RAC1B Regulation of TGFB1 Reveals an Unexpected Role of Autocrine TGFβ1 in the Suppression of Cell Motility

Simple Summary Transforming growth factor (TGF)β secreted by cancer cells and acting in an autostimulatory manner has been implicated in epithelial-mesenchymal transition and cellular invasion. However, the upstream inducers/downstream effectors, or the effects on cell migration, of endogenously produced TGFβ1 remain poorly characterized. Here, we studied whether autocrine TGFβ is regulated by the small GTPase, RAC1B, and how it impacts cell motility. We found that RAC1B induced an increase in the levels of TGFβ1 mRNA and the secreted bioactive protein in pancreatic carcinoma Panc1 and breast cancer MDA-MB-231 cells. Antibody-mediated neutralization of secreted TGFβ or inhibition of its synthesis increased cell migration and the activities of promigratory genes, but downregulated those of antimigratory genes, i.e., SMAD3. Restoration of SMAD3 protein expression in these cells was able to prevent the TGFβ1 inhibition-induced rise in migratory activity, suggesting that a RAC1B-autocrine TGFβ1-SMAD3 pathway can suppress mesenchymal transdifferentiation and cell motility. Abstract Autocrine transforming growth factor (TGF)β has been implicated in epithelial-mesenchymal transition (EMT) and invasion of several cancers including pancreatic ductal adenocarcinoma (PDAC) as well as triple-negative breast cancer (TNBC). However, the precise mechanism and the upstream inducers or downstream effectors of endogenous TGFB1 remain poorly characterized. In both cancer types, the small GTPase RAC1B inhibits cell motility induced by recombinant human TGFβ1 via downregulation of the TGFβ type I receptor, ALK5, but whether RAC1B also impacts autocrine TGFβ signaling has not yet been studied. Intriguingly, RNA interference-mediated knockdown (RNAi-KD) or CRISPR/Cas-mediated knockout of RAC1B in TGFβ1-secreting PDAC-derived Panc1 cells resulted in a dramatic decrease in secreted bioactive TGFβ1 in the culture supernatants and TGFB1 mRNA expression, while the reverse was true for TNBC-derived MDA-MB-231 cells ectopically expressing RAC1B. Surprisingly, the antibody-mediated neutralization of secreted bioactive TGFβ or RNAi-KD of the endogenous TGFB1 gene, was associated with increased rather than decreased migratory activities of Panc1 and MDA-MB-231 cells, upregulation of the promigratory genes SNAI1, SNAI2 and RAC1, and downregulation of the invasion suppressor genes CDH1 (encoding E-cadherin) and SMAD3. Intriguingly, ectopic re-expression of SMAD3 was able to rescue Panc1 and MDA-MB-231 cells from the TGFB1 KD-induced rise in migratory activity. Together, these data suggest that RAC1B favors synthesis and secretion of autocrine TGFβ1 which in a SMAD3-dependent manner blocks EMT-associated gene expression and cell motility.

[1]  J. Korkola,et al.  Cellular senescence in cancer: from mechanisms to detection , 2020, Molecular oncology.

[2]  R. Derynck,et al.  TGFβ biology in cancer progression and immunotherapy , 2020, Nature Reviews Clinical Oncology.

[3]  H. Lehnert,et al.  RAC1B Induces SMAD7 via USP26 to Suppress TGFβ1-Dependent Cell Migration in Mesenchymal-Subtype Carcinoma Cells , 2020, Cancers.

[4]  P. De,et al.  Active RAC1 Promotes Tumorigenic Phenotypes and Therapy Resistance in Solid Tumors , 2020, Cancers.

[5]  P. Jin,et al.  Grainyhead-like 2 as a double-edged sword in development and cancer. , 2020, American journal of translational research.

[6]  H. Lehnert,et al.  Negative Control of Cell Migration by Rac1b in Highly Metastatic Pancreatic Cancer Cells Is Mediated by Sequential Induction of Nonactivated Smad3 and Biglycan , 2019, Cancers.

[7]  H. Lehnert,et al.  RAC1B: A Guardian of the Epithelial Phenotype and Protector Against Epithelial-Mesenchymal Transition , 2019, Cells.

[8]  R. M. Simpson,et al.  The Outcome of TGFβ Antagonism in Metastatic Breast Cancer Models In Vivo Reflects a Complex Balance between Tumor-Suppressive and Proprogression Activities of TGFβ , 2019, Clinical Cancer Research.

[9]  H. Ungefroren Blockade of TGF-β signaling: a potential target for cancer immunotherapy? , 2019, Expert opinion on therapeutic targets.

[10]  M. Hollenberg,et al.  RAC1B Suppresses TGF-β1-Dependent Cell Migration in Pancreatic Carcinoma Cells through Inhibition of the TGF-β Type I Receptor ALK5 , 2019, Cancers.

[11]  H. Lehnert,et al.  RAC1B: A Rho GTPase with Versatile Functions in Malignant Transformation and Tumor Progression , 2019, Cells.

[12]  E. Furth,et al.  EMT Subtype Influences Epithelial Plasticity and Mode of Cell Migration. , 2018, Developmental cell.

[13]  S. Ghadiali,et al.  Myoferlin regulates epithelial cancer cell plasticity and migration through autocrine TGF-β1 signaling , 2018, Oncotarget.

[14]  D. Witte,et al.  Negative regulation of TGF-β1-induced MKK6-p38 and MEK-ERK signalling and epithelial-mesenchymal transition by Rac1b , 2017, Scientific Reports.

[15]  M. Miyazaki,et al.  Grainyhead‐like 2 (GRHL2) regulates epithelial plasticity in pancreatic cancer progression , 2017, Cancer medicine.

[16]  H. Lehnert,et al.  The role of TGF-β and its crosstalk with RAC1/RAC1b signaling in breast and pancreas carcinoma , 2017, Cell Communication and Signaling.

[17]  J. Schulzke,et al.  A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion. , 2015, Journal of the American Society of Nephrology : JASN.

[18]  C. Kuperwasser,et al.  SLUG: Critical regulator of epithelial cell identity in breast development and cancer , 2014, Cell adhesion & migration.

[19]  Bogi Andersen,et al.  Mammary morphogenesis and regeneration require the inhibition of EMT at terminal end buds by Ovol2 transcriptional repressor. , 2014, Developmental cell.

[20]  H. Lehnert,et al.  Rac1b negatively regulates TGF-β1-induced cell motility in pancreatic ductal epithelial cells by suppressing Smad signalling , 2013, Oncotarget.

[21]  C. Hill,et al.  Controlling Long-Term Signaling: Receptor Dynamics Determine Attenuation and Refractory Behavior of the TGF-β Pathway , 2013, Science Signaling.

[22]  H. Ford,et al.  Epithelial-mesenchymal transition and tumor suppression are controlled by a reciprocal feedback loop between ZEB1 and Grainyhead-like-2. , 2013, Cancer research.

[23]  Claude C. Warzecha,et al.  Complex changes in alternative pre-mRNA splicing play a central role in the epithelial-to-mesenchymal transition (EMT). , 2012, Seminars in cancer biology.

[24]  A. Hata,et al.  Targeting the TGFβ signalling pathway in disease , 2012, Nature Reviews Drug Discovery.

[25]  Deokhoon Kim,et al.  Smad3 regulates E-cadherin via miRNA-200 pathway , 2012, Oncogene.

[26]  R. Flavell,et al.  Autocrine/paracrine TGF-β1 inhibits Langerhans cell migration , 2012, Proceedings of the National Academy of Sciences.

[27]  A. Bernstein,et al.  Concentration-dependent effects of transforming growth factor β1 on corneal wound healing , 2011, Molecular vision.

[28]  Sophia Hsin-Jung Li,et al.  Paracrine and Autocrine Signals Induce and Maintain Mesenchymal and Stem Cell States in the Breast , 2011, Cell.

[29]  M. F. Shannon,et al.  An autocrine TGF-β/ZEB/miR-200 signaling network regulates establishment and maintenance of epithelial-mesenchymal transition , 2011, Molecular biology of the cell.

[30]  J. Manley,et al.  Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. , 2010, Genes & development.

[31]  T. Keck,et al.  ZEB1 in Pancreatic Cancer , 2010, Cancers.

[32]  K. Sugano,et al.  IL-13 promotes the proliferation of rat pancreatic stellate cells through the suppression of NF-kappaB/TGF-beta1 pathway. , 2010, Biochemical and biophysical research communications.

[33]  Julia Schüler,et al.  The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs , 2009, Nature Cell Biology.

[34]  J. Massagué,et al.  TGFβ in Cancer , 2008, Cell.

[35]  P. Jordan,et al.  Increased Rac1b Expression Sustains Colorectal Tumor Cell Survival , 2008, Molecular Cancer Research.

[36]  C. Theillet,et al.  Snail and Slug Play Distinct Roles during Breast Carcinoma Progression , 2006, Clinical Cancer Research.

[37]  P. Jordan,et al.  Expression of Rac1b stimulates NF-kappaB-mediated cell survival and G1/S progression. , 2005, Experimental cell research.

[38]  Brian Bierie,et al.  Effect of conditional knockout of the type II TGF-beta receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis. , 2005, Cancer research.

[39]  C. Arteaga,et al.  Autocrine Transforming Growth Factor-β Signaling Mediates Smad-independent Motility in Human Cancer Cells* , 2003, The Journal of Biological Chemistry.

[40]  Xiu-fen Lei,et al.  Autocrine TGFβ supports growth and survival of human breast cancer MDA-MB-231 cells , 2002, Oncogene.

[41]  T. Gress,et al.  TGF‐β–induced invasiveness of pancreatic cancer cells is mediated by matrix metalloproteinase‐2 and the urokinase plasminogen activator system , 2001, International journal of cancer.

[42]  T. Lüscher,et al.  Bimodal effects of angiotensin II on migration of human and rat smooth muscle cells. Direct stimulation and indirect inhibition via transforming growth factor-beta 1. , 1997, Arteriosclerosis, thrombosis, and vascular biology.

[43]  M. Sporn,et al.  Transforming growth factor beta 1 positively regulates its own expression in normal and transformed cells. , 1988, The Journal of biological chemistry.

[44]  D. Rades,et al.  Ionizing radiation induces a motile phenotype in human carcinoma cells in vitro through hyperactivation of the TGF-beta signaling pathway , 2015, Cellular and Molecular Life Sciences.

[45]  Xiu-fen Lei,et al.  Autocrine TGFbeta supports growth and survival of human breast cancer MDA-MB-231 cells. , 2002, Oncogene.