101
|
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: 9.5] [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
|
102
|
Shukla AK, Kim J, Ahn S, Xiao K, Shenoy SK, Liedtke W, Lefkowitz RJ. Arresting a transient receptor potential (TRP) channel: beta-arrestin 1 mediates ubiquitination and functional down-regulation of TRPV4. J Biol Chem 2010; 285:30115-25. [PMID: 20650893 PMCID: PMC2943294 DOI: 10.1074/jbc.m110.141549] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
β-Arrestins, originally discovered to desensitize activated G protein-coupled receptors, (aka seven-transmembrane receptors, 7TMRs) also mediate 7TMR internalization and G protein-independent signaling via these receptors. More recently, several regulatory roles of β-arrestins for atypical 7TMRs and non-7TM receptors have emerged. Here, we uncover an entirely novel regulatory role of β-arrestins in cross-talk between the angiotensin receptor (AT1aR) and a member of the transient receptor potential (TRP) ion channel family, TRPV4. AT1aR and TRPV4 form a constitutive complex in the plasma membrane, and angiotensin stimulation leads to recruitment of β-arrestin 1 to this complex. Surprisingly, angiotensin stimulation results in ubiquitination of TRPV4, a process that requires β-arrestin 1, and subsequently to internalization and functional down-regulation of TRPV4. β-Arrestin 1 interacts with, and acts as an adaptor for AIP4, an E3 ubiquitin ligase responsible for TRPV4 ubiquitination. Thus, our data provide the first evidence of a functional link between β-arrestins and TRPV4 and uncovers an entirely novel mechanism to maintain appropriate intracellular Ca2+ concentration to avoid excessive Ca2+ signaling.
Collapse
Affiliation(s)
- Arun K Shukla
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | |
Collapse
|
103
|
Draheim KM, Chen HB, Tao Q, Moore N, Roche M, Lyle S. ARRDC3 suppresses breast cancer progression by negatively regulating integrin beta4. Oncogene 2010; 29:5032-47. [PMID: 20603614 PMCID: PMC2997682 DOI: 10.1038/onc.2010.250] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Large-scale genetic analyses of human tumor samples have been used to identify novel oncogenes, tumor suppressors and prognostic factors, but the functions and molecular interactions of many individual genes have not been determined. In this study we examined the cellular effects and molecular mechanism of the arrestin family member, ARRDC3, a gene preferentially lost in a subset of breast cancers. Oncomine data revealed that the expression of ARRDC3 decreases with tumor grade, metastases and recurrences. ARRDC3 overexpression represses cancer cell proliferation, migration, invasion, growth in soft agar and in vivo tumorigenicity, whereas downregulation of ARRCD3 has the opposite effects. Mechanistic studies showed that ARRDC3 functions in a novel regulatory pathway that controls the cell surface adhesion molecule, β-4 integrin (ITGβ4), a protein associated with aggressive tumor behavior. Our data indicates ARRDC3 directly binds to a phosphorylated form of ITGβ4 leading to its internalization, ubiquitination and ultimate degradation. The results identify the ARRCD3-ITGβ4 pathway as a new therapeutic target in breast cancer and show the importance of connecting genetic arrays with mechanistic studies in the search for new treatments.
Collapse
Affiliation(s)
- K M Draheim
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | |
Collapse
|
104
|
Gfi-1B controls human erythroid and megakaryocytic differentiation by regulating TGF-β signaling at the bipotent erythro-megakaryocytic progenitor stage. Blood 2010; 115:2784-95. [DOI: 10.1182/blood-2009-09-241752] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Growth factor independence-1B (Gfi-1B) is a transcriptional repressor essential for erythropoiesis and megakaryopoiesis. Targeted gene disruption of GFI1B in mice leads to embryonic lethality resulting from failure to produce definitive erythrocytes, hindering the study of Gfi-1B function in adult hematopoiesis. We here show that, in humans, Gfi-1B controls the development of erythrocytes and megakaryocytes by regulating the proliferation and differentiation of bipotent erythro-megakaryocytic progenitors. We further identify in this cell population the type III transforming growth factor-β receptor gene, TGFBR3, as a direct target of Gfi-1B. Knockdown of Gfi-1B results in altered transforming growth factor-β (TGF-β) signaling as shown by the increase in Smad2 phosphorylation and its inability to associate to the transcription intermediary factor 1-γ (TIF1-γ). Because the Smad2/TIF1-γ complex is known to specifically regulate erythroid differentiation, we propose that, by repressing TGF-β type III receptor (TβRΙII) expression, Gfi-1B favors the Smad2/TIF1-γ interaction downstream of TGF-β signaling, allowing immature progenitors to differentiate toward the erythroid lineage.
Collapse
|
105
|
Looyenga BD, Wiater E, Vale W, Hammer GD. Inhibin-A antagonizes TGFbeta2 signaling by down-regulating cell surface expression of the TGFbeta coreceptor betaglycan. Mol Endocrinol 2010; 24:608-20. [PMID: 20160125 DOI: 10.1210/me.2008-0374] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inhibin is an atypical member of the TGFbeta family of signaling ligands and is classically understood to function via competitive antagonism of activin ligand binding. Inhibin-null (Inha-/-) mice develop both gonadal and adrenocortical tumors, the latter of which depend upon gonadectomy for initiation. We have previously shown that gonadectomy initiates adrenal tumorigenesis in Inha-/- mice by elevating production of LH, which drives aberrant proliferation and differentiation of subcapsular adrenocortical progenitor cells. In this study, we demonstrate that LH signaling specifically up-regulates expression of TGFbeta2 in the subcapsular region of the adrenal cortex, which coincides with regions of aberrant Smad3 activation in Inha-/- adrenal glands. Consistent with a functional interaction between inhibin and TGFbeta2, we further demonstrate that recombinant inhibin-A antagonizes signaling by TGFbeta2 in cultured adrenocortical cells. The mechanism of this antagonism depends upon the mutual affinity of inhibin-A and TGFbeta2 for the signaling coreceptor betaglycan. Although inhibin-A cannot physically displace TGFbeta2 from its binding sites on betaglycan, binding of inhibin-A to the cell surface causes endocytic internalization of betaglycan, thereby reducing the number of available binding sites for TGFbeta2 on the cell surface. The mechanism by which inhibin-A induces betaglycan internalization is clathrin independent, making it distinct from the mechanism by which TGFbeta ligands themselves induce betaglycan internalization. These data indicate that inhibin can specifically antagonize TGFbeta2 signaling in cellular contexts where surface expression of betaglycan is limiting and provide a novel mechanism for activin-independent phenotypes in Inha-/- mice.
Collapse
|
106
|
Lee JD, Hempel N, Lee NY, Blobe GC. The type III TGF-beta receptor suppresses breast cancer progression through GIPC-mediated inhibition of TGF-beta signaling. Carcinogenesis 2009; 31:175-83. [PMID: 19955393 DOI: 10.1093/carcin/bgp271] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Loss of expression of the type III transforming growth factor-beta receptor (TbetaRIII or betaglycan), a transforming growth factor-beta (TGF-beta) superfamily co-receptor, is common in human breast cancers. TbetaRIII suppresses cancer progression in vivo by reducing cancer cell migration and invasion by largely unknown mechanisms. Here, we demonstrate that the cytoplasmic domain of TbetaRIII is essential for TbetaRIII-mediated downregulation of migration and invasion in vitro and TbetaRIII-mediated inhibition of breast cancer progression in vivo. Functionally, the cytoplasmic domain of TbetaRIII is required to attenuate TGF-beta signaling, whereas TbetaRIII-mediated attenuation of TGF-beta signaling is required for TbetaRIII-mediated inhibition of migration and invasion. Mechanistically, both TbetaRIII-mediated inhibition of TGF-beta signaling and TbetaRIII-mediated inhibition of invasion occur through the interaction of the cytoplasmic domain of TbetaRIII with the scaffolding protein GAIP-interacting protein C-terminus (GIPC). Taken together, these studies support a functional role for the TbetaRIII cytoplasmic domain interacting with GIPC to suppress breast cancer progression.
Collapse
Affiliation(s)
- Jason D Lee
- Department of Medicine, Duke University Medical Center, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
107
|
Kovacs JJ, Hara MR, Davenport CL, Kim J, Lefkowitz RJ. Arrestin development: emerging roles for beta-arrestins in developmental signaling pathways. Dev Cell 2009; 17:443-58. [PMID: 19853559 DOI: 10.1016/j.devcel.2009.09.011] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Arrestins were identified as mediators of G protein-coupled receptor (GPCR) desensitization and endocytosis. However, it is now clear that they scaffold many intracellular signaling networks to modulate the strength and duration of signaling by diverse types of receptors--including those relevant to the Hedgehog, Wnt, Notch, and TGFbeta pathways--and downstream kinases such as the MAPK and Akt/PI3K cascades. The involvement of arrestins in many discrete developmental signaling events suggests an indispensable role for these multifaceted molecular scaffolds.
Collapse
Affiliation(s)
- Jeffrey J Kovacs
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
108
|
Schulte G, Schambony A, Bryja V. beta-Arrestins - scaffolds and signalling elements essential for WNT/Frizzled signalling pathways? Br J Pharmacol 2009; 159:1051-8. [PMID: 19888962 DOI: 10.1111/j.1476-5381.2009.00466.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
beta-arrestins were originally identified as negative regulators of G protein-coupled receptor signalling. Recent studies have revealed that beta-arrestins serve as intracellular scaffolds and signalling intermediates. Their diverse functions in intracellular signalling pathways provide mechanisms for achieving signal specificity that might be attacked for pharmacological intervention. Here, we summarize the importance of beta-arrestin function for WNT [wingless (from Drosophila) and the oncogene int-1]/Frizzled (FZD) signalling. WNTs are secreted lipoglycoproteins that act through the seven transmembrane-spanning receptors of the FZD family. It recently became evident that beta-arrestins are required for cellular communication by means of WNTs and FZDs both in cellular systems and in vivo. Although the overall importance of arrestin for WNT/FZD signalling remains obscure, interaction with the central phosphoprotein Dishevelled and the endocytic machinery implicates beta-arrestin as a determinant of WNT signalling specificity, a mediator of WNT/FZD desensitization and a regulator of signalling compartmentation.
Collapse
Affiliation(s)
- Gunnar Schulte
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | | | | |
Collapse
|
109
|
|
110
|
Mythreye K, Blobe GC. Proteoglycan signaling co-receptors: roles in cell adhesion, migration and invasion. Cell Signal 2009; 21:1548-58. [PMID: 19427900 PMCID: PMC2735586 DOI: 10.1016/j.cellsig.2009.05.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 05/04/2009] [Indexed: 12/17/2022]
Abstract
Signaling co-receptors are diverse, multifunctional components of most major signaling pathways, with roles in mediating and regulating signaling in both physiological and pathophysiological circumstances. Many of these signaling co-receptors, including CD44, glypicans, neuropilins, syndecans and TssRIII/betaglycan are also proteoglycans. Like other co-receptors, these proteoglycan signaling co-receptors can bind multiple ligands, promoting the formation of receptor signaling complexes and regulating signaling at the cell surface. The proteoglycan signaling co-receptors can also function as structural molecules to regulate adhesion, cell migration, morphogenesis and differentiation. Through a balance of these signaling and structural roles, proteoglycan signaling co-receptors can have either tumor promoting or tumor suppressing functions. Defining the role and mechanism of action of these proteoglycan signaling co-receptors should enable more effective targeting of these co-receptors and their respective pathways for the treatment of human disease.
Collapse
Affiliation(s)
| | - Gerard C. Blobe
- Department of Medicine, Duke University Medical Center, Durham NC 27708
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham NC 27708
| |
Collapse
|
111
|
|
112
|
Frizzled. Br J Pharmacol 2009. [DOI: 10.1111/j.1476-5381.2009.00501_25.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
113
|
Bianchi EN, Ferrari SL. Beta-arrestin2 regulates parathyroid hormone effects on a p38 MAPK and NFkappaB gene expression network in osteoblasts. Bone 2009; 45:716-25. [PMID: 19560570 PMCID: PMC2741591 DOI: 10.1016/j.bone.2009.06.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 06/09/2009] [Accepted: 06/19/2009] [Indexed: 01/13/2023]
Abstract
Interaction of the cytoplasmic adaptor molecule beta-arrestin2 with the activated parathyroid hormone (PTH)/PTHrP receptor inhibits G protein mediated signaling and triggers MAPKs signaling. In turn, the effects of both intermittent (i.) and continuous (c.) PTH on bone are altered in beta-arrestin2-deficient (Arrb2(-/-)) mice. To elucidate the expression profile of bone genes responsive to PTH and targeted for regulation by beta-arrestin2, we performed microarray analysis using total RNA from primary osteoblastic cells isolated from wild-type (WT) and Arrb2(-/-) mice. By comparing gene expression profiles in cells exposed to i.PTH, c.PTH or vehicle (Veh) for 2 weeks, we found that i.PTH specifically up-regulated 215 sequences (including beta-arrestin2) and down-regulated 200 sequences in WT cells, about two-thirds of them being under the control of beta-arrestin2. In addition, beta-arrestin2 appeared necessary to the down-regulation of a genomic cluster coding for small leucin-rich proteins (SLRPs) including osteoglycin, osteomodulin and asporin. Pathway analyses identified a main gene network centered on p38 MAPK and NFkappaB that requires beta-arrestin2 for up- or down-regulation by i.PTH, and a smaller network of PTH-regulated genes centered on TGFB1, that is normally repressed by beta-arrestin2. In contrast the expression of some known PTH gene targets regulated by the cAMP/PKA pathway was not affected by the presence or absence of beta-arrestin2 in osteoblasts. These results indicate that beta-arrestin2 targets prominently p38 MAPK- and NFkappaB-dependent expression in osteoblasts exposed to i.PTH, and delineates new molecular mechanisms to explain the anabolic and catabolic effects of PTH on bone.
Collapse
Affiliation(s)
- Estelle N Bianchi
- Department of Rehabilitation and Geriatrics, WHO Center for Osteoporosis Prevention, Geneva University Hospitals and University of Geneva, Faculty of Medicine, Switzerland.
| | | |
Collapse
|
114
|
Lee NY, Kirkbride KC, Sheu RD, Blobe GC. The transforming growth factor-beta type III receptor mediates distinct subcellular trafficking and downstream signaling of activin-like kinase (ALK)3 and ALK6 receptors. Mol Biol Cell 2009; 20:4362-70. [PMID: 19726563 DOI: 10.1091/mbc.e09-07-0539] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) signal through the BMP type I and type II receptors to regulate cellular processes, including embryonic development. The type I BMP receptors activin-like kinase (ALK)3 and ALK6 share a high degree of homology, yet possess distinct signaling roles. Here, we report that although the transforming growth factor (TGF)-beta type III receptor (TbetaRIII) enhanced both ALK3 and ALK6 signaling, TbetaRIII more potently enhanced ALK6-mediated stimulation of the BMP-responsive promoters XVent2 and 3GC2, and up-regulation of the early response gene Smad6. In contrast, TbetaRIII specifically enhanced ALK3-mediated up-regulation of the early response gene ID-1. TbetaRIII associated with ALK3 primarily through their extracellular domains, whereas its interaction with ALK6 required both the extracellular and cytoplasmic domains. TbetaRIII, along with its interacting scaffolding protein beta-arrestin2, induced the internalization of ALK6. In contrast, TbetaRIII colocalized with and resulted in the cell surface retention of ALK3, independently of beta-arrestin2. Although complex formation between TbetaRIII, ALK6, and beta-arrestin2 and TbetaRIII/ALK6 internalization resulted in maximal BMP signaling, the TbetaRIII mutant unable to interact with beta-arrestin2, TbetaRIII-T841A, was unable to do so. These studies support a novel role for TbetaRIII in mediating differential ALK3 and ALK6 subcellular trafficking resulting in distinct signaling downstream of ALK3 and ALK6.
Collapse
Affiliation(s)
- Nam Y Lee
- Department of Medicine, Duke University Medical Center, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
115
|
Escalona RM, Stenvers KL, Farnworth PG, Findlay JK, Ooi GT. Reducing betaglycan expression by RNA interference (RNAi) attenuates inhibin bioactivity in LbetaT2 gonadotropes. Mol Cell Endocrinol 2009; 307:149-56. [PMID: 19524135 DOI: 10.1016/j.mce.2009.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 10/20/2022]
Abstract
Betaglycan is an inhibin-binding protein co-receptor, the forced expression of which confers inhibin responsiveness on cells previously non-responsive to inhibin. The present study determines whether removal of betaglycan expression in otherwise inhibin-responsive cells will render the cells insensitive to inhibin. Small interfering RNAs (siRNAs) designed to the betaglycan gene were transfected into LbetaT2 gonadotrope cells to 'knock-down' betaglycan expression. To control for non-specific effects, siRNAs corresponding to an unrelated sequence (BF-1) were used. Two activin-responsive promoter constructs were used to assess inhibin bioactivity; an ovine FSHbeta promoter (oFSHbeta-lux), and a construct containing three copies of the activin-responsive sequence from the GnRHR promoter (3XpGRAS-PRL-lux). Activin stimulated the activity of both promoters 5-8-fold. Inhibin suppressed these activin-stimulated promoter activities by 52+/-11% and 51+/-7%, respectively. Similar inhibin suppression was also seen for cells co-transfected with the control BF-1 siRNAs. In contrast, inhibin's ability to suppress activin-stimulated activity was significantly reduced (33+/-3%, p<0.005 and 24+/-4%, p<0.045, respectively) in cells co-transfected with betaglycan siRNAs. These results demonstrated that endocrine effects of inhibin as a negative feedback controller of FSH production in gonadotropes are dependent on betaglycan expression.
Collapse
Affiliation(s)
- Ruth M Escalona
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
116
|
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: 12.2] [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
|
117
|
Li TT, Alemayehu M, Aziziyeh AI, Pape C, Pampillo M, Postovit LM, Mills GB, Babwah AV, Bhattacharya M. Beta-arrestin/Ral signaling regulates lysophosphatidic acid-mediated migration and invasion of human breast tumor cells. Mol Cancer Res 2009; 7:1064-77. [PMID: 19609003 DOI: 10.1158/1541-7786.mcr-08-0578] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The lipid mediator lysophosphatidic acid (LPA) plays a role in cancer progression and signals via specific G protein-coupled receptors, LPA(1-3). LPA has been shown to enhance the metastasis of breast carcinoma cells to bone. However, the mechanisms by which LPA receptors regulate breast cancer cell migration and invasion remain unclear. Breast cancer cell proliferation has been shown to be stimulated by Ral GTPases, a member of the Ras superfamily. Ral activity can be regulated by the multifunctional protein beta-arrestin. We now show that HS578T and MDA-MB-231 breast cancer cells and MDA-MB-435 melanoma cells have higher expression of beta-arrestin 1 mRNA compared with the nontumorigenic mammary MCF-10A cells. Moreover, we found that the mRNA levels of LPA1, LPA2, beta-arrestin 2, and Ral GTPases are elevated in the advanced stages of breast cancer. LPA stimulates the migration and invasion of MDA-MB-231 cells, but not of MCF-10A cells, and this is mediated by pertussis toxin-sensitive G proteins and LPA1. However, ectopic expression of LPA1 in MCF-10A cells caused these cells to acquire an invasive phenotype. Gene knockdown of either beta-arrestin or Ral proteins significantly impaired LPA-stimulated migration and invasion. Thus, our data show a novel role for beta-arrestin/Ral signaling in mediating LPA-induced breast cancer cell migration and invasion, two important processes in metastasis.
Collapse
Affiliation(s)
- Timothy T Li
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
The type III TGF-beta receptor regulates epithelial and cancer cell migration through beta-arrestin2-mediated activation of Cdc42. Proc Natl Acad Sci U S A 2009; 106:8221-6. [PMID: 19416857 DOI: 10.1073/pnas.0812879106] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Loss of expression of the TGF-beta superfamily coreceptor, the type III TGF-beta receptor (TbetaRIII or betaglycan), occurs in a broad spectrum of human cancers including breast, lung, ovarian, pancreatic, prostate, and renal cell cancer. TbetaRIII suppresses cancer progression in vivo, at least in part, by reducing cancer cell motility. However, the mechanism by which TbetaRIII regulates migration is unknown. Here, we demonstrate an unexpected TGF-beta signaling independent role for TbetaRIII in activating Cdc42, altering the actin cytoskeleton and reducing directional persistence to inhibit random migration of both cancer and normal epithelial cells. Functionally, TbetaRIII through its interaction with the scaffolding protein beta-arrestin2, activates Cdc42 and inhibits migration. These studies identify a TGF-beta independent homeostatic function for TbetaRIII in regulating cell migration.
Collapse
|
119
|
Abstract
PTH-stimulated intracellular signaling is regulated by the cytoplasmic adaptor molecule beta-arrestin. We reported that the response of cancellous bone to intermittent PTH is reduced in beta-arrestin2(-/-) mice and suggested that beta-arrestins could influence the bone mineral balance by controlling RANKL and osteoprotegerin (OPG) gene expression. Here, we study the role of beta-arrestin2 on the in vitro development and activity of bone marrow (BM) osteoclasts (OCs) and Ephrins ligand (Efn), and receptor (Eph) mRNA levels in bone in response to PTH and the changes of bone microarchitecture in wildtype (WT) and beta-arrestin2(-/-) mice in models of bone remodeling: a low calcium diet (LoCa) and ovariectomy (OVX). The number of PTH-stimulated OCs was higher in BM cultures from beta-arrestin2(-/-) compared with WT, because of a higher RANKL/OPG mRNA and protein ratio, without directly influencing osteoclast activity. In vivo, high PTH levels induced by LoCa led to greater changes in TRACP5b levels in beta-arrestin2(-/-) compared with WT. LoCa caused a loss of BMD and bone microarchitecture, which was most prominent in beta-arrestin2(-/-). PTH downregulated Efn and Eph genes in beta-arrestin2(-/-), but not WT. After OVX, vertebral trabecular bone volume fraction and trabecular number were lower in beta-arrestin2(-/-) compared with WT. Histomorphometry showed that OC number was higher in OVX-beta-arrestin2(-/-) compared with WT. These results indicate that beta-arrestin2 inhibits osteoclastogenesis in vitro, which resulted in decreased bone resorption in vivo by regulating RANKL/OPG production and ephrins mRNAs. As such, beta-arrestins should be considered an important mechanism for the control of bone remodeling in response to PTH and estrogen deprivation.
Collapse
|
120
|
You HJ, How T, Blobe GC. The type III transforming growth factor-beta receptor negatively regulates nuclear factor kappa B signaling through its interaction with beta-arrestin2. Carcinogenesis 2009; 30:1281-7. [PMID: 19325136 DOI: 10.1093/carcin/bgp071] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) increases or decreases nuclear factor kappa B (NFkappaB) signaling in a context-dependent manner through mechanisms that remain to be defined. The type III transforming growth factor-beta receptor (TbetaRIII) is a TGF-beta superfamily co-receptor with emerging roles in both mediating and regulating TGF-beta superfamily signaling. We have previously reported a novel interaction of TbetaRIII with the scaffolding protein, beta-arrestin2, which results in TbetaRIII internalization and downregulation of TGF-beta signaling. beta-arrestin2 also scaffolds interacting receptors with the mitogen-activated protein kinase and NFkappaB-signaling pathways. Here, we demonstrate that TbetaRIII, through its interaction with beta-arrestin2, negatively regulates NFkappaB signaling in MCF10A breast epithelial and MDA-MB-231 breast cancer cells. Increasing TbetaRIII expression reduced NFkappaB-mediated transcriptional activation and IkappaBalpha degradation, whereas a TbetaRIII mutant unable to interact with beta-arrestin2, TbetaRIII-T841A, had no effect. In a reciprocal manner, short hairpin RNA-mediated silencing of either TbetaRIII expression or beta-arrestin2 expression increased NFkappaB-mediated transcriptional activation and IkappaBalpha degradation. Functionally, TbetaRIII-mediated repression of NFkappaB signaling is important for TbetaRIII-mediated inhibition of breast cancer cell migration. These studies define a mechanism through which TbetaRIII regulates NFkappaB signaling and expand the roles of this TGF-beta superfamily co-receptor in regulating epithelial cell homeostasis.
Collapse
Affiliation(s)
- Hye Jin You
- Division of Basic and Applied Sciences, Carcinogenesis Branch, National Cancer Center, Jungbalsan-ro 111, Ilsandong-gu, Goyang, Gyeonggi 410-769, South Korea
| | | | | |
Collapse
|
121
|
Kim J, Ahn S, Rajagopal K, Lefkowitz RJ. Independent beta-arrestin2 and Gq/protein kinase Czeta pathways for ERK stimulated by angiotensin type 1A receptors in vascular smooth muscle cells converge on transactivation of the epidermal growth factor receptor. J Biol Chem 2009; 284:11953-62. [PMID: 19254952 PMCID: PMC2673264 DOI: 10.1074/jbc.m808176200] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recent studies in receptor-transfected cell lines have demonstrated that
extracellular signal-regulated kinase (ERK) activation by angiotensin type 1A
receptor and other G protein-coupled receptors can be mediated by both G
protein-dependent and β-arrestin-dependent mechanisms. However, few
studies have explored these mechanisms in primary cultured cells expressing
endogenous levels of receptors. Accordingly, here we utilized the
β-arrestin biased agonist for the angiotensin type 1A receptor,
SII-angiotensin (SII), and RNA interference techniques to investigate
angiotensin II (ANG)-activated β-arrestin-mediated mitogenic signaling
pathways in rat vascular smooth muscle cells. Both ANG and SII induced DNA
synthesis via the ERK activation cascade. Even though SII cannot induce
calcium influx (G protein activation) after receptor stimulation, it does
cause ERK activation, although less robustly than ANG. Activation by both
ligands is diminished by depletion of β-arrestin2 by small interfering
RNA, although the effect is more complete with SII. ERK activation at early
time points but not later time points is strongly inhibited by those protein
kinase C inhibitors that can block protein kinase Cζ. Moreover, ANG- and
SII-mediated ERK activation require transactivation of the epidermal growth
factor receptor via metalloprotease 2/9 and Src kinase. β-Arrestin2
facilitates ANG and SII stimulation of Src-mediated phosphorylation of Tyr-845
on the EGFR, a known site for Src phosphorylation. These studies delineate a
convergent mechanism by which G protein-dependent and
β-arrestin-dependent pathways can independently mediate ERK-dependent
transactivation of the EGFR in vascular smooth muscle cells thus controlling
cellular proliferative responses.
Collapse
Affiliation(s)
- Jihee Kim
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
122
|
Xiong DH, Liu XG, Guo YF, Tan LJ, Wang L, Sha BY, Tang ZH, Pan F, Yang TL, Chen XD, Lei SF, Yerges LM, Zhu XZ, Wheeler VW, Patrick AL, Bunker CH, Guo Y, Yan H, Pei YF, Zhang YP, Levy S, Papasian CJ, Xiao P, Lundberg YW, Recker RR, Liu YZ, Liu YJ, Zmuda JM, Deng HW. Genome-wide association and follow-up replication studies identified ADAMTS18 and TGFBR3 as bone mass candidate genes in different ethnic groups. Am J Hum Genet 2009; 84:388-98. [PMID: 19249006 DOI: 10.1016/j.ajhg.2009.01.025] [Citation(s) in RCA: 155] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 01/17/2009] [Accepted: 01/30/2009] [Indexed: 01/17/2023] Open
Abstract
To identify and validate genes associated with bone mineral density (BMD), which is a prominent osteoporosis risk factor, we tested 379,319 SNPs in 1000 unrelated white U.S. subjects for associations with BMD. For replication, we genotyped the most significant SNPs in 593 white U.S. families (1972 subjects), a Chinese hip fracture (HF) sample (350 cases, 350 controls), a Chinese BMD sample (2955 subjects), and a Tobago cohort of African ancestry (908 males). Publicly available Framingham genome-wide association study (GWAS) data (2953 whites) were also used for in silico replication. The GWAS detected two BMD candidate genes, ADAMTS18 (ADAM metallopeptidase with thrombospondin type 1 motif, 18) and TGFBR3 (transforming growth factor, beta receptor III). Replication studies verified the significant findings by GWAS. We also detected significant associations with hip fracture for ADAMTS18 SNPs in the Chinese HF sample. Meta-analyses supported the significant associations of ADAMTS18 and TGFBR3 with BMD (p values: 2.56 x 10(-5) to 2.13 x 10(-8); total sample size: n = 5925 to 9828). Electrophoretic mobility shift assay suggested that the minor allele of one significant ADAMTS18 SNP might promote binding of the TEL2 factor, which may repress ADAMTS18 expression. The data from NCBI GEO expression profiles also showed that ADAMTS18 and TGFBR3 genes were differentially expressed in subjects with normal skeletal fracture versus subjects with nonunion skeletal fracture. Overall, the evidence supports that ADAMTS18 and TGFBR3 might underlie BMD determination in the major human ethnic groups.
Collapse
|
123
|
Constam DB. Riding Shotgun: A Dual Role for the Epidermal Growth Factor-Cripto/FRL-1/Cryptic Protein Cripto in Nodal Trafficking. Traffic 2009; 10:783-91. [DOI: 10.1111/j.1600-0854.2009.00874.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
124
|
Abstract
Transforming growth factor-beta (TGF-beta) signaling is tightly regulated to ensure its proper physiological functions in different cells and tissues. Like other cell surface receptors, TGF-beta receptors are internalized into the cell, and this process plays an important regulatory role in TGF-beta signaling. It is well documented that TGF-beta receptors are endocytosed via clathrin-coated vesicles as TGF-beta endocytosis can be blocked by potassium depletion and the GTPase-deficient dynamin K44A mutant. TGF-beta receptors may also enter cells via cholesterol-rich membrane microdomain lipid rafts/caveolae and are found in caveolin-1-positive vesicles. Although receptor endocytosis is not essential for TGF-beta signaling, clathrin-mediated endocytosis has been shown to promote TGF-beta-induced Smad activation and transcriptional responses. Lipid rafts/caveolae are widely regarded as signaling centers for G protein-coupled receptors and tyrosine kinase receptors, but they are indicated to facilitate the degradation of TGF-beta receptors and therefore turnoff of TGF-beta signaling. This review summarizes current understanding of TGF-beta receptor endocytosis, the possible mechanisms underlying this process, and the role of endocytosis in modulation of TGF-beta signaling.
Collapse
Affiliation(s)
- Ye-Guang Chen
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
125
|
Finger EC, Lee NY, You HJ, Blobe GC. Endocytosis of the type III transforming growth factor-beta (TGF-beta) receptor through the clathrin-independent/lipid raft pathway regulates TGF-beta signaling and receptor down-regulation. J Biol Chem 2008; 283:34808-18. [PMID: 18845534 PMCID: PMC2596377 DOI: 10.1074/jbc.m804741200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 09/26/2008] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) signals through three highly conserved cell surface receptors, the type III TGF-beta receptor (T beta RIII), the type II TGF-beta receptor (T beta RII), and the type I TGF-beta receptor (T beta RI) to regulate diverse cellular processes including cell proliferation, differentiation, migration, and apoptosis. Although T beta RI and T beta RII undergo ligand-independent endocytosis by both clathrin-mediated endocytosis, resulting in enhanced signaling, and clathrin-independent endocytosis, resulting in receptor degradation, the mechanism and function of T beta RIII endocytosis is poorly understood. T beta RIII is a heparan sulfate proteoglycan with a short cytoplasmic tail that functions as a TGF-beta superfamily co-receptor, contributing to TGF-beta signaling through mechanisms yet to be fully defined. We have reported previously that T beta RIII endocytosis, mediated by a novel interaction with beta arrestin-2, results in decreased TGF-beta signaling. Here we demonstrate that T beta RIII undergoes endocytosis in a ligand and glycosaminoglycan modification-independent and cytoplasmic domain-dependent manner, with the interaction of Thr-841 in the cytoplasmic domain of T beta RIII with beta-arrestin2 enhancing T beta RIII endocytosis. T beta RIII undergoes both clathrin-mediated and clathrin-independent endocytosis. Importantly, inhibition of the clathrin-independent, lipid raft pathway, but not of the clathrin-dependent pathway, results in decreased TGF-beta1 induced Smad2 and p38 phosphorylation, supporting a specific role for clathrin-independent endocytosis of T beta RIII in regulating both Smad-dependent and Smad-independent TGF-beta signaling.
Collapse
Affiliation(s)
- Elizabeth C. Finger
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Nam Y. Lee
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Hye-jin You
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Gerard C. Blobe
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| |
Collapse
|
126
|
Patel PA, Tilley DG, Rockman HA. Physiologic and cardiac roles of beta-arrestins. J Mol Cell Cardiol 2008; 46:300-8. [PMID: 19103204 DOI: 10.1016/j.yjmcc.2008.11.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 11/10/2008] [Accepted: 11/12/2008] [Indexed: 01/08/2023]
Abstract
Beta-arrestin1 and beta-arrestin2 were initially identified by sequence homology to visual arrestins and by their ability to bind to and inactivate signaling of the beta-2-adrenergic receptor in a process known as desensitization. While the role of beta-arrestins in desensitization has been known for some time, more recent evidence has revealed that beta-arrestins are multifunctional scaffolding proteins that are involved in numerous aspects of G protein-coupled receptor (GPCR) signaling. Interestingly, exciting new data shows that beta-arrestins can mediate signaling in their own right independent of classical second messenger mediated signaling, and that this beta-arrestin-mediated signaling may be cardioprotective. Identifying novel ligands for GPCRs that can block G protein-mediated signaling while simultaneously promoting beta-arrestin-mediated signaling could provide powerful new therapies for cardiac disease.
Collapse
Affiliation(s)
- Priyesh A Patel
- Duke University School of Medicine, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
127
|
Margulis V, Maity T, Zhang XY, Cooper SJ, Copland JA, Wood CG. Type III transforming growth factor-beta (TGF-beta) receptor mediates apoptosis in renal cell carcinoma independent of the canonical TGF-beta signaling pathway. Clin Cancer Res 2008; 14:5722-30. [PMID: 18794080 DOI: 10.1158/1078-0432.ccr-08-0546] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE Alterations in transforming growth factor-beta (TGF-beta) signaling occur early during malignant transformation of renal epithelial cells and are associated with loss of type III TGF-beta receptor (TbetaRIII) expression. We evaluated the role of TbetaRIII in mediation of apoptosis using in vitro cell culture and in vivo animal models of clear cell renal cell carcinoma. EXPERIMENTAL DESIGN TbetaR3 expression was manipulated with adenoviral gene vector delivery system in vitro and in vivo. Induction of apoptosis and signaling through the Smad and mitogen-activated protein kinase (MAPK) pathways were examined at various time points after infection. To study viral oncolysis in vivo, human renal cell carcinoma cells were implanted s.c. in the flanks of nude mice and treated with intratumoral injections of adenovirus. RESULTS Restoring TbetaRIII expression in clear cell renal cell carcinoma resulted in a marked induction of apoptosis using in vitro cell culture and in vivo animal models. The expression of the cytoplasmic domain, but not the extracellular domain, of TbetaRIII mimicked the induction of apoptosis by full-length TbetaRIII in cell culture and the growth inhibition of tumors in athymic nude mice. TbetaRIII-associated apoptosis was not dependent on signaling through the canonical TGF-beta/Smad pathway but was mediated through p38 MAPK. CONCLUSION These findings indicate a novel mechanistic antitumor function for TbetaRIII and further support its role as an important tumor suppressor in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Vitaly Margulis
- Department of Urology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
128
|
An essential function for beta-arrestin 2 in the inhibitory signaling of natural killer cells. Nat Immunol 2008; 9:898-907. [PMID: 18604210 DOI: 10.1038/ni.1635] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 06/17/2008] [Indexed: 11/09/2022]
Abstract
The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.
Collapse
|
129
|
ALK1 opposes ALK5/Smad3 signaling and expression of extracellular matrix components in human chondrocytes. J Bone Miner Res 2008; 23:896-906. [PMID: 18333754 DOI: 10.1359/jbmr.080209] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION TGF-beta is a multifunctional regulator of chondrocyte proliferation, differentiation, and extracellular matrix production. Dysregulation of TGF-beta action has been implicated in cartilage diseases such as osteoarthritis. TGF-beta signaling is transduced through a pair of transmembrane serine/threonine kinases, known as the type I (ALK5) and type II receptors. However, recent studies on endothelial cells have identified ALK1 as a second type I TGF-beta receptor and have shown that ALK1 and ALK5 have opposing functions in these cells. Here we examined ALK1 expression and its regulation of TGF-beta signaling and responses in human chondrocytes. MATERIALS AND METHODS ALK1 expression in human chondrocytes was examined by RT-PCR and Western blot. The ability of ALK1 to form complexes with other TGF-beta receptors was determined by affinity labeling/immunoprecipitation and by immunoprecipitation followed by Western blot. The effect of ALK1 on TGF-beta1-induced signaling and responses was determined by varying ALK1 expression levels and measuring transcriptional activity using promoter/luciferase assays, Smad1/5 and Smad3 phosphorylation, and expression of type II collagen, PAI-1, and fibronectin. RESULTS Our results indicate that ALK1 is expressed in human chondrocytes and that it is a component of the TGF-beta receptor system, associating with ALK5, type II TGF-beta receptor, endoglin, and betaglycan. Furthermore, we show that both ALK1 and ALK5 are needed for TGF-beta-induced phosphorylation of intracellular mediators Smad1/5, whereas only ALK5 is essential for TGF-beta1-induced phosphorylation of Smad3. In addition, our results show that ALK1 inhibits, whereas ALK5 potentiates, TGF-beta-induced Smad3-driven transcriptional activity and the expression of PAI-1, fibronectin, and type II collagen in chondrocytes. CONCLUSIONS Our results suggest that ALK1 and ALK5 display opposing functions in human chondrocytes, implicating an essential role for ALK1 in the regulation of TGF-beta signaling and function in these cells.
Collapse
|
130
|
Bernabeu C, Conley BA, Vary CPH. Novel biochemical pathways of endoglin in vascular cell physiology. J Cell Biochem 2008; 102:1375-88. [PMID: 17975795 DOI: 10.1002/jcb.21594] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The broad role of the transforming growth factor beta (TGFbeta) signaling pathway in vascular development, homeostasis, and repair is well appreciated. Endoglin is emerging as a novel, complex, and poorly understood regulatory component of the TGFbeta receptor complex, whose importance is underscored by its recognition as the site of mutations causing hereditary hemorrhagic telangiectasia (HHT) [McAllister et al., 1994]. Extensive analyses of endoglin function in normal developmental mouse models [Bourdeau et al., 1999; Li et al., 1999; Arthur et al., 2000] and in HHT animal models [Bourdeau et al., 2000; Torsney et al., 2003] exemplify the importance of understanding endoglin's biochemical functions. However, novel mechanisms underlying the regulation of these pathways continue to emerge. These mechanisms include modification of TGFbeta receptor signaling at the ligand and receptor activation level, direct effects of endoglin on cell adhesion and migration, and emerging roles for endoglin in the determination of stem cell fate and tissue patterning. The purpose of this review is to highlight the cellular and molecular studies that underscore the central role of endoglin in vascular development and disease.
Collapse
Affiliation(s)
- Carmelo Bernabeu
- Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), 28040 Madrid, Spain
| | | | | |
Collapse
|
131
|
Kirkbride KC, Townsend TA, Bruinsma MW, Barnett JV, Blobe GC. Bone Morphogenetic Proteins Signal through the Transforming Growth Factor-β Type III Receptor. J Biol Chem 2008; 283:7628-37. [DOI: 10.1074/jbc.m704883200] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
132
|
Hempel N, How T, Cooper SJ, Green TR, Dong M, Copland JA, Wood CG, Blobe GC. Expression of the type III TGF-beta receptor is negatively regulated by TGF-beta. Carcinogenesis 2008; 29:905-12. [PMID: 18299279 DOI: 10.1093/carcin/bgn049] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The type III transforming growth factor-beta receptor (TbetaRIII or betaglycan) is a ubiquitously expressed transforming growth factor-beta (TGF-beta) superfamily coreceptor with essential roles in embryonic development. Recent studies have defined a role for TbetaRIII in the pathogenesis of human cancers, with frequent loss of TbetaRIII expression at the message and protein level. Mechanisms for the loss of TbetaRIII expression remain to be fully defined. Advanced human cancers often have elevated circulating levels of TGF-beta1. Here, we define a specific role for TGF-beta1 in negatively regulating TbetaRIII at the message level in breast and ovarian cancer models. TGF-beta1 decreased TbetaRIII message and protein levels in ovarian (Ovca420) and breast cancer (MDA-MB-231) cell lines in both a dose- and time-dependent manner. TGF-beta1-mediated TbetaRIII repression is mediated by the type I TGF-beta receptor/Smad2/3 pathway as the activin receptor-like kinase 5 (ALK5) inhibitor, SB431542, abrogated this effect, while the expression of constitutively active ALK5 was sufficient to repress TbetaRIII expression. Mechanistically, TGF-beta1 does not affect TbetaRIII messenger RNA (mRNA) stability, but instead directly regulates the TbetaRIII promoter. We define alternative promoters for the TGFBR3 gene, a distal and proximal promoter. Although both promoters are active, only the proximal promoter was responsive and negatively regulated by TGF-beta1 and constitutively active ALK5. Taken together, these studies define TGF-beta1-mediated downregulation of TbetaRIII mRNA expression through effects on the ALK5/Smad2/3 pathway on the TGFBR3 gene proximal promoter as a potential mechanism for decreased TbetaRIII expression in human cancers.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Gordon KJ, Blobe GC. Role of transforming growth factor-beta superfamily signaling pathways in human disease. Biochim Biophys Acta Mol Basis Dis 2008; 1782:197-228. [PMID: 18313409 DOI: 10.1016/j.bbadis.2008.01.006] [Citation(s) in RCA: 504] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 01/22/2008] [Accepted: 01/23/2008] [Indexed: 12/14/2022]
Abstract
Transforming growth factor beta (TGF-beta) superfamily signaling pathways are ubiquitous and essential regulators of cellular processes including proliferation, differentiation, migration, and survival, as well as physiological processes, including embryonic development, angiogenesis, and wound healing. Alterations in these pathways, including either germ-line or somatic mutations or alterations in the expression of members of these signaling pathways often result in human disease. Appropriate regulation of these pathways is required at all levels, particularly at the ligand level, with either a deficiency or an excess of specific TGF-beta superfamily ligands resulting in human disease. TGF-beta superfamily ligands and members of these TGF-beta superfamily signaling pathways also have emerging roles as diagnostic, prognostic or predictive markers for human disease. Ongoing studies will enable targeting of TGF-beta superfamily signaling pathways for the chemoprevention and treatment of human disease.
Collapse
Affiliation(s)
- Kelly J Gordon
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | | |
Collapse
|
134
|
Hill JA, Feuerer M, Tash K, Haxhinasto S, Perez J, Melamed R, Mathis D, Benoist C. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity 2008; 27:786-800. [PMID: 18024188 DOI: 10.1016/j.immuni.2007.09.010] [Citation(s) in RCA: 502] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 08/07/2007] [Accepted: 09/26/2007] [Indexed: 12/13/2022]
Abstract
The CD4(+)CD25(+) lineage of regulatory T (Treg) cells plays a key role in controlling immune and autoimmune responses and is characterized by a unique transcriptional signature. The transcription factor Foxp3 had been thought to determine the Treg cell lineage, a hypothesis challenged by recent observations. We have performed a cross-sectional analysis of the Treg cell signature in Treg-like cells generated under a number of conditions, with or without Foxp3, to delineate the elements that can be ascribed to T cell activation, interleukin-2, transforming growth factor-beta (TGF-beta) signaling, or Foxp3 itself. These influences synergized to determine many of the signature's components. Much of the Treg cell signature was not ascribable to Foxp3 because it contained gene clusters that are coregulated with, but not transactivated by, Foxp3. Thus, a higher level of regulation upstream of Foxp3 determines the lineage, distinct from elements downstream of Foxp3 that are essential for its regulatory properties.
Collapse
Affiliation(s)
- Jonathan A Hill
- Section on Immunology and Immunogenetics, Joslin Diabetes Center; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Finger EC, Turley RS, Dong M, How T, Fields TA, Blobe GC. TbetaRIII suppresses non-small cell lung cancer invasiveness and tumorigenicity. Carcinogenesis 2008; 29:528-35. [PMID: 18174241 DOI: 10.1093/carcin/bgm289] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The transforming growth factor-beta (TGF-beta) superfamily has essential roles in lung development, regulating cell proliferation, branching morphogenesis, differentiation and apoptosis. Although most lung cancers become resistant to the tumor suppressor effects of TGF-beta, and loss or mutation of one of the components of the TGF-beta signaling pathway, including TbetaRII, Smad2 and Smad4 have been reported, mutations are not common in non-small cell lung cancer (NSCLC). Here we demonstrate that the TGF-beta superfamily co-receptor, the type III TGF-beta receptor (TbetaRIII or betaglycan) is lost in the majority of NSCLC specimens at the mRNA and protein levels, with loss correlating with increased tumor grade and disease progression. Loss of heterozygosity at the TGFBR3 genomic locus occurs in 38.5% of NSCLC specimens and correlates with decreased TbetaRIII expression, suggesting loss of heterozygosity as one mechanism for TbetaRIII loss. In the H460 cell model of NSCLC, restoring TbetaRIII expression decreased colony formation in soft agar. In the A549 cell model of NSCLC, restoring TbetaRIII expression significantly decreased cellular migration and invasion through Matrigel, in the presence and absence of TGF-beta1, and decreased tumorigenicity in vivo. In a reciprocal manner, shRNA-mediated silencing of endogenous TbetaRIII expression enhanced invasion through Matrigel. Mechanistically, TbetaRIII functions, at least in part, through undergoing ectodomain shedding, generating soluble TbetaRIII, which is able to inhibit cellular invasiveness. Taken together, these results support TbetaRIII as a novel tumor suppressor gene that is commonly lost in NSCLC resulting in a functional increase in cellular migration, invasion and anchorage-independent growth of lung cancer cells.
Collapse
Affiliation(s)
- Elizabeth C Finger
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, 221B MSRB Research Drive, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
136
|
Gordon KJ, Dong M, Chislock EM, Fields TA, Blobe GC. Loss of type III transforming growth factor β receptor expression increases motility and invasiveness associated with epithelial to mesenchymal transition during pancreatic cancer progression. Carcinogenesis 2007; 29:252-62. [DOI: 10.1093/carcin/bgm249] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
137
|
Criswell TL, Arteaga CL. Modulation of NFkappaB activity and E-cadherin by the type III transforming growth factor beta receptor regulates cell growth and motility. J Biol Chem 2007; 282:32491-500. [PMID: 17823118 DOI: 10.1074/jbc.m704434200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor beta is growth-inhibitory in non-transformed epithelial cells but becomes growth-promoting during tumorigenesis. The role of the type I and II receptors in tumorigenesis has been extensively studied, but the role of the ubiquitously expressed type III receptor (TbetaRIII) remains elusive. We developed short hairpin RNAs directed against TbetaRIII to investigate the role of this receptor in breast cancer tumorigenesis. Nontumorigenic NMuMG mouse cells stably expressing short hairpin RNA specific to mouse TbetaRIII (NM-kd) demonstrated increased cell growth, motility, and invasion as compared with control cells expressing shRNA to human TbetaRIII (NM-con). Reconstitution of TbetaRIII expression with rat TbetaRIII abrogated the increased growth and motility seen in the NM-kd cells. In addition, the NM-kd cells exhibited marked reduction in the expression of the adherens junction protein, E-cadherin. This loss of E-cadherin was due to increased NFkappaB activity that, in turn, resulted in increased expression of the transcriptional repressors of E-cadherin such as Snail, Slug, Twist, and Sip1. Finally, NMuMG cells in which TbetaRIII had been knocked down formed invasive tumors in athymic nude mice, whereas the control cells did not. These data indicate that TbetaRIII acts as a tumor suppressor in nontumorigenic mammary epithelial cells at least in part by inhibiting NFkappaB-mediated repression of E-cadherin.
Collapse
Affiliation(s)
- Tracy L Criswell
- Department of Cancer Biology, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232-6307, USA
| | | |
Collapse
|
138
|
Eszlinger M, Jaeschke H, Paschke R. Insights from molecular pathways: potential pharmacologic targets of benign thyroid nodules. Curr Opin Endocrinol Diabetes Obes 2007; 14:393-7. [PMID: 17940470 DOI: 10.1097/med.0b013e3282ef5f96] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To describe molecular pathways that might be of relevance for a potential pharmacologic therapy of benign thyroid nodules. RECENT FINDINGS Constitutively activating thyrotropin receptor mutations have been found in about 60% of hot nodules. Its predominant role for signaling in hot nodules has been confirmed by in-vitro mutagenesis studies, thyrotropin receptor modeling and microarray studies. In contrast, the basic molecular cause of cold thyroid nodules is so far largely unknown. Defective sodium/iodide symporter trafficking, accumulation of T4-deficient, insufficiently iodinated thyroglobulin, increased oxidative stress and differential expression of several Gqalpha-protein kinase C pathway-associated genes have, however, recently been identified in cold thyroid nodules. SUMMARY As disturbed thyrotropin receptor signaling plays a central role in hot thyroid nodules, the identification of effective low-molecular-weight thyrotropin receptor ligands, such as thyrotropin receptor agonists, inverse agonists and antagonists has a pharmacologic potential in the diagnosis and treatment of thyroid cancer, Graves' disease and hot thyroid nodules, respectively. Further studies have to clarify the pharmacologic potential of the enhancement of antioxidative mechanisms and the inhibition of Gqalpha-protein kinase C signaling in cold thyroid nodules.
Collapse
|
139
|
Zou L, Yang R, Chai J, Pei G. Rapid xenograft tumor progression in beta-arrestin1 transgenic mice due to enhanced tumor angiogenesis. FASEB J 2007; 22:355-64. [PMID: 17890288 DOI: 10.1096/fj.07-9046com] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
beta-arrestins (beta-Arrs) are known to be associated with tumor signaling pathways such as transforming growth factor-beta1 (TGF-beta1), P53/Murine double minute (MDM2) and NF-kappaB. To investigate the role of beta-Arr in tumor progression in vivo, we generated beta-Arr transgenic mice by subcutaneously inoculating tumor cells in them. We found that the xenograft tumor initiated earlier and grew more rapidly in beta-Arr1 transgenic mice than in both the beta-Arr2 transgenic and wild-type mice after inoculating murine liver cancer Hepa1-6 cells or lymphoma EL4 cells. Moreover, matrix metalloproteinase 9 (MMP9) activity, vascular endothelial growth factor (VEGF) concentration in plasma and new small blood vessel formation in tumor tissues were enhanced in beta-Arr1 transgenic mice compared with those in control mice. In addition, injection of MMP9 inhibitors in beta-Arr1 transgenic mice abrogated all these effects and suppressed rapid tumor progression. Similar results were observed in human microvascular endothelial cells, where overexpressed beta-Arr1 did increase MMP9 activity and small blood vessel formation. Furthermore, phosphatidylinositol 3-kinase (PI3K) inhibitors could suppress beta-Arr1-enhanced MMP9 activity and the C-terminal 181-418 amino acids (aa) of beta-Arr1 was largely responsible for this effect. Our data reveal a functional role for beta-arrestin1 in tumor progression in vivo, in which overexpression of beta-Arr1 promotes MMP9 activity and tumor angiogenesis by providing a suitable microenvironment for tumor progression.
Collapse
Affiliation(s)
- Lin Zou
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | |
Collapse
|
140
|
Kimura T, Allen PB, Nairn AC, Caplan MJ. Arrestins and spinophilin competitively regulate Na+,K+-ATPase trafficking through association with a large cytoplasmic loop of the Na+,K+-ATPase. Mol Biol Cell 2007; 18:4508-18. [PMID: 17804821 PMCID: PMC2043564 DOI: 10.1091/mbc.e06-08-0711] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The activity and trafficking of the Na(+),K(+)-ATPase are regulated by several hormones, including dopamine, vasopressin, and adrenergic hormones through the action of G-protein-coupled receptors (GPCRs). Arrestins, GPCR kinases (GRKs), 14-3-3 proteins, and spinophilin interact with GPCRs and modulate the duration and magnitude of receptor signaling. We have found that arrestin 2 and 3, GRK 2 and 3, 14-3-3 epsilon, and spinophilin directly associate with the Na(+),K(+)-ATPase and that the associations with arrestins, GRKs, or 14-3-3 epsilon are blocked in the presence of spinophilin. In COS cells that overexpressed arrestin, the Na(+),K(+)-ATPase was redistributed to intracellular compartments. This effect was not seen in mock-transfected cells or in cells expressing spinophilin. Furthermore, expression of spinophilin appeared to slow, whereas overexpression of beta-arrestins accelerated internalization of the Na(+),K(+)-ATPase endocytosis. We also find that GRKs phosphorylate the Na(+),K(+)-ATPase in vitro on its large cytoplasmic loop. Taken together, it appears that association with arrestins, GRKs, 14-3-3 epsilon, and spinophilin may be important modulators of Na(+),K(+)-ATPase trafficking.
Collapse
Affiliation(s)
- Tohru Kimura
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026, USA
| | | | | | | |
Collapse
|
141
|
Hamdan FF, Rochdi MD, Breton B, Fessart D, Michaud DE, Charest PG, Laporte SA, Bouvier M. Unraveling G protein-coupled receptor endocytosis pathways using real-time monitoring of agonist-promoted interaction between beta-arrestins and AP-2. J Biol Chem 2007; 282:29089-100. [PMID: 17675294 DOI: 10.1074/jbc.m700577200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The most widely studied pathway underlying agonist-promoted internalization of G protein-coupled receptors (GPCRs) involves beta-arrestin and clathrin-coated pits. However, both beta-arrestin- and clathrin-independent processes have also been reported. Classically, the endocytic routes are characterized using pharmacological inhibitors and various dominant negative mutants, resulting sometimes in conflicting results and interpretational difficulties. Here, taking advantage of the fact that beta-arrestin binding to the beta2 subunit of the clathrin adaptor AP-2 (beta2-adaptin) is needed for the beta-arrestin-mediated targeting of GPCRs to clathrin-coated pits, we developed a bioluminescence resonance energy transfer-based approach directly assessing the molecular steps involved in the endocytosis of GPCRs in living cells. For 10 of the 12 receptors tested, including some that were previously suggested to internalize via clathrin-independent pathways, agonist stimulation promoted beta-arrestin 1 and 2 interaction with beta2-adaptin, indicating a beta-arrestin- and clathrin-dependent endocytic process. Detailed analyses of beta-arrestin interactions with both the receptor and beta2-adaptin also allowed us to demonstrate that recruitment of beta-arrestins to the receptor and the ensuing conformational changes are the leading events preceding AP-2 engagement and subsequent clathrin-mediated endocytosis. Among the receptors tested, only the endothelin A and B receptors failed to promote interaction between beta-arrestins and beta2-adaptin. However, both receptors recruited beta-arrestins upon agonist stimulation, suggesting a beta-arrestin-dependent but clathrin-independent route of internalization for these two receptors. In addition to providing a new tool to dissect the molecular events involved in GPCR endocytosis, the bioluminescence resonance energy transfer-based beta-arrestin/beta2-adaptin interaction assay represents a novel biosensor to assess receptor activation.
Collapse
Affiliation(s)
- Fadi F Hamdan
- Department of Biochemistry, Groupe de Recherche Universitaire sur le Médicament, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Murphy SJ, Shapira KE, Henis YI, Leof EB. A unique element in the cytoplasmic tail of the type II transforming growth factor-beta receptor controls basolateral delivery. Mol Biol Cell 2007; 18:3788-99. [PMID: 17634290 PMCID: PMC1995729 DOI: 10.1091/mbc.e06-10-0930] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor (TGF)-beta receptors stimulate diverse signaling processes that control a wide range of biological responses. In polarized epithelia, the TGFbeta type II receptor (T2R) is localized at the basolateral membranes. Sequential cytoplasmic truncations resulted in receptor missorting to apical surfaces, and they indicated an essential targeting element(s) near the receptor's C terminus. Point mutations in the full-length receptor confirmed this prediction, and a unique basolateral-targeting region was elucidated between residues 529 and 538 (LTAxxVAxxR) that was distinct, but colocalized within a clinically significant signaling domain essential for TGFbeta-dependent activation of the Smad2/3 cascade. Transfer of a terminal 84 amino-acid fragment, containing the LTAxxVAxxR element, to the apically sorted influenza hemagglutinin (HA) protein was dominant and directed basolateral HA expression. Although delivery to the basolateral surfaces was direct and independent of any detectable transient apical localization, fluorescence recovery after photobleaching demonstrated similar mobility for the wild-type receptor and a missorted mutant lacking the targeting motif. This latter finding excludes the possibility that the domain acts as a cell membrane retention signal, and it supports the hypothesis that T2R sorting occurs from an intracellular compartment.
Collapse
Affiliation(s)
- Stephen J. Murphy
- *Thoracic Diseases Research Unit, Department of Biochemistry and Molecular Biology and Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| | - Keren E. Shapira
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I. Henis
- Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Edward B. Leof
- *Thoracic Diseases Research Unit, Department of Biochemistry and Molecular Biology and Mayo Clinic Cancer Center, Mayo Clinic College of Medicine, Rochester, MN 55905; and
| |
Collapse
|
143
|
Shi Y, Feng Y, Kang J, Liu C, Li Z, Li D, Cao W, Qiu J, Guo Z, Bi E, Zang L, Lu C, Zhang JZ, Pei G. Critical regulation of CD4+ T cell survival and autoimmunity by beta-arrestin 1. Nat Immunol 2007; 8:817-24. [PMID: 17618287 DOI: 10.1038/ni1489] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 06/15/2007] [Indexed: 01/24/2023]
Abstract
CD4+ T cells are important in adaptive immunity, but their dysregulation can cause autoimmunity. Here we demonstrate that the multifunctional adaptor protein beta-arrestin 1 positively regulated naive and activated CD4+ T cell survival. We found enhanced expression of the proto-oncogene Bcl2 through beta-arrestin 1-dependent regulation of acetylation of histone H4 at the Bcl2 promoter. Mice deficient in the gene encoding beta-arrestin 1 (Arrb1) were much more resistant to experimental autoimmune encephalomyelitis, whereas overexpression of Arrb1 increased susceptibility to this disease. CD4+ T cells from patients with multiple sclerosis had much higher Arrb1 expression, and 'knockdown' of Arrb1 by RNA-mediated interference in those cells increased apoptosis induced by cytokine withdrawal. Our data demonstrate that beta-arrestin 1 is critical for CD4+ T cell survival and is a factor in susceptibility to autoimmunity.
Collapse
Affiliation(s)
- Yufeng Shi
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Lee NY, Blobe GC. The interaction of endoglin with beta-arrestin2 regulates transforming growth factor-beta-mediated ERK activation and migration in endothelial cells. J Biol Chem 2007; 282:21507-17. [PMID: 17540773 DOI: 10.1074/jbc.m700176200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In endothelial cells, transforming growth factor beta (TGF-beta) signals through two distinct pathways to regulate endothelial cell proliferation and migration, the ALK-1/Smads 1/5/8 pathway and the ALK-5/Smads 2/3 pathway. TGF-beta signaling through these pathways is further regulated in endothelial cells by the endothelial specific TGF-beta superfamily co-receptor, endoglin. The importance of endoglin, ALK-1, and ALK-5 in endothelial biology is underscored by the embryonic lethal phenotypes of knock-outs in mice due to defects in angiogenesis, and by the presence of disease-causing mutations in these genes in human vascular diseases. However, the mechanism of action of endoglin is not well defined. Here we define a novel interaction between endoglin and the scaffolding protein beta-arrestin2. Both co-immunoprecipitation and fluorescence confocal studies demonstrate the specific interaction between endoglin and beta-arrestin2 in endothelial cells, enhanced by ALK-1 and to a lesser extent by the type II TGF-beta receptor. The endoglin/beta-arrestin2 interaction results in endoglin internalization and co-accumulation of endoglin and beta-arrestin2 in endocytic vesicles. Whereas endoglin did not have a direct impact on either Smad 2/3 or Smad 1/5/8 activation, endoglin antagonized TGF-beta-mediated ERK signaling, altered the subcellular distribution of activated ERK, and inhibited endothelial cell migration in a manner dependent on the ability of endoglin to interact with beta-arrestin2. Reciprocally, small interfering RNA-mediated silencing of endogenous beta-arrestin2 expression restored TGF-beta-mediated ERK activation and increased endothelial cell migration in an endoglin-dependent manner. These studies define a novel function for endoglin, and further expand the roles mediated by the ubiquitous scaffolding protein beta-arrestin2.
Collapse
Affiliation(s)
- Nam Y Lee
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
145
|
Hempel N, How T, Dong M, Murphy SK, Fields TA, Blobe GC. Loss of betaglycan expression in ovarian cancer: role in motility and invasion. Cancer Res 2007; 67:5231-8. [PMID: 17522389 DOI: 10.1158/0008-5472.can-07-0035] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transforming growth factor-beta (TGF-beta) superfamily members, TGF-beta, activin, and inhibin, all have prominent roles in regulating normal ovarian function. Betaglycan, or the type III TGF-beta receptor, is a coreceptor that regulates TGF-beta, activin, and inhibin signaling. Here, we show that betaglycan expression is frequently decreased or lost in epithelial derived ovarian cancer at both the mRNA and protein level, with the degree of loss correlating with tumor grade. Treatment of ovarian cancer cell lines with the methyltransferase inhibitor 5-aza-2-deoxycytidine and the histone deacetylase inhibitor trichostatin A resulted in significant synergistic induction of betaglycan message levels and increased betaglycan protein expression, indicating that epigenetic silencing may play a role in the loss of betaglycan expression observed in ovarian cancer. Although restoring betaglycan expression in Ovca429 ovarian cancer cells is not sufficient to restore TGF-beta-mediated inhibition of proliferation, betaglycan significantly inhibits ovarian cancer cell motility and invasiveness. Furthermore, betaglycan specifically enhances the antimigratory effects of inhibin and the ability of inhibin to repress matrix metalloproteinase levels in these cells. These results show, for the first time, epigenetic regulation of betaglycan expression in ovarian cancer, and a novel role for betaglycan in regulating ovarian cancer motility and invasiveness.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
146
|
Eszlinger M, Krohn K, Kukulska A, Jarzab B, Paschke R. Perspectives and limitations of microarray-based gene expression profiling of thyroid tumors. Endocr Rev 2007; 28:322-38. [PMID: 17353294 DOI: 10.1210/er.2006-0047] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Microarray technology has become a powerful tool to analyze the gene expression of tens of thousands of genes simultaneously. Microarray-based gene expression profiles are available for malignant thyroid tumors (i.e., follicular thyroid carcinoma, and papillary thyroid carcinoma), and for benign thyroid tumors (such as autonomously functioning thyroid nodules and cold thyroid nodules). In general, the two main foci of microarray investigations are improved understanding of the pathophysiology/molecular etiology of thyroid neoplasia and the detection of genetic markers that could improve the differential diagnosis of thyroid tumors. Their results revealed new features, not known from one-gene studies. Simultaneously, the increasing number of microarray analyses of different thyroid pathologies raises the demand to efficiently compare the data. However, the use of different microarray platforms complicates cross-analysis. In addition, there are other important differences between these studies: 1) some studies use intraindividual comparisons, whereas other studies perform interindividual comparisons; 2) the reference tissue is defined as strictly nonnodular healthy tissue or also contains benign lesions such as goiter, follicular adenoma, and hyperplastic nodules in some studies; and 3) the widely used Affymetrix GeneChip platform comprises several GeneChip generations that are only partially compatible. Moreover, the different studies are characterized by strong differences in data analysis methods, which vary from simple empiric filters to sophisticated statistic algorithms. Therefore, this review summarizes and compares the different published reports in the context of their study design. It also illustrates perspectives and solutions for data set integration and meta-analysis, as well as the possibilities to combine array analysis with other genetic approaches.
Collapse
Affiliation(s)
- Markus Eszlinger
- III. Medical Department, University of Leipzig, Ph.-Rosenthal-Str. 27, D-04103 Leipzig, Germany
| | | | | | | | | |
Collapse
|
147
|
Abstract
Upon their discovery, beta-arrestins 1 and 2 were named for their capacity to sterically hinder the G protein coupling of agonist-activated seven-transmembrane receptors, ultimately resulting in receptor desensitization. Surprisingly, recent evidence shows that beta-arrestins can also function to activate signaling cascades independently of G protein activation. By serving as multiprotein scaffolds, the beta-arrestins bring elements of specific signaling pathways into close proximity. beta-Arrestin regulation has been demonstrated for an ever-increasing number of signaling molecules, including the mitogen-activated protein kinases ERK, JNK, and p38 as well as Akt, PI3 kinase, and RhoA. In addition, investigators are discovering new roles for beta-arrestins in nuclear functions. Here, we review the signaling capacities of these versatile adapter molecules and discuss the possible implications for cellular processes such as chemotaxis and apoptosis.
Collapse
Affiliation(s)
- Scott M DeWire
- Howard Hughes Medical Institute and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
148
|
Abstract
To ensure that extracellular stimuli are translated into intracellular signals of appropriate magnitude and specificity, most signaling cascades are tightly regulated. One of the major mechanisms involved in the regulation of G protein-coupled receptors (GPCRs) involves their endocytic trafficking. GPCR endocytic trafficking entails the targeting of receptors to discrete endocytic sites at the plasma membrane, followed by receptor internalization and intracellular sorting. This regulates the level of cell surface receptors, the sorting of receptors to degradative or recycling pathways, and in some cases the specific signaling pathways. In this chapter we discuss the mechanisms that regulate receptor endocytic trafficking, emphasizing the role of GPCR kinases (GRKs) and arrestins in this process.
Collapse
Affiliation(s)
- Catherine A C Moore
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | |
Collapse
|
149
|
Ruiz-Gómez A, Molnar C, Holguín H, Mayor F, de Celis JF. The cell biology of Smo signalling and its relationships with GPCRs. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1768:901-12. [PMID: 17094938 DOI: 10.1016/j.bbamem.2006.09.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 09/20/2006] [Accepted: 09/25/2006] [Indexed: 12/11/2022]
Abstract
The Smoothened (Smo) signalling pathway participates in many developmental processes, contributing to the regulation of gene expression by controlling the activity of transcription factors belonging to the Gli family. The key elements of the pathway were identified by means of genetic screens carried out in Drosophila, and subsequent analysis in other model organisms revealed a high degree of conservation in both the proteins involved and in their molecular interactions. Recent analysis of the pathway, using a combination of biochemical and cell biological approaches, is uncovering the intricacies of Smo signalling, placing its elements in particular cellular compartments and qualifying the molecular processes involved. These include the synthesis, secretion and diffusion of the ligand, the activation of the receptor and the modifications in the activity of nuclear effectors. In this review we discuss recent advances in understanding biochemical and cellular aspects of Smo signalling, with particular focus in the similarities in the mechanism of signal transduction between Smo and other transmembrane proteins belonging to the G-Protein coupled receptors superfamily (GPCR).
Collapse
Affiliation(s)
- Ana Ruiz-Gómez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.
| | | | | | | | | |
Collapse
|
150
|
Turley RS, Finger EC, Hempel N, How T, Fields TA, Blobe GC. The Type III Transforming Growth Factor-β Receptor as a Novel Tumor Suppressor Gene in Prostate Cancer. Cancer Res 2007; 67:1090-8. [PMID: 17283142 DOI: 10.1158/0008-5472.can-06-3117] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transforming growth factor-beta (TGF-beta) signaling pathway has an important role in regulating normal prostate epithelium, inhibiting proliferation, differentiation, and both androgen deprivation-induced and androgen-independent apoptosis. During prostate cancer formation, most prostate cancer cells become resistant to these homeostatic effects of TGF-beta. Although the loss of expression of either the type I (TbetaRI) or type II (TbetaRII) TGF-beta receptor has been documented in approximately 30% of prostate cancers, most prostate cancers become TGF-beta resistant without mutation or deletion of TbetaRI, TbetaRII, or Smads2, 3, and 4, and thus, the mechanism of resistance remains to be defined. Here, we show that type III TGF-beta receptor (TbetaRIII or betaglycan) expression is decreased or lost in the majority of human prostate cancers as compared with benign prostate tissue at both the mRNA and protein level. Loss of TbetaRIII expression correlates with advancing tumor stage and a higher probability of prostate-specific antigen (PSA) recurrence, suggesting a role in prostate cancer progression. The loss of TbetaRIII expression is mediated by the loss of heterozygosity at the TGFBR3 genomic locus and epigenetic regulation of the TbetaRIII promoter. Functionally, restoring TbetaRIII expression in prostate cancer cells potently decreases cell motility and cell invasion through Matrigel in vitro and prostate tumorigenicity in vivo. Taken together, these studies define the loss of TbetaRIII expression as a common event in human prostate cancer and suggest that this loss is important for prostate cancer progression through effects on cell motility, invasiveness, and tumorigenicity.
Collapse
Affiliation(s)
- Ryan S Turley
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|