1
|
Danek V, Tureckova J, Huebner K, Erlenbach-Wuensch K, Baranova P, Dobes J, Balounova J, Simova M, Novosadova V, Madureira Trufen CE, Prochazkova M, Talacko P, Harant K, Barinka C, Beck IM, Schneider-Stock R, Sedlacek R, Prochazka J. CUL4A exhibits tumor-suppressing role via regulation of HUWE1-mediated SMAD3 intracellular shuttling. Cancer Lett 2025; 621:217663. [PMID: 40120800 DOI: 10.1016/j.canlet.2025.217663] [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: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Changes in cellular physiology and proteomic homeostasis accompanied the initiation and progression of colorectal cancer. Thus, ubiquitination represents a central regulatory mechanism in proteome dynamics. However, the complexity of the ubiquitinating network involved in carcinogenesis remains unclear. This study revealed the tumor-suppressive role of the ubiquitin ligase Cullin4A (CUL4A) in the intestine. We showed that simultaneous loss of CUL4A and hyperactivation of the Wnt pathway promotes tumor development in the distal colon. This tumor development is caused by an accumulation of the inactive SMAD3, a TGF-β pathway mediator. Depletion of CUL4A resulted in stabilization of HUWE1, which attenuated SMAD3 function. We showed a correlation between the intracellular localization of CUL4A and colorectal cancer progression, where nuclear CUL4A localization correlates with advanced colorectal cancer progression. In summary, we identified CUL4A as an important regulator of SMAD3 signal transduction competence in a HUWE1-dependent manner and demonstrated a critical role for the crosstalk between ubiquitination and the Wnt/TGF-β signaling pathways in gastrointestinal homeostasis.
Collapse
Affiliation(s)
- Veronika Danek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Jolana Tureckova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Kerstin Huebner
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | | | - Petra Baranova
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, 252 50, Czech Republic
| | - Jan Dobes
- Department of Cell Biology, Faculty of Science, Charles University, 128 00, Prague, Czech Republic
| | - Jana Balounova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Michaela Simova
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Vendula Novosadova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Carlos Eduardo Madureira Trufen
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Michaela Prochazkova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Pavel Talacko
- BIOCEV Proteomics Core Facility, Faculty of Science, Charles University, Vestec, 252 50, Czech Republic
| | - Karel Harant
- BIOCEV Proteomics Core Facility, Faculty of Science, Charles University, Vestec, 252 50, Czech Republic
| | - Cyril Barinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, 252 50, Czech Republic
| | - Inken M Beck
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Animal Research Centre, Ulm University, Ulm, Germany
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, 91054, Erlangen, Germany; Institute of Pathology, FAU Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic
| | - Jan Prochazka
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic; Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20, Prague, Czech Republic.
| |
Collapse
|
2
|
Soto ME, Rodríguez-Brito M, Pérez-Torres I, Herrera-Alarcon V, Martínez-Hernández H, Hernández I, Castrejón-Téllez V, Peña-Ocaña BA, Alvarez-Leon E, Manzano-Pech L, Gamboa R, Fuentevilla-Alvarez G, Huesca-Gómez C. Analysis of FBN1, TGFβ2, TGFβR1 and TGFβR2 mRNA as Key Molecular Mechanisms in the Damage of Aortic Aneurysm and Dissection in Marfan Syndrome. Int J Mol Sci 2025; 26:3067. [PMID: 40243722 PMCID: PMC11989073 DOI: 10.3390/ijms26073067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Marfan syndrome (MFS) is an inherited connective tissue disorder, with aortic root aneurysm and/or dissection being the most severe and life-threatening complication. These conditions have been linked to pathogenic variants in the FBN1 gene and dysregulated TGFβ signaling. Our objective was to evaluate the mRNA expression of FBN1, TGFBR1, TGFBR2, and TGFB2 in aortic tissue from MFS patients undergoing surgery for aortic dilation. This prospective study (2014-2023) included 20 MFS patients diagnosed according to the 2010 Ghent criteria, who underwent surgery for aneurysm or dissection based on Heart Team recommendations, along with 20 non-MFS controls. RNA was extracted, and mRNA levels were quantified using RT-qPCR. Patients with dissection showed significantly higher FBN1 mRNA levels [79 (48.1-110.1)] compared to controls [37.2 (25.1-79)] (p = 0.03). Conversely, TGFB2 expression was significantly lower in MFS patients [12.17 (6.54-24.70)] than in controls [44.29 (25.85-85.36)] (p = 0.029). A positive correlation was observed between higher FBN1 expression and a larger sinotubular junction diameter (r = 0.42, p = 0.07), while increased FBN1 expression was particularly evident in MFS patients with dissection. Additionally, TGFB2 expression showed an inverse correlation with ascending aortic diameter (r = 0.53, p = 0.01). In aortic tissue, we found decreased TGFB2 and receptor levels alongside increased FBN1 mRNA levels. These molecular alterations may reflect compensatory mechanisms in response to tissue damage caused by mechanical stress, leading to dysregulation of physiological signaling pathways and ultimately contributing to aortic dilation in MFS.
Collapse
Affiliation(s)
- María Elena Soto
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Mexico City 14080, Mexico;
- Cardiovascular Line in American British Cowdray (ABC) Medical Center, PAI ABC Sur 136 No. 16, Col. Las Américas, Mexico City 01120, Mexico
| | - Myrlene Rodríguez-Brito
- Cardiothoracic Surgery Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (M.R.-B.); (V.H.-A.); (H.M.-H.); (I.H.)
| | - Israel Pérez-Torres
- Cardiovascular Biomedicine Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Mexico City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - Valentín Herrera-Alarcon
- Cardiothoracic Surgery Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (M.R.-B.); (V.H.-A.); (H.M.-H.); (I.H.)
| | - Humberto Martínez-Hernández
- Cardiothoracic Surgery Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (M.R.-B.); (V.H.-A.); (H.M.-H.); (I.H.)
| | - Iván Hernández
- Cardiothoracic Surgery Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (M.R.-B.); (V.H.-A.); (H.M.-H.); (I.H.)
| | - Vicente Castrejón-Téllez
- Physiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (V.C.-T.)
| | - Betsy Anaid Peña-Ocaña
- Biochemistry Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Mexico City 14080, Mexico;
- Tecnológico Nacional de México, Instituto Tecnológico de Tuxtla Gutiérrez, Tuxtla Gutiérrez 29050, Mexico
| | - Edith Alvarez-Leon
- Sub-Directorate of Basic Research, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Mexico City 14080, Mexico;
| | - Linaloe Manzano-Pech
- Cardiovascular Biomedicine Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. Sección XVI, Mexico City 14080, Mexico; (I.P.-T.); (L.M.-P.)
| | - Ricardo Gamboa
- Physiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (V.C.-T.)
| | - Giovanny Fuentevilla-Alvarez
- Endocrinology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico
| | - Claudia Huesca-Gómez
- Physiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1, Col. 4 Sección XVI, Mexico City 14080, Mexico; (V.C.-T.)
| |
Collapse
|
3
|
Nair A, Singh R, Gautam N, Saxena S, Mittal S, Shah S, Talegaonkar S. Multifaceted role of phytoconstituents based nano drug delivery systems in combating TNBC: A paradigm shift from chemical to natural. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9207-9226. [PMID: 38953968 DOI: 10.1007/s00210-024-03234-0] [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: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024]
Abstract
Triple negative breast cancer is considered to be a malignancy of grave concern with limited routes of treatment due to the absence of specific breast cancer markers and ambiguity of other potential drug targets. Poor prognosis and inadequate survival rates have prompted further research into the understanding of the molecular pathophysiology and targeting of the disease. To overcome the recurrence and resistance mechanisms of the TNBC cells, various approaches have been devised, and are being continuously evaluated to enhance their efficacy and safety. Chemo-Adjuvant therapy is one such treatment modality being employed to improve the efficiency of standard chemotherapy. Combining chemo-adjuvant therapy with other upcoming approaches of cancer therapeutics such as phytoconstituents and nanotechnology has yielded promising results in the direction of improving the prognosis of TNBC. Numerous nanoformulations have been proven to substantially enhance the specificity and cellular uptake of drugs by cancer cells, thus reducing the possibility of unintended systemic side effects within cancer patients. While phytoconstituents offer a wide variety of beneficial active constituents useful in cancer therapeutics, most favorable outcomes have been observed within the scope of polyphenols, isoquinoline alkaloids and isothiocyanates. With an enhanced understanding of the molecular mechanisms of TNBC and the advent of newer targeting technologies and novel phytochemicals of medicinal importance, a new era of cancer theranostic treatments can be explored. This review hopes to instantiate the current body of research regarding the role of certain phytoconstituents and their potential nanoformulations in targeting specific TNBC pathways for treatment and diagnostic purposes.
Collapse
Affiliation(s)
- Anandita Nair
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Roshni Singh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Namrata Gautam
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Shilpi Saxena
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India
| | - Saurabh Mittal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, U.P, Noida, 201303, India.
| | - Sadia Shah
- Department of Pharmacology, Era College of Pharmacy, Era University, Lucknow, 226003, India.
| | - Sushama Talegaonkar
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 17, Delhi, India.
| |
Collapse
|
4
|
Jafarinia H, Khalilimeybodi A, Barrasa-Fano J, Fraley SI, Rangamani P, Carlier A. Insights gained from computational modeling of YAP/TAZ signaling for cellular mechanotransduction. NPJ Syst Biol Appl 2024; 10:90. [PMID: 39147782 PMCID: PMC11327324 DOI: 10.1038/s41540-024-00414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.
Collapse
Affiliation(s)
- Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Stephanie I Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA.
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
5
|
Najafiyan B, Bokaii Hosseini Z, Esmaelian S, Firuzpour F, Rahimipour Anaraki S, Kalantari L, Hheidari A, Mesgari H, Nabi-Afjadi M. Unveiling the potential effects of resveratrol in lung cancer treatment: Mechanisms and nanoparticle-based drug delivery strategies. Biomed Pharmacother 2024; 172:116207. [PMID: 38295754 DOI: 10.1016/j.biopha.2024.116207] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 03/03/2024] Open
Abstract
Lung cancer ranks among the most prevalent forms of cancer and remains a significant factor in cancer-related mortality across the world. It poses significant challenges to healthcare systems and society as a whole due to its high incidence, mortality rates, and late-stage diagnosis. Resveratrol (RV), a natural compound found in various plants, has shown potential as a nanomedicine for lung cancer treatment. RV has varied effects on cancer cells, including promoting apoptosis by increasing pro-apoptotic proteins (Bax and Bak) and decreasing anti-apoptotic proteins (Bcl-2). It also hinders cell proliferation by influencing important signaling pathways (MAPK, mTOR, PI3K/Akt, and Wnt/β-catenin) that govern cancer progression. In addition, RV acts as a potent antioxidant, diminishing oxidative stress and safeguarding cells against DNA damage. However, using RV alone in cancer treatment has drawbacks, such as low bioavailability, lack of targeting ability, and susceptibility to degradation. In contrast, nanoparticle-based delivery systems address these limitations and hold promise for improving treatment outcomes in lung cancer; nanoparticle formulations of RV offer advantages such as improved drug delivery, increased stability, controlled release, and targeted delivery to lung cancer cells. This article will provide an overview of lung cancer, explore the potential of RV as a therapeutic agent, discuss the benefits and challenges of nanoparticle-based drug delivery, and highlight the promise of RV nanoparticles for cancer treatment, including lung cancer. By optimizing these systems for clinical application, future studies aim to enhance overall treatment outcomes and improve the prognosis for lung cancer patients.
Collapse
Affiliation(s)
- Behnam Najafiyan
- Faculty of Pharmacy, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Samar Esmaelian
- Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Faezeh Firuzpour
- Student of Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
6
|
Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: From bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024; 48:87-127. [PMID: 37859532 DOI: 10.1002/cbin.12097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor-β (TGF-β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF-β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF-β causes extracellular matrix deposition, epithelial-mesenchymal transition, cancer-associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF-β in inflammation, fibrosis, and carcinogenesis. As TGF-β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF-β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor-suppressor functions. There is a need for careful dosing of TGF-β drugs for therapeutic use and patient selection.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
7
|
Ishii S, Fujiwara T, Yagi H, Takeda N, Ando M, Yamauchi H, Inuzuka R, Taniguchi Y, Hatano M, Komuro I. A novel pathogenic variant located just upstream of the C-terminal Ser423-X-Ser425 phosphorylation motif in SMAD3 causing Loeys-Dietz syndrome. Mol Genet Genomic Med 2023; 11:e2257. [PMID: 37864304 PMCID: PMC10724497 DOI: 10.1002/mgg3.2257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 04/21/2023] [Accepted: 07/14/2023] [Indexed: 10/22/2023] Open
Abstract
OBJECTIVE Loeys-Dietz syndrome (LDS) is a heritable disorder of connective tissue closely related to Marfan syndrome (MFS). LDS is caused by loss-of-function variants of genes that encode components of transforming growth factor-β (TGF-β) signaling; nevertheless, LDS type 1/2 caused by TGFBR1/2 pathogenic variants is frequently found to have paradoxical increases in TGF-β signaling in the aneurysmal aortic wall. Here, we present a Japanese LDS family having a novel SMAD3 variant. METHODS The proband was tested via clinical, genetic, and histological analyses. In vitro analysis was performed for pathogenic evaluation. RESULTS The novel heterozygous missense variant of SMAD3 [c.1262G>A, p.(Cys421Tyr)], located just upstream of the C-terminal Ser423-X-Ser425 phosphorylation motif, was found in this instance of LDS type 3. This variant led to reduced phospho-SMAD3 (Ser423/Ser425) levels and transcription activity in vitro; however, a paradoxical upregulation of TGF-β signaling was evident in the aortic wall. CONCLUSIONS Our results revealed the presence of TGF-β paradox in this case with the novel loss-of-function SMAD3 variant. The precise mechanism underlying the paradox is unknown, but further research is warranted to clarify the influence of the SMAD3 variant type and location on the LDS3 phenotype as well as the molecular mechanism leading to LDS3 aortopathy.
Collapse
Affiliation(s)
- Satoshi Ishii
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
| | - Takayuki Fujiwara
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
- Department of Computational Diagnostic Radiology and Preventive MedicineThe University of Tokyo HospitalTokyoJapan
| | - Hiroki Yagi
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
| | - Norifumi Takeda
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
| | - Masahiko Ando
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
- Department of Cardiac SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Haruo Yamauchi
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
- Department of Cardiac SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Ryo Inuzuka
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
- Department of PediatricsThe University of Tokyo HospitalTokyoJapan
| | - Yuki Taniguchi
- Marfan Syndrome CenterThe University of Tokyo HospitalTokyoJapan
- Department of Orthopedic SurgeryThe University of Tokyo HospitalTokyoJapan
| | - Masaru Hatano
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
- Department of Advanced Medical Center for Heart FailureThe University of Tokyo HospitalTokyoJapan
| | - Issei Komuro
- Department of Cardiovascular MedicineThe University of Tokyo HospitalTokyoJapan
| |
Collapse
|
8
|
Singh S, Gouri V, Samant M. TGF-β in correlation with tumor progression, immunosuppression and targeted therapy in colorectal cancer. Med Oncol 2023; 40:335. [PMID: 37855975 DOI: 10.1007/s12032-023-02204-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Colorectal cancer (CRC) is a complex malignancy responsible for the second-highest cancer deaths worldwide. TGF-β maintains normal cellular homeostasis by inhibiting the cell cycle and inducing apoptosis, but its elevated level is correlated with colorectal cancer progression, as TGF-β is a master regulator of the epithelial-to-mesenchymal transition, a critical step of metastasis. Tumors, including CRC, use elevated TGF-β levels to avoid immune surveillance by modulating immune cell differentiation, proliferation, and effector function. Presently, the treatment of advanced CRC is mainly based on chemotherapy, with multiple adverse effects. Thus, there is a need to develop alternate tactics because CRC continue to be mostly resistant to the present therapeutic regimen. TGF-β blockade has emerged as a promising therapeutic target in cancer therapy. Blocking TGF-β with phytochemicals and other molecules, such as antisense oligonucleotides, monoclonal antibodies, and bifunctional traps, alone or in combination, may be a safer and more effective way to treat CRC. Furthermore, combination immunotherapy comprising TGF-β blockers and immune checkpoint inhibitors is gaining popularity because both molecules work synergistically to suppress the immune system. Here, we summarize the current understanding of TGF-β as a therapeutic target for managing CRC and its context-dependent tumor-promoting or tumor-suppressing nature.
Collapse
Affiliation(s)
- Sumeet Singh
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Vinita Gouri
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
- Department of Zoology, Kumaun University, Nainital, Uttarakhand, India
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
9
|
Miranda P, Mirisis AA, Kukushkin NV, Carew TJ. Pattern detection in the TGFβ cascade controls the induction of long-term synaptic plasticity. Proc Natl Acad Sci U S A 2023; 120:e2300595120. [PMID: 37748056 PMCID: PMC10556637 DOI: 10.1073/pnas.2300595120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023] Open
Abstract
Transforming growth factor β (TGFβ) is required for long-term memory (LTM) for sensitization in Aplysia. When LTM is induced using a two-trial training protocol, TGFβ inhibition only blocks LTM when administrated at the second, not the first trial. Here, we show that TGFβ acts as a "repetition detector" during the induction of two-trial LTM. Secretion of the biologically inert TGFβ proligand must coincide with its proteolytic activation by the Bone morphogenetic protein-1 (BMP-1/Tolloid) metalloprotease, which occurs specifically during trial two of our two-trial training paradigm. This paradigm establishes long-term synaptic facilitation (LTF), the cellular correlate of LTM. BMP-1 application paired with a single serotonin (5HT) pulse induced LTF, whereas neither a single 5HT pulse nor BMP-1 alone effectively did so. On the other hand, inhibition of endogenous BMP-1 activity blocked the induction of two-trial LTF. These results suggest a unique role for TGFβ in the interaction of repeated trials: during learning, repeated stimuli engage separate steps of the TGFβ cascade that together are necessary for the induction of long-lasting memories.
Collapse
Affiliation(s)
- Paige Miranda
- Center for Neural Science, New York University, New York, NY10003
| | | | - Nikolay V. Kukushkin
- Center for Neural Science, New York University, New York, NY10003
- Liberal Studies, New York University, New York, NY10003
| | - Thomas J. Carew
- Center for Neural Science, New York University, New York, NY10003
| |
Collapse
|
10
|
Piekarska K, Bonowicz K, Grzanka A, Jaworski ŁM, Reiter RJ, Slominski AT, Steinbrink K, Kleszczyński K, Gagat M. Melatonin and TGF-β-Mediated Release of Extracellular Vesicles. Metabolites 2023; 13:metabo13040575. [PMID: 37110233 PMCID: PMC10142249 DOI: 10.3390/metabo13040575] [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: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The immune system, unlike other systems, must be flexible and able to "adapt" to fully cope with lurking dangers. The transition from intracorporeal balance to homeostasis disruption is associated with activation of inflammatory signaling pathways, which causes modulation of the immunology response. Chemotactic cytokines, signaling molecules, and extracellular vesicles act as critical mediators of inflammation and participate in intercellular communication, conditioning the immune system's proper response. Among the well-known cytokines allowing for the development and proper functioning of the immune system by mediating cell survival and cell-death-inducing signaling, the tumor necrosis factor α (TNF-α) and transforming growth factor β (TGF-β) are noteworthy. The high bloodstream concentration of those pleiotropic cytokines can be characterized by anti- and pro-inflammatory activity, considering the powerful anti-inflammatory and anti-oxidative stress capabilities of TGF-β known from the literature. Together with the chemokines, the immune system response is also influenced by biologically active chemicals, such as melatonin. The enhanced cellular communication shows the relationship between the TGF-β signaling pathway and the extracellular vesicles (EVs) secreted under the influence of melatonin. This review outlines the findings on melatonin activity on TGF-β-dependent inflammatory response regulation in cell-to-cell communication leading to secretion of the different EV populations.
Collapse
Affiliation(s)
- Klaudia Piekarska
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Klaudia Bonowicz
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Łukasz M Jaworski
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA
| | - Andrzej T Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Maciej Gagat
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| |
Collapse
|
11
|
The Twofold Role of Osteogenic Small Molecules in Parkinson's Disease Therapeutics: Crosstalk of Osteogenesis and Neurogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3813541. [PMID: 36545269 PMCID: PMC9763015 DOI: 10.1155/2022/3813541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/17/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022]
Abstract
Deemed one of the most problematic neurodegenerative diseases in the elderly population, Parkinson's disease remains incurable to date. Ongoing diagnostic studies, however, have revealed that a large number of small molecule drugs that trigger the BMP2-Smad signaling pathway with an osteogenic nature may be effective in Parkinson's disease treatment. Although BMP2 and Smad1, 3, and 5 biomolecules promote neurite outgrowth and neuroprotection in dopaminergic cells as well, small molecules are quicker at crossing the BBB and reaching the damaged dopaminergic neurons located in the substantia nigra due to a molecular weight less than 500 Da. It is worth noting that osteogenic small molecules that inhibit Smurf1 phosphorylation do not offer therapeutic opportunities for Parkinson's disease; whereas, osteogenic small molecules that trigger Smad1, 3, and 5 phosphorylation may have strong therapeutic implications in Parkinson's disease by increasing the survival rate of dopaminergic cells and neuritogenesis. Notably, from a different perspective, it might be said that osteogenic small molecules can possibly put forth therapeutic options for Parkinson's disease by improving neuritogenesis and cell survival.
Collapse
|
12
|
Cinnamomum bejolghota Extract Inhibits Colorectal Cancer Cell Metastasis and TGF-β1-Induced Epithelial-Mesenchymal Transition via Smad and Non-Smad Signaling Pathway. Sci Pharm 2022. [DOI: 10.3390/scipharm90020030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cinnamomum bejolghota, used in Thai traditional medicine remedies, has several biological activities including antimicrobial, antifungal, and anticancer. In colorectal cancer, epithelial-mesenchymal transition (EMT) is an initial step of cancer metastasis. Thus, this study investigated the effects of C. bejolghota bark extract (CBE) on colorectal cancer cell metastasis and transforming growth factor-β1 (TGF-β1) induced EMT in LoVo cells. The results showed that CBE could reduce cell migration, invasion, and adhesion of LoVo cells in a dose-dependent manner. In addition, our studies also showed that CBE could reverse TGF-β1-induced morphological changes as well as increase an epithelial marker, E-cadherin, while the expression of the mesenchymal marker, N-cadherin, was decreased in TGF-β1-treated LoVo cells. MMP-2 expression was effectively decreased but TIMP-1 and TIMP-2 expression was increased by the CBE treatment in LoVo cells. CBE also inhibited Smad2/3 phosphorylation and nuclear translocation as well as decreased the expression of Snail, Slug, and TCF8/ZEB1 transcription factors in LoVo cells. Moreover, CBE could inhibit TGF-β1-induced Smad-independent signaling pathway by decreased phosphorylation of ERK1/2, p38, and Akt. These findings suggest that CBE inhibited TGF-β1-induced EMT in LoVo cells via both Smad-dependent and Smad-independent pathways. Therefore, CBE may function as an alternative therapeutic treatment for colorectal cancer metastasis.
Collapse
|
13
|
A Tight Control of Non-Canonical TGF-β Pathways and MicroRNAs Downregulates Nephronectin in Podocytes. Cells 2022; 11:cells11010149. [PMID: 35011710 PMCID: PMC8750045 DOI: 10.3390/cells11010149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/22/2021] [Accepted: 01/01/2022] [Indexed: 02/01/2023] Open
Abstract
Nephronectin (NPNT) is an extracellular matrix protein in the glomerular basement membrane that is produced by podocytes and is important for the integrity of the glomerular filtration barrier. Upregulated transforming growth factor β (TGF-β) and altered NPNT are seen in different glomerular diseases. TGF-β downregulates NPNT and upregulates NPNT-targeting microRNAs (miRs). However, the pathways involved were previously unknown. By using selective inhibitors of the canonical, SMAD-dependent, and non-canonical TGF-β pathways, we investigated NPNT transcription, translation, secretion, and regulation through miRs in podocytes. TGF-β decreased NPNT mRNA and protein in cultured human podocytes. TGF-β-dependent regulation of NPNT was meditated through intracellular signaling pathways. Under baseline conditions, non-canonical pathways predominantly regulated NPNT post-transcriptionally. Podocyte NPNT secretion, however, was not dependent on canonical or non-canonical TGF-β pathways. The canonical TGF-β pathway was also dispensable for NPNT regulation after TGF-β stimulation, as TGF-β was still able to downregulate NPNT in the presence of SMAD inhibitors. In contrast, in the presence of different non-canonical pathway inhibitors, TGF-β stimulation did not further decrease NPNT expression. Moreover, distinct non-canonical TGF-β pathways mediated TGF-β-induced upregulation of NPNT-targeting miR-378a-3p. Thus, we conclude that post-transcriptional fine-tuning of NPNT expression in podocytes is mediated predominantly through non-canonical TGF-β pathways.
Collapse
|
14
|
Labibi B, Bashkurov M, Wrana JL, Attisano L. Modeling the Control of TGF-β/Smad Nuclear Accumulation by the Hippo Pathway Effectors, Taz/Yap. iScience 2020; 23:101416. [PMID: 32798968 PMCID: PMC7452192 DOI: 10.1016/j.isci.2020.101416] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/25/2020] [Accepted: 07/27/2020] [Indexed: 01/29/2023] Open
Abstract
Integration of transforming growth factor β (TGF-β) signals with those of other pathways allows for precise temporal and spatial control of gene expression patterns that drive development and homeostasis. The Hippo pathway nuclear effectors, Taz/Yap, interact with the TGF-β transcriptional mediators, Smads, to control Smad activity. Key to TGF-β signaling is the nuclear localization of Smads. Thus, to investigate the role of Taz/Yap in Smad nuclear accumulation, we developed mathematical models of Hippo and TGF-β cross talk. The models were based on experimental measurements of TGF-β-induced changes in Taz/Yap and Smad subcellular localization obtained using high-throughput immunofluorescence (IF) imaging in the mouse mammary epithelial cell line, EpH4. Bayesian MCMC DREAM parameter estimation was used to quantify the uncertainty in estimates of the kinetic parameters. Variation of the model parameters and statistical analysis show that our modeling predicts that Taz/Yap can alter TGF-β receptor activity and directly or indirectly act as nuclear retention factors.
Taz/Yap modulate TGF-β-induced nuclear accumulation of Smad2/3 and Smad4 TGF-β does not affect Taz/Yap localization when Hippo activity is constant Taz/Yap loss may alter activity of both Receptor and Smad nuclear retention factors The mediator complex regulates Smad nuclear accumulation
Collapse
Affiliation(s)
- Bita Labibi
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Mikhail Bashkurov
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
15
|
He Z, Deng F, Xiong S, Cai Y, He Z, Wang X, Li S, Yang D, Yan T. Expression and regulation of Smad2 by gonadotropins in the protogynous hermaphroditic ricefield eel (Monopterus albus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1155-1165. [PMID: 32128659 DOI: 10.1007/s10695-020-00778-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Smad2, a receptor-activated Smad, plays a critical role in regulating gametogenesis. In this study, a smad2 homologue was identified and sequenced from ricefield eel ovary cDNA, and its mRNA and protein expression levels were analysed during oocyte development. The cDNA sequence of ricefield eel smad2 consisted of 1863 bp encoding a 467-amino acid protein that had high sequence homology with Smad proteins in other teleosts, especially in Poeciliopsis prolifica. The results of real-time quantitative PCR (RT-qPCR) analysis revealed that smad2 is expressed in the ovary during gonad development, increased continuously until the early vitellogenic stage in the ovaries, and then decreased with ovary maturation. Smad2 protein immunoreactivity was localized in the cytoplasm of follicular cells, oogonia, and primary growth stage oocytes. In vitro experiments revealed that follicle-stimulating hormone (FSH) and human chorionic gonadotropin (hCG) promoted smad2 expression in ovary tissue in a time- and dose-dependent manner, respectively. In summary, Smad2 plays a potentially vital role in ricefield eel ovary development.
Collapse
Affiliation(s)
- Zhi He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Faqiang Deng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Sen Xiong
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yueping Cai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhide He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiongyan Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Song Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Deying Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Taiming Yan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
16
|
Bartoli-Leonard F, Wilkinson FL, Langford-Smith AWW, Alexander MY, Weston R. The Interplay of SIRT1 and Wnt Signaling in Vascular Calcification. Front Cardiovasc Med 2018; 5:183. [PMID: 30619890 PMCID: PMC6305318 DOI: 10.3389/fcvm.2018.00183] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Vascular calcification is a major health risk and is highly correlated with atherosclerosis, diabetes, and chronic kidney disease. The development of vascular calcification is an active and complex process linked with a multitude of signaling pathways, which regulate promoters and inhibitors of osteogenesis, the balance of which become deregulated in disease conditions. SIRT1, a protein deacetylase, known to be protective in inhibiting oxidative stress and inflammation within the vessel wall, has been shown as a possible key player in modulating the cell-fate determining canonical Wnt signaling pathways. Suppression of SIRT1 has been reported in patients suffering with cardiovascular pathologies, suggesting that the sustained acetylation of osteogenic factors could contribute to their activation and in turn, lead to the progression of calcification. There is clear evidence of the synergy between β-Catenin and elevated Runx2, and with Wnt signaling being β-Catenin dependent, further understanding is needed as to how these molecular pathways converge and interact, in order to provide novel insight into the mechanism by which smooth muscle cells switch to an osteogenic differentiation programme. Therefore, this review will describe the current concepts of pathological soft tissue mineralization, with a focus on the contribution of SIRT1 as a regulator of Wnt signaling and its targets, discussing SIRT1 as a potential target for manipulation and therapy.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Fiona L Wilkinson
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alex W W Langford-Smith
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - M Y Alexander
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ria Weston
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
17
|
Yu C, Jeremy RW. Angiotensin, transforming growth factor β and aortic dilatation in Marfan syndrome: Of mice and humans. IJC HEART & VASCULATURE 2018; 18:71-80. [PMID: 29876507 PMCID: PMC5988480 DOI: 10.1016/j.ijcha.2018.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/21/2018] [Accepted: 02/28/2018] [Indexed: 01/09/2023]
Abstract
Marfan syndrome is consequent upon mutations in FBN1, which encodes the extracellular matrix microfibrillar protein fibrillin-1. The phenotype is characterised by development of thoracic aortic aneurysm. Current understanding of the pathogenesis of aneurysms in Marfan syndrome focuses upon abnormal vascular smooth muscle cell signalling through the transforming growth factor beta (TGFβ) pathway. Angiotensin II (Ang II) can directly induce aortic dilatation and also influence TGFβ synthesis and signalling. It has been hypothesised that antagonism of Ang II signalling may protect against aortic dilatation in Marfan syndrome. Experimental studies have been supportive of this hypothesis, however results from multiple clinical trials are conflicting. This paper examines current knowledge about the interactions of Ang II and TGFβ signalling in the vasculature, and critically interprets the experimental and clinical findings against these signalling interactions.
Collapse
Affiliation(s)
- Christopher Yu
- Sydney Medical School, University of Sydney, Sydney 2006, Australia
| | | |
Collapse
|
18
|
Wu L. Functional characteristics of a novel SMAD4 mutation from thoracic aortic aneurysms (TAA). Gene 2017; 628:129-133. [PMID: 28716708 DOI: 10.1016/j.gene.2017.07.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 01/12/2023]
Abstract
SMAD4 is as an essential mediator of the transforming growth factor β (TGF-β) signaling pathway, and dysregulated TGF-β signaling is linked with thoracic aortic aneurysms (TAAs). In this study, we functionally characterized the Smad4 S271N mutation (the mutation c. 812G>A in Smad4 results in the amino acid substitution Ser271Asn) that was isolated from TAA individuals. We first constructed wild-type human Smad4 and Smad4 S271N plasmids. These constructs were then transiently transfected into HEK293T cells, and subsequent real-time PCR and western blotting demonstrated that wild-type Smad4 and Smad4 S271N were successfully expressed in 293T cells. We found that HEK293T cells overexpressing Smad4 S271N showed a strong increase in both cytoplasmic and nuclear Smad4 protein levels in response to TGF-β1. Although TGF-β signaling was the same in wild-type Smad4- and Smad4 S271N-transfected cells following TGF-β1 exposure, interestingly, we observed that transient Smad4 S271N expression in HEK293T cells caused a significant basal activation of TGF-β signaling. These results indicated that Smad4 may not directly induce TAA; rather it may contribute to TAA in combination with other risk factors.
Collapse
Affiliation(s)
- Lifei Wu
- Basic Medical College, Shanxi Medical University, 56 Xinjian S Rd, Taiyuan, Shanxi 30001, China.
| |
Collapse
|
19
|
Hur W, Lee HY, Min HS, Wufuer M, Lee CW, Hur JA, Kim SH, Kim BK, Choi TH. Regeneration of full-thickness skin defects by differentiated adipose-derived stem cells into fibroblast-like cells by fibroblast-conditioned medium. Stem Cell Res Ther 2017; 8:92. [PMID: 28427476 PMCID: PMC5399413 DOI: 10.1186/s13287-017-0520-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 01/26/2017] [Accepted: 02/24/2017] [Indexed: 01/08/2023] Open
Abstract
Background Fibroblasts are ubiquitous cells in the human body and are absolutely necessary for wound healing such as for injured skin. This role of fibroblasts was the reason why we aimed to differentiate human adipose-derived stem cells (hADSCs) into fibroblasts and to test their wound healing potency. Recent reports on hADSC-derived conditioned medium have indicated stimulation of collagen synthesis as well as migration of dermal fibroblasts in wound sites with these cells. Similarly, human fibroblast-derived conditioned medium (F-CM) was reported to contain a variety of factors known to be important for growth of skin. However, it remains unknown whether and how F-CM can stimulate hADSCs to secrete type I collagen. Methods In this study, we obtained F-CM from the culture of human skin fibroblast HS27 cells in DMEM media. For an in-vivo wound healing assay using cell transplantation, balb/c nude mice with full-thickness skin wound were used. Results Our data showed that levels of type I pro-collagen secreted by hADSCs cultured in F-CM increased significantly compared with hADSCs kept in normal medium for 72 h. In addition, from a Sircol collagen assay, the amount of collagen in F-CM-treated hADSC conditioned media (72 h) was markedly higher than both the normal medium-treated hADSC conditioned media (72 h) and the F-CM (24 h). We aimed to confirm that hADSCs in F-CM would differentiate into fibroblast cells in order to stimulate wound healing in a skin defect model. To investigate whether F-CM induced hADSCs into fibroblast-like cells, we performed FACS analysis and verified that both F-CM-treated hADSCs and HS27 cells contained similar expression patterns for CD13, CD54, and CD105, whereas normal medium-treated hADSCs were significantly different. mRNA level analysis for Nanog, Oct4A, and Sox2 as undifferentiation markers and vimentin, HSP47, and desmin as matured fibroblast markers supported the characterization that hADSCs in F-CM were highly differentiated into fibroblast-like cells. To discover the mechanism of type I pro-collagen expression in hADSCs in F-CM, we observed that phospho-smad 2/3 levels were increased in the TGF-β/Smad signaling pathway. For in-vivo analysis, we injected various cell types into balb/c nude mouse skin carrying a 10-mm punch wound, and observed a significantly positive wound healing effect in this full-thickness excision model with F-CM-treated hADSCs rather than with untreated hADSCs or the PBS injected group. Conclusions We differentiated F-CM-treated hADSCs into fibroblast-like cells and demonstrated their efficiency in wound healing in a skin wound model.
Collapse
Affiliation(s)
- Woojune Hur
- Department of Plastic and Reconstructive Surgery, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Hoon Young Lee
- Department of Plastic and Reconstructive Surgery, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea
| | - Hye Sook Min
- Department of Preventive Medicine, Graduate School of Public Health, Seoul National University, Seoul, 152-742, Republic of Korea
| | - Maierdanjiang Wufuer
- Department of Plastic and Reconstructive Surgery, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Chang-Won Lee
- Department of Naval Architecture and Ocean Engineering, College of Engineering, Seoul National University 110-744, Seoul, Republic of Korea
| | - Ji An Hur
- Department of Internal Medicine, School of Medicine, Yeungnam University, Daegu, 712-749, Republic of Korea
| | - Sang Hyon Kim
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, 700-712, Republic of Korea
| | - Byeung Kyu Kim
- Department of Plastic and Reconstructive Surgery, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Tae Hyun Choi
- Department of Plastic and Reconstructive Surgery, Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, 110-799, Republic of Korea.
| |
Collapse
|
20
|
Witkowska M, Majchrzak A, Cebula-Obrzut B, Wawrzyniak E, Robak T, Smolewski P. The distribution and potential prognostic value of SMAD protein expression in chronic lymphocytic leukemia. Tumour Biol 2017; 39:1010428317694551. [DOI: 10.1177/1010428317694551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The SMAD proteins are responsible for transducing signals from activated transforming growth factor-beta. This is the first study assessing the expression of SMAD-1/8, SMAD-2/3, SMAD-4, and SMAD-7 in chronic lymphocytic leukemia cells with regard to their clinical significance and potential prognostic value. Overexpression of SMAD-1/8 was observed in 160 chronic lymphocytic leukemia patients compared to 42 healthy volunteers (p = 0.023) and was associated with a more progressive course of the disease (p = 0.016). Moreover, the high expression of SMAD-1/8 correlated with other, well-established prognostic factors, including clinical stage (p = 0.010) and lymphocyte doubling time (p = 0.021). The expression of SMAD-4 was lower in chronic lymphocytic leukemia patients compared with the control group (p = 0.003). Importantly, lower SMAD-4 levels correlated with longer progression-free survival (p = 0.009), progressive course of the disease (p = 0.002), advanced clinical stage (p = 0.0004), elevated beta-2-microglobulin and lactate dehydrogenase levels (p < 0.05), shorter lymphocyte doubling time (p = 0.009), and CD38 antigen expression (p = 0.039). In addition, lower SMAD-4 expression correlated with lower apoptotic index (p = 0.0007) and lower expression of receptors for vascular endothelial growth factors VEGFR-1 and VEGFR-2. A significant association was found between the low expression of inhibitory protein SMAD-7 and both zeta-chain-associated protein kinase 70–negative cells (p = 0.04) and lower apoptotic index (p = 0.004). No differences were observed in SMAD-2/3 expression. In conclusion, our results demonstrate a significant correlation between greater SMAD-1/8 and lower SMAD-4 expression in chronic lymphocytic leukemia cells, as well as more progressive outcome and poor prognosis. These data provide supporting evidence that the expression of SMAD proteins plays an important role in disease development and may be considered as a novel, biologic prognostic factor in this disease.
Collapse
Affiliation(s)
- Magdalena Witkowska
- Department of Experimental Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Agata Majchrzak
- Department of Experimental Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Barbara Cebula-Obrzut
- Department of Experimental Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Ewa Wawrzyniak
- Department of Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
21
|
Wu Y, Yu X, Yi X, Wu K, Dwabe S, Atefi M, Elshimali Y, Kemp KT, Bhat K, Haro J, Sarkissyan M, Vadgama JV. Aberrant Phosphorylation of SMAD4 Thr277-Mediated USP9x-SMAD4 Interaction by Free Fatty Acids Promotes Breast Cancer Metastasis. Cancer Res 2017; 77:1383-1394. [PMID: 28115363 DOI: 10.1158/0008-5472.can-16-2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/22/2016] [Accepted: 12/10/2016] [Indexed: 12/22/2022]
Abstract
Obesity increases the risk of distant metastatic recurrence and reduces breast cancer survival. However, the mechanisms behind this pathology and identification of relevant therapeutic targets are poorly defined. Plasma free fatty acids (FFA) levels are elevated in obese individuals. Here we report that TGFβ transiently activates ERK and subsequently phosphorylates SMAD4 at Thr277, which facilitates a SMAD4-USP9x interaction, SMAD4 nuclear retention, and stimulates TGFβ/SMAD3-mediated transcription of Twist and Snail. USP9x inhibited the E3 ubiquitin-protein ligase TIF1γ from binding and monoubiquitinating SMAD4, hence maintaining the SMAD4 nuclear retention. FFA further facilitated TGFβ-induced ERK activation, SMAD4 phosphorylation, and nuclear retention, promoting TGFβ-dependent cancer progression. Inhibition of ERK and USP9x suppressed obesity-induced metastasis. In addition, clinical data indicated that phospho-ERK and -SMAD4 levels correlate with activated TGFβ signaling and metastasis in overweight/obese patient breast cancer specimens. Altogether, we demonstrate the vital interaction of USP9x and SMAD4 for governing TGFβ signaling and dyslipidemia-induced aberrant TGFβ activation during breast cancer metastasis. Cancer Res; 77(6); 1383-94. ©2017 AACR.
Collapse
Affiliation(s)
- Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California. .,David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
| | - Xiaoting Yu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xianghua Yi
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,Center for Animal Experiment/ABSL-3 Laboratory, Wuhan University, Hubei, China
| | - Sami Dwabe
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Mohammad Atefi
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Yahya Elshimali
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Kevin T Kemp
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Kruttika Bhat
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Jesse Haro
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Marianna Sarkissyan
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Jaydutt V Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California. .,David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
| |
Collapse
|
22
|
Hong T, Fung ES, Zhang L, Huynh G, Monuki ES, Nie Q. Semi-adaptive response and noise attenuation in bone morphogenetic protein signalling. J R Soc Interface 2016; 12:rsif.2015.0258. [PMID: 25972436 DOI: 10.1098/rsif.2015.0258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Temporal dynamics of morphogen-driven signalling events are critical for proper embryonic development. During development, cells translate extracellular bone morphogenetic protein (BMP) gradients, often subject to noise, into graded intracellular tail-phosphorylated SMAD (TP-SMAD) levels. Using modelling and experimental approaches, we found that BMPs induce TP-SMAD responses in neural precursor cells in a concentration-dependent manner, which are semi-adaptive within a specific intermediate range of BMP concentration. These semi-adaptive TP-SMAD responses involve an intrinsically slow deactivation of BMP receptors, which attenuates noise by prolonging SMAD deactivation time after BMP withdrawal, but increases response time. Interestingly, negative feedback on BMP receptors is also required for semi-adaptation, which benefits both noise attenuation and response time, and therefore balances the trade-off seen with slow BMP receptor deactivation. These results highlight the rich dynamics of SMAD regulation in response to graded BMP concentration, and elucidate general design principles for balancing noise attenuation and activation speed in signalling systems.
Collapse
Affiliation(s)
- Tian Hong
- Department of Mathematics, University of California, Irvine, CA, USA Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Ernest S Fung
- Center for Complex Biological Systems, University of California, Irvine, CA, USA Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Lei Zhang
- Beijing International Center for Mathematical Research, Peking University, Beijing, China
| | - Grace Huynh
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Edwin S Monuki
- Center for Complex Biological Systems, University of California, Irvine, CA, USA Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, CA, USA Center for Complex Biological Systems, University of California, Irvine, CA, USA
| |
Collapse
|
23
|
Tian Y, Yang Y, Gao L, Zhao H, Peng X, Zhang Z, Wu G. Expression of histone deacetylase-1 and p300 in aristolochic acid nephropathy models. Toxicol Mech Methods 2014; 24:377-84. [PMID: 24796935 DOI: 10.3109/15376516.2014.920448] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Aristolochic acid nephropathy (AAN) is mainly caused by aristolochic acid I (AAI), but the actual mechanism is still uncertain. The current study explored the correlation among the expression of Smad7, p300, histone deacetylase-1 (HDAC1) and the development of AAN using transmission electron microscopy (TEM), RT-PCR, and western blotting in the AAN mouse model and in the AAN cell model. TEM revealed that the renal tubular epithelial cells from the AAI-treated mice presented organelle damages and nuclear deformation. We found that a certain dose of AAI caused renal fibrosis and induced renal tubular epithelial cells to differentiate into myofibroblasts. There was a gradual increase in the expression of HDAC1 mRNA and protein observed using RT-PCR and western blotting in the AAN cell model compared with the control group. Gradual decrease in the expression of Smad7 and p300 mRNA and protein was revealed in the AAN mouse and cell models compared with the control group. These results suggest that AAI dose dependently contributed to the development of AAN, and HDAC1 and p300 participate in the modulation of TGF-β/Smad pathway-mediated renal interstitial fibrosis.
Collapse
Affiliation(s)
- Yahui Tian
- Laboratory of Pharmacology of Chinese Veterinary Medicine, Department of Animal Science and Technology, College of Animal Science and Technology, Beijing University of Agriculture , Beijing , China
| | | | | | | | | | | | | |
Collapse
|
24
|
The microRNA networks of TGFβ signaling in cancer. Tumour Biol 2013; 35:2857-69. [PMID: 24323563 DOI: 10.1007/s13277-013-1481-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 11/26/2013] [Indexed: 01/24/2023] Open
Abstract
In metazoans, the transforming growth factor β (TGFβ) signaling regulates a host of activities ranging from embryonic development to tissue homeostasis. The normal as well as tumor cells respond to this cytokine signaling pathway in a highly context-dependent manner. It acts as a potent tumor suppressor initially by inducing cell cycle arrest and apoptosis. But advanced tumors often misuse TGFβ signaling for tumor progression by selectively disabling the tumor suppressor arm and using other properties of TGFβ signaling such as induction of angiogenesis, epithelial to mesenchymal transition, and metastases. This dual role of TGFβ in cancer remained a mystery until recently. But recent advances in the field of microRNA provided a deeper understanding about this dual nature of TGFβ signaling in cancers. In the present review, we present an account of the role of microRNAs in deregulating TGFβ signaling and modulating cancer cell behavior during tumor initiation and cancer progression. This review also includes a discussion on the recent advances in the deregulation of TGFβ signaling in carcinogenesis.
Collapse
|
25
|
Seamons A, Treuting PM, Brabb T, Maggio-Price L. Characterization of dextran sodium sulfate-induced inflammation and colonic tumorigenesis in Smad3(-/-) mice with dysregulated TGFβ. PLoS One 2013; 8:e79182. [PMID: 24244446 PMCID: PMC3823566 DOI: 10.1371/journal.pone.0079182] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/21/2013] [Indexed: 12/22/2022] Open
Abstract
There are few mouse models that adequately mimic large bowel cancer in humans or the gastrointestinal inflammation which frequently precedes it. Dextran sodium sulphate (DSS)-induces colitis in many animal models and has been used in combination with the carcinogen azoxymethane (AOM) to induce cancer in mice. Smad3−/− mice are deficient in the transforming growth factor beta (TGFβ) signaling molecule, SMAD3, resulting in dysregulation of the cellular pathway most commonly affected in human colorectal cancer, and develop inflammation-associated colon cancer. Previous studies have shown a requirement for a bacterial trigger for the colitis and colon cancer phenotype in Smad3−/− mice. Studies presented here in Smad3−/− mice detail disease induction with DSS, without the use of AOM, and show a) Smad3−/− mice develop a spectrum of lesions ranging from acute and chronic colitis, crypt herniation, repair, dysplasia, adenomatous polyps, disseminated peritoneal adenomucinosis, adenocarcinoma, mucinous adenocarcinoma (MAC) and squamous metaplasia; b) the colon lesions have variable galactin-3 (Mac2) staining c) increased DSS concentration and duration of exposure leads to increased severity of colonic lesions; d) heterozygosity of SMAD3 does not confer increased susceptibility to DSS-induced disease and e) disease is partially controlled by the presence of T and B cells as Smad3−/−Rag2−/− double knock out (DKO) mice develop a more severe disease phenotype. DSS-induced disease in Smad3−/− mice may be a useful animal model to study not only inflammation-driven MAC but other human diseases such as colitis cystica profunda (CCP) and pseudomyxomatous peritonei (PMP).
Collapse
Affiliation(s)
- Audrey Seamons
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Piper M. Treuting
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Thea Brabb
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lillian Maggio-Price
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
26
|
Shen H, Gelberman RH, Silva MJ, Sakiyama-Elbert SE, Thomopoulos S. BMP12 induces tenogenic differentiation of adipose-derived stromal cells. PLoS One 2013; 8:e77613. [PMID: 24155967 PMCID: PMC3796462 DOI: 10.1371/journal.pone.0077613] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/04/2013] [Indexed: 12/26/2022] Open
Abstract
Adipose-derived stromal cells (ASCs) are pluripotent cells that have the capacity to differentiate into tendon fibroblasts (TFs). They are abundant in adults, easy to access, and are therefore an ideal cell source for tendon tissue engineering. Despite this potential, the molecular cues necessary for tenogenic differentiation of ASCs are unknown. Unlike other bone morphogenetic proteins (BMPs), BMP12, BMP13, and BMP14 have been reported to be less osteo-chondrogenic and to induce tendon rather than bone formation in vivo. This study investigated the effects of BMP12 and BMP14 on ASC differentiation in vitro. In canine ASCs, BMP12 effectively increased the expression of the tendon markers scleraxis and tenomodulin at both mRNA and protein levels. Consistent with these results, BMP12 induced scleraxis promoter driven-GFP and tenomodulin protein expression in mouse ASCs. Although BMP12 also enhanced the expression of the cartilage matrix gene aggrecan in ASCs, the resulting levels remained considerably lower than those detected in tendon fibroblasts. In addition, BMP12 reduced expression of the bone marker osteocalcin, but not the osteogenic transcription factor runx-2. BMP14 exhibited similar, but marginally less potent and selective effects, compared to BMP12. BMPs are known to signal through the canonical Smad pathway and the non-canonical mitogen-activated protein kinase (MAPK) pathway. BMP12 triggered robust phosphorylation of Smad1/5/8 but not Smad2/3 or p38 MAPK in ASCs. The effect was likely conveyed by type I receptors ALK2/3/6, as phosphorylation of Smad1/5/8 was blocked by the ALK2/3/6 inhibitor LDN-193189 but not by the ALK4/5/7 inhibitor SB-505124. Moreover, ALK6 was found to be the most abundant type I receptor in ASCs, with mRNA expression 100 to 10,000 times that of any other type I receptor. Collectively, results support the conclusion that BMP12 induces tenogenic differentiation of ASCs via the Smad1/5/8 pathway.
Collapse
Affiliation(s)
- Hua Shen
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, United States of America
| | - Richard H. Gelberman
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, United States of America
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, United States of America
| | - Shelly E. Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Stavros Thomopoulos
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
27
|
Yang FR, Fang BW, Lou JS. Effects of Fufang Biejia Ruangan Pills on hepatic fibrosis in vivo and in vitro. World J Gastroenterol 2013; 19:5326-5333. [PMID: 23983437 PMCID: PMC3752568 DOI: 10.3748/wjg.v19.i32.5326] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/02/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the protective effect and the relevant mechanisms of Fufang Biejia Ruangan Pills (FFBJRGP) on hepatic fibrosis in vivo and in vitro.
METHODS: Hepatic fibrosis was induced by carbon tetrachloride composite factors. Adult Wistar rats were randomly divided into four groups: normal control group; hepatic fibrosis model group; FFBJRGP-treated group at a daily dose of 0.55 g/kg; and colchicine-treated group at a daily dose of 0.1 g/kg. The effects of FFBJRGP on liver function, serum levels of hyaluronic acid (HA), type IV collagen (CIV), type III procollagen (PC III), laminin (LN), histopathology, and expression of transforming growth factor (TGF-β1) and Smad3 in hepatic fibrosis were evaluated in vivo. The effects of FFBJRGP on survival rate, hydroxyproline content and cell cycle distribution were further detected in vitro.
RESULTS: Compared with the hepatic fibrosis model group, rats treated with FFBJRGP showed a reduction in hepatic collagen deposition and improvement in hepatic lesions. Compared with those of the model group, the activities of alanine aminotransferase (62.0 ± 23.7 U/L) and aspartate aminotransferase (98.8 ± 40.0 U/L) in the FFBJRGP-treated group were decreased (50.02 ± 3.7 U/L and 57.2 ± 30.0 U/L, respectively, P < 0.01). Compared with those in the model group, the levels of PCIII (35.73 ± 17.90 μg/mL), HA (563.82 ± 335.54 ng/mL), LN (89.57 ± 7.59 ng/mL) and CIV (29.20 ± 6.17 ng/mL) were decreased to 30.18 ± 9.41, 456.18 ± 410.83, 85.46 ± 7.51 and 28.02 ± 9.45 ng/mL, respectively. Reverse-transcriptase polymerase chain reaction and Western blotting also revealed that expression of TGF-β1 and Smad3 were down-regulated in vivo. Cell proliferation was inhibited, the level of hydroxyproline was decreased compared with the control group (P < 0.01), and the cell cycle was redistributed when exposed to FFBJRGP in vitro.
CONCLUSION: FFBJRGP inhibits hepatic fibrosis in vivo and in vitro, which is probably associated with downregulation of fibrogenic signal transduction of the TGF-β-Smad pathway.
Collapse
|
28
|
Sivadas VP, George NA, Kattoor J, Kannan S. Novel mutations and expression alterations inSMAD3/TGFBR2genes in oral carcinoma correlate with poor prognosis. Genes Chromosomes Cancer 2013; 52:1042-52. [DOI: 10.1002/gcc.22099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 07/10/2013] [Indexed: 01/10/2023] Open
Affiliation(s)
- Vadakke Peringode Sivadas
- Laboratory of Cell Cycle Regulation & Molecular Oncology, Division of Cancer Research, Regional Cancer Centre; Thiruvananthapuram 695 011 Kerala India
| | - Nebu Abraham George
- Division of Surgical Oncology, Regional Cancer Centre; Thiruvananthapuram 695 011 Kerala India
| | - Jayasree Kattoor
- Division of Cytopathology, Regional Cancer Centre; Thiruvananthapuram 695 011 Kerala India
| | - S. Kannan
- Laboratory of Cell Cycle Regulation & Molecular Oncology, Division of Cancer Research, Regional Cancer Centre; Thiruvananthapuram 695 011 Kerala India
| |
Collapse
|
29
|
Boerma M, Wang J, Sridharan V, Herbert JM, Hauer-Jensen M. Pharmacological induction of transforming growth factor-beta1 in rat models enhances radiation injury in the intestine and the heart. PLoS One 2013; 8:e70479. [PMID: 23936211 PMCID: PMC3723823 DOI: 10.1371/journal.pone.0070479] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 06/20/2013] [Indexed: 12/30/2022] Open
Abstract
Radiation therapy in the treatment of cancer is dose limited by radiation injury in normal tissues such as the intestine and the heart. To identify the mechanistic involvement of transforming growth factor-beta 1 (TGF-β1) in intestinal and cardiac radiation injury, we studied the influence of pharmacological induction of TGF-β1 with xaliproden (SR 57746A) in rat models of radiation enteropathy and radiation-induced heart disease (RIHD). Because it was uncertain to what extent TGF-β induction may enhance radiation injury in heart and intestine, animals were exposed to irradiation schedules that cause mild to moderate (acute) radiation injury. In the radiation enteropathy model, male Sprague-Dawley rats received local irradiation of a 4-cm loop of rat ileum with 7 once-daily fractions of 5.6 Gy, and intestinal injury was assessed at 2 weeks and 12 weeks after irradiation. In the RIHD model, male Sprague-Dawley rats received local heart irradiation with a single dose of 18 Gy and were followed for 6 months after irradiation. Rats were treated orally with xaliproden starting 3 days before irradiation until the end of the experiments. Treatment with xaliproden increased circulating TGF-β1 levels by 300% and significantly induced expression of TGF-β1 and TGF-β1 target genes in the irradiated intestine and heart. Various radiation-induced structural changes in the intestine at 2 and 12 weeks were significantly enhanced with TGF-β1 induction. Similarly, in the RIHD model induction of TGF-β1 augmented radiation-induced changes in cardiac function and myocardial fibrosis. These results lend further support for the direct involvement of TGF-β1 in biological mechanisms of radiation-induced adverse remodeling in the intestine and the heart.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America.
| | | | | | | | | |
Collapse
|
30
|
Abstract
INTRODUCTION The transforming growth factor-β (TGF-β) signaling pathway has a pivotal role in tumor suppression and yet, paradoxically, in tumor promotion. Functional context dependent insights into the TGF-β pathway are crucial in developing TGF-β-based therapeutics for cancer. AREAS COVERED This review discusses the molecular mechanism of the TGF-β pathway and describes the different ways of tumor suppression by TGF-β. It is then explained how tumors can evade these effects and how TGF-β contributes to further growing and spreading of some of the tumors. In the last part of the review, the data on targeting TGF-β pathway for cancer treatment is assessed. This review focuses on anti-TGF-β based treatment and other options targeting activated pathways in tumors where the TGF-β tumor suppressor pathway is lost. Pre-clinical as well up to date results of the most recent clinical trials are given. EXPERT OPINION Targeting the TGF-β pathway can be a promising direction in cancer treatment. However, several challenges still exist, the most important are differentiating between the carcinogenic effects of TGF-β and its other physiological roles, and delineating the tumor suppressive versus the tumor promoting roles of TGF-β in each specific tumor. Future studies are needed in order to find safer and more effective TGF-β-based drugs.
Collapse
Affiliation(s)
- Lior H Katz
- Visiting Scientist, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Houston, TX, USA
| | - Ying Li
- Assistant Professor (Research), The University of Texas, M. D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Dr. Lopa Mishra’s Lab, Houston, TX, USA
| | - Jiun-Sheng Chen
- Research Assistant II, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Dr. Lopa Mishra’s Lab, Houston, TX, USA
| | - Nina M Muñoz
- Research Scientist, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Dr. Lopa Mishra’s Lab, Houston, TX, USA
| | - Avijit Majumdar
- Postdoctoral Fellow, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Dr.Lopa Mishra’s Lab, Houston, TX, USA
| | - Jian Chen
- Instructor, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Houston, TX, USA
| | - Lopa Mishra
- Del and Dennis McCarthy Distinguished Professor and Chair, The University of Texas, M.D. Anderson Cancer Center, Department of Gastroenterology, Hepatology, & Nutrition, Houston, TX, USA, Tel: +1 713 794 3221; Fax: +1 713 745 1886
| |
Collapse
|
31
|
Wang X, Qian Y, Jin R, Wo Y, Chen J, Wang C, Wang D. Effects of TRAP-1-like protein (TLP) gene on collagen synthesis induced by TGF-β/Smad signaling in human dermal fibroblasts. PLoS One 2013; 8:e55899. [PMID: 23418473 PMCID: PMC3572169 DOI: 10.1371/journal.pone.0055899] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 01/04/2013] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Hypertrophic scars are pathologic proliferations of the dermal skin layer resulting from excessive collagen deposition during the healing process of cutaneous wounds. Current research suggests that the TGF-β/Smad signaling pathway is closely associated with normal scar and hypertrophic scar formation. TRAP-1-like protein (TLP), a cytoplasmic protein, has been reported to efficiently regulate Smad2- and Smad3-dependent signal expression in the TGF-β pathway. The relationship between TLP and Type I/III collagen (Col I/III) synthesis explored in the present study provides an effective target for wound healing and gene therapy of hypertrophic scarring. OBJECTIVE To investigate the effects of TLP on collagen synthesis in human dermal fibroblasts. METHODS Lentiviral vectors encoding TLP was constructed to transfect fibroblasts derived from normal human skin. The expression of Col I/III and phosphorylation of Smad2 and Smad3 in fibroblasts were examined after TLP treatment. In addition, the comparison of TLP expression in normal skin tissues and in hypertrophic scar tissues was performed, and the effect of TLP on cell viability was analyzed by MTT assay. RESULTS TLP expression in hypertrophic scar tissue was markedly higher than in normal skin tissue. The Real Time PCR and Western blot test results both revealed that the synthesis of Col I/III was positively correlated with the expression of TLP. TLP also facilitate Smad2 phosphorylation while, conversely, inhibiting Smad3 phosphorylation. TLP may play a cooperative role, along with the cytokine TGF-β1, in improving the overall cell viability of skin fibroblasts. CONCLUSIONS TLP likely acts as a molecular modulator capable of altering the balance of Smad3- and Smad2-dependent signaling through regulation of phosphorylation, thus facilitating collagen synthesis in fibroblasts. Based on genetic variation in TLP levels in different tissues, these results suggest that TLP plays a key role in the process of TGF-β1/Smad3 signaling that contributes to wound healing and genesis of pathologic scars.
Collapse
Affiliation(s)
- Xue Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yunliang Qian
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Rong Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Wo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Jun Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Danru Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
32
|
Huang Q, Li Y, Zhang S, Huang R, Zheng L, Wei L, He M, Liao M, Li L, Zhuo L, Lin X. Effect and mechanism of methyl helicterate isolated from Helicteres angustifolia (Sterculiaceae) on hepatic fibrosis induced by carbon tetrachloride in rats. JOURNAL OF ETHNOPHARMACOLOGY 2012; 143:889-895. [PMID: 22967666 DOI: 10.1016/j.jep.2012.08.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 08/10/2012] [Accepted: 08/17/2012] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Methyl helicterate is a triterpenoid isolated from Helicteres angustifolia (Sterculiaceae), one of the valuable traditional Chinese herbs. Antifibrotic activities of H. angustifolia have been extensively proved. AIM OF THE STUDY The purpose of this study was to investigate the effect of methyl helicterate (MH) on liver fibrosis in rats induced by carbon tetrachloride (CCl(4)) and to explore its underlying mechanism. MATERIALS AND METHODS Hepatic fibrosis was induced in male Sprague-Dawley (SD) rats by intragastric administration with 2 ml/kg CCl(4) (mixed 1:1 in peanut oil) twice a week for 12 weeks. To evaluate the effect of MH (16.72, 33.45, 66.90 mg/kg) on hepatic fibrosis, liver function, histological study and hepatic fibrosis evaluation were performed. Liver function was assessed by determining the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (Alb) and total protein (TP). The biomarkers such as hydroxyproline (Hyp), hyaluronic acid (HA), type III precollagen (PCIII) and laminin (LN) were examined for the evaluation of hepatic fibrosis. The underlying mechanism was investigated by measuring oxidative stress level and detecting the expression of TGF-β1 mRNA and Smad3 protein. RESULTS MH (33.45, 66.90 mg/kg) treatment significantly inhibited the loss of body weight and the increase of liver index in rats induced by CCl(4). MH also improved the liver function as indicated by decreasing serum enzymatic activities of ALT, AST, TP and Alb (P<0.05). Histological results indicated that MH alleviated liver damage and reduced the formation of fibrous septa. Moreover, MH significantly decreased liver Hyp, HA, LN and PCIII (P<0.05). Research on mechanism showed that MH could markedly reduce liver malondialdehyde (MDA) concentration, increase activities of liver superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and inhibit the expression of TGF-β1 mRNA and Smad3 protein (P<0.05). CONCLUSIONS Our findings indicated that MH can inhibit CCl(4)-induced hepatic fibrosis, which may be ascribed to its radical scavenging action, antioxidant activity, and modulation of TGF-β-Smad3 signaling pathway.
Collapse
Affiliation(s)
- Quanfang Huang
- First Affiliated Hospital of Guangxi Traditional Chinese Medicine University, Nanning 530023, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gahr SA, Weber GM, Rexroad CE. Identification and expression of Smads associated with TGF-β/activin/nodal signaling pathways in the rainbow trout (Oncorhynchus mykiss). FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:1233-1244. [PMID: 22290475 DOI: 10.1007/s10695-012-9611-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 01/19/2012] [Indexed: 05/31/2023]
Abstract
The Smad proteins are essential components of the TGF-β/activin/nodal family signaling pathway. We report the identification and expression of transcripts representing three receptor Smads (Smad2a, Smad2b, and Smad3), two common Smads (Smad4a and Smad4b), and one inhibitory Smad (Smad7). Phylogenetic analysis suggests this gene family evolved through the combination of ancient and more recent salmonid genome duplication events. Tissue distribution, embryonic expression, and expression in growth hormone (GH) treated fish were assessed by reverse transcription PCR or qPCR. All six Smad transcripts were ubiquitously expressed in adult tissues. We observed the highest expression of the receptor Smads in unfertilized eggs, generally decreasing during early embryonic development and slightly increasing around 11 days post-fertilization (dpf). Smad7 expression was low for most of embryonic development, with a dramatic increase at the onset of muscle development (6 dpf), and at hatch (24 dpf). Smad4 expression was low during early embryonic development and increased after 14 dpf. The increased expression of Smad4 and Smad7 during late embryonic development may indicate modulation of gene expression by GH axis, which initiates activity during late embryonic development. These data were supported by the modulation of these Smads in the gill filament, stomach, and muscle following a GH treatment. Additionally, these changes are concurrent with the modulation of expression of TGF-β family members. Most significantly, the increased expression of Smad7 in the muscle is simultaneous with increased expression of MSTN1A and not MSTN1B during both embryonic development and following GH treatment. These data indicate a promyogenic role for Smad7 as previously identified in other non-fish species.
Collapse
Affiliation(s)
- Scott A Gahr
- Biology Department, St. Vincent College, 300 Fraser Purchase Rd., Latrobe, PA 15650, USA.
| | | | | |
Collapse
|
34
|
Bruce DL, Sapkota GP. Phosphatases in SMAD regulation. FEBS Lett 2012; 586:1897-905. [PMID: 22576046 DOI: 10.1016/j.febslet.2012.02.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/02/2012] [Accepted: 02/02/2012] [Indexed: 11/28/2022]
Abstract
SMAD transcription factors are key mediators of the transforming growth factor-beta (TGFß) family of cytokines. Reversible phosphorylation of SMAD proteins plays a key role in regulating their function. Several phosphatases have been proposed to act on SMAD proteins to influence TGFß/BMP signalling. Here we provide an overview of the SMAD regulation by different protein phosphatases and review the evidence supporting each phosphatase as a candidate SMAD-phosphatase.
Collapse
Affiliation(s)
- David L Bruce
- MRC Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dow Street, DD1 5EH Dundee, Scotland, UK
| | | |
Collapse
|
35
|
Tripathi P, Wang Y, Casey AM, Chen F. Absence of canonical Smad signaling in ureteral and bladder mesenchyme causes ureteropelvic junction obstruction. J Am Soc Nephrol 2012; 23:618-28. [PMID: 22282597 DOI: 10.1681/asn.2011060566] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Obstruction of the ureteropelvic junction (UPJ) is a common congenital anomaly frequently associated with ureteral defects. To study the molecular mechanisms that modulate ureteral development, we inactivated Smad4, the common Smad critical for transcriptional responses to TGF-β and Bmp signaling, in the ureteral and bladder mesenchyme during embryogenesis. Loss of canonical Smad signaling in these tissues caused bilateral UPJ obstruction and severe hydronephrosis beginning at embryonic day 17.5. Despite a reduction in quantity of ureteral smooth muscle, differentiation proceeded without Smad4, producing a less severe phenotype than Bmp4 mutants; this finding suggests that at least some Bmp4 functions in ureteral smooth muscle may be Smad-independent. The absence of canonical Smad signaling in the ureteral mesenchyme, but not in the urothelium itself, led to urothelial disorganization, highlighting the importance of mesenchymal support for epithelial development. Transcript profiling revealed altered expression in known Bmp targets, smooth muscle-specific genes, and extracellular matrix-related genes in mutant ureters before the onset of hydronephrosis. Expression of the Bmp target Id2 was significantly lower in Smad4 mutants, consistent with the observation that Id2 mutants develop UPJ obstruction. In summary, Smad4 deficiency reduces the number and contractility of ureteral smooth muscle cells, leading to abnormal pyeloureteral peristalsis and functional obstruction. The subsequent bending and luminal constriction of the ureter at the UPJ marks the transition from a functional obstruction to a more intractable physical obstruction, suggesting that early intervention for this disease may prevent more irreversible damage to the urinary tract.
Collapse
Affiliation(s)
- Piyush Tripathi
- Renal Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
36
|
Soond SM, Chantry A. How ubiquitination regulates the TGF-β signalling pathway: new insights and new players: new isoforms of ubiquitin-activating enzymes in the E1-E3 families join the game. Bioessays 2012; 33:749-58. [PMID: 21932223 DOI: 10.1002/bies.201100057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ubiquitination of protein species in regulating signal transduction pathways is universally accepted as of fundamental importance for normal development, and defects in this process have been implicated in the progression of many human diseases. One pathway that has received much attention in this context is transforming growth factor-beta (TGF-β) signalling, particularly during the regulation of epithelial-mesenchymal transition (EMT) and tumour progression. While E3-ubiquitin ligases offer themselves as potential therapeutic targets, much remains to be unveiled regarding mechanisms that culminate in their regulation. With this in mind, the focus of this review highlights the regulation of the ubiquitination pathway and the significance of a recently described group of NEDD4 E3-ubiquitin ligase isoforms in the context of TGF-β pathway regulation. Moreover, we now broaden these observations to incorporate a growing number of protein isoforms within the ubiquitin ligase superfamily as a whole, and discuss their relevance in defining a new 'iso-ubiquitinome'.
Collapse
Affiliation(s)
- Surinder M Soond
- University of East Anglia, School Of Biological Sciences, Norwich, Norfolk, UK.
| | | |
Collapse
|
37
|
Mechanism and regulation of nucleocytoplasmic trafficking of smad. Cell Biosci 2011; 1:40. [PMID: 22204445 PMCID: PMC3292837 DOI: 10.1186/2045-3701-1-40] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 12/28/2011] [Indexed: 11/24/2022] Open
Abstract
Smad proteins are the intracellular mediators of transforming growth factor β (TGF-β) signaling. Smads function as transcription factors and their activities require carboxyl-terminal phosphorylation by TGF-β receptor kinases which are embedded in the cell membrane. Therefore, the translocation of activated Smads from the cytoplasm into the nucleus is a rate-limiting step in TGF-β signal transduction into the nucleus. On the other hand, the export of Smads out of the nucleus turns off TGF-β effect. Such spatial control of Smad ensures a tight regulation of TGF-β target genes. Several cross-talk pathways have been shown to affect TGF-β signaling by impairing nuclear translocation of Smad, exemplifying the biological importance of the nuclear transport process. Many laboratories have investigated the underlying molecular mechanism of Smad nucleocytoplasmic translocation, combining genetics, biochemistry and sophisticated live cell imaging approaches. The last few years have witnessed the elucidation of several key players in Smad nuclear transport, most importantly the karyopherins that carry Smads across the nuclear envelope and nuclear pore proteins that facilitate the trans-nuclear envelope movement. The foundation is now set to further elucidate how the nuclear transport process is regulated and exploit such knowledge to manipulate TGF-β signaling. In this review we will discuss the current understanding of the molecular machinery responsible for nuclear import and export of Smads.
Collapse
|
38
|
Huang D, Wang Y, Wang L, Zhang F, Deng S, Wang R, Zhang Y, Huang K. Poly(ADP-ribose) polymerase 1 is indispensable for transforming growth factor-β Induced Smad3 activation in vascular smooth muscle cell. PLoS One 2011; 6:e27123. [PMID: 22073128 PMCID: PMC3205050 DOI: 10.1371/journal.pone.0027123] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 10/11/2011] [Indexed: 12/31/2022] Open
Abstract
Background Transforming growth factor type-β (TGF-β)/Smad pathway plays an essential role in vascular fibrosis. Reactive oxygen species (ROS) generation also mediates TGF-β signaling-induced vascular fibrosis, suggesting that some sort of interaction exists between Smad and redox pathways. However, the underlying molecular mechanism is largely unknown. This study aims to investigate the influence of poly(ADP-ribose) polymerase 1 (PARP1), a downstream effector of ROS, on TGF-β signaling transduction through Smad3 pathway in rat vascular smooth muscle cells (VSMCs). Methods and Results TGF-β1 treatment promoted PARP1 activation through induction of ROS generation in rat VSMCs. TGF-β1-induced phosphorylation and nuclear accumulation of Smad3 was prevented by treatment of cells with PARP inhibitor, 3-aminobenzamide (3AB) or N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetami (PJ34), or PARP1 siRNA. TGF-β1 treatment promoted poly(ADP-ribosy)lation of Smad3 via activation of PARP1 in the nucleus. Poly(ADP-ribosy)lation enhanced Smad-Smad binding element (SBE) complex formation in nuclear extracts and increased DNA binding activity of Smad3. Pretreatment with 3AB, PJ34, or PARP1 siRNA prevented TGF-β1-induced Smad3 transactivation and expression of Smad3 target genes, including collagen Iα1, collagen IIIα1 and tissue inhibitor of metalloproteinase 1, in rat VSMCs. Conclusions PARP1 is indispensable for TGF-β1 induced Smad3 activation in rat VSMCs. Targeting PARP1 may be a promising therapeutic approach against vascular diseases induced by dysregulation of TGF-β/Smad3 pathway.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Blotting, Southwestern
- Blotting, Western
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Electrophoretic Mobility Shift Assay
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Fluorescent Antibody Technique
- Immunoenzyme Techniques
- Immunoprecipitation
- Luciferases/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Phenanthrenes/pharmacology
- Phosphorylation/drug effects
- Poly(ADP-ribose) Polymerase Inhibitors
- Poly(ADP-ribose) Polymerases/genetics
- Poly(ADP-ribose) Polymerases/metabolism
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/genetics
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Signal Transduction/drug effects
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Trans-Activators
- Transcription, Genetic
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Dan Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Central Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Deng
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| | - Kai Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| |
Collapse
|
39
|
Zu X, Zhang Q, Cao R, Liu J, Zhong J, Wen G, Cao D. Transforming growth factor-β signaling in tumor initiation, progression and therapy in breast cancer: an update. Cell Tissue Res 2011; 347:73-84. [PMID: 21845401 DOI: 10.1007/s00441-011-1225-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 07/22/2011] [Indexed: 01/06/2023]
Abstract
Transforming growth factor-β (TGF-β) is a ubiquitous cytokine playing an essential role in cell proliferation, differentiation, apoptosis, adhesion and invasion, as well as in cellular microenvironment. In malignant diseases, TGF-β signaling features a growth inhibitory effect at an early stage but aggressive oncogenic activity at the advanced malignant state. Here, we update the current understanding of TGF-β signaling in cancer development and progression with a focus on breast cancer. We also review the current approaches of TGF-β signaling-targeted therapeutics for human malignancies.
Collapse
Affiliation(s)
- Xuyu Zu
- Clinical Research Institution, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Dingwall M, Marchildon F, Gunanayagam A, Louis CS, Wiper-Bergeron N. Retinoic acid-induced Smad3 expression is required for the induction of osteoblastogenesis of mesenchymal stem cells. Differentiation 2011; 82:57-65. [PMID: 21612856 DOI: 10.1016/j.diff.2011.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 04/28/2011] [Accepted: 05/09/2011] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells are pluripotent precursor cells that can differentiate into osteoblasts, adipocytes, chondrocytes and myocytes. Despite their important therapeutic potential little is known about the transcriptional cascades that govern lineage decisions in these cells. Treatment of C3H10T1/2 mouse mesenchymal stem cells with retinoic acid (RA) inhibits adipogenesis and enhances osteoblastogenesis. In particular, RA treatment stimulates the expression of the osteoblast master regulator, runt-related transcription factor 2 (Runx2), whose expression is necessary for the formation of bone. We have shown previously in mesenchymal stem cells that RA acts to stimulate osteoblastogenesis by interfering with the actions of the bzip transcription factor CCAAT/Enhancer Binding Protein beta (C/EBPβ), where it binds to a negative regulatory element within the Runx2 promoter and inhibits its expression. Herein we show that Smad3, whose expression is stimulated by RA, relays the effects of RA on differentiation by initiating the displacement of C/EBPβ from the Runx2 promoter. In addition to stimulating Smad3 expression, RA also stimulated the nuclear localization of this factor, such that in the absence of RA, ectopic Smad3 was unable to drive osteoblastogenesis. While not sufficient to promote osteoblastogenesis, knockdown of Smad3 using a specific shRNA prevented the RA-mediated stimulation of differentiation and displacement of C/EBPβ from the Runx2 P1 promoter. Taken together, these data indicate that Smad3 is an important mediator of RA activity during mesenchymal stem cell differentiation and is necessary for the stimulation of osteoblastogenesis.
Collapse
Affiliation(s)
- Molly Dingwall
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | | | | | | | | |
Collapse
|
41
|
Wang L, Liu YT, Hao R, Chen L, Chang Z, Wang HR, Wang ZX, Wu JW. Molecular mechanism of the negative regulation of Smad1/5 protein by carboxyl terminus of Hsc70-interacting protein (CHIP). J Biol Chem 2011; 286:15883-94. [PMID: 21454478 PMCID: PMC3091198 DOI: 10.1074/jbc.m110.201814] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The transforming growth factor-β (TGF-β) superfamily of ligands signals along two intracellular pathways, Smad2/3-mediated TGF-β/activin pathway and Smad1/5/8-mediated bone morphogenetic protein pathway. The C terminus of Hsc70-interacting protein (CHIP) serves as an E3 ubiquitin ligase to mediate the degradation of Smad proteins and many other signaling proteins. However, the molecular mechanism for CHIP-mediated down-regulation of TGF-β signaling remains unclear. Here we show that the extreme C-terminal sequence of Smad1 plays an indispensable role in its direct association with the tetratricopeptide repeat (TPR) domain of CHIP. Interestingly, Smad1 undergoes CHIP-mediated polyubiquitination in the absence of molecular chaperones, and phosphorylation of the C-terminal SXS motif of Smad1 enhances the interaction and ubiquitination. We also found that CHIP preferentially binds to Smad1/5 and specifically disrupts the core signaling complex of Smad1/5 and Smad4. We determined the crystal structures of CHIP-TPR in complex with the phosphorylated/pseudophosphorylated Smad1 peptides and with an Hsp70/Hsc70 C-terminal peptide. Structural analyses and subsequent biochemical studies revealed that the distinct CHIP binding affinities of Smad1/5 or Smad2/3 result from the nonconservative hydrophobic residues at R-Smad C termini. Unexpectedly, the C-terminal peptides from Smad1 and Hsp70/Hsc70 bind in the same groove of CHIP-TPR, and heat shock proteins compete with Smad1/5 for CHIP interaction and concomitantly suppress, rather than facilitate, CHIP-mediated Smad ubiquitination. Thus, we conclude that CHIP inhibits the signaling activities of Smad1/5 by recruiting Smad1/5 from the functional R-/Co-Smad complex and further promoting the ubiquitination/degradation of Smad1/5 in a chaperone-independent manner.
Collapse
Affiliation(s)
- Le Wang
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and
| | - Yi-Tong Liu
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and
| | - Rui Hao
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and
| | - Lei Chen
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and
| | - Zhijie Chang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing 100084, China and
| | - Hong-Rui Wang
- MOE Key Laboratory of Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhi-Xin Wang
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and
| | - Jia-Wei Wu
- From the MOE Key Laboratory of Bioinformatics, School of Life Sciences, and , To whom correspondence should be addressed. Tel.: 86-10-62789387; Fax: 86-10-62792826; E-mail:
| |
Collapse
|
42
|
Shepherd RD, Kos SM, Rinker KD. Flow-dependent Smad2 phosphorylation and TGIF nuclear localization in human aortic endothelial cells. Am J Physiol Heart Circ Physiol 2011; 301:H98-H107. [PMID: 21490324 DOI: 10.1152/ajpheart.00668.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Endothelial cells respond to fluid flow stimulation through transient and sustained signal pathway activation. Smad2 is a signaling molecule and transcription factor in the Smad signaling pathway, traditionally associated with TGF-β. Although phosphorylation of Smad2 in the receptor-dependent COOH-terminal region is the most appreciated way Smad2 is activated to affect gene expression, phosphorylation may also occur in the MH1-MH2 linker region (L-psmad2). Here, we show that in human aortic endothelial cells (HAEC), Smad2 was both preferentially phosphorylated in the linker region and localized to the nucleus in a flow-dependent manner. The Smad corepressor transforming growth interacting factor (TGIF) was also found to have flow-dependent nuclear localization. Tissue studies confirmed this L-psmad2 generation trend in rat aorta, indicating likely importance in arterial tissue. HAEC-based inhibitor studies demonstrated that L-psmad2 levels were not related to MAPK phosphorylation, but instead followed the pattern of pAkt(473), both with and without the phosphatidylinositol 3-kinase inhibitor PI-103. Akt and Smad species were also shown to directly interact under flow relative to static controls. To further evaluate impacts of PI-103 treatment, expression profiles for two TGF-β and shear stress-dependent genes were determined and showed that mRNAs were lower from untreated 10 dyn/cm(2) than 2 dyn/cm(2) average shear stress cultures. However, upon exposure to PI-103, this trend was reversed, with a stronger response observed at 10 dyn/cm(2). Taken together, the results of this work suggest that fluid flow exposure may influence endothelial gene expression by a novel mechanism involving Akt, L-psmad2, and TGIF.
Collapse
Affiliation(s)
- Robert D Shepherd
- Univ. of Calgary, 2500 Univ. Dr. N.W., Centre for Bioengineering Research and Education, CCIT 208, Calgary AB, Canada T2N 1N4
| | | | | |
Collapse
|
43
|
Zhang ZF, Zhang YG, Hu DH, Shi JH, Liu JQ, Zhao ZT, Wang HT, Bai XZ, Cai WX, Zhu HY, Tang CW. Smad interacting protein 1 as a regulator of skin fibrosis in pathological scars. Burns 2011; 37:665-72. [PMID: 21236582 DOI: 10.1016/j.burns.2010.12.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 11/20/2010] [Accepted: 12/05/2010] [Indexed: 01/07/2023]
Abstract
Keloids and hypertrophic scars are significant symptomatic clinical problems characterized by the excessive and abnormal deposition of collagen-based extracellular matrix (ECM) components. However, the molecular basis of keloid and hypertrophic scar formation has not been fully elucidated. Here, we demonstrated that down-regulation of the transcription factor Smad interacting protein 1 (SIP1) could be relevant to keloid and hypertrophic scar formation. The results of the present study show that the level of SIP1 mRNA is significantly decreased in pathological scar tissues and in normal skin and pathological scar fibroblasts treated with transforming growth factor β1 (TGF-β1). In contrast, the expression of SIP1 mRNA is not decreased in normotrophic scar samples. The SIP1 mRNA level inversely correlates with the mRNA level of type I collagen (COL1A2) and directly correlates with the mRNA level of matrix metalloproteinase-1 (MMP1). Overexpression of SIP1 in keloid and hypertrophic scar fibroblasts represses TGF-β1-stimulated COL1A2 expression and induces MMP1 expression. Alternatively, knockdown of SIP1 in normal skin fibroblasts enhance TGF-β1-induced COL1A2 levels. These findings suggest that SIP1 could be a regulator of skin fibrosis, and depletion of SIP1 in pathological scar tissues could result in an up-regulation of collagen and down-regulation of matrix metalloproteinase, leading to an abnormal accumulation of ECM along with fibrosis and pathological scar formation.
Collapse
Affiliation(s)
- Zhan-Feng Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Leonardi A, Di Stefano A, Motterle L, Zavan B, Abatangelo G, Brun P. Transforming growth factor-β/Smad - signalling pathway and conjunctival remodelling in vernal keratoconjunctivitis. Clin Exp Allergy 2010; 41:52-60. [PMID: 20977504 DOI: 10.1111/j.1365-2222.2010.03626.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Vernal keratoconjunctivitis (VKC) is a chronic ocular allergic inflammation characterized by corneal complications and the formation of giant papillae. Sma- and Mad-related proteins (Smad) modulate extracellular matrix gene expression during wound healing, inflammation and tissue remodelling. OBJECTIVE To investigate the relationship between allergic inflammation and TGF-β/Smad signalling pathway, expression in VKC patients and in primary cultured conjunctival fibroblasts exposed to mediators found previously over-expressed in VKC. METHODS Smad-2, -3, -7, phospho-(p)Smads, TGF-β1 and -β2 were evaluated in the conjunctiva of normal subjects (CT) and VKC patients by immunohistochemistry. The expression of Smads, pro-collagen I (PIP), TGF-β1, -β2, mitogen-activated protein kinase (p38/MAPK), c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK1/2) were also determined in conjunctival fibroblast cultures exposed to histamine, IL-4, -13, TGF-β1, IFN-γ and TNF-α using immunostaining or RT-PCR. RESULTS Immunostaining for Smad-2, -3, pSmad-2, -3, TGF-β1, -β2 and PIP was significantly increased in VKC stroma compared with CT. In conjunctival fibroblast cultures, Smad-3 and PIP were stimulated by histamine, IL-4, -13 and TGF-β1 exposure, while PIP was reduced by IFN-γ, and TNF-α mRNA expression of Smad-3 was increased by histamine, while Smad-7 was reduced by IL-4. In addition, histamine, IL-4 and TNF-α increased JNK and ERK1/2 expression. CONCLUSION AND CLINICAL RELEVANCE The TGF-β/Smad signalling pathway is over-expressed in VKC tissues and modulated in conjunctival fibroblasts by histamine, IL-4, TGF-β1 and TNF-α. These mechanisms may be involved in fibrillar collagen production, giant papillae formation and tissue remodelling typical of VKC and might provide new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- A Leonardi
- Ophthalmology Unit, Department of Neuroscience, University of Padua, Padua, Italy.
| | | | | | | | | | | |
Collapse
|
45
|
McCullagh E, Seshan A, El-Samad H, Madhani HD. Coordinate control of gene expression noise and interchromosomal interactions in a MAP kinase pathway. Nat Cell Biol 2010; 12:954-62. [PMID: 20852627 PMCID: PMC2948760 DOI: 10.1038/ncb2097] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 08/19/2010] [Indexed: 11/29/2022]
Abstract
In the Saccharomyces cerevisiae pheromone-response pathway, the transcription factor Ste12 is inhibited by two mitogen-activated protein (MAP)-kinase-responsive regulators, Dig1 and Dig2. These two related proteins bind to distinct regions of Ste12 but are redundant in their inhibition of Ste12-dependent gene expression. Here we describe three functions for Dig1 that are non-redundant with those of Dig2. First, the removal of Dig1 results in a specific increase in intrinsic and extrinsic noise in the transcriptional outputs of the mating pathway. Second, in dig1Δ cells, Ste12 relocalizes from the nucleoplasmic distribution seen in wild-type cells into discrete subnuclear foci. Third, genome-wide insertional chromatin immunoprecipitation studies revealed that Ste12-dependent genes have increased interchromosomal interactions in dig1Δ cells. These findings suggest that the regulation of gene expression through long-range gene interactions, a widely observed phenomenon, comes at the cost of increased noise. Consequently, cells may have evolved mechanisms to suppress noise by controlling these interactions.
Collapse
Affiliation(s)
- Emma McCullagh
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16 St. San Francisco, CA 94158
| | - Anupama Seshan
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16 St. San Francisco, CA 94158
| | - Hana El-Samad
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16 St. San Francisco, CA 94158
| | - Hiten D. Madhani
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16 St. San Francisco, CA 94158
| |
Collapse
|
46
|
Ishikawa M, Nishijima N, Shiota J, Sakagami H, Tsuchida K, Mizukoshi M, Fukuchi M, Tsuda M, Tabuchi A. Involvement of the serum response factor coactivator megakaryoblastic leukemia (MKL) in the activin-regulated dendritic complexity of rat cortical neurons. J Biol Chem 2010; 285:32734-32743. [PMID: 20709749 DOI: 10.1074/jbc.m110.118745] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dynamic changes in neuronal morphology and transcriptional regulation play crucial roles in the neuronal network and function. Accumulating evidence suggests that the megakaryoblastic leukemia (MKL) family members, which function not only as actin-binding proteins but also as serum response factor (SRF) transcriptional coactivators, regulate neuronal morphology. However, the extracellular ligands and signaling pathways, which activate MKL-mediated morphological changes in neurons, remain unresolved. Here, we demonstrate that in addition to MKL1, MKL2, highly enriched in the forebrain, strongly contributes to the dendritic complexity, and this process is triggered by stimulation with activin, a member of the transforming growth factor β (TGF-β) superfamily. Activin promoted dendritic complexity in a SRF- and MKL-dependent manner without drastically affecting MKL localization and protein levels. In contrast, activin promoted the nuclear export of suppressor of cancer cell invasion (SCAI), which is a corepressor for SRF and MKL. Furthermore, overexpression of SCAI blocked activin-induced SRF transcriptional responses and dendritic complexity. Collectively, these results strongly suggest that activin-SCAI-MKL signaling is a novel pathway that regulates the dendritic morphology of rat cortical neurons by excluding SCAI from the nucleus and activating MKL/SRF-mediated gene expression.
Collapse
Affiliation(s)
- Mitsuru Ishikawa
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Naoki Nishijima
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Jun Shiota
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa 228-8555, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Miho Mizukoshi
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Mamoru Fukuchi
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masaaki Tsuda
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Akiko Tabuchi
- From the Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
47
|
Seong HA, Jung H, Ha H. Murine protein serine/threonine kinase 38 stimulates TGF-beta signaling in a kinase-dependent manner via direct phosphorylation of Smad proteins. J Biol Chem 2010; 285:30959-70. [PMID: 20659902 DOI: 10.1074/jbc.m110.138370] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study demonstrated that murine protein serine/threonine kinase 38 (MPK38) coimmunoprecipitates with Smad proteins (Smad2, -3, -4, and -7) and that this association is mediated by the catalytic kinase domain of MPK38. The association between MPK38 and Smad2, -3, and -4 was significantly increased by TGF-β or ASK1 signals, whereas these signals decreased association of MPK38 with Smad7. MPK38 stimulated TGF-β-induced transcription required for TGF-β-mediated biological functions, such as apoptosis and cell growth arrest, in a kinase-dependent manner. Knockdown of endogenous MPK38 showed an opposite effect, inhibiting TGF-β signaling. MPK38-mediated phosphorylation of Smad proteins (Ser(245) of Smad2, Ser(204) of Smad3, Ser(343) of Smad4, and Thr(96) of Smad7) was also found to be crucial to the positive regulation of TGF-β signaling induced by MPK38. In addition, MPK38 enhanced nuclear translocation of Smad3, as well as redistribution of Smad7 from the nucleus to the cytoplasm, in response to TGF-β. Together, these results indicate that MPK38 functions as a stimulator of TGF-β signaling through direct interaction with and phosphorylation of Smad proteins.
Collapse
Affiliation(s)
- Hyun-A Seong
- Department of Biochemistry, School of Life Sciences, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | | | | |
Collapse
|
48
|
Zeng S, Chen J, Shen H. Controlling of bone morphogenetic protein signaling. Cell Signal 2010; 22:888-93. [DOI: 10.1016/j.cellsig.2009.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 12/31/2009] [Indexed: 01/13/2023]
|
49
|
Mangone FRR, Walder F, Maistro S, Pasini FS, Lehn CN, Carvalho MB, Brentani MM, Snitcovsky I, Federico MHH. Smad2 and Smad6 as predictors of overall survival in oral squamous cell carcinoma patients. Mol Cancer 2010; 9:106. [PMID: 20462450 PMCID: PMC2885344 DOI: 10.1186/1476-4598-9-106] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 05/12/2010] [Indexed: 12/19/2022] Open
Abstract
Background To test if the expression of Smad1-8 mRNAs were predictive of survival in patients with oral squamous cell carcinoma (SCC). Patients and Methods We analyzed, prospectively, the expression of Smad1-8, by means of Ribonuclease Protection Assay in 48 primary, operable, oral SCC. In addition, 21 larynx, 10 oropharynx and 4 hypopharynx SCC and 65 matched adjacent mucosa, available for study, were also included. For survival analysis, patients were categorized as positive or negative for each Smad, according to median mRNA expression. We also performed real-time quantitative PCR (QRTPCR) to asses the pattern of TGFβ1, TGFβ2, TGFβ3 in oral SCC. Results Our results showed that Smad2 and Smad6 mRNA expression were both associated with survival in Oral SCC patients. Cox Multivariate analysis revealed that Smad6 positivity and Smad2 negativity were both predictive of good prognosis for oral SCC patients, independent of lymph nodal status (P = 0.003 and P = 0.029, respectively). In addition, simultaneously Smad2- and Smad6+ oral SCC group of patients did not reach median overall survival (mOS) whereas the mOS of Smad2+/Smad6- subgroup was 11.6 months (P = 0.004, univariate analysis). Regarding to TGFβ isoforms, we found that Smad2 mRNA and TGFβ1 mRNA were inversely correlated (p = 0.05, R = -0.33), and that seven of the eight TGFβ1+ patients were Smad2-. In larynx SCC, Smad7- patients did not reach mOS whereas mOS of Smad7+ patients were only 7.0 months (P = 0.04). No other correlations were found among Smad expression, clinico-pathological characteristics and survival in oral, larynx, hypopharynx, oropharynx or the entire head and neck SCC population. Conclusion Smad6 together with Smad2 may be prognostic factors, independent of nodal status in oral SCC after curative resection. The underlying mechanism which involves aberrant TGFβ signaling should be better clarified in the future.
Collapse
Affiliation(s)
- Flavia R R Mangone
- Disciplina de Oncologia, Departamento de Radiologia, LIM 24, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Avenida Dr Arnaldo 455, São Paulo, Brasil.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ding CC, Thong KJ, Krishna A, Telfer EE. Activin A inhibits activation of human primordial follicles in vitro. J Assist Reprod Genet 2010; 27:141-7. [PMID: 20198416 PMCID: PMC2854985 DOI: 10.1007/s10815-010-9395-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 01/27/2010] [Indexed: 10/25/2022] Open
Abstract
PURPOSE To determine whether Activin A affects the activation and survival of human primordial follicles in vitro. METHODS Ovarian cortical biopsies from eight women undergoing elective caesarean sections or benign gynaecological procedures were taken and cut into small pieces (1-3 mm(3)), cultured in serum-free medium for 7 days with/without human recombinant Activin A at a concentration of either 50 or 100 ng/ml. Ovarian tissue were analysed by histology for follicle viability, development and density. RESULT(S) Significant activation of primordial follicles within cultured cortical tissue was observed after 7 days in control medium. However, medium supplemented with Activin A at 50 ng/ml resulted in significant inhibition of follicular activation. Increasing the concentration of Activin A to 100 ng/ml reversed the inhibitory effect. The effect of Activin A appeared to be specific to activation of non-growing (primordial) follicles into the growing population since no significant differences in follicle viability was observed between treatment groups. CONCLUSION(S) Activin A at a concentration of 50 ng/ml can inhibit the spontaneous activation of human primordial follicles in vitro indicating that this may be a component of the signalling mechanisms that maintain follicular quiescence.
Collapse
Affiliation(s)
- Chi Christina Ding
- Reproductive Medicine Unit, Liverpool Women’s Hospital, Liverpool, UK
- Assisted Conception Unit, Edinburgh Fertility and Reproductive Endocrine Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - K. Joo Thong
- Assisted Conception Unit, Edinburgh Fertility and Reproductive Endocrine Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | | - Evelyn E. Telfer
- Institute of Cell Biology, The Darwin Building, University of Edinburgh, The King’s Buildings, Mayfield Road, Edinburgh, UK
| |
Collapse
|