Int J Mol Sci 2020; 21:ijms21114158. [PMID: 32532126 PMCID: PMC7312018 DOI: 10.3390/ijms21114158] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
In chronic peritoneal diseases, mesothelial-mesenchymal transition is determined by cues from the extracellular environment rather than just the cellular genome. The transformation of peritoneal mesothelial cells and other host cells into myofibroblasts is mediated by cell membrane receptors, Transforming Growth Factor β1 (TGF-β1), Src and Hypoxia-inducible factor (HIF). This article provides a narrative review of the reprogramming of mesothelial mesenchymal transition in chronic peritoneal diseases, drawing on the similarities in pathophysiology between encapsulating peritoneal sclerosis and peritoneal metastasis, with a particular focus on TGF-β1 signaling and estrogen receptor modulators. Estrogen receptors act at the cell membrane/cytosol as tyrosine kinases that can phosphorylate Src, in a similar way to other receptor tyrosine kinases; or can activate the estrogen response element via nuclear translocation. Tamoxifen can modulate estrogen membrane receptors, and has been shown to be a potent inhibitor of mesothelial-mesenchymal transition (MMT), peritoneal mesothelial cell migration, stromal fibrosis, and neoangiogenesis in the treatment of encapsulating peritoneal sclerosis, with a known side effect and safety profile. The ability of tamoxifen to inhibit the transduction pathways of TGF-β1 and HIF and achieve a quiescent peritoneal stroma makes it a potential candidate for use in cancer treatments. This is relevant to tumors that spread to the peritoneum, particularly those with mesenchymal phenotypes, such as colorectal CMS4 and MSS/EMT gastric cancers, and pancreatic cancer with its desmoplastic stroma. Morphological changes observed during mesothelial mesenchymal transition can be treated with estrogen receptor modulation and TGF-β1 inhibition, which may enable the regression of encapsulating peritoneal sclerosis and peritoneal metastasis.
Collapse
|
35
|
Hu J, Chen Y, Huang Y, Su Y. Human umbilical cord mesenchymal stem cell-derived exosomes suppress dermal fibroblasts-myofibroblats transition via inhibiting the TGF-β1/Smad 2/3 signaling pathway. Exp Mol Pathol 2020; 115:104468. [PMID: 32445750 DOI: 10.1016/j.yexmp.2020.104468] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/08/2020] [Accepted: 05/17/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Exosomes originated from mesenchymal stem cells (MSCs) benefit wound healing. This study investigated effects of exosomes originated from human umbilical cord MSCs (hUC-MSCs) on dermal fibroblasts-myofibroblasts transition via the TGF-β1/Smad2/3 signaling pathway. METHODS Firstly, hUC-MSCs were collected and identified. Alizarin red, oil red O staining and toluidine blue staining were used to determine the osteogenic, adipogenic and chondrogenic differentiation abilities of hUC-MSCs. Then exosomes from hUC-MSCs were extracted and identified. To figure out the roles of exosomes and TGF-β1 in dermal fibroblasts-myofibroblasts transition, dermal fibroblasts were treated with TGF-β1 or/and exosomes at different concentrations. RT-qPCR, Western blot analyses were employed to examine levels of Collagen I, Collagen III, α-smooth muscle actin (α-SMA), and Smad2/3 phosphorylation, and immunofluorescence was employed to test α-SMA content and the localization and nucleation of Smad2/3 protein in cells. RESULTS hUC-MSCs and exosomes were successfully cultured and extracted. Levels of Collagen I, Collagen III, α-SMA, and Smad2/3, and Smad2/3 phosphorylation in fibroblasts treated with exosomes decreased markedly. After treatment with exosomes and TGF-β1 together, levels of Collagen I, Collagen III, α-SMA, and Smad2/3, and Smad2/3 phosphorylation in fibroblasts decreased significantly as compared to TGF-β1-treated fibroblasts. Exosome treatment reduced the entry of Smad2/3 into fibroblasts. CONCLUSION Our data suggested that hUC-MSCs-derived exosomes could inhibit dermal fibroblasts-myofibroblasts transition by inhibiting the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Jian Hu
- Department of Burn and Plastic Surgery, the People's Hospital of Bao'an Shenzhen, Shenzhen 518101, PR China
| | - Yuanwen Chen
- Department of Burn and Plastic Surgery, the People's Hospital of Bao'an Shenzhen, Shenzhen 518101, PR China
| | - Yubin Huang
- Department of Burn and Plastic Surgery, the People's Hospital of Bao'an Shenzhen, Shenzhen 518101, PR China
| | - Yongsheng Su
- Department of Burn and Plastic Surgery, the People's Hospital of Bao'an Shenzhen, Shenzhen 518101, PR China.
| |
Collapse
|
36
|
Liu X, Zhang T, Zhang C. Sitagliptin Inhibits Extracellular Matrix Accumulation and Proliferation in Lung Fibroblasts. Med Sci Monit 2020; 26:e922644. [PMID: 32301442 PMCID: PMC7191949 DOI: 10.12659/msm.922644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Fibroblasts activation-induced fibrosis can cause idiopathic pulmonary fibrosis (IPF). Excessive activation of fibroblasts contributes to poor healing or severe visceral fibrosis and even organ dysfunction. Sitagliptin acts as a dipeptidyl peptidase 4 inhibitor to reduce glucose level in type 2 diabetes, but its role in fibrosis of lung fibroblasts is elusive. We investigated the mechanism of sitagliptin in TGF-ß-activated lung fibroblasts and evaluated the efficacy of sitagliptin in extracellular matrix accumulation and fibroblasts proliferation. MATERIAL AND METHODS By in vitro lung fibroblasts culture, we assessed the expression of lung fibroblasts biomarker (alpha-SMA) and extracellular matrix (Col-1, Col-3, fibronectin) following TGF-ß stimulation and treatment with sitagliptin. Mechanistically, the phosphorylation level of Smad-3 protein in cells was analyzed using Western blotting, and the apoptosis level was assessed by Western blotting and flow cytometry. The degree of proliferation was determined using immunofluorescence and scratch-healing assay. RESULTS We found that treatment with sitagliptin attenuates fibroblasts activation following TGF-ß stimulation. Furthermore, the extracellular matrix was decreased by sitagliptin treatment by suppressing the phosphorylation level of Smad-3 protein. We found that sitagliptin does not affect apoptosis in fibroblasts, but it does affect the degree of proliferation of lung fibroblasts, thus ameliorating fibrosis after TGF-ß stimulation. CONCLUSIONS Sitagliptin inhibits fibrosis in TGF-ß-induced lung fibroblasts activation, which restrains extracellular matrix formation and cell proliferation in fibroblasts. Therefore, sitagliptin appears to have promise as a treatment of fibroproliferative disease caused by activation and proliferation of fibroblasts.
Collapse
Affiliation(s)
- Xiuwu Liu
- Department of Internal Medicine, Linyi People's Hospital, Linyi, Shandong, China (mainland)
| | - Tao Zhang
- Department of Respiratory Medicine, Linyi Jinluo Hospital, Linyi, Shandong, China (mainland)
| | - Chengcai Zhang
- Department of Respiratory and Critical Care Medicine, Linyi High-Tech Hospital, Linyi, Shandong, China (mainland)
| |
Collapse
|
37
|
Farrell MR, Ziegelmann MJ, Levine LA. Minimally invasive therapies for Peyronie's disease: the current state of the art. Transl Androl Urol 2020; 9:S269-S283. [PMID: 32257868 PMCID: PMC7108992 DOI: 10.21037/tau.2019.08.06] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 07/25/2019] [Indexed: 01/18/2023] Open
Abstract
Peyronie's disease (PD) is a relatively common condition that can result in significant penile deformity, sexual dysfunction, and psychological bother. Surgical straightening offers the highest probability of success during the stable phase of the disease. However, for men in the acute phase of PD or for those with less severe deformity who elect to avoid surgery, a variety of non-surgical treatment options are available. Oral therapies, including L-citrulline and pentoxifylline, are most useful as part of a combination regimen rather than as monotherapy. Intralesional therapy with IFN-α2b, verapamil, and collagenase clostridium histolyticum (CCH) can cause significant reduction in penile curvature, yet these results may not be clinically significant for men with more severe curvature. Further investigation into the timing of administration and optimal patient characteristics is required. Penile traction therapy offers a clinically significant improvement in penile length and curvature. However, this has traditionally required hours of daily therapy. Overall, a combination of oral, topical, injection and traction therapies may provide the most significant benefit among the non-surgical modalities for PD.
Collapse
Affiliation(s)
- M Ryan Farrell
- Division of Urology, Rush University Medical Center, Chicago, IL, USA
| | | | - Laurence A Levine
- Division of Urology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Hu LF, Feng J, Dai X, Sun Y, Xiong M, Lai L, Zhong S, Yi C, Chen G, Li H, Yang Q, Kuang Q, Long T, Zhan J, Tang T, Ge C, Tan J, Xu M. Oral flavonoid fisetin treatment protects against prolonged high-fat-diet-induced cardiac dysfunction by regulation of multicombined signaling. J Nutr Biochem 2019; 77:108253. [PMID: 31835147 DOI: 10.1016/j.jnutbio.2019.108253] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 08/03/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
Excess high-fat diet (HFD) intake predisposes the occurrence of obesity-associated heart injury, but the mechanism is elusive. Fisetin (FIS), as a natural flavonoid, has potential activities to alleviate obesity-induced metabolic syndrome. However, the underlying molecular mechanisms of FIS against HFD-induced cardiac injury remain unclear. The present study was to explore the protective effects of FIS on cardiac dysfunction in HFD-fed mice. We found that FIS alleviated HFD-triggered metabolic disorder by reducing body weight, fasting blood glucose and insulin levels, and insulin resistance. Moreover, FIS supplements significantly alleviated dyslipidemia in both mouse hearts and cardiomyocytes stimulated by metabolic stress. FIS treatment abolished HFD-induced inflammatory response in heart tissues through suppressing TNF receptor-1/TNF receptor-associated factor-2 (Tnfr-1/Traf-2) signaling. Furthermore, FIS induced a strong reduction in the expression of fibrosis-related genes, contributing to the inhibition of fibrosis by inactivating transforming growth factor (Tgf)-β1/Smads/Erk1/2 signaling. Collectively, these results demonstrated that FIS could be a promising therapeutic strategy for the treatment of obesity-associated cardiac injury.
Collapse
Affiliation(s)
- Lin-Feng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Jing Feng
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Xianling Dai
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Yan Sun
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Mingxin Xiong
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Lili Lai
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Shaoyu Zhong
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Chao Yi
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Geng Chen
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Huanhuan Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Qiufeng Yang
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Qin Kuang
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Tingting Long
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Jianxia Zhan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Tingting Tang
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China
| | - Chenxu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China.
| | - Minxuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing 400067, PR China.
| |
Collapse
|
39
|
Dewidar B, Meyer C, Dooley S, Meindl-Beinker N. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019; 8:cells8111419. [PMID: 31718044 PMCID: PMC6912224 DOI: 10.3390/cells8111419] [Citation(s) in RCA: 525] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is an advanced liver disease condition, which could progress to cirrhosis and hepatocellular carcinoma. To date, there is no direct approved antifibrotic therapy, and current treatment is mainly the removal of the causative factor. Transforming growth factor (TGF)-β is a master profibrogenic cytokine and a promising target to treat fibrosis. However, TGF-β has broad biological functions and its inhibition induces non-desirable side effects, which override therapeutic benefits. Therefore, understanding the pleiotropic effects of TGF-β and its upstream and downstream regulatory mechanisms will help to design better TGF-β based therapeutics. Here, we summarize recent discoveries and milestones on the TGF-β signaling pathway related to liver fibrosis and hepatic stellate cell (HSC) activation, emphasizing research of the last five years. This comprises impact of TGF-β on liver fibrogenesis related biological processes, such as senescence, metabolism, reactive oxygen species generation, epigenetics, circadian rhythm, epithelial mesenchymal transition, and endothelial-mesenchymal transition. We also describe the influence of the microenvironment on the response of HSC to TGF-β. Finally, we discuss new approaches to target the TGF-β pathway, name current clinical trials, and explain promises and drawbacks that deserve to be adequately addressed.
Collapse
Affiliation(s)
- Bedair Dewidar
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, 31527 Tanta, Egypt
| | - Christoph Meyer
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Nadja Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Correspondence: ; Tel.: +49-621-383-4983; Fax: +49-621-383-1467
| |
Collapse
|
40
|
Birnhuber A, Biasin V, Schnoegl D, Marsh LM, Kwapiszewska G. Transcription factor Fra-2 and its emerging role in matrix deposition, proliferation and inflammation in chronic lung diseases. Cell Signal 2019; 64:109408. [PMID: 31473307 DOI: 10.1016/j.cellsig.2019.109408] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
Fos-related antigen-2 (Fra-2) belongs to the activator protein 1 (AP-1) family of transcription factors and is involved in a broad variety of cellular processes, such as proliferation or differentiation. Aberrant expression of Fra-2 or regulation can lead to severe growth defects or diverse pathologies. Elevated Fra-2 expression has been described in several chronic lung diseases, such as pulmonary fibrosis, chronic obstructive pulmonary disease and asthma. However, the pathomechanisms behind the Fra-2-induced pulmonary remodelling are still not fully elucidated. Fra-2 overexpressing mice were initially described as a model of systemic sclerosis associated organ fibrosis, with predominant alterations in the lung. High levels of Fra-2 expression give rise to profound inflammation with severe remodelling of the parenchyma and the vasculature, resulting in fibrosis and pulmonary hypertension, respectively, but also alters bronchial function. In this review we discuss the central role of Fra-2 connecting inflammation, cellular proliferation and extracellular matrix deposition underlying chronic lung diseases and what we can learn for future therapeutic options.
Collapse
Affiliation(s)
- A Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - V Biasin
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - D Schnoegl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - L M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - G Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria; Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
| |
Collapse
|
41
|
Li X, Clappier C, Kleiter I, Heuchel R. Tamoxifen affects chronic pancreatitis-related fibrogenesis in an experimental mouse model: an effect beyond Cre recombination. FEBS Open Bio 2019; 9:1756-1768. [PMID: 31380604 PMCID: PMC6768287 DOI: 10.1002/2211-5463.12714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/13/2019] [Accepted: 08/02/2019] [Indexed: 01/20/2023] Open
Abstract
Tamoxifen is very successfully used for the induction of CreERT‐mediated genomic recombination in conditional mouse models. Recent studies, however, indicated that tamoxifen might also affect the fibrotic response in several disease models following administration, both in vitro and in vivo. In order to investigate a possible effect of tamoxifen on pancreatic fibrogenesis and to evaluate an optimal treatment scheme in an experimental pancreatitis mouse model, we administered tamoxifen by oral gavage to both male and female C57BL/6J mice and then waited for different periods of time before inducing chronic pancreatitis by cerulein. We observed a sex‐specific and time‐dependent effect of tamoxifen on the fibrotic response as measured by collagen deposition and the number of myofibroblasts and macrophages. The findings of in vitro studies, in which cerulein was administrated with or without 4‐hydroxytamoxifen to stimulate primary murine female and male pancreatic stellate cells, supported our in vivo observations. Real‐time PCR also indicated that this effect may be related to differences in ERα expression between female and male stellate cells. Our data demonstrate that tamoxifen administration has unignorable side effects, which affect the experimental outcome in a cerulein‐based model of chronic pancreatitis in mice. We suggest a 2‐week waiting period before cerulein administration to reduce side effects to a minimum for the described fibrosis model in female mice.
Collapse
Affiliation(s)
- Xuan Li
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Clappier
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ingo Kleiter
- Department of Neurology, Ruhr-Universität Bochum, Germany
| | - Rainer Heuchel
- Pancreas Cancer Research (PaCaRes) Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Wu B, Shah SN, Lu P, Bollinger LE, Blaeser A, Sparks S, Harper AD, Lu QL. Long-Term Treatment of Tamoxifen and Raloxifene Alleviates Dystrophic Phenotype and Enhances Muscle Functions of FKRP Dystroglycanopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 188:1069-1080. [PMID: 29571322 DOI: 10.1016/j.ajpath.2017.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/14/2017] [Accepted: 12/19/2017] [Indexed: 01/06/2023]
Abstract
The third most common form of limb-girdle muscular dystrophies is caused by mutations of the Fukutin-related protein (FKRP) gene, with no effective therapy available. Selective estrogen receptor modulators, tamoxifen and raloxifene, have been widely used for human conditions for their anti-inflammatory, antifibrosis, prevention of bone loss, and muscle building effects (essential features for muscular dystrophy therapies). We evaluated therapeutic values of tamoxifen and raloxifene in FKRPP448L mutant mouse with severe dystrophic phenotype. The mice were treated with the drugs for 1 year through daily gavage. We demonstrate that tamoxifen and raloxifene significantly ameliorated the disease progression. The improvement includes increase in grip force production, extended running time and distance in treadmill test, and enhancement in cardiac and respiratory functions. Significant reduction in muscle pathology includes diminished fibrosis and fiber degeneration. Tamoxifen and raloxifene also significantly mitigated bone loss. Tamoxifen, but not raloxifene, caused severe adverse effects on male reproductive organs. The results demonstrate that tamoxifen and raloxifene hold significant potential for treating FKRP-related muscular dystrophy and probably other muscular dystrophies. Sex-related differential effects of the drugs call for a careful consideration for the drug and dosage selection in male and female patient populations.
Collapse
Affiliation(s)
- Bo Wu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina.
| | - Sapana N Shah
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Peijuan Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Lauren E Bollinger
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Anthony Blaeser
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Susan Sparks
- Clinical Genetics/Department of Pediatrics, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Amy D Harper
- Clinical Genetics/Department of Pediatrics, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina
| | - Qi L Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Levine Children's Hospital, Carolinas Medical Center, Charlotte, North Carolina.
| |
Collapse
|
43
|
Zhang Y, Wang J, Zhou S, Xie Z, Wang C, Gao Y, Zhou J, Zhang X, Li Q. Flavones hydroxylated at 5, 7, 3' and 4' ameliorate skin fibrosis via inhibiting activin receptor-like kinase 5 kinase activity. Cell Death Dis 2019; 10:124. [PMID: 30741930 PMCID: PMC6370799 DOI: 10.1038/s41419-019-1333-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/10/2018] [Accepted: 12/21/2018] [Indexed: 01/10/2023]
Abstract
Skin fibrosis is mainly characterized by excessive collagen deposition. Studies have recently identified a number of flavonoids with variable structures that have the potency of inhibiting collagen synthesis and thus attenuating organ fibrosis. In this study, we found that flavones with 5, 7, 3', 4' hydroxy substitution reduced collagen expression most efficiently. Among those flavones, luteolin, quercetin, and myricetin were selected for follow-up. In vivo, the three compounds ameliorated skin fibrosis and reduced collagen deposition. Further analysis showed the compounds had significant inhibition on the proliferation, activation and contractile ability of dermal fibroblasts in vitro and in vivo. More importantly, we revealed that luteolin, quercetin, and myricetin selectively downregulated the phosphorylation of Smad2/3 in TGF-β/Smads signaling via binding to activin receptor-like kinase 5 (ALK5) and impairing its catalytic activity. We also found flavones with 5, 7, 3', 4' hydroxy substitution showed stronger affinity with ALK5 compared with other flavonoids. Herein, we identified at least in part the underlying molecular basis as well as the critical structures that contribute to the antifibrotic bioactivity of flavones, which might benefit drug design and modification.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sizheng Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhibo Xie
- Department of Pancreatic Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuandong Wang
- Stem Cell and Regenerative Medicine Lab Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Gao
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Zhou
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoling Zhang
- Stem Cell and Regenerative Medicine Lab Department of Orthopedic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingfeng Li
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
44
|
Ilg MM, Mateus M, Stebbeds WJ, Milenkovic U, Christopher N, Muneer A, Albersen M, Ralph DJ, Cellek S. Antifibrotic Synergy Between Phosphodiesterase Type 5 Inhibitors and Selective Oestrogen Receptor Modulators in Peyronie's Disease Models. Eur Urol 2019; 75:329-340. [DOI: 10.1016/j.eururo.2018.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/04/2018] [Indexed: 01/09/2023]
|
45
|
Mitz CA, Viloria-Petit AM. TGF-beta signalling in bovine mammary gland involution and a comparative assessment of MAC-T and BME-UV1 cells as in vitro models for its study. PeerJ 2019; 6:e6210. [PMID: 30671288 PMCID: PMC6338098 DOI: 10.7717/peerj.6210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022] Open
Abstract
The goal of the dairy industry is ultimately to increase lactation persistency, which is the length of time during which peak milk yield is sustained. Lactation persistency is determined by the balance of cell apoptosis and cell proliferation; when the balance is skewed toward the latter, this results in greater persistency. Thus, we can potentially increase milk production in dairy cows through manipulating apoptogenic and antiproliferative cellular signaling that occurs in the bovine mammary gland. Transforming growth factor beta 1 (TGFβ1) is an antiproliferative and apoptogenic cytokine that is upregulated during bovine mammary gland involution. Here, we discuss possible applications of TGFβ1 signaling for the purposes of increasing lactation persistency. We also compare the features of mammary alveolar cells expressing SV-40 large T antigen (MAC-T) and bovine mammary epithelial cells-clone UV1 (BME-UV1) cells, two extensively used bovine mammary epithelial cell lines, to assess their appropriateness for the study of TGFβ1 signaling. TGFβ1 induces apoptosis and arrests cell growth in BME-UV1 cells, and this was reported to involve suppression of the somatotropic axis. Conversely, there is no proof that exogenous TGFβ1 induces apoptosis of MAC-T cells. In addition to TGFβ1's different effects on apoptosis in these cell lines, hormones and growth factors have distinct effects on TGFβ1 secretion and synthesis in MAC-T and BME-UV1 cells as well. MAC-T and BME-UV1 cells may behave differently in response to TGFβ1 due to their contrasting phenotypes; MAC-T cells have a profile indicative of both myoepithelial and luminal populations, while the BME-UV1 cells exclusively contain a luminal-like profile. Depending on the nature of the research question, the use of these cell lines as models to study TGFβ1 signaling should be carefully tailored to the questions asked.
Collapse
Affiliation(s)
- Charlotte Alexandra Mitz
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | |
Collapse
|
46
|
Fix C, Carver-Molina A, Chakrabarti M, Azhar M, Carver W. Effects of the isothiocyanate sulforaphane on TGF-β1-induced rat cardiac fibroblast activation and extracellular matrix interactions. J Cell Physiol 2019; 234:13931-13941. [PMID: 30609032 DOI: 10.1002/jcp.28075] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/11/2018] [Indexed: 12/30/2022]
Abstract
An important step in many pathological conditions, particularly tissue and organ fibrosis, is the conversion of relatively quiescent cells into active myofibroblasts. These are highly specialized cells that participate in normal wound healing but also contribute to pathogenesis. These cells possess characteristics of smooth muscle cells and fibroblasts, have enhanced synthetic activity secreting abundant extracellular matrix components, cytokines, and growth factors, and are capable of generating contractile force. As such, these cells have become potential therapeutic targets in a number of disease settings. Transforming growth factor β (TGF-β) is a potent stimulus of fibrosis and myofibroblast formation and likewise is an important therapeutic target in several disease conditions. The plant-derived isothiocyanate sulforaphane has been shown to have protective effects in several pathological models including diabetic cardiomyopathy, carcinogenesis, and fibrosis. These studies suggest that sulforaphane may be an attractive preventive agent against disease progression, particularly in conditions involving alterations of the extracellular matrix and activation of myofibroblasts. However, few studies have evaluated the effects of sulforaphane on cardiac fibroblast activation and their interactions with the extracellular matrix. The present studies were carried out to determine the potential effects of sulforaphane on the conversion of quiescent cardiac fibroblasts to an activated myofibroblast phenotype and associated alterations in signaling, expression of extracellular matrix receptors, and cellular physiology following stimulation with TGF-β1. These studies demonstrate that sulforaphane attenuates TGF-β1-induced myofibroblast formation and contractile activity. Sulforaphane also reduces expression of collagen-binding integrins and inhibits canonical and noncanonical TGF-β signaling pathways.
Collapse
Affiliation(s)
- Charity Fix
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Amanda Carver-Molina
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Mrinmay Chakrabarti
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Wayne Carver
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina
| |
Collapse
|
47
|
|
48
|
Sun L, Guo Z, Sun J, Li J, Dong Z, Zhang Y, Chen J, Kan Q, Yu Z. MiR-133a acts as an anti-oncogene in Hepatocellular carcinoma by inhibiting FOSL2 through TGF-β/Smad3 signaling pathway. Biomed Pharmacother 2018; 107:168-176. [DOI: 10.1016/j.biopha.2018.07.151] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 01/27/2023] Open
|
49
|
Stewart E, McEvoy J, Wang H, Chen X, Honnell V, Ocarz M, Gordon B, Dapper J, Blankenship K, Yang Y, Li Y, Shaw TI, Cho JH, Wang X, Xu B, Gupta P, Fan Y, Liu Y, Rusch M, Griffiths L, Jeon J, Freeman BB, Clay MR, Pappo A, Easton J, Shurtleff S, Shelat A, Zhou X, Boggs K, Mulder H, Yergeau D, Bahrami A, Mardis ER, Wilson RK, Zhang J, Peng J, Downing JR, Dyer MA. Identification of Therapeutic Targets in Rhabdomyosarcoma through Integrated Genomic, Epigenomic, and Proteomic Analyses. Cancer Cell 2018; 34:411-426.e19. [PMID: 30146332 PMCID: PMC6158019 DOI: 10.1016/j.ccell.2018.07.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/09/2018] [Accepted: 07/25/2018] [Indexed: 12/13/2022]
Abstract
Personalized cancer therapy targeting somatic mutations in patient tumors is increasingly being incorporated into practice. Other therapeutic vulnerabilities resulting from changes in gene expression due to tumor specific epigenetic perturbations are progressively being recognized. These genomic and epigenomic changes are ultimately manifest in the tumor proteome and phosphoproteome. We integrated transcriptomic, epigenomic, and proteomic/phosphoproteomic data to elucidate the cellular origins and therapeutic vulnerabilities of rhabdomyosarcoma (RMS). We discovered that alveolar RMS occurs further along the developmental program than embryonal RMS. We also identified deregulation of the RAS/MEK/ERK/CDK4/6, G2/M, and unfolded protein response pathways through our integrated analysis. Comprehensive preclinical testing revealed that targeting the WEE1 kinase in the G2/M pathway is the most effective approach in vivo for high-risk RMS.
Collapse
Affiliation(s)
- Elizabeth Stewart
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Justina McEvoy
- Departments of Molecular and Cellular Biology and Pediatrics, BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Hong Wang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Integrated Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Victoria Honnell
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Monica Ocarz
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA
| | - Brittney Gordon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA
| | - Jason Dapper
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA; Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kaley Blankenship
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanling Yang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yuxin Li
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Proteomics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Timothy I Shaw
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Proteomics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ji-Hoon Cho
- Proteomics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xusheng Wang
- Proteomics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Pankaj Gupta
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yu Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lyra Griffiths
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA
| | - Jongrye Jeon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA
| | - Burgess B Freeman
- Preclinical Pharmacokinetics Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael R Clay
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alberto Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sheila Shurtleff
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anang Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kristy Boggs
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Mulder
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Donald Yergeau
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elaine R Mardis
- The McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA; Department of Genetics, Washington University, St. Louis, MO 63108, USA; Department of Medicine, Washington University, St. Louis, MO 63108, USA
| | - Richard K Wilson
- The McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA; Department of Genetics, Washington University, St. Louis, MO 63108, USA; Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63108, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - James R Downing
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 323, Memphis, TN 38105-3678, USA; Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
50
|
Zent J, Guo LW. Signaling Mechanisms of Myofibroblastic Activation: Outside-in and Inside-Out. Cell Physiol Biochem 2018; 49:848-868. [PMID: 30184544 DOI: 10.1159/000493217] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Myofibroblasts are central mediators of fibrosis. Typically derived from resident fibroblasts, myofibroblasts represent a heterogeneous population of cells that are principally defined by acquired contractile function and high synthetic ability to produce extracellular matrix (ECM). Current literature sheds new light on the critical role of ECM signaling coupled with mechanotransduction in driving myofibroblastic activation. In particular, transforming growth factor β1 (TGF-β1) and extra domain A containing fibronectin (EDA-FN) are thought to be the primary ECM signaling mediators that form and also induce positive feedback loops. The outside-in and inside-out signaling circuits are transmitted and integrated by TGF-β receptors and integrins at the cell membrane, ultimately perpetuating the abundance and activities of TGF-β1 and EDA-FN in the ECM. In this review, we highlight these conceptual advances in understanding myofibroblastic activation, in hope of revealing its therapeutic anti-fibrotic implications.
Collapse
Affiliation(s)
- Joshua Zent
- Medical Scientist Training Program, the Ohio State University, Columbus, Columbus, Ohio, USA
| | - Lian-Wang Guo
- Department of Surgery, Department of Physiology & Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, Wexner Medical Center, the Ohio State University, Columbus, Ohio, USA
| |
Collapse
|