Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Molecules ; 27(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36080255

ABSTRACT

The ATX-LPA-LPAR1 signaling pathway plays a universal role in stimulating diverse cellular responses, including cell proliferation, migration, survival, and invasion in almost every cell type. The ATX-LPAR1 axis is linked to several metabolic and inflammatory diseases including cancer, fibrosis, and rheumatoid arthritis. Numerous selective ATX or LPAR1 inhibitors have been developed and so far, their clinical efficacy has only been evaluated in idiopathic pulmonary fibrosis. None of the ATX and LPAR1 inhibitors have advanced to clinical trials for cancer and rheumatoid arthritis. Nonetheless, several research groups, including ours, have shown considerable benefit of simultaneous ATX and LPAR1 inhibition through combination therapy. Recent research suggests that dual-targeting therapies are superior to combination therapies that use two selective inhibitors. However, limited reports are available on ATX-LPAR1 dual inhibitors, potentially due to co-expression of multiple different LPARs with close structural similarities at the same target. In this review, we discuss rational design and future directions of dual ATX-LPAR1 inhibitors.


Subject(s)
Arthritis, Rheumatoid , Idiopathic Pulmonary Fibrosis , Neoplasms , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism
2.
Article in English | MEDLINE | ID: mdl-26005700

ABSTRACT

The role of the lysophospholipase D autotaxin (ATX) and lysophosphatidic acid (LPA) in cancer is emerging and represents two key players in regulating cancer progression. In this brief review, we will discuss some of our recent findings, which highlight a central role that LPA and its receptor plays in orchestrating melanoma-stroma interactions in the establishment of lung metastases. In particular, we evaluated not only the function of LPA receptors on tumor cells but also their role on host tissues and how they can influence melanoma growth and metastasis. Using the syngeneic B16F10 murine melanoma model, we made three key observations. First, our in vitro findings demonstrate that LPA receptors, specifically LPA2 and LPA5 expressed in B16F10 cells appear to have opposing roles in cell invasion; the former seems to be responsible for the high basal invasion rate of B16F10 cells while the latter is anti-invasive upon exogenous LPA stimulation. Second, we observed a profound reduction in the incidence of pulmonary melanoma metastasis in LPA1- and LPA5-knockout (KO) mice, respectively, when compared to wild-type (WT) mice. Third, no differences in terms of subcutaneous tumor growth between LPA1KO, LPA5KO and WT mice were observed. These findings suggest that LPA receptors exert different functions in melanoma cells versus host tissues in terms of invasion and metastasis.

3.
Chem Biol ; 22(2): 206-16, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25619933

ABSTRACT

Pharmacological mitigation of injuries caused by high-dose ionizing radiation is an unsolved medical problem. A specific nonlipid agonist of the type 2 G protein coupled receptor for lysophosphatidic acid (LPA2) 2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl]benzoic acid (DBIBB) when administered with a postirradiation delay of up to 72 hr reduced mortality of C57BL/6 mice but not LPA2 knockout mice. DBIBB mitigated the gastrointestinal radiation syndrome, increased intestinal crypt survival and enterocyte proliferation, and reduced apoptosis. DBIBB enhanced DNA repair by augmenting the resolution of γ-H2AX foci, increased clonogenic survival of irradiated IEC-6 cells, attenuated the radiation-induced death of human CD34(+) hematopoietic progenitors and enhanced the survival of the granulocyte/macrophage lineage. DBIBB also increased the survival of mice suffering from the hematopoietic acute radiation syndrome after total-body irradiation. DBIBB represents a drug candidate capable of mitigating acute radiation syndrome caused by high-dose γ-radiation to the hematopoietic and gastrointestinal system.


Subject(s)
Apoptosis/drug effects , Lysophospholipids/pharmacology , Naphthalimides/pharmacology , Receptors, Lysophosphatidic Acid/agonists , Sulfonamides/pharmacology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Acute Radiation Syndrome/prevention & control , Animals , Apoptosis/radiation effects , Binding Sites , Caspase 8/metabolism , Cell Line , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Gamma Rays , Histones/metabolism , Humans , Lysophospholipids/chemistry , Lysophospholipids/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Naphthalimides/chemistry , Naphthalimides/therapeutic use , Protein Structure, Tertiary , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Sulfonamides/chemistry , Sulfonamides/therapeutic use
4.
Mol Cancer Res ; 13(1): 174-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25158955

ABSTRACT

UNLABELLED: Autotaxin (ENPP2/ATX) and lysophosphatidic acid (LPA) receptors represent two key players in regulating cancer progression. The present study sought to understand the mechanistic role of LPA G protein-coupled receptors (GPCR), not only in the tumor cells but also in stromal cells of the tumor microenvironment. B16F10 melanoma cells predominantly express LPA5 and LPA2 receptors but lack LPA1. LPA dose dependently inhibited invasion of cells across a Matrigel layer. RNAi-mediated knockdown of LPA5 relieved the inhibitory effect of LPA on invasion without affecting basal invasion. This suggests that LPA5 exerts an anti-invasive action in melanoma cells in response to LPA. In addition, both siRNA-mediated knockdown and pharmacologic inhibition of LPA2 reduced the basal rate invasion. Unexpectedly, when probing the role of this GPCR in host tissues, it was found that the incidence of melanoma-derived lung metastasis was greatly reduced in LPA5 knockout (KO) mice compared with wild-type (WT) mice. LPA1-KO but not LPA2-KO mice also showed diminished melanoma-derived lung metastasis, suggesting that host LPA1 and LPA5 receptors play critical roles in the seeding of metastasis. The decrease in tumor cell residence in the lungs of LPA1-KO and LPA5-KO animals was apparent 24 hours after injection. However, KO of LPA1, LPA2, or LPA5 did not affect the subcutaneous growth of melanoma tumors. IMPLICATIONS: These findings suggest that tumor and stromal LPA receptors, in particular LPA1 and LPA5, play different roles in invasion and the seeding of metastasis.


Subject(s)
Lung Neoplasms/genetics , Melanoma, Experimental/genetics , Receptors, Lysophosphatidic Acid/genetics , Animals , Carcinogenesis/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Phosphoric Diester Hydrolases/genetics , Signal Transduction/genetics , Tumor Microenvironment
5.
J Cancer Sci Ther ; 7(2): 34-43, 2015.
Article in English | MEDLINE | ID: mdl-26819680

ABSTRACT

OBJECTIVE: Ovarian cancer is a gynecological malignancy that has a high mortality rate in women due to metastatic progression and recurrence. miRNAs are small, endogenous, noncoding RNAs that function as tumor suppressors or oncogenes in various human cancers by selectively suppressing the expression of target genes. The objective of this study is to investigate the role of miR-203 in ovarian cancer. METHODS: miR-203 was expressed in ovarian cancer SKOV3 and OVCAR3 cells using lentiviral vector and cell proliferation, migration, invasion were examined using MTT, transwell and Matrigel assays, respectively. Tumor growth was examined using Xenograft mouse model. RESULTS: miR-203 expression was downregulated, whereas expression of its target gene Snai2 was upregulated in human ovarian serous carcinoma tissue as compared to normal ovaries. In addition, high miR-203 expression was associated with long-term survival rate of ovarian cancer patients. miR-203 overexpression inhibited cell proliferation, migration, and invasion of SKOV3 and OVCAR3 ovarian cancer cells. Furthermore, miR-203 overexpression inhibited the epithelial to mesenchymal transition (EMT) in ovarian cancer cells. Silencing Snai2 with lentiviral short hairpin (sh) RNA mimics miR-203-mediated inhibition of EMT and tumor cell invasion. Xenografts of miR-203-overexpressing ovarian cancer cells in immunodeficient mice exhibited a significantly reduced tumor growth. CONCLUSION: miR-203 functions as a tumor suppressor by down regulating Snai2 in ovarian cancer.

6.
Blood ; 124(20): 3141-50, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25277122

ABSTRACT

Autotaxin (ATX), through its lysophospholipase D activity controls physiological levels of lysophosphatidic acid (LPA) in blood. ATX is overexpressed in multiple types of cancers, and together with LPA generated during platelet activation promotes skeletal metastasis of breast cancer. However, the pathophysiological sequelae of regulated interactions between circulating LPA, ATX, and platelets remain undefined in cancer. In this study, we show that ATX is stored in α-granules of resting human platelets and released upon tumor cell-induced platelet aggregation, leading to the production of LPA. Our in vitro and in vivo experiments using human breast cancer cells that do not express ATX (MDA-MB-231 and MDA-B02) demonstrate that nontumoral ATX controls the early stage of bone colonization by tumor cells. Moreover, expression of a dominant negative integrin αvß3-Δ744 or treatment with the anti-human αvß3 monoclonal antibody LM609, completely abolished binding of ATX to tumor cells, demonstrating the requirement of a fully active integrin αvß3 in this process. The present results establish a new mechanism for platelet contribution to LPA-dependent metastasis of breast cancer cells, and demonstrate the therapeutic potential of disrupting the binding of nontumor-derived ATX with the tumor cells for the prevention of metastasis.


Subject(s)
Blood Platelets/immunology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Integrin alphaVbeta3/immunology , Phosphoric Diester Hydrolases/immunology , Animals , Blood Platelets/pathology , Bone Neoplasms/blood , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone and Bones/immunology , Bone and Bones/pathology , Breast/immunology , Breast/pathology , Breast Neoplasms/blood , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Lysophospholipids/immunology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Platelet Activation
7.
PLoS One ; 9(8): e105331, 2014.
Article in English | MEDLINE | ID: mdl-25137052

ABSTRACT

Ovarian cancer presents therapeutic challenges due to its typically late detection, aggressive metastasis, and therapeutic resistance. The transcription factor Krüppel-like factor 4 (KLF4) has been implicated in human cancers as a tumor suppressor or oncogene, although its role depends greatly on the cellular context. The role of KLF4 in ovarian cancer has not been elucidated in mechanistic detail. In this study, we investigated the role of KLF4 in ovarian cancer cells by transducing the ovarian cancer cell lines SKOV3 and OVCAR3 with a doxycycline-inducible KLF4 lentiviral vector. Overexpression of KLF4 reduced cell proliferation, migration, and invasion. The epithelial cell marker gene E-cadherin was significantly upregulated, whereas the mesenchymal cell marker genes vimentin, twist1 and snail2 (slug) were downregulated in both KLF4-expressing SKOV3 and OVCAR3 cells. KLF4 inhibited the transforming growth factor ß (TGFß)-induced epithelial to mesenchymal transition (EMT) in ovarian cancer cells. Taken together, our data demonstrate that KLF4 functions as a tumor suppressor gene in ovarian cancer cells by inhibiting TGFß-induced EMT.


Subject(s)
Doxycycline/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Kruppel-Like Transcription Factors/genetics , Cell Movement , Cell Proliferation , Female , Genetic Vectors , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lentivirus/genetics , MCF-7 Cells , Ovarian Neoplasms , Transcriptional Activation , Transforming Growth Factor beta/physiology
8.
Biochim Biophys Acta ; 1831(1): 117-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127512

ABSTRACT

Due to its antiapoptotic action, derivatives of the lipid mediator lysophosphatidic acid (LPA) provide potential therapeutic utility in diseases associated with programmed cell death. Apoptosis is one of the major pathophysiological processes elicited by radiation injury to the organism. Consequently, therapeutic explorations applying compounds that mimic the antiapoptotic action of LPA have begun. Here we present a brief account of our decade-long drug discovery effort aimed at developing LPA mimics with a special focus on specific agonists of the LPA(2) receptor subtype, which was found to be highly effective in protecting cells from apoptosis. We describe new evidence that 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), a prototypic nonlipid agonist specific to the LPA(2) receptor subtype, rescues apoptotically condemned cells in vitro and in vivo from injury caused by high-dose γ-irradiation. GRI977143 shows the features of a radiomitigator because it is effective in rescuing the lives of mice from deadly levels of radiation when administered 24h after radiation exposure. Our findings suggest that by specifically activating LPA(2) receptors GRI977143 activates the ERK1/2 prosurvival pathway, effectively reduces Bax translocation to the mitochondrion, attenuates the activation of initiator and effector caspases, reduces DNA fragmentation, and inhibits PARP-1 cleavage associated with γ-irradiation-induced apoptosis. GRI977143 also inhibits bystander apoptosis elicited by soluble proapoptotic mediators produced by irradiated cells. Thus, GRI977143 can serve as a prototype scaffold for lead optimization paving the way to more potent analogs amenable for therapeutic exploration. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.


Subject(s)
Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Receptors, Lysophosphatidic Acid/metabolism , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/pathology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bystander Effect/drug effects , Bystander Effect/radiation effects , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Cytoprotection/drug effects , Cytoprotection/radiation effects , DNA Fragmentation/drug effects , DNA Fragmentation/radiation effects , Embryo, Mammalian/cytology , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/radiation effects , Gamma Rays , Lysophospholipids/chemistry , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Knockout , Organophosphorus Compounds/pharmacology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Injuries/pathology , Survival Analysis
9.
Mol Pharmacol ; 82(6): 1162-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22968304

ABSTRACT

Lysophosphatidic acid (LPA) is a highly potent endogenous lipid mediator that protects and rescues cells from programmed cell death. Earlier work identified the LPA2 G protein-coupled receptor subtype as an important molecular target of LPA mediating antiapoptotic signaling. Here we describe the results of a virtual screen using single-reference similarity searching that yielded compounds 2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (NSC12404), 2-((3-(1,3-dioxo-1H-benzo[de]isoquinolin-2(3H)-yl)propyl)thio)benzoic acid (GRI977143), 4,5-dichloro-2-((9-oxo-9H-fluoren-2-yl)carbamoyl)benzoic acid (H2L5547924), and 2-((9,10-dioxo-9,10-dihydroanthracen-2-yl)carbamoyl) benzoic acid (H2L5828102), novel nonlipid and drug-like compounds that are specific for the LPA2 receptor subtype. We characterized the antiapoptotic action of one of these compounds, GRI977143, which was effective in reducing activation of caspases 3, 7, 8, and 9 and inhibited poly(ADP-ribose)polymerase 1 cleavage and DNA fragmentation in different extrinsic and intrinsic models of apoptosis in vitro. Furthermore, GRI977143 promoted carcinoma cell invasion of human umbilical vein endothelial cell monolayers and fibroblast proliferation. The antiapoptotic cellular signaling responses were present selectively in mouse embryonic fibroblast cells derived from LPA(1&2) double-knockout mice reconstituted with the LPA2 receptor and were absent in vector-transduced control cells. GRI977143 was an effective stimulator of extracellular signal-regulated kinase 1/2 activation and promoted the assembly of a macromolecular signaling complex consisting of LPA2, Na⁺ - H⁺ exchange regulatory factor 2, and thyroid receptor interacting protein 6, which has been shown previously to be a required step in LPA-induced antiapoptotic signaling. The present findings indicate that nonlipid LPA2-specific agonists represent an excellent starting point for development of lead compounds with potential therapeutic utility for preventing the programmed cell death involved in many types of degenerative and inflammatory diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing/metabolism , Animals , CHO Cells , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , DNA Fragmentation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , LIM Domain Proteins/metabolism , Lysophospholipids/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Neoplasm Invasiveness , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex , Rats , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Sodium-Hydrogen Exchangers/metabolism , Transcription Factors/metabolism , bcl-2-Associated X Protein/metabolism
10.
Biochem Soc Trans ; 40(1): 31-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22260662

ABSTRACT

LPA (lysophosphatidic acid, 1-acyl-2-hydroxy-sn-glycero-3-phosphate), is a growth factor-like lipid mediator that regulates many cellular functions, many of which are unique to malignantly transformed cells. The simple chemical structure of LPA and its profound effects in cancer cells has attracted the attention of the cancer therapeutics field and drives the development of therapeutics based on the LPA scaffold. In biological fluids, LPA is generated by ATX (autotaxin), a lysophospholipase D that cleaves the choline/serine headgroup from lysophosphatidylcholine and lysophosphatidylserine to generate LPA. In the present article, we review some of the key findings that make the ATX-LPA signalling axis an emerging target for cancer therapy.


Subject(s)
Neoplasms/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Lysophospholipids/metabolism , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasms/drug therapy , Neoplasms/pathology , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases/genetics , Signal Transduction , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...