Insights Into the Transforming Growth Factor-Β Signaling Pathway in Cutaneous Melanoma

Total Page:16

File Type:pdf, Size:1020Kb

Insights Into the Transforming Growth Factor-Β Signaling Pathway in Cutaneous Melanoma Insights into Transforming Growth Factor-β Signaling Pathway in Cutaneous Melanoma Ann Dermatol Vol. 25, No. 2, 2013 http://dx.doi.org/10.5021/ad.2013.25.2.135 REVIEW ARTICLE Insights into the Transforming Growth Factor-β Signaling Pathway in Cutaneous Melanoma Carole Yolande Perrot1-3, Delphine Javelaud1-3, Alain Mauviel1-3 1Institut Curie, Team “TGF-β and Oncogenesis”, Equipe Labellisée Ligue Contre le Cancer, 2INSERM U1021, 3CNRS UMR 3347, Orsay, France Transforming growth factor-β (TGF-β) is a pleiotropic gro- oncogenes, including NRAS, BRAF, CKIT and cyclin- wth factor with broad tissue distribution that plays critical dependent kinase (CDK) and tumor suppressors such as roles during embryonic development, normal tissue home- CDKN2A (p16Ink4A) and p53, are linked to melanoma ostasis, and cancer. While its cytostatic activity on normal development3-5. BRAF mutations are found in 70% of all epithelial cells initially defined TGF-β signaling as a tumor melanomas and more than 90% of these mutations carry a suppressor pathway, there is ample evidence indicating that single missense mutation consisting in an activating TGF-β is a potent pro-tumorigenic agent, acting via auto- substitution of glutamate for valine at position 600 crine and paracrine mechanisms to promote peri-tumoral (BRAFV600E) within the kinase domain of this protein6,7. angiogenesis, together with tumor cell migration, immune CDKN2A is a gene involved in familial melanoma escape, and dissemination to metastatic sites. This review pathogenesis and a germline mutation is found among summarizes the current knowledge on the implication of melanoma-prone families8. Additionally, rare germline TGF-β signaling in melanoma. (Ann Dermatol 25(2) 135∼ mutations of CDK4 are exclusively found in individuals 144, 2013) with genetic predisposition to melanoma9. Increased survival of melanoma patients has been obser- -Keywords- ved over the recent decades, which may be due to earlier Melanoma, Metastasis, TGF-beta signaling, Therapeutic- detection (and subsequent surgical resection) of primary targets melanoma lesions. Yet, therapeutic options for unresec- table or metastatic melanoma remain extremely limited. Until recently, dacarbazine and interleukin-2 (IL-2) were INTRODUCTION the only two agents approved by the US Food and Drug Administration (FDA) for the treatment of metastatic mela- Melanoma, a neoplasm of the skin that arises from the noma. In 2011, two additional drugs were approved by the TM transformation of melanocytes, is the most lethal of cu- FDA: Vemurafenib (Plexxikon/Zelboraf ; Roche, Basel, taneous malignancies1. Many factors are now known to Switzerland) and Ipilimumab. Vemurafenib is an oral agent contribute to the formation of melanoma, including expo- that targets mutant BRAF, while Ipilimumab is a fully sure to ultra-violet radiation, skin type, hair/eye color, the human monoclonal antibody against cytotoxic T-lympho- presence of dysplastic nevi and/or increased nevi number, cyte antigen 4 (CTLA-4) that is expressed on the surface of and family history of melanoma2. Mutations of several activated T cells. This novel immunotherapy increases cell-mediated immunity by blocking the key immune che- Corresponding author: Alain Mauviel, Institut Curie, University Center, Bldg ckpoint molecule CTLA-4, thus enhances natural anti- 110, 91400 Orsay, France. Tel: 33-1-69-863043, Fax: 33-1-69-863051, tumor immune response. Despite early therapeutic benefit E-mail: [email protected] of these drugs, materialized by rapid and spectacular This is an Open Access article distributed under the terms of the regression of tumors, resistance rapidly occurs, with brutal Creative Commons Attribution Non-Commercial License (http:// tumor relapse. creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, Melanoma displays multifactorial etiology and its genetic provided the original work is properly cited. and immunological background has not yet been fully Vol. 25, No. 2, 2013 135 CY Perrot, et al elucidated. In vitro studies indicate that cultured melano- regulates the transcription of various target genes. ma cell lines produce excessive levels of cytokines and SMAD7, an inhibitory SMAD, interferes with TGF-β growth factors with pleiotropic biological activities10. signaling by several mechanisms: it can bind TβRI to Among them, Transforming growth factor β (TGF-β) fun- prevent SMAD2/3 phosphorylation and activation, or ctions as an autocrine and paracrine factor that drives recruit the protein phosphatase PP1/GADD34 or the E3 many cellular processes including tumor growth, invasion, ubiquitin ligases SMURF1 and SMURF2 to the receptor escape from immune surveillance, angiogenesis and meta- complex, leading to either receptor dephosphorylation or stasis11. We herein provide in-depth analysis of the rela- proteasomal degradation, respectively11,12. TGF-β path- tionship between melanoma progression and TGF-β way potentially activates other signaling pathways in a signaling. non-canonical manner, including the mitogen-activated protein kinases (MAPKs) and PI3K/AKT pathways11. TGF-β SIGNALING TGF-β IN MELANOCYTES AND MELANOMA The canonical TGF-β signaling cascade is well charac- terized. Briefly, TGF-β ligands bind to the type II trans- In normal epithelial tissues, TGF-β acts as a potent cell membrane receptor serine/threonine kinases (TβRII), whi- growth inhibitor. Thus, it is generally accepted that during ch in turn assembles with, phosphorylates and activates early stages of carcinogenesis, tumor suppressive effects of type I receptor (TβRI; ALK5). Activated TβRI phosphory- TGF-β are mainly driven by the induction of a cytostatic lates the downstream effectors SMAD2 and SMAD3, program. TGF-β upregulates cyclin-dependent kinase which then associate with SMAD4 (Fig. 1). This new (CDK) inhibitors (p21, p15, p27Kip1 and p57Kip2), while complex accumulates in the cell nucleus where it repressing pro-proliferative factors including c-MYC and inhibitor of differentiation (ID) family members11. In addi- tion to its cytostatic effects, the TGF-β/SMAD cascade also regulates the expression of several pro-apoptotic genes13, which also contribute to its tumor suppressor activities. In advanced tumors, TGF-β signaling favors cancer cell proliferation and dissemination, peri-tumoral angiogene- sis, epithelial to mesenchymal transition (EMT) of tumor cells and tumor escape from immune surveillance, thus acting as tumor promoter. In normal melanocytes, TGF-β acts as a potent inhibitor of proliferation and differentiation14. TGF-β1 represses the paired-box transcription factor PAX3, which coopera- tes with the homeobox transcription factor SOX10 to induce melanocyte differentiation15. TGF-β may also in- terfere with melanocyte maturation by inhibiting of microphtalmia transcription factor (M-MITF), a master transcription factor specific of the melanocytic lineage that controls cell survival, migration and differentiation16. Cell autonomous activation of the TGF-β pathway in me- lanoma cell lines has been well documented17,18. While Fig. 1. The transforming growth factor (TGF)-β signaling cascade. TGF-β binds to the type II receptor that recruits, phosphorylates normal melanocytes only express TGF-β1, melanoma cells and activates type I receptor (TβRI). TβRI in turn phosphorylates express and secrete high quantities of the three TGF-β SMAD2/3 which then associate with the co-SMAD SMAD4 to isoforms, TGF-β1, TGF-β2 and TGF-β312. TGF-β ligan- form a heterocomplex that accumulates in the nucleus to regulate ds are able to induce their own expression through a target gene transcription. Inhibitory SMAD7 binds activated TβRI to prevent SMAD2/3 phosphorylation, or recruits E3 ubiquitin positive amplification loop. Furthermore, treatment of me- ligases (SMURF1/2, WW1) or phosphatase GADD34/PP1 to the lanoma cell lines with exogenous TGF-β1 induces the receptor complexes, inducing proteasomal degradation or de- secretion of active TGF-β1 and TGF-β219. Consistently, phosphorylation of the latter, respectively. C-SKI and SNON act β as transcriptional co-repressors to repress TGF-β-induced, plasma levels of TGF- are found elevated in melanoma 20 SMAD-driven, transcription. TβRII: TGF-βreceptor type II. patients, in parallel with metastatic progression . Also, 136 Ann Dermatol Insights into Transforming Growth Factor-β Signaling Pathway in Cutaneous Melanoma TGF-β2 expression was found to correlate with tumor miR-21129. Yet, TGF-β has been implicated in the meta- thickness and invasiveness21. static spread of melanoma12, suggesting that increased Genome-wide expression analysis of nearly one hundred TGF-β signaling through TβRII expression might enhan- human melanoma cell lines identified two groups with ce melanoma invasion and motility when miR-211 levels very distinct gene expression profiles associated with are low. distinct behaviors in vitro: the first group is characterized by high expression of neural crest and melanocytic di- MELANOMA RESPONSIVENESS TO TGF-β fferentiation markers and exhibits a proliferative, poorly Melanoma resistance to TGF-β-mediated growth supp- invasive phenotype, while the other group is charac- ression terized by the expression of a number of genes whose concomitant expression is reminiscent of a TGF-β signa- While normal melanocytes are growth-inhibited by TGF-β, ture, and associated with a highly invasive phenotype and melanoma cells exhibit a variable degree of resistance to its low
Recommended publications
  • Recombinant Human TGF-Beta 2 Protein
    ACROBiosystems Recombinant Human TGF-Beta 2 Protein Cat. # TG2-H4213 For Research and Further Cell Culture Manufacturing Use Source: Bioactivity: Recombinant Human TGF Beta 2 Protein (rhTGFB2) The bio-activity was determined by its ability to Ala 303 - Ser 414 (Accession # NP_003229.1) was inhibit IL-4 induced HT-2 cell proliferation. ED50<0.1 produced in human HEK293 cells at ACRObiosystems. ng/ml, corresponding to a specific activity of >1X107 Unit/mg Molecular Characterization: Formulation: rhTGFB2 contains no “tags” and has a calculated MW of 12.7 kDa (monomer). DTT-reduced protein migrates Lyophilized from 0.22 μm filtered solution in TFA, as a 13 kDa polypeptide and the non-reduced cystine- acetonitrile. Normally Mannitol or Trehalose are linked homodimer migrates as a 25 kDa protein. added as protectants before lyophilization. Contact us for customized product format or Endotoxin: formulation. Less than 1.0 EU per μg of the rhTGFB2 by the LAL Reconstitution : method. See Certificate of Analysis for details of Purity: reconstitution instruction and specific concentration. >98% as determined by SDS-PAGE of reduced (+) and Storage: non-reduced (-) rhTGFB2. Avoid repeated freeze-thaw cycles. No activity loss was observed after storage at: SDS-PAGE: • 4-8℃ for 1 year in lyophilized state The purity of rhTGFB2 • 4-8℃ for 1 month under sterile conditions after was determined by SDS- reconstitution PAGE of reduced (+) and • -20 ℃ to -70 ℃ for 3 months under sterile non-reduced (-) rhTGFB2 conditions after reconstitution and staining overnight with Coomassie Blue. Background: Transforming growth factor beta 2 ( TGFB2) is also known as TGF-β2, TGF-beta2, Glioblastoma-derived T-cell suppressor factor, G-TSF, BSC-1 cell growth inhibitor, Polyergin, Cetermin, and is a polypeptide member of the transforming growth factor beta superfamily of cytokines.
    [Show full text]
  • Role of TGF-Β3 in the Regulation of Immune Responses T
    Role of TGF-β3 in the regulation of immune responses T. Okamura1,2, K. Morita1, Y. Iwasaki1, M. Inoue1, T. Komai1, K. Fujio1, K. Yamamoto1 1Department of Allergy and ABSTRACT ences, indicating their non-redundancy Rheumatology, Graduate School of Transforming growth factor-betas (12-16). The TGF-βs have opposite ef- Medicine, The University of Tokyo, (TGF-βs) are multifunctional cytokines fects on tissue fibrosis. Wound-healing Tokyo, Japan; that have been implicated in the regu- experiments revealed that TGF-β1 2Max Planck-The University of Tokyo Center for Integrative Inflammology, lation of a broad range of biological and TGF-β2 cause fibrotic scarring The University of Tokyo, Tokyo, Japan. processes, including cell proliferation, responses and that TGF-β3 induces a Tomohisa Okamura, MD, PhD cell survival, and cell differentiation. scar-free response (17). Both TGF-β1 Kaoru Morita, MD The three isoforms identified in mam- and -β2 activate the collagen α2 (I) Yukiko Iwasaki, MD, PhD mals, TGF-β1, TGF-β2, and TGF-β3, gene promoter, resulting in increased Mariko Inoue, MD have high sequence homology, bind to collagen synthesis (18). It was also re- Toshihiko Komai, MD the same receptors, and show similar ported that TGF-β1, but not TGF-β3, is Keishi Fujio, MD, PhD biological functions in many in vitro a crucial factor in the development of Kazuhiko Yamamoto, MD, PhD studies. However, analysis of the in vivo pulmonary fibrosis (19). Most of the in- Please address correspondence to: functions of the three isoforms and mice formation on the immunological activ- Keishi Fujio, MD, PhD, deficient for each cytokine reveals strik- Department of Allergy and ity of TGF-βs derives from studies of Rheumatology, ing differences, illustrating their unique TGF-β1 and, in part, TGF-β2, whereas Graduate School of Medicine, biological importance and functional recent investigations have begun to il- The University of Tokyo, non-redundancy.
    [Show full text]
  • Transforming Growth Factor-Β Soluble Receptor III (TGF-Β Sriii)
    Transforming Growth Factor-b Soluble Receptor III (TGF-b sRIII) Human, Recombinant Expressed in mouse NSO cells Product Number T4567 Product Description Reagent Transforming Growth Factor-b soluble Receptor III Recombinant human TGF-b sRIII is supplied as (TGF-b sRIII) is produced from a DNA sequence approximately 100 mg of protein lyophilized from a encoding the amino terminal (781 amino acid residue) 0.2 mm filtered solution in phosphate buffered saline extracellular domain of human TGF-b receptor type III (PBS) containing 5 mg of bovine serum albumin. protein.1 Mature human TGF-b sRIII, a 760 amino acid residue protein generated after cleavage of a 21 amino Preparation Instructions acid residue signal peptide, has a predicted molecular Reconstitute the contents of the vial using sterile mass of approximately 84 kDa. As a result of glycosy- phosphate-buffered saline (PBS) containing at least lation, recombinant TGF-b sRIII migrates as a 100 kDa 0.1% human serum albumin or bovine serum albumin. protein in SDS-PAGE. Prepare a stock solution of no less than 20 mg/ml. The transforming growth factor-b (TGF-b) family of Storage/Stability cytokines are multifunctional peptides; capable of Store at -20 °C. Upon reconstitution, store at 2 °C to influencing cell proliferation, growth, differentiation, and 8 °C for one month. For extended storage, freeze in other functions in a wide range of cell types. Most working aliquots. Repeated freezing and thawing is not mammalian cells express three abundant high affinity recommended. Do not store in a frost-free freezer. TGF receptors, which can bind and be cross-linked to 2 TGF-b.
    [Show full text]
  • Augmented and Accelerated Nephrogenesis in TGF-␤2 Heterozygous Mutant Mice
    0031-3998/08/6306-0607 Vol. 63, No. 6, 2008 PEDIATRIC RESEARCH Printed in U.S.A. Copyright © 2008 International Pediatric Research Foundation, Inc. Augmented and Accelerated Nephrogenesis in TGF-␤2 Heterozygous Mutant Mice SUNDER SIMS-LUCAS, GEORGINA CARUANA, JOHN DOWLING, MICHELLE M. KETT, AND JOHN F. BERTRAM Department of Anatomy and Developmental Biology [S.S.-L., G.C., J.F.B.], Monash University, Melbourne 3800, Australia; Department of Anatomical Pathology [J.D.], Alfred Hospital, Prahran 3181, Australia; Department of Physiology [M.M.K.], Monash University, Melbourne 3800, Australia ABSTRACT: Several members of the transforming growth factor-␤ development than TGF-␤1 and TGF-␤3 (21). TGF-␤2 gene (TGF-␤) superfamily play key roles in kidney development, either expression has been detected in the kidney at embryonic day directly or indirectly regulating nephron number. Although low (E) 11.5 (22) and the protein localized in and around the nephron number is a risk factor for cardiovascular and renal disease, ureteric epithelium and nephron structures from E12.5 the implications of increased nephron number has not been examined (21,23). TGF-␤2 homozygous null mutant mice (TGF-␤2Ϫ/Ϫ) due to the absence of appropriate animal models. Here, using unbi- die within 24 h of birth due to defective lung development ased stereology we demonstrated that kidneys from TGF-␤2 het- erozygous (TGF-␤2ϩ/Ϫ) mice have approximately 60% more (24). The kidneys of these mice display progressive deterio- nephrons than wild-type mice at postnatal day 30. To determine ration of the ureteric tubules and an enlarged renal pelvis after whether augmented nephron number involved accelerated ureteric E15.
    [Show full text]
  • Growth Factors and Signaling Proteins in Craniofacial Development Robert Spears and Kathy K.H
    Growth Factors and Signaling Proteins in Craniofacial Development Robert Spears and Kathy K.H. Svoboda Regulation of growth and development is controlled by the interactions of cells with each other and the extracellular environment through signal transduction pathways that control the differentiation process by stimulating proliferation or causing cell death. This review will define the common signaling molecules and provide an overview of the general principles of signal transduction events. We also review the signal transduction pathways controlling one specific mechanism found in craniofacial development termed epithelial- mesenchymal transformation (EMT) employed during gastrulation, cranial neural crest migration, and secondary palate formation. Semin Orthod 11:184–198 © 2005 Elsevier Inc. All rights reserved. Growth Factors and receptors (GPCR, Fig 1A), ion-channel receptors, tyrosine Signal Transduction kinase-linked receptors, and receptors with intrinsic enzy- matic activity.2 The GPCRs are characterized by multiple ntracellular signaling is usually triggered by a cell surface transmembrane domains (usually seven) that wind the pro- Ievent such as a specific protein (ligand) binding to a cell tein in and out of the membrane in a serpentine conformation surface receptor to form a receptor-ligand interaction. Cells (Fig 1A, GPCR). The ion-channel receptors are closely re- contacting neighboring cells or their surrounding noncellular lated and actually open a membrane channel when the tissue are termed cell-cell or cell-extracellular matrix (ECM) ligand binds. Many cytokine receptors are in the tyrosine contacts (Fig 1A).1 Interactions of cells with other cells or the kinase-linked class as they lack intrinsic activity; but when ECM can stimulate many reactions, including: increased cell the ligand binds, intracellular tyrosine kinases become division, cell movement, differentiation, and even pro- activated to generate cellular changes.
    [Show full text]
  • Transforming Growth Factor-Β Gene Silencing Using Adenovirus Expressing TGF-Β1 Or TGF-Β2 Shrna
    Cancer Gene Therapy (2013) 20, 94–100 & 2013 Nature America, Inc. All rights reserved 0929-1903/13 www.nature.com/cgt ORIGINAL ARTICLE Transforming growth factor-b gene silencing using adenovirus expressing TGF-b1 or TGF-b2 shRNA SOh1,2, E Kim1,2, D Kang1,3, M Kim1, J-H Kim4 and JJ Song1 Tumor cells secrete a variety of cytokines to outgrow and evade host immune surveillance. In this context, transforming growth factor-b1 (TGF-b1) is an extremely interesting cytokine because it has biphasic effects in cancer cells and normal cells. TGF-b1 acts as a growth inhibitor in normal cells, whereas it promotes tumor growth and progression in tumor cells. Overexpression of TGF-b1 in tumor cells also provides additional oncogenic activities by circumventing the host immune surveillance. Therefore, this study ultimately aimed to test the hypothesis that suppression of TGF-b1 in tumor cells by RNA interference can have antitumorigenic effects. However, we demonstrated here that the interrelation between TGF-b isotypes should be carefully considered for the antitumor effect in addition to the selection of target sequences with highest efficacy. The target sequences were proven to be highly specific and effective for suppressing both TGF-b1 mRNA and protein expression in cells after infection with an adenovirus expressing TGF-b1 short hairpin RNA (shRNA). A single base pair change in the shRNA sequence completely abrogated the suppressive effect on TGF-b1. Surprisingly, the suppression of TGF-b1 induced TGF-b3 upregulation, and the suppression of TGF-b2 induced another unexpected downregulation of both TGF-b1 and TGF-b3.
    [Show full text]
  • Tgf-Β Receptor Ii
    CONTEXT SPECIFIC SIGNALING OF TGF-β RECEPTOR II DISSERTATION ZUR ERLANGUNG DES AKADEMISCHEN GRADES DES DOKTORS DER NATURWISSENSCHAFTEN (DR. RER. NAT.) EINGEREICHT IM FACHBEREICH BIOLOGIE,CHEMIE,PHARMAZIE DER FREIEN UNIVERSITÄT BERLIN vorgelegt von DANIEL WALTER HORBELT AUS ERLENBACH AM MAIN 2010 Diese Arbeit wurde von Oktober 2004 bis August 2010 in der Arbeitsgruppe von Prof. Petra Knaus am Institut für Chemie-Biochemie der Freien Universität Berlin angefertigt. 1. GUTACHTER:PROF.DR.PETRA KNAUS 2. GUTACHTER: PD DR.PETER N. ROBINSON Disputation am 12.11.2010 . Contents List of Figures V List of Tables VII Summary IX Zusammenfassung XI 1 Introduction 1 1.1 The TGF-β superfamily . .1 1.2 Mechanisms of TGF-β signaling . .3 1.2.1 Secretion and activation of TGF-β ...................3 1.2.2 Ligand binding and receptor activation of TGF-β receptors . .3 1.2.3 TGF-β isoform specificity . .6 1.2.4 The TGF-β receptor II isoform TβRII-B and aim of the project: "Antibodies against TβRII-B" . .8 1.2.5 Activation of Smad signaling by TGF-β receptors . .9 1.2.6 TGF-β signaling through Smads . 12 1.2.7 Non-Smad signaling by TGF-β ..................... 13 1.2.8 Endocytic regulation of TGF-β signaling . 15 1.3 Marfan Syndrome and MFS-related diseases . 17 1.3.1 Classical Marfan Syndrome . 17 1.3.2 Marfan syndrome type II, Loeys-Dietz syndrome and Thoracic Aortic Aneurysms and Dissections syndrome . 19 1.4 Pathological remodeling in the aortic media and the development of aortic aneurysms . 21 1.5 Role of TGF-β in vascular remodeling .
    [Show full text]
  • TGF-B1 Suppresses Apoptosis Via Differential Regulation of MAP Kinases and Ceramide Production
    Cell Death and Differentiation (2003) 10, 516–527 & 2003 Nature Publishing Group All rights reserved 1350-9047/03 $25.00 www.nature.com/cdd TGF-b1 suppresses apoptosis via differential regulation of MAP kinases and ceramide production H-H Chen1, S Zhao1 and J-G Song*,1 Introduction 1 Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, The balance between cell proliferation, differentiation, and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, apoptosis is controlled by various internal and external stimuli, Shanghai, People’s Republic of China which play crucial roles in normal development and home- * Corresponding author: J Song, Laboratory of Molecular Cell Biology, Institute ostasis of living cells and organisms. Disruption of this of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, balance by different factors may lead to abnormal cell death, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, growth, and many pathological events, including cancer, People’s Republic of China. E-mail: [email protected] immune and developmental diseases. Studies on the under- lying mechanisms through which the balance between cell Received 22.7.02; revised 18.10.02; accepted 29.10.02 Edited by JYJ Wang proliferation, differentiation, and apoptosis are maintained or disrupted are important in understanding various physiologi- cal and pathological processes. It is now known that complex Abstract signaling network, which is formed by the interaction or ‘crosstalk’ among various signaling molecules and pathways, Serum deprivation induces apoptosis in NIH3T3 cells, which is implicated in the subtle and general control of cell is associated with increased intracellular ceramide genera- proliferation, differentiation, and apoptosis.
    [Show full text]
  • Transforming Growth Factor-β in Stem Cells and Tissue Homeostasis
    Bone Research www.nature.com/boneres REVIEW ARTICLE OPEN Transforming growth factor-β in stem cells and tissue homeostasis Xin Xu1, Liwei Zheng2, Quan Yuan3, Gehua Zhen4, Janet L. Crane4,5, Xuedong Zhou1 and Xu Cao4 TGF-β 1–3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF- β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/ fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
    [Show full text]
  • The Potential Role of Transforming Growth Factor Beta Family Ligand Interactions in Prostate Cancer
    AIMS Molecular Science, 4(1): 41-61. DOI: 10.3934/molsci.2017.1.41 Received 19 October 2016, Accepted 25 January 2017, Published 13 February 2017 http://www.aimspress.com/journal/Molecular Review The potential role of transforming growth factor beta family ligand interactions in prostate cancer Kit P. Croxford, Karen L. Reader, and Helen D. Nicholson* Department of Anatomy, University of Otago, Dunedin 9054, New Zealand * Correspondence: Email: [email protected]; Tel: +64 3 479 5295. Abstract: The transforming growth factor beta (TGF-β) family plays an important role in embryonic development and control of the cell cycle. Members of the TGF-β family have pleiotropic functions and are involved in both the inhibition and progression of various cancers. In particular, deregulation of the TGF-β family has been associated with prostate cancer, as both a mechanism of disease progression and a possible therapeutic target. This review concentrates on the TGF-βs, activins and inhibins, bone morphogenetic proteins and NODAL and their connection to prostate cancer. Whilst most studies examine the family members in isolation, there are multiple interactions that may occur between members which can alter their function. Such interactions include ligand competition for receptor binding and shared intracellular pathways such as the Mothers against decapentaplegic (SMAD) proteins. Another mechanism for interaction within the TGF-β family is facilitated by their dimeric structure; heterodimers can form which exhibit different functional capabilities to their homodimeric counterparts. The potential formation of TGF-β family heterodimers has not been well examined in prostate cancer. The multiple methods of interrelations between members highlights the need for gross analysis of the TGF-β family and related factors in association with prostate cancer, in order to discover possible future avenues for TGF-β based diagnosis and treatments of the disease.
    [Show full text]
  • The Effects of Transforming Growth Factor-B2 on the Expression of Follistatin and Activin a in Normal and Glaucomatous Human Trabecular Meshwork Cells and Tissues
    Biochemistry and Molecular Biology The Effects of Transforming Growth Factor-b2 on the Expression of Follistatin and Activin A in Normal and Glaucomatous Human Trabecular Meshwork Cells and Tissues Ashley M. Fitzgerald, Cecilia Benz, Abbot F. Clark, and Robert J. Wordinger PURPOSE. To compare follistatin (FST) and activin (Act) expres- in irreversible blindness and is estimated to affect more than 60 sion in normal and glaucomatous trabecular meshwork (TM) million people.2 Important risk factors for POAG include age, cells and tissues and determine if exogenous TGF-b2 regulates race, and elevated IOP. Elevated IOP results from increased the expression of FST and Act in TM cells. resistance of aqueous humor (AH) outflow through the METHODS. Total RNA was isolated from TM cell strains, and mRNA trabecular meshwork (TM) due to excess accumulation of expression for FST 317/344 isoforms and Act was determined via extracellular matrix (ECM) proteins.4–6 RT-PCR and quantitative PCR (qPCR). Western immunoblotting TGF-b2isthemostabundantTGF-b isoform in the eye.7,8 A and immunocytochemistry determined FST and Act A protein number of studies have reported elevated levels of TGF-b2(2– levels in normal TM (NTM) and glaucomatous TM (GTM) cells. 5ng/mL) in the AH of patients with POAG.7,9–11,51 Endoge- Cells were treated with recombinant human TGF-b2 protein at 0 nous TGF-b2 levels are elevated in both cultured glaucoma- to 10 ng/mL for 0 to 72 hours. qPCR, Western immunoblotting, tous TM (GTM) cell stains and GTM tissues.12,48,49 In other immunocytochemistry, and ELISA immunoassay were utilized to tissues, TGF-b signaling has been shown to mediate fibrotic determine changes in FST and Act A mRNA and protein levels.
    [Show full text]
  • Cardiac-Specific Transcription Factor Genes Smad4 and Gata4 Cooperatively Regulate Cardiac Valve Development
    Corrections MEDICAL SCIENCES DEVELOPMENTAL BIOLOGY Correction for “Irradiation induces bone injury by damaging Correction for “Cardiac-specific transcription factor genes Smad4 bone marrow microenvironment for stem cells,” by Xu Cao, and Gata4 cooperatively regulate cardiac valve development,” by Xiangwei Wu, Deborah Frassica, Bing Yu, Lijuan Pang, Lingling Ivan P. Moskowitz, Jun Wang, Michael A. Peterson, William Xian, Mei Wan, Weiqi Lei, Michael Armour, Erik Tryggestad, T. Pu, Alexander C. Mackinnon, Leif Oxburgh, Gerald John Wong, Chun Yi Wen, William Weijia Lu, and Frank C. Chu, Molly Sarkar, Charles Berul, Leslie Smoot, Elizabeth J. Frassica, which appeared in issue 4, January 25, 2011, of Proc J. Robertson, Robert Schwartz, Jonathan G. Seidman, and Natl Acad Sci USA (108:1609–1614; first published January 10, Christine E. Seidman, which appeared in issue 10, March 8, 2011; 10.1073/pnas.1015350108). 2011, of Proc Natl Acad Sci USA (108:4006–4011; first published The authors note that their conflict of interest statement was February 17, 2011; 10.1073/pnas.1019025108). omitted during publication. The authors declare that John Wong The authors note that the title appeared incorrectly. The title has a research service contract with Gulmay Medical, Inc. in the should instead appear as “Transcription factor genes Smad4 and transfer of the SARRP technology from Johns Hopkins to the Gata4 cooperatively regulate cardiac valve development.” The company. The SARRP was used by Dr. Xu Cao to irradiate his online version has been corrected. study animals, and was only peripherally related to the subject matter of the manuscript. Additionally, the specific unit was www.pnas.org/cgi/doi/10.1073/pnas.1103973108 constructed at Johns Hopkins with grant support from the National Cancer Institute (US), and not by Gulmay.
    [Show full text]