1
|
Choi AS, Jenkins-Lane LM, Barton W, Kumari A, Lancaster C, Raulerson C, Ji H, Altomare D, Starr MD, Whitaker R, Phaeton R, Arend R, Shtutman M, Nixon AB, Hempel N, Lee NY, Mythreye K. Heparan sulfate modifications of betaglycan promote TIMP3-dependent ectodomain shedding to fine-tune TGF-β signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555364. [PMID: 37693479 PMCID: PMC10491198 DOI: 10.1101/2023.08.29.555364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
In pathologies such as cancer, aberrant Transforming Growth Factor-β (TGF-β) signaling exerts profound tumor intrinsic and extrinsic consequences. Intense clinical endeavors are underway to target this pivotal pathway. Central to the success of these interventions is pinpointing factors that decisively modulate the TGF-β responses. Betaglycan/type III TGF-β receptor (TβRIII), is an established co-receptor for the TGF-β superfamily known to bind directly to TGF-βs 1-3 and inhibin A/B. While betaglycan can be membrane-bound, it can also undergo ectodomain cleavage to produce soluble-betaglycan that can sequester its ligands. The extracellular domain of betaglycan undergoes heparan sulfate and chondroitin sulfate glycosaminoglycan modifications, transforming betaglycan into a proteoglycan. Here we report the unexpected discovery that the heparan sulfate modifications are critical for the ectodomain shedding of betaglycan. In the absence of such modifications, betaglycan is not shed. Such shedding is indispensable for the ability of betaglycan to suppress TGF-β signaling and the cells' responses to exogenous TGF-β ligands. Using unbiased transcriptomics, we identified TIMP3 as a key regulator of betaglycan shedding and thereby TGF-β signaling. Our results bear significant clinical relevance as modified betaglycan is present in the ascites of patients with ovarian cancer and can serve as a marker for predicting patient outcomes and TGF-β signaling responses. These studies are the first to demonstrate a unique reliance on the glycosaminoglycan modifications of betaglycan for shedding and influence on TGF-β signaling responses. Dysregulated shedding of TGF-β receptors plays a vital role in determining the response and availability of TGF-βs', which is crucial for prognostic predictions and understanding of TGF-β signaling dynamics.
Collapse
|
2
|
Zhou C, Li J, Lin L, Shu R, Dong B, Cao D, Li Q, Wang Z. A targeted transforming growth factor-beta (TGF-β) blocker, TTB, inhibits tumor growth and metastasis. Oncotarget 2018; 9:23102-23113. [PMID: 29796175 PMCID: PMC5955403 DOI: 10.18632/oncotarget.24562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/13/2017] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor beta (TGF-β) promotes cancer growth in late stage cancers. To inhibit the TGF-β pathway, we investigated a tumor-targeting TGF-β receptor blocker, TTB, and its role in tumor progress. The targeted TTB comprised of the extracellular domain of the TGF-β receptor II, the endoglin domain of TGF-β receptor III, and the human immuno-globin IgG1 constant fragment (Fc). To enhance tumor microenvironment targeting, a RGD peptide was fused at the N-terminal of TTB. The targeted TTB exhibited potent TGF-β neutralization activities, and inhibited cancer cell migration and invasion as well as colony formation. In xenograft models, the TTB had potent tumor inhibition activities. The TTB also attenuated the TGF-β1-induced Smad2 phosphorylation and epithelial to mesenchymal transformation (EMT), and suppressed breast cancer metastasis. Thus, the TTB is an effective TGF-β blocker with a potential for blocking excessive TGF-β induced pathogenesis in vivo.
Collapse
Affiliation(s)
- Changhua Zhou
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jing Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Limin Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Rui Shu
- Ying Rui Inc., Guangzhou, Guangdong, 510009, China
| | - Bin Dong
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510009, China
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Qing Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Zhong Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
3
|
Qin T, Barron L, Xia L, Huang H, Villarreal MM, Zwaagstra J, Collins C, Yang J, Zwieb C, Kodali R, Hinck CS, Kim SK, Reddick RL, Shu C, O'Connor-McCourt MD, Hinck AP, Sun LZ. A novel highly potent trivalent TGF-β receptor trap inhibits early-stage tumorigenesis and tumor cell invasion in murine Pten-deficient prostate glands. Oncotarget 2018; 7:86087-86102. [PMID: 27863384 PMCID: PMC5349899 DOI: 10.18632/oncotarget.13343] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
The effects of transforming growth factor beta (TGF-β) signaling on prostate tumorigenesis has been shown to be strongly dependent on the stage of development, with TGF-β functioning as a tumor suppressor in early stages of disease and as a promoter in later stages. To study in further detail the paradoxical tumor-suppressive and tumor-promoting roles of the TGF-β pathway, we investigated the effect of systemic treatment with a TGF-β inhibitor on early stages of prostate tumorigenesis. To ensure effective inhibition, we developed and employed a novel trivalent TGF-β receptor trap, RER, comprised of domains derived from the TGF-β type II and type III receptors. This trap was shown to completely block TβRII binding, to antagonize TGF-β1 and TGF-β3 signaling in cultured epithelial cells at low picomolar concentrations, and it showed equal or better anti-TGF-β activities than a pan TGF-β neutralizing antibody and a TGF-β receptor I kinase inhibitor in various prostate cancer cell lines. Systemic administration of RER inhibited prostate tumor cell proliferation as indicated by reduced Ki67 positive cells and invasion potential of tumor cells in high grade prostatic intraepithelial neoplasia (PIN) lesions in the prostate glands of Pten conditional null mice. These results provide evidence that TGF-β acts as a promoter rather than a suppressor in the relatively early stages of this spontaneous prostate tumorigenesis model. Thus, inhibition of TGF-β signaling in early stages of prostate cancer may be a novel therapeutic strategy to inhibit the progression as well as the metastatic potential in patients with prostate cancer.
Collapse
Affiliation(s)
- Tai Qin
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Vascular Surgery, Second Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Lindsey Barron
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lu Xia
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Department of Gynecology and Obstetrics, Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria M Villarreal
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - John Zwaagstra
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Cathy Collins
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Junhua Yang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA
| | - Christian Zwieb
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ravindra Kodali
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Sun Kyung Kim
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Chang Shu
- Department of Vascular Surgery, Second Xiangya Hospital and Xiangya School of Medicine, Central South University, Hunan, China
| | - Maureen D O'Connor-McCourt
- National Research Council Human Health Therapeutics Portfolio, Montréal, Quebec, Canada, Maureen O'Connor-McCourt is currently affiliated with Formation Biologics, Montréal, Quebec, Canada
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, Texas, USA
| |
Collapse
|
4
|
Paracrine Secretion of Transforming Growth Factor β by Ductal Cells Promotes Acinar-to-Ductal Metaplasia in Cultured Human Exocrine Pancreas Tissues. Pancreas 2017; 46:1202-1207. [PMID: 28902792 PMCID: PMC6192250 DOI: 10.1097/mpa.0000000000000913] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE We aimed to evaluate the contribution of acinar-to-ductal metaplasia (ADM) to the accumulation of cells with a ductal phenotype in cultured human exocrine pancreatic tissues and reveal the underlying mechanism. METHODS We sorted and cultured viable cell populations in human exocrine pancreatic tissues with a flow cytometry-based lineage tracing method to evaluate possible mechanisms of ADM. Cell surface markers, gene expression pattern, and sphere formation assay were used to examine ADM. RESULTS A large proportion of acinar cells gained CD133 expression during the 2-dimensional culture and showed down-regulation of acinar markers and up-regulation of ductal markers, assuming an ADM phenotype. In a serum-free culture condition, ADM induction was mainly dependent on transforming growth factor β (TGF-β) secreted from cultured ductal cells. Human acinar cells when cultured alone for a week in a serum-free condition do not undergo ADM. However, serum may contain other factors besides TGF-β to induce ADM in human acinar cells. In addition, we found that TGF-β cannot induce ADM of murine acinar cells. CONCLUSIONS Ductal cells are the major source of TGF-β that induces ADM in cultured human exocrine pancreatic tissues. This culture system might be a useful model to investigate the mechanism of ADM in human cells.
Collapse
|
5
|
Xu YT, Shen MH, Jin AY, Li H, Zhu R. Maternal circulating levels of transforming growth factor-β superfamily and its soluble receptors in hypertensive disorders of pregnancy. Int J Gynaecol Obstet 2017; 137:246-252. [PMID: 28281288 DOI: 10.1002/ijgo.12142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 12/03/2016] [Accepted: 03/06/2017] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To assess circulating levels of transforming growth factor (TGF)-β superfamily members and their soluble receptors in hypertensive disorders of pregnancy, and to investigate associations with clinical manifestations. METHODS A retrospective study was conducted using data for women admitted to a center in China for delivery between May 2011 and April 2013. Women with severe pre-eclampsia, mild pre-eclampsia, and gestational hypertension were included, along with a control group. Serum levels of activin A, inhibin A, TGF-β1, soluble endoglin (sEng), and soluble betaglycan (sBG) were measured. RESULTS Women with severe pre-eclampsia (n = 17) had higher mean levels of activin A (23.5±2.1 μg/L), inhibin A (1.7±0.2 μg/L), sEng (32.1±3.2 μg/L), and sBG (84.1±9.4 μg/L) than did normotensive controls (n = 18), women with gestational hypertension (n = 15), and those with mild pre-eclampsia (n = 14; all P<0.05). Women with early-onset pre-eclampsia (n = 13) had higher levels of these serum markers than did preterm normotensive controls (n = 8; all P<0.001). Women with severe or early-onset pre-eclampsia had the lowest TGF-β1 levels. Activin A, inhibin A, sEng, and sBG levels were positively correlated with mean arterial pressure and proteinuria (all P<0.01). CONCLUSION Pre-eclampsia is associated with an imbalance of members of the TGF-β superfamily and their soluble receptors, which might contribute to the development of pre-eclampsia and help to predict onset and severity.
Collapse
Affiliation(s)
- Yan-Ting Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min-Hong Shen
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ai-Ying Jin
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Li
- Center for Human Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Rui Zhu
- Center for Human Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
6
|
Soluble CD109 binds TGF-β and antagonizes TGF-β signalling and responses. Biochem J 2015; 473:537-47. [PMID: 26621871 DOI: 10.1042/bj20141488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 11/30/2015] [Indexed: 12/16/2022]
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine implicated in many diseases, including tissue fibrosis and cancer. TGF-β mediates diverse biological responses by signalling through type I and II TGF-β receptors (TβRI and TβRII). We have previously identified CD109, a glycosylphosphatidylinositol (GPI)-anchored protein, as a novel TGF-β co-receptor that negatively regulates TGF-β signalling and responses and demonstrated that membrane-anchored CD109 promotes TGF-β receptor degradation via a SMAD7/Smurf2-mediated mechanism. To determine whether CD109 released from the cell surface (soluble CD109 or sCD109) also acts as a TGF-β antagonist, we determined the efficacy of recombinant sCD109 to interact with TGF-β and inhibit TGF-β signalling and responses. Our results demonstrate that sCD109 binds TGF-β with high affinity as determined by surface plasmon resonance (SPR) and cell-based radioligand binding and affinity labelling competition assays. SPR detected slow dissociation kinetics between sCD109 and TGF-β at low concentrations, indicating a stable and effective interaction. In addition, sCD109 antagonizes TGF-β-induced Smad2/3 phosphorylation, transcription and cell migration. Together, our results suggest that sCD109 can bind TGF-β, inhibit TGF-β binding to its receptors and decrease TGF-β signalling and TGF-β-induced cellular responses.
Collapse
|
7
|
A Systems Biology-Based Investigation into the Pharmacological Mechanisms of Sheng-ma-bie-jia-tang Acting on Systemic Lupus Erythematosus by Multi-Level Data Integration. Sci Rep 2015; 5:16401. [PMID: 26560501 PMCID: PMC4642335 DOI: 10.1038/srep16401] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/12/2015] [Indexed: 11/08/2022] Open
Abstract
Sheng-ma-bie-jia-tang (SMBJT) is a Traditional Chinese Medicine (TCM) formula that is widely used for the treatment of Systemic Lupus Erythematosus (SLE) in China. However, molecular mechanism behind this formula remains unknown. Here, we systematically analyzed targets of the ingredients in SMBJT to evaluate its potential molecular mechanism. First, we collected 1,267 targets from our previously published database, the Traditional Chinese Medicine Integrated Database (TCMID). Next, we conducted gene ontology and pathway enrichment analyses for these targets and determined that they were enriched in metabolism (amino acids, fatty acids, etc.) and signaling pathways (chemokines, Toll-like receptors, adipocytokines, etc.). 96 targets, which are known SLE disease proteins, were identified as essential targets and the rest 1,171 targets were defined as common targets of this formula. The essential targets directly interacted with SLE disease proteins. Besides, some common targets also had essential connections to both key targets and SLE disease proteins in enriched signaling pathway, e.g. toll-like receptor signaling pathway. We also found distinct function of essential and common targets in immune system processes. This multi-level approach to deciphering the underlying mechanism of SMBJT treatment of SLE details a new perspective that will further our understanding of TCM formulas.
Collapse
|
8
|
Gatza CE, Elderbroom JL, Oh SY, Starr MD, Nixon AB, Blobe GC. The balance of cell surface and soluble type III TGF-β receptor regulates BMP signaling in normal and cancerous mammary epithelial cells. Neoplasia 2015; 16:489-500. [PMID: 25077702 PMCID: PMC4198744 DOI: 10.1016/j.neo.2014.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/12/2014] [Accepted: 05/19/2014] [Indexed: 12/20/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily that are over-expressed in breast cancer, with context dependent effects on breast cancer pathogenesis. The type III TGF-β receptor (TβRIII) mediates BMP signaling. While TβRIII expression is lost during breast cancer progression, the role of TβRIII in regulating BMP signaling in normal mammary epithelium and breast cancer cells has not been examined. Restoring TβRIII expression in a 4T1 murine syngeneic model of breast cancer suppressed Smad1/5/8 phosphorylation and inhibited the expression of the BMP transcriptional targets, Id1 and Smad6, in vivo. Similarly, restoring TβRIII expression in human breast cancer cell lines or treatment with sTβRIII inhibited BMP-induced Smad1/5/8 phosphorylation and BMP-stimulated migration and invasion. In normal mammary epithelial cells, shRNA-mediated silencing of TβRIII, TβRIII over-expression, or treatment with sTβRIII inhibited BMP-mediated phosphorylation of Smad1/5/8 and BMP induced migration. Inhibition of TβRIII shedding through treatment with TAPI-2 or expression of a non-shedding TβRIII mutant rescued TβRIII mediated inhibition of BMP induced Smad1/5/8 phosphorylation and BMP induced migration and/or invasion in both in normal mammary epithelial cells and breast cancer cells. Conversely, expression of a TβRIII mutant, which exhibited increased shedding, significantly reduced BMP-mediated Smad1/5/8 phosphorylation, migration, and invasion. These data demonstrate that TβRIII regulates BMP-mediated signaling and biological effects, primarily through the ligand sequestration effects of sTβRIII in normal and cancerous mammary epithelial cells and suggest that the ratio of membrane bound versus sTβRIII plays an important role in mediating these effects.
Collapse
Affiliation(s)
| | - Jennifer L Elderbroom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Sun Young Oh
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Mark D Starr
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC.
| |
Collapse
|
9
|
Elderbroom JL, Huang JJ, Gatza CE, Chen J, How T, Starr M, Nixon AB, Blobe GC. Ectodomain shedding of TβRIII is required for TβRIII-mediated suppression of TGF-β signaling and breast cancer migration and invasion. Mol Biol Cell 2014; 25:2320-32. [PMID: 24966170 PMCID: PMC4142606 DOI: 10.1091/mbc.e13-09-0524] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The type III TGF-β receptor (TβRIII) undergoes ectodomain shedding, with surface TβRIII enhancing and soluble TβRIII inhibiting TGF-β signaling. TβRIII mutants with impaired or enhanced shedding are used to demonstrate that the ratio of soluble to membrane-bound TβRIII regulates TβRIII/TGF-β–mediated signaling and biology in vitro and in vivo. The type III transforming growth factor β (TGF-β) receptor (TβRIII), also known as betaglycan, is the most abundantly expressed TGF-β receptor. TβRIII suppresses breast cancer progression by inhibiting migration, invasion, metastasis, and angiogenesis. TβRIII binds TGF-β ligands, with membrane-bound TβRIII presenting ligand to enhance TGF-β signaling. However, TβRIII can also undergo ectodomain shedding, releasing soluble TβRIII, which binds and sequesters ligand to inhibit downstream signaling. To investigate the relative contributions of soluble and membrane-bound TβRIII on TGF-β signaling and breast cancer biology, we defined TβRIII mutants with impaired (ΔShed-TβRIII) or enhanced ectodomain shedding (SS-TβRIII). Inhibiting ectodomain shedding of TβRIII increased TGF-β responsiveness and abrogated TβRIII's ability to inhibit breast cancer cell migration and invasion. Conversely, expressing SS-TβRIII, which increased soluble TβRIII production, decreased TGF-β signaling and increased TβRIII-mediated inhibition of breast cancer cell migration and invasion. Of importance, SS-TβRIII–mediated increases in soluble TβRIII production also reduced breast cancer metastasis in vivo. Taken together, these studies suggest that the ratio of soluble TβRIII to membrane-bound TβRIII is an important determinant for regulation of TβRIII- and TGF-β–mediated signaling and biology.
Collapse
Affiliation(s)
| | - Jennifer J Huang
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708
| | | | - Jian Chen
- Department of Medicine, Duke University, Durham, NC 27708
| | - Tam How
- Department of Medicine, Duke University, Durham, NC 27708
| | - Mark Starr
- Department of Medicine, Duke University, Durham, NC 27708
| | - Andrew B Nixon
- Department of Medicine, Duke University, Durham, NC 27708
| | - Gerard C Blobe
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708Department of Medicine, Duke University, Durham, NC 27708
| |
Collapse
|
10
|
Biswas T, Gu X, Yang J, Ellies LG, Sun LZ. Attenuation of TGF-β signaling supports tumor progression of a mesenchymal-like mammary tumor cell line in a syngeneic murine model. Cancer Lett 2013; 346:129-38. [PMID: 24368187 DOI: 10.1016/j.canlet.2013.12.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/11/2013] [Accepted: 12/14/2013] [Indexed: 02/07/2023]
Abstract
Previous studies have suggested that TGF-β functions as a tumor promoter in metastatic, mesenchymal-like breast cancer cells and that TGF-β inhibitors can effectively abrogate tumor progression in several of these models. Here we report a novel observation with the use of genetic and pharmacological approaches, and murine mammary cell injection models in both syngeneic and immune compromised mice. We found that TGF-β receptor II (TβRII) knockdown in the MMTV-PyMT derived Py8119, a mesenchymal-like murine mammary tumor cell line, resulted in increased orthotopic tumor growth potential in a syngeneic background and a similar trend in an immune compromised background. Systemic treatment with a small-molecule TGF-β receptor I kinase inhibitor induced a trend towards increased metastatic colonization of distant organs following intracardiac inoculation of Py8119 cells, with little effect on the colonization of luminal-like Py230 cells, also derived from MMTV-PyMT tumors. Taken together, our data suggest that the attenuation of TGF-β signaling in mesenchymal-like mammary tumors does not necessarily inhibit their malignant potential, and anti-TGF-β therapeutic intervention requires greater precision in identifying molecular markers in tumors with an indication of functional TGF-β signaling.
Collapse
Affiliation(s)
- Tanuka Biswas
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiang Gu
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Junhua Yang
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lesley G Ellies
- Department of Pathology, University of California at San Diego, La Jolla, CA, USA
| | - Lu-Zhe Sun
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
11
|
Zwaagstra JC, Sulea T, Baardsnes J, Lenferink AEG, Collins C, Cantin C, Paul-Roc B, Grothe S, Hossain S, Richer LP, L'Abbé D, Tom R, Cass B, Durocher Y, O'Connor-McCourt MD. Engineering and therapeutic application of single-chain bivalent TGF-β family traps. Mol Cancer Ther 2012; 11:1477-87. [PMID: 22562986 DOI: 10.1158/1535-7163.mct-12-0060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Deregulation of TGF-β superfamily signaling is a causative factor in many diseases. Here we describe a protein engineering strategy for the generation of single-chain bivalent receptor traps for TGF-β superfamily ligands. Traps were assembled using the intrinsically disordered regions flanking the structured binding domain of each receptor as "native linkers" between two binding domains. This yields traps that are approximately threefold smaller than antibodies and consists entirely of native receptor sequences. Two TGF-β type II receptor-based, single-chain traps were designed, termed (TβRII)2 and (TβRIIb)2, that have native linker lengths of 35 and 60 amino acids, respectively. Both single-chain traps exhibit a 100 to 1,000 fold higher in vitro ligand binding and neutralization activity compared with the monovalent ectodomain (TβRII-ED), and a similar or slightly better potency than pan-TGF-β-neutralizing antibody 1D11 or an Fc-fused receptor trap (TβRII-Fc). Despite its short in vivo half-life (<1 hour), which is primarily due to kidney clearance, daily injections of the (TβRII)2 trap reduced the growth of 4T1 tumors in BALB/c mice by 50%, an efficacy that is comparable with 1D11 (dosed thrice weekly). In addition, (TβRII)2 treatment of mice with established 4T1 tumors (100 mm(3)) significantly inhibited further tumor growth, whereas the 1D11 antibody did not. Overall, our results indicate that our rationally designed bivalent, single-chain traps have promising therapeutic potential.
Collapse
Affiliation(s)
- John C Zwaagstra
- Biotechnology Research Institute, National Research Council Canada, Montreal, Quebec, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Mishra S, Tang Y, Wang L, deGraffenried L, Yeh IT, Werner S, Troyer D, Copland JA, Sun LZ. Blockade of transforming growth factor-beta (TGFβ) signaling inhibits osteoblastic tumorigenesis by a novel human prostate cancer cell line. Prostate 2011; 71:1441-54. [PMID: 21321980 PMCID: PMC3108007 DOI: 10.1002/pros.21361] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 01/19/2011] [Indexed: 11/08/2022]
Abstract
BACKGROUND The skeleton is the most common site of prostate cancer metastasis, which often results in osteoblastic lesions. The role of transforming growth factor-beta (TGFβ) signaling in prostate cancer-induced osteoblastic metastasis is not clear. We investigated the role of TGFβ signaling in prostate cancer-induced bone metastasis using a novel human prostate cancer cell line, PacMetUT1. METHODS We injected PacMetUT1/Luc-GFP cells in male nude mice by intracardiac and intratibia injections and then investigated the effect of TGFβ signaling abrogation on osteoblastic tumor growth and incidence in vivo by using fluorescence and bioluminescence imaging analysis and quantifying bone and tumor volume by histomorphometry analysis. Osteoclasts were counted using TRAP assay. RESULTS Osteoblastic bone metastasis in skull, rib, and femur was detected after 10-16 weeks of intracardiac injection of the PacMetUT1 cells. Stable knockdown of TGFβ1 with an shRNA resulted in decreased tumor incidence and bone formation when the cells were directly injected into the tibiae. Systemic administration of either a small inhibitor of TGFβ type I receptor kinase or a pan TGFβ binding protein (BG(E) RII) also decreased bone tumor growth and osteoblastic bone formation in vivo after 7 weeks of treatment. CONCLUSIONS Our results for the first time indicate that blockade of TGFβ signaling in the PacMetUT1 model significantly inhibits osteoblastic bone formation and tumor incidence. Thus, TGFβ signaling pathway may be a viable target for the prevention and treatment of prostate cancer-induced bone metastasis.
Collapse
Affiliation(s)
- Sweta Mishra
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Yuping Tang
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Long Wang
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | | | - I-Tien Yeh
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Sherry Werner
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Dean Troyer
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Lu-Zhe Sun
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
- Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas
- Corresponding author: Lu-Zhe Sun, Ph.D., Department of Cellular & Structural Biology, University of Texas Health Science Center, 7703 Floyd Curl Drive, Mail Code 7762, San Antonio, TX 78229-3900, Tel: (210) 567-5746; Fax: (210) 567-3803,
| |
Collapse
|
13
|
TGF-β signalling is mediated by two autonomously functioning TβRI:TβRII pairs. EMBO J 2011; 30:1263-76. [PMID: 21423151 DOI: 10.1038/emboj.2011.54] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 02/04/2011] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor (TGF)-βs are dimeric polypeptides that have vital roles in regulating cell growth and differentiation. They signal by assembling a receptor heterotetramer composed of two TβRI:TβRII heterodimers. To investigate whether the two heterodimers bind and signal autonomously, one of the TGF-β protomers was substituted to block receptor binding. The substituted dimer, TGF-β3 WD, bound the TβRII extracellular domain and recruited the TβRI with affinities indistinguishable from TGF-β3, but with one-half the stoichiometry. TGF-β3 WD was further shown to retain one-quarter to one-half the signalling activity of TGF-β3 in three established assays for TGF-β function. Single-molecule fluorescence imaging with GFP-tagged receptors demonstrated a measurable increase in the proportion of TβRI and TβRII dimers upon treatment with TGF-β3, but not with TGF-β3 WD. These results provide evidence that the two TβRI:TβRII heterodimers bind and signal in an autonomous manner. They further underscore how the TGF-βs diverged from the bone morphogenetic proteins, the ancestral ligands of the TGF-β superfamily that signal through a RI:RII:RII heterotrimer.
Collapse
|
14
|
Gatza CE, Oh SY, Blobe GC. Roles for the type III TGF-beta receptor in human cancer. Cell Signal 2010; 22:1163-74. [PMID: 20153821 PMCID: PMC2875339 DOI: 10.1016/j.cellsig.2010.01.016] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 01/16/2010] [Indexed: 12/20/2022]
Abstract
Transforming growth factor beta (TGF-beta) superfamily ligands have important roles in regulating cellular homeostasis, embryonic development, differentiation, proliferation, immune surveillance, angiogenesis, motility, and apoptosis in a cell type and context specific manner. TGF-beta superfamily signaling pathways also have diverse roles in human cancer, functioning to either suppress or promote cancer progression. The TGF-beta superfamily co-receptor, the type III TGF-beta receptor (TbetaRIII, also known as betaglycan) mediates TGF-beta superfamily ligand dependent as well as ligand independent signaling to both Smad and non-Smad signaling pathways. Loss of TbetaRIII expression during cancer progression and direct effects of TbetaRIII on regulating cell migration, invasion, proliferation, and angiogenesis support a role for TbetaRIII as a suppressor of cancer progression and/or as a metastasis suppressor. Defining the physiological function and mechanism of TbetaRIII action and alterations in TbetaRIII function during cancer progression should enable more effective targeting of TbetaRIII and TbetaRIII mediated functions for the diagnosis and treatment of human cancer.
Collapse
Affiliation(s)
| | - Sun Young Oh
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Gerard C. Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
15
|
Mendoza V, Vilchis-Landeros MM, Mendoza-Hernández G, Huang T, Villarreal MM, Hinck AP, López-Casillas F, Montiel JL. Betaglycan has two independent domains required for high affinity TGF-beta binding: proteolytic cleavage separates the domains and inactivates the neutralizing activity of the soluble receptor. Biochemistry 2010; 48:11755-65. [PMID: 19842711 DOI: 10.1021/bi901528w] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Betaglycan is a coreceptor for members of the transforming growth factor beta (TGF-beta) superfamily. Mutagenesis has identified two ligand binding regions, one at the membrane-distal and the other at the membrane-proximal half of the betaglycan ectodomain. Here we show that partial plasmin digestion of soluble betaglycan produces two proteolysis-resistant fragments of 45 and 55 kDa, consistent with the predicted secondary structure, which indicates an intervening nonstructured linker region separating the highly structured N- and C-terminal domains. Amino terminal sequencing indicates that the 45 and 55 kDa fragments correspond, respectively, to the membrane-distal and -proximal regions. Plasmin treatment of membrane betaglycan results in the production of equivalent proteolysis-resistant fragments. The 45 and 55 kDa fragments, as well as their recombinant soluble counterparts, Sol Delta10 and Sol Delta11, bind TGF-beta, but nonetheless, compared to intact soluble betaglycan, have a severely diminished ability to block TGF-beta activity. Surface plasmon resonance (SPR) analysis indicates that soluble betaglycan has K(d)'s in the low nanomolar range for the three TGF-beta isoforms, while those for Sol Delta10 and Sol Delta11 are 1-2 orders of magnitude higher. SPR analysis further shows that the K(d)'s of Sol Delta11 are not changed in the presence of Sol Delta10, indicating that the high affinity of soluble betaglycan is a consequence of tethering the domains together. Overall, these results suggest that betaglycan ectodomain exhibits a bilobular structure in which each lobule folds independently and binds TGF-beta through distinct nonoverlapping interfaces and that linker modification may be an approach to improve soluble betaglycan's TGF-beta neutralizing activity.
Collapse
Affiliation(s)
- Valentín Mendoza
- Departmento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de Mexico, D.F. 04510, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Bernabeu C, Lopez-Novoa JM, Quintanilla M. The emerging role of TGF-beta superfamily coreceptors in cancer. Biochim Biophys Acta Mol Basis Dis 2009; 1792:954-73. [PMID: 19607914 DOI: 10.1016/j.bbadis.2009.07.003] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2009] [Revised: 07/02/2009] [Accepted: 07/06/2009] [Indexed: 12/23/2022]
Abstract
The transforming growth factor beta (TGF-beta) signaling pathway plays a key role in different physiological processes such as development, cellular proliferation, extracellular matrix synthesis, angiogenesis or immune responses and its deregulation may result in tumor development. The TGF-beta coreceptors endoglin and betaglycan are emerging as modulators of the TGF-beta response with important roles in cancer. Endoglin is highly expressed in the tumor-associated vascular endothelium with prognostic significance in selected neoplasias and with potential to be a prime vascular target for antiangiogenic cancer therapy. On the other hand, the expression of endoglin and betaglycan in tumor cells themselves appears to play an important role in the progression of cancer, influencing cell proliferation, motility, invasiveness and tumorigenicity. In addition, experiments in vitro and in vivo in which endoglin or betaglycan expression is modulated have provided evidence that they act as tumor suppressors. The purpose of this review was to highlight the potential of membrane and soluble forms of the endoglin and betaglycan proteins as molecular targets in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Carmelo Bernabeu
- Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), 28040 Madrid, Spain.
| | | | | |
Collapse
|