Editorial Open Access
Copyright ©2010 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Biol Chem. Dec 26, 2010; 1(12): 353-361
Published online Dec 26, 2010. doi: 10.4331/wjbc.v1.i12.353
CARMA3: A novel scaffold protein in regulation of NF-κB activation and diseases
Jiyuan Sun, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, TX 77030, United States
Author contributions: Sun J solely contributed to this paper.
Supported by The National Institutes of Health through MD Anderson’s Cancer Center Support Grant, No. CA016672
Correspondence to: Jiyuan Sun, MD, PhD, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Unit 1002, 1515 Holcombe Blvd., Houston, TX 77030, United States. ssmu@hotmail.com
Telephone: +1-713-7971828 Fax: +1-713-7453120
Received: September 17, 2010
Revised: October 18, 2010
Accepted: October 25, 2010
Published online: December 26, 2010

Abstract

CARD recruited membrane associated protein 3 (CARMA3) is a novel scaffold protein. It belongs to the CARMA protein family, and is known to activate nuclear factor (NF)-κB. However, it is still unknown which receptor functions upstream of CARMA3 to trigger NF-κB activation. Recently, several studies have demonstrated that CARMA3 serves as an indispensable adaptor protein in NF-κB signaling under some G protein-coupled receptors (GPCRs), such as lysophosphatidic acid (LPA) receptor and angiotensin (Ang) II receptor. Mechanistically, CARMA3 recruits its essential downstream molecules Bcl10 and MALT1 to form the CBM (CARMA3-Bcl10-MALT1) signalosome whereby it triggers NF-κB activation. GPCRs and NF-κB play pivotal roles in the regulation of various cellular functions, therefore, aberrant regulation of the GPCR/NF-κB signaling axis leads to the development of many types of diseases, such as cancer and atherogenesis. Recently, the GPCR/CARMA3/NF-κB signaling axis has been confirmed in these specific diseases and it plays crucial roles in the pathogenesis of disease progression. In ovarian cancer cell lines, knockdown of CARMA3 abolishes LPA receptor-induced NF-κB activation, and reduces LPA-induced ovarian cancer invasion. In vascular smooth cells, downregulation of CARMA3 substantially impairs Ang-II-receptor-induced NF-κB activation, and in vivo studies have confirmed that Bcl10-deficient mice are protected from developing Ang-II-receptor-induced atherosclerosis and aortic aneurysms. In this review, we summarize the biology of CARMA3, describe the role of the GPCR/CARMA3/NF-κB signaling axis in ovarian cancer and atherogenesis, and speculate about the potential roles of this signaling axis in other types of cancer and diseases. With a significant increase in the identification of LPA- and Ang-II-like ligands, such as endothelin-1, which also activates NF-κB via CARMA3 and contributes to the development of many diseases, CARMA3 is emerging as a novel therapeutic target for various types of cancer and other diseases.

Key Words: G protein-coupled receptor, β-arrestin, CARD recruited membrane associated protein 3, Nuclear factor-κB, Cancer, Atherogenesis



INTRODUCTION

CARD recruited membrane associated protein 3 (CARMA3) is a novel scaffold protein. CARMA3 belongs to the CARMA protein family, which includes CARMA1, CARMA2, and CARMA3[1,2]. CARMA protein is also referred to as Bimp (Bcl10-interacting membrane protein), because it was first identified as a protein that interacts with Bcl10, another CARD domain adaptor protein. Although the biology and function of the CARMA protein family has not been completely elucidated, it has been shown that all of them play important roles in nuclear factor (NF)-κB activation[1-3]. NF-κB was first identified as a transcription factor of immunoglobulin κ light chain in B cells and is characterized by its important roles in the immune system[4]. NF-κB is now known to be ubiquitously expressed in all cell types and has prominent roles in tumorigenesis and the development of neural, heart and immune diseases[4-8]. The NF-κB family has five members: p50, p52, RelA (p65), RelB, and c-Rel[4]. In resting cells, all five members form homodimers or heterodimers and are sequestered in the cytoplasm via coupling with the inhibitor of κB (IκB) proteins, such as IκBα[9]. IκB masks the nuclear localization signal of NF-κB and inhibits its function.

NF-κB is activated via the classical or the alternative pathway[4]. Most receptors, such as the T-cell receptor (TCR) and B-cell receptor (BCR)[4], activate NF-κB through the classical pathway. Only a small number of receptors activate NF-κB through the alternative pathway[4,10]. Upon receptor activation, downstream adapters bind to these receptors and recruit kinases to activate the IκB kinase (IKK) complex[11,12]. IKK comprises IKKα, IKKβ, and IKKγ [NF-κB essential modulator (NEMO)] in the classical pathway and an IKKα dimer in the alternative pathway. The IKK complex directly phosphorylates IκBα at serines 32 and 36, which leads to IκBα polyubiquitination by the E3 ubiquitin ligase[4]. IκBα is then degraded by the 26S proteasome, and the NF-κB dimer is released from the cytoplasm and translocated to the nucleus, where it transactivates its target genes[13].

Although the CARMA protein family has been shown to activate NF-κB, it is still unknown which receptors function upstream of CAMRA3 to trigger NF-κB activation. Recently, it has been shown that some G protein-coupled receptors (GPCRs), like lysophosphatidic acid (LPA) and angiotensin (Ang) II receptors trigger NF-κB activation via CARMA3. GPCRs comprise a large protein family of transmembrane receptors that sense molecules outside the cell and activate inside signal transduction pathways, and ultimately, cellular responses. GPCRs are the largest cell surface receptors. Up to 2% of the human genome encodes GPCRs[14]. GPCRs are expressed throughout the body, including the central nervous system, cardiovascular system, gastrointestinal tract, musculoskeletal system, genitourinary system, reproductive system, and almost all organs controlled by the autonomic nervous system[15]. GPCRs are activated by a diverse array of ligands and play crucial roles in physiology. Furthermore, they are involved in almost all types of stimulus-response pathways and are important targets of 40%-50% of modern drugs[16].

GPCRs signal via heterotrimeric G proteins (Gα, Gβ, and Gγ) or β-arrestins[17,18]. G proteins are heterotrimeric and include eighteen α subunits that are classified into four groups (Gs, Gi, Gq, and G12/13), twelve β subunits, and five γ subunits[19]. These G proteins independently or cooperatively activate their downstream signaling cascades[19]. β-arrestins also function to relay signals rather than simply desensitize GPCR-induced signals[20]. Upon activation, GPCRs activate numerous downstream effectors. One important target is NF-κB. Constitutive activation of NF-κB contributes to various diseases, including cancer and atherogenesis[5,21]. In this review, we summarize the biology of CARMA3 and the CARMA protein family, discuss the role of the GPCR/CARMA3/NF-κB signaling axis in ovarian cancer and atherosclerosis, and speculate about the potential roles of this signaling axis in other types of cancer and diseases.

CARMA PROTEIN FAMILY
Structure

CARMA proteins are caspase recruitment domain (CARD)-containing members of the membrane-associated guanylate kinase (GUK) family. The CARMA protein family has three members: CARMA1, CARMA2, and CARMA3. The three members share similar structures (Figure 1A): an N-terminal CARD followed by a coiled-coil domain; a linker region; a PDZ domain; an Src homology 3 (SH3) domain; and a GUK-like domain[1,3]. The CARD domain is found in a variety of proteins, especially those involved in apoptosis and inflammation. This domain consists of six or seven antiparallel α helices with a hydrophobic core and outer surface. It mediates the interaction of larger protein complexes by association with different individual CARD domains[22-24]. The coiled-coil domain mediates dimerization[25]. The linker region contains crucial phosphorylation sites[26]. Upon phosphorylation of the linker region, CARMA protein is activated, unfolds, and recruits downstream molecules (Figure 1B). The PDZ, SH3 and GUK domains are membrane-associated domains that act in membrane localization. Therefore, they are also known as membrane-associated GUK domains[27]. Although the structure of the GUK domain is similar to that of GUK itself, it does not have any kinase activity.

Figure 1
Figure 1 CARD recruited membrane associated protein family members. A: The CARD recruited membrane associated (CARMA) protein family has three members: CARMA1, CARMA2 and CARMA3. Each member shares similar structures: an N-terminal caspase-recruitment domain (CARD), followed by a coiled-coil domain (CC), a linker region, a PDZ domain, an SH3 domain, and a GUK-like domain. Although all CARMA protein members share similar structures, they are transcribed by distinct genes, and expressed in different tissues. Specifically, CARMA1 is predominantly expressed in spleen, thymus, and peripheral blood leukocytes; CARMA2 is expressed only in placenta; and CARMA3 is expressed in a broad range of tissues, especially highly in liver, kidney, heart, and brain, but not in spleen, thymus, or peripheral blood lymphocytes; B: Upon activation, the linker region is phosphorylated. The CARD domain of CARMA protein interacts with Bcl10, which further binds MALT1, while PDZ and SH3 domains associate with the IκB kinase complex via NF-κB essential modulator (NEMO). Additionally, different CARMA proteins also interact with other unique signaling molecules. For example, CARMA3 interacts with β-arrestin 2, whereas CARMA1 associates with ADAP.
Distribution

Although the three CARMA proteins have similar structures, they are transcribed by different genes and expressed in different tissues[1,3,28]. Specifically, CARMA1 is predominantly expressed in the spleen, thymus, and peripheral blood leukocytes[3]; CARMA2 is expressed only in the placenta[1]; and CARMA3 is expressed in a broad range of tissues, especially at high levels in the liver, kidney, heart, and brain, but is not expressed in the spleen, thymus, or peripheral blood lymphocytes[28].

Function

Overexpression of CARMA proteins induces robust NF-κB activation[1,2]. However, the receptors that employ CARMA proteins to activate NF-κB remain unknown. Recently, studies have demonstrated that CARMA1 is required for TCR- and BCR-induced NF-κB activation[29-31]. Also, we and other groups have shown that CARMA3 is indispensable for induction of NF-κB activation by some GPCR ligands (LPA, endothelin-1, and Ang II)[32,33]. CARMA1 and CARMA3 activate NF-κB by recruiting the same downstream molecules: Bcl10 (B-cell chronic lymphocytic leukemia-lymphoma 10), MALT1 (mucosa-associated lymphoid tissue lymphoma translocation gene 1), and TRAF6 (tumor necrosis factor receptor-associated factor 6)[34-36]. Bcl10 and MALT1 are two indispensable proteins thought to synergize in NF-κB activation. TRAF6 is an E3 ubiquitin ligase that catalyzes the formation of polyubiquitin chains on IKK and facilitates NF-κB activation[37,38]. In addition, overexpression of CARMA2 activates NF-κB. However, because CARMA2 is only expressed in placental tissues, its function remains undetermined.

Mechanism of activation

Upon receptor activation, CARMA proteins are recruited to the membrane proximal regions of receptors by adaptor proteins, where they can be further phosphorylated by specific protein kinase C (PKC) isoforms, which results in activation and recruitment of downstream effectors. In T cells, adhesion- and degranulation-promoting adaptor protein (ADAP) links CARMA1 with the membrane proximal region of TCR and facilitates its phosphorylation and activation by PKC[39]. In ADAP-deficient T cells, TCR-stimulated assembly of the CARMA1/Bcl10/MALT1 complex and activation of NF-κB are substantially impaired[39]. Upon GPCR activation, CARMA3 is linked with the GPCR viaβ-arrestin 2. In β-arrestin-2-deficient mouse embryonic fibroblasts, GPCR (LPA)-induced NF-κB activation is completely abolished[40]. Although it has been reported that β-arrestins inhibit GPCR-induced NF-κB activation[41,42], it is proposed that the phosphorylation status of β-arrestin 2 might critically regulate and determine its function in NF-κB activation.

After CARMA proteins are linked to the receptor proximal region, PKC is engaged in phosphorylation of CARMA proteins. In T and B cells, PKCθ and PKCβ, respectively, phosphorylate CARMA1 and play indispensable roles in TCR- and BCR-induced NF-κB activation. In both pathways, PKCθ and PKCβ phosphorylate similar residues on the link region of CARMA1 and contribute to NF-κB activation[43,44]. PKC also functions in GPCR-induced NF-κB activation. Several groups have revealed that PKCα or PKCδ might be required for GPCR-induced NF-κB activation[33,45]. Also, PKCδ deficiency reportedly impairs LPA-induced NF-κB-dependent interleukin (IL)-8 secretion[46], and dominant-negative PKCα substantially attenuates LPA-induced NF-κB activation[47], which indicates that PKCδ or PKCα is the key PKC isoform in phosphorylation and activation of CARMA3.

In particular, in response to TCR and BCR activation, PKCθ and PKCβ are activated and phosphorylate S552 and S564/S649/S657, respectively, on the CARMA1 linker region. Mutations on these residues abolish TCR- and BCR-induced NF-κB activation[48,49]. Similar to CARMA1, it has been proposed that some PKC isoforms activate NF-κB through CARMA3 in the presence of GPCR[32,33,47]. In determining which residue of CARMA3 is phosphorylated, we have demonstrated that CARMA3 mutant S520A, an analog of CARMA1 S552A, does not rescue TCR-induced NF-κB activation[49]; however, wild-type CARMA3 does rescue TCR-induced NF-κB activation in CARMA1-deficient Jurkat T cells[49]. Therefore, CARMA3 S520 might be the crucial site for CARMA3 phosphorylation and activation.

Upon phosphorylation, CARMA1 and CARMA3 contribute to NF-κB activation by regulating the activity of the IKK complex through IKK NEMO polyubiquitination[33,38]. Although reports have suggested that phosphorylation of IKK indicates its activation, we have shown that phosphorylation of IKK is not sufficient to induce its kinase activity[33]. Only after both IKKα and IKKβ are phosphorylated and IKK NEMO is ubiquitinated, is IKK activated and able to phosphorylate downstream IκBα[33,38]. In GPCR-induced NF-κB signaling, IKKα/β phosphorylation is controlled by a PKC-dependent but CARMA3-independent pathway, and IKK NEMO polyubiquitination is controlled by a CARMA3-dependent pathway (Figure 2). Therefore, in CARMA3-deficient murine embryonic fibroblasts, IKKα/β phosphorylation remains intact, but IKK NEMO polyubiquitination is impaired. Consequently, IKK is not activated and is unable to phosphorylate IκBα, thus, LPA-induced NF-κB activation is completely abolished[33,38].

Figure 2
Figure 2 Working model of CARD recruited membrane associated protein 3-dependent nuclear factor-κB activation in the G protein-coupled receptor (lysophosphatidic acid) signaling pathways. G protein-coupled receptor (GPCR) [lysophosphatidic acid (LPA)]-induced nuclear factor (NF)-κB activation involves the recruitment of CARD recruited membrane associated protein 3 (CARMA3) to the receptor by β-arrestin 2, which leads to formation of the CARMA3/Bcl10/MALT1/TRAF6 complex, which results in polyubiquitination of the IκB kinase (IKK) complex. A CARMA3-independent, PKC-dependent signal induces phosphorylation of the IKK complex by an unknown kinase in the presence of GPCR. After IKK is both polyubiquitinated [NF-κB essential modulator (NEMO)] and phosphorylated (IKK), it is activated, which leads to NF-κB activation. In the absence of CARMA3, GPCR (LPA)-induced polyubiquitination of the IKK complex is defective, which results in defects in IKK and NF-κB activation. WT: Wild-type; Ub: Ubiquitin.
THE GPCR/CARMA3/NF-κB SIGNALING AXIS

At the molecular level, GPCR activates CARMA3, which in turn further activates NF-κB via multiple pathways[19]. However, which pathway is important in relaying signals to NF-κB via CAMRA3 remains to be determined. Upon ligand binding to receptors, G proteins such as Gαq, Gαi, and Gα12/13 are activated[50]. Gαq then activates phospholipase Cβ, which hydrolyzes phosphatidylinositol 4 5-bisphosphate. With consequent production of diacylglycerol and release of calcium from endoplasm, PKC is activated[51], thereby leading to NF-κB activation. This pathway promotes cell survival. In addition, Gαi activates the phosphatidylinositol 3-kinase (PI3K)/AKT and SOS/RAS/ERK pathways[52,53], which activate NF-κB and promote cell spread, migration, invasion, and DNA synthesis. Furthermore, Gα12/13 activates NF-κB via the G12/13/RHO/GEF/RHOA pathway and contributes to contraction and cell rounding[54].

CARMA3 is an indispensable signaling component in GPCR-induced NF-κB activation, therefore, it plays a crucial role in the development of diseases that result from the aberrant regulation of GPCR/NF-κB signaling, such as tumor progression and atherogenesis. GPCR activates NF-κB via CARMA3, and NF-κB in turn upregulates the expression of numerous genes that are involved in cell proliferation, anti-apoptosis, angiogenesis, migration, invasion, metastasis and inflammation, such as cyclin D1[55], bcl-2[56], vascular endothelial growth factor (VEGF)[57,58], cyclooxygenase-2[59], matrix metalloproteinase (MMP)-2[58], MMP-9[58], urokinase plasminogen activator (uPA)[57,58,60], growth-regulated oncogene α[61-63], IL-6[64], and IL-8[58,65]. Thus, sustained NF-κB activity has emerged as a hallmark of many diseases[5,21].

Recently, we have discovered that CARMA3 is indispensable for GPCR-induced NF-κB activation in murine embryonic fibroblasts[33]. However, whether the GPCR/CARMA3/NF-κB signaling axis is found in some specific diseases has yet to be completely elucidated. Below, we briefly describe the role of the GPCR/CARMA3/NF-κB signaling axis in ovarian cancer and atherogenesis, and speculate about its potential roles in other types of cancer and diseases (Figure 3).

Figure 3
Figure 3 Proposed working model of G protein-coupled receptor/CARD recruited membrane associated protein 3/nuclear factor-κB signaling pathways in cancer, cardiovascular diseases, and other diseases. G protein-coupled receptor (GPCR) [lysophosphatidic acid (LPA), angiotensin (Ang) II] activates CARD recruited membrane associated protein 3 (CARMA3), which in turn triggers nuclear factor (NF)-κB activation. NF-κB plays an important role in regulation of many physiological and pathological processes. Aberrant regulation of the GPCR/CARMA3/NF-κB signaling axis results in cancer, cardiovascular diseases, and probably other diseases. Mechanistically, it promotes cell proliferation, angiogenesis and metastasis, and inhibits apoptosis. In addition, it also induces inflammation. CARMA3 is indispensable for GPCR (LPA, Ang II)-induced NF-κB activation. Consequently, it plays pivotal roles in GPCR-induced tumor progression and cardiovascular diseases. Full definition of GPCR/CARMA3/NF-κB signaling events could aid the discovery of new drug targets and production of profoundly significant clinical therapies for cancer, cardiovascular diseases, and many other diseases. TNF: Tumor necrosis factor; IL: Interleukin; VEGF: Vascular endothelial growth factor; uPA: Urokinase plasminogen activator; MMPs: Matrix metalloproteinases.
GPCR/CARMA3/NF-κB signaling axis in ovarian cancer

Ovarian cancer is among the four most common cancers worldwide. In 90% of patients, LPA expression level is significantly elevated[66]. LPA is a typical GPCR ligand that activates NF-κB and leads to tumor progression. LPA receptors LPA1, LPA2, and LPA3 are aberrantly expressed in ovarian cancer cells[50]. Consequently, LPA serves as a diagnostic marker for ovarian cancer[67].

As a major active constituent of serum, LPA is a water-soluble phospholipid derivative of an intermediate in intracellular metabolism[50] or it is produced extracellularly from lysophosphatidylcholine by phospholipase A1/A2 or autotaxin (lysophospholipase D)[50,68,69]. Autotaxin is a widely expressed extracellular exophosphodiesterase that contributes to synthesis of LPA and promotes tumor invasion and metastasis[70]. LPA activates NF-κB and exerts striking wide hormone- and growth-factor-like effects, such as proliferation, apoptosis, differentiation, and chemotaxis[50]. Mechanistically, LPA activates NF-κB and transactivates numerous NF-κB target genes, such as cyclin D1, VEGF, uPA, IL-6, and IL-8. All of these genes play crucial roles in tumor progression.

Hu et al[55] have demonstrated that, at concentrations found in ascitic fluid, LPA can directly promote ovarian tumor growth by increasing the expression of cyclin D1, a key G1-phase checkpoint regulator, which results in cell proliferation. In addition, LPA stimulates secretion of VEGF[71] and promotes ovarian cancer angiogenesis, migration, and invasion[72]. Furthermore, LPA enhances secretion of IL-6, a pleiotropic cytokine that is involved in ovarian carcinogenesis via the Gi/PI3K/AKT/NF-κB pathway[64].

Recently, Li et al[60] have shown that LPA-induced, NF-κB-mediated ovarian cancer migration and invasion is partially dependent on expression of the NF-κB target gene uPA. Mutation of an NF-κB binding site in the uPA promoter region results in reduction of LPA-induced activation of the uPA promoter by > 80%. Li et al[60] have concluded that the Gi/Ras/Raf/NF-κB/uPA signaling cascade is responsible for LPA-induced ovarian cancer cell migration and invasion.

More recently, Mahanivong et al[47] have demonstrated that the LPA/CARMA3/NF-κB signaling axis is found in ovarian cancer cells. In this study, they observed that CARMA3 nucleated the LPA/NF-κB signaling pathway. LPA-induced NF-κB activation and ovarian cancer cell migration and invasion are substantially attenuated upon silencing CARMA3, Bcl10, and MALT1 with specific siRNAs. Mechanistically, Mahanivong et al[47] have found that the Ras/PKCα signaling cascade is involved and PKC might phosphorylate CARMA3. Thus, they delineated the entire GPCR/CARMA3/NF-κB signaling pathway in ovarian cancer cells.

Before the discovery of CARMA3, accumulating evidence suggested that the LPA/NF-κB signaling axis contributes to ovarian cancer tumorigenesis and progression, whereas the precise signaling components and mechanisms are not well defined. The study by Mahanivong et al[47] has provided the first evidence that the LPA/CARMA3/NF-κB signaling axis exists in ovarian cancer, plays important roles in ovarian cancer cell progression, and is a novel therapeutic target for ovarian cancer. In addition, it offers insight into other types of cancer. For example, aberrant regulation of LPA/NF-κB/IL-6/8 signaling pathways has been confirmed in breast cancer[57,61,73-82], colon cancer[83-88], prostate cancer[89-94], and lung cancer[95,96]. Inhibition of LPA activation or NF-κB signaling has been shown to prevent tumor progression and enhance sensitivity of chemotherapy[82]. Although the roles of CARMA3 in tumor progression have yet to be confirmed in these cancer types, the LPA/CARMA3/NF-κB signaling axis might also be found in these cancer types due to the high conservation of signaling pathways in most cell types. Future research will focus on the role of CARMA3 in these cancer types.

GPCR/CARMA3/NF-κB signaling axis in atherogenesis

Ang II is another type of GPCR ligand. It is a seven-amino-acid oligopeptide that is derived from Ang I and angiotensinogen. Ang II is a powerful hormone in the blood and regulates blood pressure[97]. In addition, it plays a crucial role in atherogenesis[98]. Atherogenesis results from vascular inflammation[99-101]. Epidemiologically, the hallmark of vascular inflammation is the elevation of IL-6[21]. IL-6 leads to the recruitment of circulating leukocytes and macrophages into the vascular wall, thereby leading to oxidation of lipoprotein, and atherogenesis[21]. It has been shown that Ang II infusion induces IL-6 production, which results in atherogenesis and vascular diseases[102]. IL-6 is an NF-κB targeted gene. Accumulating evidence has also illuminated the central role of NF-κB as a signal regulator that controls the process of vascular inflammation[21]. Therefore, the NF-κB/IL-6 signaling pathway plays a crucial role in atherogenesis and vascular inflammation.

Although Ang II is known to activate NF-κB and IL-6, the detailed molecular mechanism has remained elusive. Recently, McAllister-Lucas et al[32] have revealed that CARMA3 is an essential mediator of Ang-II-dependent NF-κB signaling. They have shown that all components of the CARMA3/Bcl10/MALT1 signaling pathway are present within the liver in the Ang-II-responsive HepG2 hepatocyte cell model. Ang-II-induced NF-κB activation is significantly abolished, upon knocking down of CARMA3, Bcl10 or MALT1. This study first provided the evidence that the CARMA3/Bcl10/Malt1 signalosome does exist and is indispensable for NF-κB activation upon Ang II receptor activation in hepatic cells.

To explore further the physiological and pathological function of CARMA3 signaling in the cardiovascular system, McAllister-Lucas et al[98] recently have revealed that CARMA3, Bcl10 and MALT1 are also expressed in vascular tissues. Consistent with previous results, Ang-II-induced NF-κB activation was significantly impaired upon knocking down of CARMA3, Bcl10, and MALT1 in vascular smooth muscle cells. Most importantly, when they crossed the Bcl10-/- and ApoE-/- mice, they found that Bcl10 deficiency protected ApoE-/- mice from Ang-II-dependent atherosclerosis and aortic aneurysms. Mechanistically, they revealed that serum levels of several pro-inflammatory mediators, which have all been implicated in the pathogenesis of atherogenesis, were also lower in ApoE-/- Bcl10-/- mice than in ApoE-/- mice (P < 0.01)[98]. Together, these results first demonstrated that Ang II/CARMA3/NF-κB signaling also exists and plays an important role in atherogenesis in addition to cancer. Further research will focus on its function and therapeutic application in atherogenesis and other vascular diseases, such as LPA- and NF-κB-induced ischemia-reperfusion injury[103,104] and coronary artery disease[104,105].

OUTLOOK

The GPCR/CARMA3/NF-κB signaling axis is a novel signaling pathway. GPCRs belong to a large family, which comprises more than 1000 receptors. Characterization of the role of CARMA3 in the GPCR-induced NF-κB activation signaling pathway will help create a holistic view of GPCR-induced NF-κB activation in the progression of various types of cancer (ovarian, colon, prostate, breast, and head and neck)[50,84], atherogenesis, as well as other diseases, such as LPA- and NF-κB-induced ischemia-reperfusion injury[103,104] and coronary artery disease[104,105]. With a strongly increasing tendency to identify more LPA-like ligands, such as the recently identified Ang II and endothelin-1, which also activate NF-κB via CARMA3 and contribute to many diseases, CARMA3 is expected to play crucial roles in a broad range of physiological and pathological conditions.

Future research will define the molecular mechanisms underlying induction of NF-κB activation by GPCR, β-arrestin 2, CARMA3, PKC, IKK, and whether CARMA3 signaling induces NF-κB activation under non-GPCR receptors. Also, we will determine what other novel signaling pathways CARMA3 mediates, and investigate the aberrant regulation of signaling cascades in diseases. Characterization of the roles and mechanisms of CARMA3 signaling will aid the discovery of new drug targets and be of major significance for many diseases and therapies.

Footnotes

Peer reviewers: Pedro A Lazo, MD, PhD, Profesor de Investigación CSIC, Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; Hiroaki Itamochi, MD, PhD, Junior Associate Professor, Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishicho, Yonago City 683-8504, Tottori, Japan; Seong-Wook Lee, PhD, Professor, Department of Molecular BiologyInstitute of Nanosensor and Biotechnology, Dankook University, 126, Jukjeon-dong, Suji-gu, Yongin-si, Gyeonggi-do, 448-701, South Korea

S- Editor Cheng JX L- Editor Kerr C E- Editor Zheng XM

References
1.  Bertin J, Wang L, Guo Y, Jacobson MD, Poyet JL, Srinivasula SM, Merriam S, DiStefano PS, Alnemri ES. CARD11 and CARD14 are novel caspase recruitment domain (CARD)/membrane-associated guanylate kinase (MAGUK) family members that interact with BCL10 and activate NF-kappa B. J Biol Chem. 2001;276:11877-11882.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Wang L, Guo Y, Huang WJ, Ke X, Poyet JL, Manji GA, Merriam S, Glucksmann MA, DiStefano PS, Alnemri ES. Card10 is a novel caspase recruitment domain/membrane-associated guanylate kinase family member that interacts with BCL10 and activates NF-kappa B. J Biol Chem. 2001;276:21405-21409.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Gaide O, Martinon F, Micheau O, Bonnet D, Thome M, Tschopp J. Carma1, a CARD-containing binding partner of Bcl10, induces Bcl10 phosphorylation and NF-kappaB activation. FEBS Lett. 2001;496:121-127.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Hayden MS, Ghosh S. Signaling to NF-kappaB. Genes Dev. 2004;18:2195-2224.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Aggarwal BB. Nuclear factor-kappaB: the enemy within. Cancer Cell. 2004;6:203-208.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Lukiw WJ, Zhao Y, Cui JG. An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J Biol Chem. 2008;283:31315-31322.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Singh MV, Kapoun A, Higgins L, Kutschke W, Thurman JM, Zhang R, Singh M, Yang J, Guan X, Lowe JS. Ca2+/calmodulin-dependent kinase II triggers cell membrane injury by inducing complement factor B gene expression in the mouse heart. J Clin Invest. 2009;119:986-996.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Panzer U, Steinmetz OM, Turner JE, Meyer-Schwesinger C, von Ruffer C, Meyer TN, Zahner G, Gómez-Guerrero C, Schmid RM, Helmchen U. Resolution of renal inflammation: a new role for NF-kappaB1 (p50) in inflammatory kidney diseases. Am J Physiol Renal Physiol. 2009;297:F429-F439.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Rothwarf DM, Karin M. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Sci STKE. 1999;1999:RE1.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Xiao G, Rabson AB, Young W, Qing G, Qu Z. Alternative pathways of NF-kappaB activation: a double-edged sword in health and disease. Cytokine Growth Factor Rev. 2006;17:281-293.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Häcker H, Karin M. Regulation and function of IKK and IKK-related kinases. Sci STKE. 2006;2006:re13.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Karin M. How NF-kappaB is activated: the role of the IkappaB kinase (IKK) complex. Oncogene. 1999;18:6867-6874.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Bhoj VG, Chen ZJ. Ubiquitylation in innate and adaptive immunity. Nature. 2009;458:430-437.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Keating CD, Kriek N, Daniels M, Ashcroft NR, Hopper NA, Siney EJ, Holden-Dye L, Burke JF. Whole-genome analysis of 60 G protein-coupled receptors in Caenorhabditis elegans by gene knockout with RNAi. Curr Biol. 2003;13:1715-1720.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Bruns IR, Chhum S, Dinh AT, Doerr H, Dunn NR, Ly YT, Mitman CL, Rickards HD, Sol C, Wan EW. A potential novel strategy to separate therapeutic- and side-effects that are mediated via the same receptor: beta-arrestin2/G-protein coupling antagonists. J Clin Pharm Ther. 2006;31:119-128.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Soltanshahi F, Mansley TE, Choi S, Clark RD. Balancing focused combinatorial libraries based on multiple GPCR ligands. J Comput Aided Mol Des. 2006;20:529-538.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Dromey JR, Pfleger KD. G protein coupled receptors as drug targets: the role of beta-arrestins. Endocr Metab Immune Disord Drug Targets. 2008;8:51-61.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Blumer JB, Smrcka AV, Lanier SM. Mechanistic pathways and biological roles for receptor-independent activators of G-protein signaling. Pharmacol Ther. 2007;113:488-506.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Ye RD. Regulation of nuclear factor kappaB activation by G-protein-coupled receptors. J Leukoc Biol. 2001;70:839-848.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Lefkowitz RJ, Whalen EJ. beta-arrestins: traffic cops of cell signaling. Curr Opin Cell Biol. 2004;16:162-168.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Brasier AR. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res. 2010;86:211-218.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Bouchier-Hayes L, Martin SJ. CARD games in apoptosis and immunity. EMBO Rep. 2002;3:616-621.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Hong GS, Jung YK. Caspase recruitment domain (CARD) as a bi-functional switch of caspase regulation and NF-kappaB signals. J Biochem Mol Biol. 2002;35:19-23.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Hiscott J, Lin R, Nakhaei P, Paz S. MasterCARD: a priceless link to innate immunity. Trends Mol Med. 2006;12:53-56.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Tanner MJ, Hanel W, Gaffen SL, Lin X. CARMA1 coiled-coil domain is involved in the oligomerization and subcellular localization of CARMA1 and is required for T cell receptor-induced NF-kappaB activation. J Biol Chem. 2007;282:17141-17147.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Rueda D, Thome M. Phosphorylation of CARMA1: the link(er) to NF-kappaB activation. Immunity. 2005;23:551-553.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Beresewicz M. [Scaffold proteins (MAGUK, Shank and Homer) in postsynaptic density in the central nervous system]. Postepy Biochem. 2007;53:188-197.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  McAllister-Lucas LM, Inohara N, Lucas PC, Ruland J, Benito A, Li Q, Chen S, Chen FF, Yamaoka S, Verma IM. Bimp1, a MAGUK family member linking protein kinase C activation to Bcl10-mediated NF-kappaB induction. J Biol Chem. 2001;276:30589-97.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Gaide O, Favier B, Legler DF, Bonnet D, Brissoni B, Valitutti S, Bron C, Tschopp J, Thome M. CARMA1 is a critical lipid raft-associated regulator of TCR-induced NF-kappa B activation. Nat Immunol. 2002;3:836-843.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Wang D, You Y, Case SM, McAllister-Lucas LM, Wang L, DiStefano PS, Nuñez G, Bertin J, Lin X. A requirement for CARMA1 in TCR-induced NF-kappa B activation. Nat Immunol. 2002;3:830-835.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Egawa T, Albrecht B, Favier B, Sunshine MJ, Mirchandani K, O'Brien W, Thome M, Littman DR. Requirement for CARMA1 in antigen receptor-induced NF-kappa B activation and lymphocyte proliferation. Curr Biol. 2003;13:1252-1258.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  McAllister-Lucas LM, Ruland J, Siu K, Jin X, Gu S, Kim DS, Kuffa P, Kohrt D, Mak TW, Nuñez G. CARMA3/Bcl10/MALT1-dependent NF-kappaB activation mediates angiotensin II-responsive inflammatory signaling in nonimmune cells. Proc Natl Acad Sci USA. 2007;104:139-144.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Grabiner BC, Blonska M, Lin PC, You Y, Wang D, Sun J, Darnay BG, Dong C, Lin X. CARMA3 deficiency abrogates G protein-coupled receptor-induced NF-{kappa}B activation. Genes Dev. 2007;21:984-996.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Margeta-Mitrovic M, Jan YN, Jan LY. A trafficking checkpoint controls GABA(B) receptor heterodimerization. Neuron. 2000;27:97-106.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, Barnes AA, Emson P, Foord SM, Marshall FH. Heterodimerization is required for the formation of a functional GABA(B) receptor. Nature. 1998;396:679-682.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Wang D, You Y, Lin PC, Xue L, Morris SW, Zeng H, Wen R, Lin X. Bcl10 plays a critical role in NF-kappaB activation induced by G protein-coupled receptors. Proc Natl Acad Sci USA. 2007;104:145-150.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Stilo R, Liguoro D, Di Jeso B, Formisano S, Consiglio E, Leonardi A, Vito P. Physical and functional interaction of CARMA1 and CARMA3 with Ikappa kinase gamma-NFkappaB essential modulator. J Biol Chem. 2004;279:34323-34331.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Shambharkar PB, Blonska M, Pappu BP, Li H, You Y, Sakurai H, Darnay BG, Hara H, Penninger J, Lin X. Phosphorylation and ubiquitination of the IkappaB kinase complex by two distinct signaling pathways. EMBO J. 2007;26:1794-1805.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Medeiros RB, Burbach BJ, Mueller KL, Srivastava R, Moon JJ, Highfill S, Peterson EJ, Shimizu Y. Regulation of NF-kappaB activation in T cells via association of the adapter proteins ADAP and CARMA1. Science. 2007;316:754-758.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Sun J, Lin X. Beta-arrestin 2 is required for lysophosphatidic acid-induced NF-kappaB activation. Proc Natl Acad Sci USA. 2008;105:17085-17090.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Witherow DS, Garrison TR, Miller WE, Lefkowitz RJ. beta-Arrestin inhibits NF-kappaB activity by means of its interaction with the NF-kappaB inhibitor IkappaBalpha. Proc Natl Acad Sci USA. 2004;101:8603-8607.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Gao H, Sun Y, Wu Y, Luan B, Wang Y, Qu B, Pei G. Identification of beta-arrestin2 as a G protein-coupled receptor-stimulated regulator of NF-kappaB pathways. Mol Cell. 2004;14:303-317.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Guo B, Su TT, Rawlings DJ. Protein kinase C family functions in B-cell activation. Curr Opin Immunol. 2004;16:367-373.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Monks CR, Kupfer H, Tamir I, Barlow A, Kupfer A. Selective modulation of protein kinase C-theta during T-cell activation. Nature. 1997;385:83-86.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Klemm S, Zimmermann S, Peschel C, Mak TW, Ruland J. Bcl10 and Malt1 control lysophosphatidic acid-induced NF-kappaB activation and cytokine production. Proc Natl Acad Sci USA. 2007;104:134-138.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Cummings R, Zhao Y, Jacoby D, Spannhake EW, Ohba M, Garcia JG, Watkins T, He D, Saatian B, Natarajan V. Protein kinase Cdelta mediates lysophosphatidic acid-induced NF-kappaB activation and interleukin-8 secretion in human bronchial epithelial cells. J Biol Chem. 2004;279:41085-41094.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Mahanivong C, Chen HM, Yee SW, Pan ZK, Dong Z, Huang S. Protein kinase C alpha-CARMA3 signaling axis links Ras to NF-kappa B for lysophosphatidic acid-induced urokinase plasminogen activator expression in ovarian cancer cells. Oncogene. 2008;27:1273-1280.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Sommer K, Guo B, Pomerantz JL, Bandaranayake AD, Moreno-García ME, Ovechkina YL, Rawlings DJ. Phosphorylation of the CARMA1 linker controls NF-kappaB activation. Immunity. 2005;23:561-574.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Matsumoto R, Wang D, Blonska M, Li H, Kobayashi M, Pappu B, Chen Y, Wang D, Lin X. Phosphorylation of CARMA1 plays a critical role in T Cell receptor-mediated NF-kappaB activation. Immunity. 2005;23:575-585.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer. 2003;3:582-591.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  van Corven EJ, Groenink A, Jalink K, Eichholtz T, Moolenaar WH. Lysophosphatidate-induced cell proliferation: identification and dissection of signaling pathways mediated by G proteins. Cell. 1989;59:45-54.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Taghavi P, Verhoeven E, Jacobs JJ, Lambooij JP, Stortelers C, Tanger E, Moolenaar WH, van Lohuizen M. In vitro genetic screen identifies a cooperative role for LPA signaling and c-Myc in cell transformation. Oncogene. 2008;27:6806-6816.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Caverzasio J, Palmer G, Suzuki A, Bonjour JP. Evidence for the involvement of two pathways in activation of extracellular signal-regulated kinase (Erk) and cell proliferation by Gi and Gq protein-coupled receptors in osteoblast-like cells. J Bone Miner Res. 2000;15:1697-1706.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Kranenburg O, Poland M, van Horck FP, Drechsel D, Hall A, Moolenaar WH. Activation of RhoA by lysophosphatidic acid and Galpha12/13 subunits in neuronal cells: induction of neurite retraction. Mol Biol Cell. 1999;10:1851-1857.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Hu YL, Albanese C, Pestell RG, Jaffe RB. Dual mechanisms for lysophosphatidic acid stimulation of human ovarian carcinoma cells. J Natl Cancer Inst. 2003;95:733-740.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Deng W, Wang DA, Gosmanova E, Johnson LR, Tigyi G. LPA protects intestinal epithelial cells from apoptosis by inhibiting the mitochondrial pathway. Am J Physiol Gastrointest Liver Physiol. 2003;284:G821-G829.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Pustilnik TB, Estrella V, Wiener JR, Mao M, Eder A, Watt MA, Bast RC Jr, Mills GB. Lysophosphatidic acid induces urokinase secretion by ovarian cancer cells. Clin Cancer Res. 1999;5:3704-3710.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Fang X, Schummer M, Mao M, Yu S, Tabassam FH, Swaby R, Hasegawa Y, Tanyi JL, LaPushin R, Eder A. Lysophosphatidic acid is a bioactive mediator in ovarian cancer. Biochim Biophys Acta. 2002;1582:257-264.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Symowicz J, Adley BP, Woo MM, Auersperg N, Hudson LG, Stack MS. Cyclooxygenase-2 functions as a downstream mediator of lysophosphatidic acid to promote aggressive behavior in ovarian carcinoma cells. Cancer Res. 2005;65:2234-2242.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Li H, Ye X, Mahanivong C, Bian D, Chun J, Huang S. Signaling mechanisms responsible for lysophosphatidic acid-induced urokinase plasminogen activator expression in ovarian cancer cells. J Biol Chem. 2005;280:10564-10571.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Bobrovnikova-Marjon EV, Marjon PL, Barbash O, Vander Jagt DL, Abcouwer SF. Expression of angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 is highly responsive to ambient glutamine availability: role of nuclear factor-kappaB and activating protein-1. Cancer Res. 2004;64:4858-4869.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Loukinova E, Chen Z, Van Waes C, Dong G. Expression of proangiogenic chemokine Gro 1 in low and high metastatic variants of Pam murine squamous cell carcinoma is differentially regulated by IL-1alpha, EGF and TGF-beta1 through NF-kappaB dependent and independent mechanisms. Int J Cancer. 2001;94:637-644.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Lee Z, Swaby RF, Liang Y, Yu S, Liu S, Lu KH, Bast RC Jr, Mills GB, Fang X. Lysophosphatidic acid is a major regulator of growth-regulated oncogene alpha in ovarian cancer. Cancer Res. 2006;66:2740-2748.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Chou CH, Wei LH, Kuo ML, Huang YJ, Lai KP, Chen CA, Hsieh CY. Up-regulation of interleukin-6 in human ovarian cancer cell via a Gi/PI3K-Akt/NF-kappaB pathway by lysophosphatidic acid, an ovarian cancer-activating factor. Carcinogenesis. 2005;26:45-52.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  So J, Navari J, Wang FQ, Fishman DA. Lysophosphatidic acid enhances epithelial ovarian carcinoma invasion through the increased expression of interleukin-8. Gynecol Oncol. 2004;95:314-322.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Sedláková I, Vávrová J, Tosner J, Hanousek L. Lysophosphatidic acid: an ovarian cancer marker. Eur J Gynaecol Oncol. 2008;29:511-514.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Ren J, Xiao YJ, Singh LS, Zhao X, Zhao Z, Feng L, Rose TM, Prestwich GD, Xu Y. Lysophosphatidic acid is constitutively produced by human peritoneal mesothelial cells and enhances adhesion, migration, and invasion of ovarian cancer cells. Cancer Res. 2006;66:3006-3014.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishimoto T, Mizuno K, Saku K, Taguchi R, Arai H. Serum lysophosphatidic acid is produced through diverse phospholipase pathways. J Biol Chem. 2002;277:48737-48744.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Song J, Clair T, Noh JH, Eun JW, Ryu SY, Lee SN, Ahn YM, Kim SY, Lee SH, Park WS. Autotaxin (lysoPLD/NPP2) protects fibroblasts from apoptosis through its enzymatic product, lysophosphatidic acid, utilizing albumin-bound substrate. Biochem Biophys Res Commun. 2005;337:967-975.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Clair T, Aoki J, Koh E, Bandle RW, Nam SW, Ptaszynska MM, Mills GB, Schiffmann E, Liotta LA, Stracke ML. Autotaxin hydrolyzes sphingosylphosphorylcholine to produce the regulator of migration, sphingosine-1-phosphate. Cancer Res. 2003;63:5446-5453.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Jeon ES, Heo SC, Lee IH, Choi YJ, Park JH, Choi KU, Park do Y, Suh DS, Yoon MS, Kim JH. Ovarian cancer-derived lysophosphatidic acid stimulates secretion of VEGF and stromal cell-derived factor-1 alpha from human mesenchymal stem cells. Exp Mol Med. 2010;42:280-293.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  So J, Wang FQ, Navari J, Schreher J, Fishman DA. LPA-induced epithelial ovarian cancer (EOC) in vitro invasion and migration are mediated by VEGF receptor-2 (VEGF-R2). Gynecol Oncol. 2005;97:870-878.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  David M, Wannecq E, Descotes F, Jansen S, Deux B, Ribeiro J, Serre CM, Grès S, Bendriss-Vermare N, Bollen M. Cancer cell expression of autotaxin controls bone metastasis formation in mouse through lysophosphatidic acid-dependent activation of osteoclasts. PLoS One. 2010;5:e9741.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br J Cancer. 2010;102:941-946.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Xu Y, Fang XJ, Casey G, Mills GB. Lysophospholipids activate ovarian and breast cancer cells. Biochem J. 1995;309:933-940.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Kitayama J, Shida D, Sako A, Ishikawa M, Hama K, Aoki J, Arai H, Nagawa H. Over-expression of lysophosphatidic acid receptor-2 in human invasive ductal carcinoma. Breast Cancer Res. 2004;6:R640-R646.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Boucharaba A, Serre CM, Guglielmi J, Bordet JC, Clézardin P, Peyruchaud O. The type 1 lysophosphatidic acid receptor is a target for therapy in bone metastases. Proc Natl Acad Sci USA. 2006;103:9643-9648.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Cicek M, Oursler MJ. Breast cancer bone metastasis and current small therapeutics. Cancer Metastasis Rev. 2006;25:635-644.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, Nordgren H, Farmer P, Praz V, Haibe-Kains B. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006;98:262-272.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, Viale G, Delorenzi M, Zhang Y, d'Assignies MS. Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007;13:3207-3214.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Liu S, Umezu-Goto M, Murph M, Lu Y, Liu W, Zhang F, Yu S, Stephens LC, Cui X, Murrow G. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell. 2009;15:539-550.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Samadi N, Gaetano C, Goping IS, Brindley DN. Autotaxin protects MCF-7 breast cancer and MDA-MB-435 melanoma cells against Taxol-induced apoptosis. Oncogene. 2009;28:1028-1039.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Shida D, Kitayama J, Yamaguchi H, Okaji Y, Tsuno NH, Watanabe T, Takuwa Y, Nagawa H. Lysophosphatidic acid (LPA) enhances the metastatic potential of human colon carcinoma DLD1 cells through LPA1. Cancer Res. 2003;63:1706-1711.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Yang M, Zhong WW, Srivastava N, Slavin A, Yang J, Hoey T, An S. G protein-coupled lysophosphatidic acid receptors stimulate proliferation of colon cancer cells through the {beta}-catenin pathway. Proc Natl Acad Sci USA. 2005;102:6027-6032.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Mori K, Kitayama J, Shida D, Yamashita H, Watanabe T, Nagawa H. Lysophosphatidic acid-induced effects in human colon carcinoma DLD1 cells are partially dependent on transactivation of epidermal growth factor receptor. J Surg Res. 2006;132:56-61.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Shida D, Fang X, Kordula T, Takabe K, Lépine S, Alvarez SE, Milstien S, Spiegel S. Cross-talk between LPA1 and epidermal growth factor receptors mediates up-regulation of sphingosine kinase 1 to promote gastric cancer cell motility and invasion. Cancer Res. 2008;68:6569-6577.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Lin S, Wang D, Iyer S, Ghaleb AM, Shim H, Yang VW, Chun J, Yun CC. The absence of LPA2 attenuates tumor formation in an experimental model of colitis-associated cancer. Gastroenterology. 2009;136:1711-1720.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Sun H, Ren J, Zhu Q, Kong FZ, Wu L, Pan BR. Effects of lysophosphatidic acid on human colon cancer cells and its mechanisms of action. World J Gastroenterol. 2009;15:4547-4555.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Xie Y, Gibbs TC, Mukhin YV, Meier KE. Role for 18:1 lysophosphatidic acid as an autocrine mediator in prostate cancer cells. J Biol Chem. 2002;277:32516-32526.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Raj GV, Sekula JA, Guo R, Madden JF, Daaka Y. Lysophosphatidic acid promotes survival of androgen-insensitive prostate cancer PC3 cells via activation of NF-kappaB. Prostate. 2004;61:105-113.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Sivashanmugam P, Tang L, Daaka Y. Interleukin 6 mediates the lysophosphatidic acid-regulated cross-talk between stromal and epithelial prostate cancer cells. J Biol Chem. 2004;279:21154-21159.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Guo R, Kasbohm EA, Arora P, Sample CJ, Baban B, Sud N, Sivashanmugam P, Moniri NH, Daaka Y. Expression and function of lysophosphatidic acid LPA1 receptor in prostate cancer cells. Endocrinology. 2006;147:4883-4892.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Hwang YS, Hodge JC, Sivapurapu N, Lindholm PF. Lysophosphatidic acid stimulates PC-3 prostate cancer cell Matrigel invasion through activation of RhoA and NF-kappaB activity. Mol Carcinog. 2006;45:518-529.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Hao F, Tan M, Xu X, Han J, Miller DD, Tigyi G, Cui MZ. Lysophosphatidic acid induces prostate cancer PC3 cell migration via activation of LPA(1), p42 and p38alpha. Biochim Biophys Acta. 2007;1771:883-892.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Xu X, Prestwich GD. Inhibition of tumor growth and angiogenesis by a lysophosphatidic acid antagonist in an engineered three-dimensional lung cancer xenograft model. Cancer. 2010;116:1739-1750.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Yamada T, Obo Y, Furukawa M, Hotta M, Yamasaki A, Honoki K, Fukushima N, Tsujiuchi T. Mutations of lysophosphatidic acid receptor-1 gene during progression of lung tumors in rats. Biochem Biophys Res Commun. 2009;378:424-427.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Mehta PK, Griendling KK. Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am J Physiol Cell Physiol. 2007;292:C82-C97.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  McAllister-Lucas LM, Jin X, Gu S, Siu K, McDonnell S, Ruland J, Delekta PC, Van Beek M, Lucas PC. The CARMA3-Bcl10-MALT1 signalosome promotes angiotensin II-dependent vascular inflammation and atherogenesis. J Biol Chem. 2010;285:25880-25884.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Sugiyama S, Kugiyama K, Nakamura S, Kataoka K, Aikawa M, Shimizu K, Koide S, Mitchell RN, Ogawa H, Libby P. Characterization of smooth muscle-like cells in circulating human peripheral blood. Atherosclerosis. 2006;187:351-362.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Maiellaro K, Taylor WR. The role of the adventitia in vascular inflammation. Cardiovasc Res. 2007;75:640-648.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Tieu BC, Lee C, Sun H, Lejeune W, Recinos A 3rd, Ju X, Spratt H, Guo DC, Milewicz D, Tilton RG. An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice. J Clin Invest. 2009;119:3637-3651.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Recinos A 3rd, LeJeune WS, Sun H, Lee CY, Tieu BC, Lu M, Hou T, Boldogh I, Tilton RG, Brasier AR. Angiotensin II induces IL-6 expression and the Jak-STAT3 pathway in aortic adventitia of LDL receptor-deficient mice. Atherosclerosis. 2007;194:125-133.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  de Vries B, Matthijsen RA, van Bijnen AA, Wolfs TG, Buurman WA. Lysophosphatidic acid prevents renal ischemia-reperfusion injury by inhibition of apoptosis and complement activation. Am J Pathol. 2003;163:47-56.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Valen G. Signal transduction through nuclear factor kappa B in ischemia-reperfusion and heart failure. Basic Res Cardiol. 2004;99:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Goetzl EJ, Graeler M, Huang MC, Shankar G. Lysophospholipid growth factors and their G protein-coupled receptors in immunity, coronary artery disease, and cancer. Sci World J. 2002;2:324-338.  [PubMed]  [DOI]  [Cited in This Article: ]