Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Bioenerg Biomembr ; 56(4): 475-482, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38886303

ABSTRACT

Lysophosphatidic acid (LPA) is a simple lipid which is endogenously synthesized from lysophosphatidylcholine (LPC) by autotaxin (ATX). LPA mediates a variety of cellular responses through the binding of G protein-coupled LPA receptors (LPA1 to LPA6). It is considered that LPA receptor-mediated signaling plays an important role in the pathogenesis of human malignancy. Genetic alterations and epigenetic changes of LPA receptors have been detected in some cancer cells as well as LPA per se. Moreover, LPA receptors contribute to the promotion of tumor progression, including cell proliferation, invasion, metastasis, tumorigenicity, and angiogenesis. In recent studies, the activation of LPA receptor-mediated signaling regulates chemoresistance and radiosensitivity in cancer cells. This review provides an updated overview on the roles of LPA receptor-mediated signaling in the regulation of cancer cell functions and its potential utility as a molecular target for novel therapies in clinical cancer approaches.


Subject(s)
Neoplasms , Receptors, Lysophosphatidic Acid , Signal Transduction , Humans , Receptors, Lysophosphatidic Acid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Lysophospholipids/metabolism , Animals
2.
Biochem Biophys Res Commun ; 727: 150322, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38945064

ABSTRACT

The tumor microenvironment (TME) comprises cancer and non-cancerous stromal cells, including fibroblasts. Free fatty acids (FFAs) regulate various biological responses by binding to G protein-coupled FFA receptors (FFARs). In this study, we examined the impact of FFAR1 and FFAR4 on the cell migration of pancreatic cancer PANC-1 cells co-cultured with 3T3 fibroblast cells under hypoxic conditions. PANC-1 cells cultured at 1 % O2 exhibited elevated FFAR1 expression and decreased FFAR4 expression compared to those at 21 % O2. Cell migration of PANC-1 cells was reduced under 1 % O2 conditions. FFAR1 knockdown enhanced PANC-1 cell migration, whereas FFAR4 knockdown inhibited it. Co-culture of PANC-1 cells with 3T3 cells at 1 % O2 significantly increased FFAR4 expression, while FFAR1 expression remained unchanged. To evaluate the effects of FFAR1 and FFAR4 on PANC-1 cell migration in co-culture with 3T3 cells, we conducted a wound healing assay using the Culture-Insert 2 Well. PANC-1 and 3T3 cells were individually seeded into the two wells and incubated at both 21 % and 1 % O2 for 13 h. The cell migration of PANC-1 cells co-cultured with 3T3 cells at 1 % O2 was notably higher compared to 21 % O2. TUG-770 reduced and TUG-891 enhanced the cell migration of PANC-1 cells co-cultured with 3T3 cells under both 21 % and 1 % O2 conditions. These findings suggest that FFAR1 and FFAR4 play important roles in regulating the cell migration of PANC-1 cells co-cultured with 3T3 cells under hypoxic conditions.

3.
Pathol Res Pract ; 260: 155385, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38875757

ABSTRACT

BACKGROUND: Tamoxifen (TAM) is a selective estrogen receptor modulator and has anti-estrogenic activity. Breast cancer cells acquire drug resistance to TAM as a consequence of long-term treatment. Lysophosphatidic acid (LPA) receptor-mediated signaling contributes to the promotion of tumor progression. This study aimed to evaluate the role of LPA receptors in the modulation of biological functions by long-term TAM treatment in breast cancer MCF-7 cells under hypoxic and estrogen-deprived conditions. METHODS: Long-term TAM treated (MCF-TAM) cells were generated from MCF-7 cells. Cells were cultured in estrogen-free medium at 1 % O2. LPA receptor expressions were measured by quantitative real-time RT-PCR analysis. Cell motile activity was investigated using Cell Culture Inserts. The CCK-8 kit was used to determine the cell proliferation rate. RESULTS: LPAR1 and LPAR3 expressions were elevated in MCF-TAM cells. MCF-TAM cell motility was enhanced by culturing at 1 % O2, compared with MCF-7 cells. When cells were cultured in estrogen-deprived medium at 1 % O2, the cell proliferation rate of MCF-TAM cells was significantly higher than that of MCF-7 cells. CONCLUSION: These results suggest that LPA receptor-mediated signaling plays an important role in the acquisition of malignant properties in long-term TAM treated MCF-7 cells under hypoxic and estrogen-deprived conditions.

4.
Biochem Biophys Res Commun ; 715: 149982, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38676998

ABSTRACT

The tumor microenvironment is an extremely complex composed of cancer cells and various non-cancer cells, including lymphatic endothelial cells. Lysophosphatidic acid (LPA) receptors (LPA1 to LPA6) activate a variety of malignant properties in human malignancies. In the present study, we examined the roles of LPA receptor-mediated signaling in biological responses of lymphatic endothelial SVEC4-10 cells induced by hypoxia. Lpar1, Lpar2 and Lpar3 expressions were decreased in SVEC4-10 cells cultured at hypoxic conditions (1 % O2). LPA had no impact on the cell growth activity of SVEC4-10 cells in 21 % O2 culture conditions. Conversely, the cell growth activity of SVEC4-10 cells in 1 % O2 culture conditions was reduced by LPA. The cell motile activity of SVEC4-10 cells was elevated by 1 % O2 culture conditions. GRI-977143 (LPA2 agonist) and (2S)-OMPT (LPA3 agonist) stimulated SVEC4-10 cell motility as well as AM966 (LPA1 antagonist). In tube formation assay, the tube formation of SVEC4-10 cells in 1 % O2 culture conditions was markedly increased, in comparison with 21 % O2. GRI-977143 and (2S)-OMPT elevated the tube formation of SVEC4-10 cells. Furthermore, the tube formation of SVEC4-10 cells was increased by AM966. These results suggest that LPA receptor-mediated signaling contributes to the modulation of hypoxic-induced biological functions of lymphatic endothelial cells.


Subject(s)
Cell Hypoxia , Cell Movement , Endothelial Cells , Lysophospholipids , Receptors, Lysophosphatidic Acid , Animals , Humans , Mice , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Lysophospholipids/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism
5.
Pathol Res Pract ; 257: 155293, 2024 May.
Article in English | MEDLINE | ID: mdl-38615508

ABSTRACT

Lysophosphatidic acid (LPA) binds to its specific G protein-coupled LPA receptors (LPA1 to LPA6), resulting in the activation of various cellular functions. LPA receptor-mediated signaling facilitates tumor progression in human malignancies. In the present study, we investigated whether LPA receptor-mediated signaling contributes to cellular responses to X-ray irradiation in osteosarcoma MG-63 cells. After X-ray irradiation (2, 4 and 8 Gy), LPAR2 and LPAR3 expression levels in MG-63 cells were significantly elevated in a dose-dependent manner, but no change of LPAR1 expression level was observed. The cell growth activities of MG-63 cells irradiated with X-rays (2, 4 and 8 Gy) were reduced by LPA. Conversely, LPA3 agonist (2 S)-OMPT enhanced the cell growth activities of X-ray irradiated MG-63 cells. The cell movement of MG-63 cells exposed to X-ray irradiation (8 Gy) was inhibited by (2 S)OMPT. In cell survival assay, (2 S)-OMPT suppressed the cell survival to cisplatin (CDDP) of MG-63 cells irradiated with X-rays (8 Gy). The cell survival to CDDP of X-ray irradiated cells was elevated by LPA3 knockdown. Moreover, we evaluated the effects of LPA2 on the cell survival to CDDP of MG-63 cells exposed to X-ray irradiation (8 Gy). The cell survival to CDDP of X-ray irradiated cells was increased by LPA2 agonist GRI-977143 and reduced by LPA2 knockdown. These results suggest that LPA receptor-signaling participates in the modulation of cellular functions induced by X-ray irradiation in osteosarcoma cells.


Subject(s)
Bone Neoplasms , Osteosarcoma , Receptors, Lysophosphatidic Acid , Humans , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Lysophospholipids/pharmacology , Lysophospholipids/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/radiotherapy , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/radiation effects , Signal Transduction/drug effects , Signal Transduction/radiation effects , X-Rays
6.
Pathol Res Pract ; 255: 155192, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38367602

ABSTRACT

BACKGROUND: In the tumor environment, malignant characteristics of cancer cells are promoted by stromal cells under hypoxia. It is unknown whether lysophosphatidic acid (LPA) receptor-mediated signaling is involved in the regulation of cellular functions by endothelial cells in pancreatic cancer cells under hypoxic conditions. METHODS: Pancreatic cancer (PANC-1) cells were co-cultured with endothelial (F2) cells and F2 cell supernatants at 21% and 1% O2. The Cell Culture Insert was used to assess the cell motile activity. The cell growth and viability to cisplatin (CDDP) were measured, using the Cell Counting Kit-8. RESULTS: LPA receptor expression levels were changed in PANC-1 cells co-cultured with F2 cells at 21% and 1% O2. The cell motile activities of PANC-1 cells co-cultured with F2 cells at 21% and 1% O2 were markedly elevated, compared with PANC-1 cells alone. The cell viabilities to CDDP of PANC-1 cells co-cultured with F2 cell supernatants at 21% and 1% O2 were regulated by the activation of LPA receptors. CONCLUSION: These results suggest that LPA receptor-mediated signaling plays an important role in the modulation of pancreatic cancer cell functions by endothelial cells under hypoxic conditions.


Subject(s)
Endothelial Cells , Lysophospholipids , Pancreatic Neoplasms , Humans , Endothelial Cells/pathology , Receptors, Lysophosphatidic Acid/metabolism , Cell Movement , Cisplatin/pharmacology , Pancreatic Neoplasms/pathology , Hypoxia/metabolism
7.
Adv Biol Regul ; 92: 101029, 2024 05.
Article in English | MEDLINE | ID: mdl-38377635

ABSTRACT

Lysophosphatidic acid (LPA) is a simple physiological lipid and structurally consists of a fatty, a phosphate and a glycerol. LPA binds to G protein-coupled LPA receptors (LPA1 to LPA6). LPA receptor-mediated signaling mediates a variety of biological responses, such as cell growth, migration, morphogenesis, differentiation and protection from apoptosis. It is considered that LPA receptor-mediated signaling plays an important role in the pathogenesis of human malignancies. So far, genetic and epigenetic alterations of LPA receptors have been found in several cancer cells as well as abnormal LPA production. In addition, LPA receptor-mediated signaling regulates the promotion of malignant behaviors, including chemo- and/or radiation-resistance. Chemotherapy and radiotherapy are the common approaches to the treatments of cancers. However, resistance to anticancer drugs and irradiation is the most critical limitation for chemotherapy and radiotherapy. In this review, we provide the roles of LPA receptor-mediated signaling in the regulation of cellular responses induced by chemotherapeutic agents and irradiation and its biological utility as a possible molecular target for improving cancer cell responses to chemotherapy and radiotherapy.


Subject(s)
Antineoplastic Agents , Lysophospholipids , Neoplasms , Receptors, Lysophosphatidic Acid , Signal Transduction , Humans , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lysophospholipids/metabolism , Animals
8.
Biochem Biophys Res Commun ; 699: 149554, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38280308

ABSTRACT

In the tumor environment, hypoxia promotes tumor progression, such as cancer cell growth, migration and chemoresistance. This study aimed to evaluate the roles of free fatty acid receptors (FFARs) in the regulation of cancer cell functions under hypoxic conditions, using fibrosarcoma HT1080 cells. HT1080 cells expressed FFAR1, FFAR2 and FFAR3 genes, but not FFAR4 gene. FFAR1, FFAR2 and FFAR3 expression levels in HT1080 cells cultured at 1 % O2 were elevated, compared with 21 % O2. The cell growth activities of HT1080 cells cultured at 21 % O2 were inhibited by acetic acid (AA) and propanoic acid (PA), but not 1 % O2. HT1080 cell motility was markedly reduced by culturing at 1 % O2. The cell growth and motility of HT1080 cells were enhanced by FFAR2 knockdown. The cell viability to cisplatin (CDDP) of HT1080 cells cultured at 1 % O2 was increased, compared with 21 % O2. FFAR2 knockdown suppressed the cell viability to CDDP of HT1080 cells. On the other hand, the cell motility and viability to CDDP of HT1080 cells cultured at 21 % O2 were suppressed by TUG-770. When HT1080 cells were cultured at 1 % O2, the cell motility and viability to CDDP were decreased, correlating with FFAR1 expression level. Moreover, FFAR1 knockdown increased the cell viability to CDDP of HT1080 cells cultured at 1 % O2. These results suggest that FFAR-mediated signaling plays an important role in the modulation of cellular functions of HT1080 cells under hypoxic conditions.


Subject(s)
Fatty Acids, Nonesterified , Fibrosarcoma , Humans , Receptors, G-Protein-Coupled/metabolism , Cisplatin/pharmacology , Signal Transduction , Fibrosarcoma/metabolism , Cell Movement
9.
Adv Biol Regul ; 89: 100978, 2023 08.
Article in English | MEDLINE | ID: mdl-37603941

ABSTRACT

Hydrogen peroxide (H2O2) is one of reactive oxygen species (ROS) and promotes malignant properties of cancer cells. Lysophosphatidic acid (LPA) signaling via LPA receptor (LPA1 to LPA6) regulates a variety of cellular functions, such as cell growth, migration and differentiation. This study aimed to evaluate the effects of LPA receptors on the cell motility and survival to anticancer drugs by H2O2 in colon cancer DLD-1 cells. To obtain H2O2 treated (DLD- H2O2) cells, cells were maintained in culture medium containing H2O2 (60 µM) for 2 months. LPAR2 and LPAR4 gene expressions were markedly elevated in DLD-H2O2 cells. The cell motility of DLD-H2O2 cells was significantly lower than that of DLD-1 cells. DLD-H2O2 cell motility was suppressed by LPA2 knockdown and stimulated by LPA4 knockdown. The cell survival rates to fluorouracil (5-FU), irinotecan (CPT-11) and oxaliplatin (L-OHP) of DLD-H2O2 cells were significantly higher than those of DLD-1 cells. The cell survival rate to 5-FU of DLD-H2O2 cells was decreased by LPA2 knockdown. Conversely, LPA4 knockdown enhanced the cell survival rate to 5-FU of DLD-H2O2 cells. In the tumor microenvironment, high levels of H2O2 production are observed under hypoxic conditions. The cell survival rate to 5-FU of DLD-H2O2 cells cultured at 1% O2 was significantly higher than that of DLD-1 cells cultured at 1% O2, correlating with LPAR2 gene expression. The present results suggest that the induction of LPA receptor-mediated signaling plays an important role in regulating cellular functions of DLD-1 cells treated with H2O2.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Hydrogen Peroxide/pharmacology , Receptors, Lysophosphatidic Acid/genetics , Fluorouracil , Cell Movement , Antineoplastic Agents/pharmacology
10.
Int J Radiat Biol ; 99(12): 1925-1933, 2023.
Article in English | MEDLINE | ID: mdl-37523658

ABSTRACT

PURPOSE: Lysophosphatidic acid (LPA) receptor-mediated signaling regulates various biological functions in cancer cells. This study aimed to evaluate the roles of LPA receptor-2 (LPA2) in cellular responses induced by X-ray irradiation in pancreatic cancer PANC-1 cells. Since X-ray irradiation generates reactive oxygen species (ROS), PANC-1 cells were treated with hydrogen peroxide (H2O2). H2O2 is a key member of ROS. METHODS: To investigate the cell survival rate to X-ray irradiation, PANC-1 cells were irradiated with X-rays (2.5-15 Gy). LPAR2 expression levels were measured by quantitative real-time RT-PCR analysis. The effects of LPA2 on the cell survival and motility were evaluated using LPA2 knockdown cells. To establish H2O2 treated cells, PANC-1 cells were cultured in 10% FBS-DMEM with H2O2 (30 µM) for 2 weeks. The cell motility and survival rate to cisplatin (CDDP) of H2O2 treated cells were examined. RESULTS: LPAR2 expression was significantly increased in PANC-1 cells irradiated with X-rays. PANC-1 cell motility was markedly decreased by X-ray irradiation. The reduced cell motility activity by X-ray irradiation was enhanced by LPA2 knockdown. The cell survival to X-ray irradiation was elevated in PANC-1 cells treated with GRI-977143 (LPA2 agonist) and suppressed by LPA2 knockdown. On the other hand, LPAR2 expression was markedly higher in H2O2 treated cells than in H2O2 untreated cells. H2O2 treated cells showed the high cell survival to CDDP in comparison with H2O2 untreated cells. GRI-977143 increased the cell survival to CDDP of H2O2 treated cells, while LPA2 knockdown suppressed. CONCLUSIONS: The present results suggest that the activation of LPA2-mediated signaling plays an important role in the modulation of cellular functions induced by X-ray irradiation and H2O2 in PANC-1 cells.


Subject(s)
Hydrogen Peroxide , Pancreatic Neoplasms , Humans , Hydrogen Peroxide/pharmacology , X-Rays , Reactive Oxygen Species , Cell Movement , Cisplatin/pharmacology , Pancreatic Neoplasms/radiotherapy , Pancreatic Neoplasms
11.
J Bioenerg Biomembr ; 55(3): 169-177, 2023 06.
Article in English | MEDLINE | ID: mdl-37219648

ABSTRACT

The tumor microenvironment (TME) consists of various cell types, including fibroblasts. The TME plays a central role in the promotion of tumor progression. In the present study, we investigated whether lysophosphatidic acid (LPA) receptor-mediated signaling regulates cellular functions by the TME of pancreatic cancer PANC-1 cells. To obtain fibroblast 3T3 cell supernatants, 3T3 cells were cultured in 5% charcoal stripped FCS-DMEM for 48 h. LPAR2 and LPAR3 expression levels were elevated in PANC-1 cells cultured in 3T3 cell supernatants. While PANC-1 cell motility was decreased by 3T3 cell supernatants, the cell survival to cisplatin (CDDP) of PANC-1 cells was markedly enhanced. Moreover, the cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants was increased by GRI-977,143 (LPA2 agonist) and (2 S)-OMPT (LPA3 agonist). Since hypoxia is caused by the restriction of adequate vascular networks to deliver oxygen into solid tumors, PANC-1 cells were cultured in 3T3 cell supernatants at 1% O2 conditions. The cell survival to CDDP of PANC-1 cells cultured in 3T3 cell supernatants at 1% O2 was significantly elevated, correlating with LPAR2 and LPAR3 expressions. These results suggest that LPA signaling via LPA2 and LPA3 is involved in the promotion of malignant properties by the TME in PANC-1 cells.


Subject(s)
Pancreatic Neoplasms , Receptors, Lysophosphatidic Acid , Mice , Animals , Humans , Receptors, Lysophosphatidic Acid/metabolism , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Cisplatin/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Cell Movement , Hypoxia/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
12.
Biochem Biophys Res Commun ; 661: 21-27, 2023 06 18.
Article in English | MEDLINE | ID: mdl-37084489

ABSTRACT

In tumor microenvironment, cancer cells can adapt to low conditions of nutrients and oxygen. Lysophosphatidic acid (LPA) receptor-mediated signaling is involved in the promotion of malignant properties in cancer cells. In the present study, to examine the roles of LPA receptors in the regulation of cell motility and survival to cisplatin (CDDP) of pancreatic cancer PANC-1 cells under glucose-deprived and hypoxic conditions, cells were cultured in 4500 mg/L high glucose (HG)-DMEM, 500 mg/L middle glucose (MG)-DMEM and 100 mg/L low glucose (LG)-DMEM at 21% and 1% O2. The expression levels of LPAR1 and LPAR2 genes in cells cultured in MG-DMEM and LG-DMEM were significantly elevated, compared with HG-DMEM cells. The cell motility and survival rate to CDDP of cells cultured in MG-DMEM and LG-DMEM were significantly lower than those of cells cultured in HG-DMEM. The cell survival to CDDP was enhanced by LPA1 knockdown and suppressed by LPA2 knockdown. Under hypoxic conditions (1% O2), LPAR1, LPAR2 and LPAR3 expressions were markedly higher in cells cultured in MG-DMEM and LG-DMEM than in cells cultured in HG-DMEM. The cell survival rates to CDDP of cells cultured in MG-DMEM and LG-DMEM were elevated in comparison with HG-DMEM. The cell survival to CDDP was reduced by LPA3 knockdown. These results suggest that LPA receptor-mediated signaling is involved in the regulation of malignant properties of PANC-1 cells under glucose-deprived and hypoxic conditions.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms , Humans , Receptors, Lysophosphatidic Acid/metabolism , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Pancreatic Neoplasms/pathology , Cell Movement , Lysophospholipids/pharmacology , Tumor Microenvironment , Pancreatic Neoplasms
13.
Biochem Biophys Res Commun ; 646: 56-62, 2023 02 26.
Article in English | MEDLINE | ID: mdl-36706706

ABSTRACT

G protein coupled free fatty acid receptors (FFARs) are involved in the pathogenesis of several human diseases. FFAR2 and FFAR3 are activated by the binding of short-chain fatty acids (SCFAs). This study aimed to evaluate the roles of FFAR2 in the regulation of cellular functions in osteosarcoma HOS cells, using acetic acid and propanoic acid as FFAR2 and FFAR3 agonists. FFAR2 and FFAR3 genes were expressed in HOS cells. The cell motile activity of HOS cells was significantly stimulated by acetic acid and propanoic acid. In contrast, acetic acid and propanoic acid had no impact on the activation of matrix metalloproteinase-2 (MMP-2) and MMP-9. In cell survival assay, the cell survival rate to cisplatin (CDDP) of HOS cells was elevated by acetic acid and propanoic acid. To assess the effects of FFAR2 on cellular functions, FFAR2 knockdown (HOS-FFAR2) cells were generated from HOS cells. The cell motile activity of HOS-FFAR2 cells was enhanced by acetic acid and propanoic acid. In the presence of acetic acid and propanoic acid, MMP-2 and MMP-9 activities were reduced in HOS-FFAR2 cells, compared with control cells. When cells were treated with acetic acid and propanoic acid, the cell survival rate to CDDP of HOS-FFAR2 cells was significantly lower than that of control cells. These results suggest that activation of FFAR2-mediated signaling is involved in the modulation of cellular functions in HOS cells.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Propionates/pharmacology , Matrix Metalloproteinase 2/genetics , Receptors, G-Protein-Coupled/metabolism , Fatty Acids, Nonesterified , Matrix Metalloproteinase 9/genetics , Fatty Acids, Volatile/pharmacology , Osteosarcoma/genetics , Cisplatin/pharmacology , Acetic Acid , Bone Neoplasms/genetics
14.
J Ocul Pharmacol Ther ; 27(4): 353-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21574866

ABSTRACT

PURPOSE: Glucocorticoids exert their actions via the glucocorticoid receptor through at least 2 intracellular mechanisms, known as transrepression and transactivation. It has been hypothesized that transrepression is the basis of their anti-inflammatory effects, whereas transactivation has been assumed to cause their side effects. ZK209614, a recently identified, novel selective glucocorticoid receptor agonist, exerts strong transrepression and weak transactivation. The objective of this study was to determine whether its pharmacological effects can be dissociated from its side effects. For this, we employed in vitro assays and topical in vivo models. METHODS: ZK209614 and dexamethasone were used in in vitro transrepression and transactivation assays. To evaluate anti-inflammatory and antiallergic activities in vivo, ZK209614 and betamethasone phosphate were tested in the carrageenan-induced conjunctivitis model and allergic conjunctivitis model in rats. To evaluate side effects in vivo, treatments with ZK209614 and betamethasone phosphate were tested for the ocular hypertensive effects in a feline model, each drug being administered topically. RESULTS: ZK209614 showed strong transrepression and weak transactivation in the in vitro assays. When given as eyedrops, ZK209614 and betamethasone phosphate each had an inhibitory effect on edema weight in the rat carrageenan-induced conjunctivitis model. In the rat allergic conjunctivitis model, ZK209614 reduced the elevated vascular permeability at a concentration of 0.1%. In the feline intraocular pressure (IOP)-elevation experiment, topically administered betamethasone phosphate elevated IOP, but ZK209614 had no effect on IOP. CONCLUSION: The present investigations suggest that ZK209614 eyedrops have both anti-inflammatory and antiallergic effects, but no unwanted IOP-elevating effect. On that basis, ZK209614 might be a promising candidate as an ophthalmic drug with a better therapeutic index than classic glucocorticoids.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzofurans/pharmacology , Benzoxazines/pharmacology , Conjunctivitis/drug therapy , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/agonists , Administration, Topical , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/toxicity , Benzofurans/administration & dosage , Benzofurans/toxicity , Benzoxazines/administration & dosage , Benzoxazines/toxicity , Betamethasone/administration & dosage , Betamethasone/analogs & derivatives , Betamethasone/pharmacology , Betamethasone/toxicity , Carrageenan , Cats , Cell Line, Tumor , Cells, Cultured , Conjunctivitis, Allergic/drug therapy , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dexamethasone/toxicity , Disease Models, Animal , Glucocorticoids/administration & dosage , Glucocorticoids/toxicity , Humans , Intraocular Pressure/drug effects , Male , Ophthalmic Solutions , Rats , Rats, Inbred BN , Rats, Wistar , Toxicity Tests , Transcriptional Activation/drug effects
15.
BMC Musculoskelet Disord ; 10: 23, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19228423

ABSTRACT

BACKGROUND: Knock-in mice (gp130F759) with a Y759F point mutation in gp130, a signal transducing receptor subunit shared by members of the IL-6 cytokine family, show sustained activation of STAT3, enhanced acute-phase or immune responses, and autoimmune arthritis. We conducted a detailed analysis of collagen-induced arthritis (CIA) in gp130F759 with a DBA/1J background (D/J.gp130F759). METHODS: We backcrossed gp130F759 to C57BL/6 and DBA/1J, and compared the pathologic changes, including occurrence of arthritis, in the two distinct genetic backgrounds. We analyzed CIA in D/J.gp130F759 and investigated the effects of methotrexate (MTX) on CIA. RESULTS: C57BL/6 background gp130F759 mice, but not D/J.gp130F759, spontaneously developed polyarthritis and glomerulonephritis. On the other hand, keratitis of the eyes only developed in D/J.gp130F759, indicating the influence of genetic background on disease development in gp130F759 mice. Resistance of the DBA/1J background against spontaneous arthritis urged us to examine CIA in D/J.gp130F759. CIA in D/J.gp130F759 was more severe, with greater bone destruction, than the control mice. After collagen immunization, splenomegaly and serum levels of rheumatoid factor and anti-DNA antibody were augmented in D/J.gp130F759. Bio-Plex analysis of serum cytokines revealed increased IL-12p40 and PDGF-BB before immunization, and increased levels of IFN-gamma, IL-17, TNF-alpha, IL-9, and MIP-1beta 8 days after the booster dose. IL-6 and PDGF-BB in D/J.gp130F759 showed distinct kinetics from the other cytokines; higher levels were observed after arthritis development. MTX partially attenuated the development of arthritis and inhibited bone destruction in D/J.gp130F759, with reduction of anti-type II collagen antibody levels, suggesting that MTX mainly affects antigen-specific immune responses in CIA. CONCLUSION: The Tyr-759 point mutation of the IL-6 family cytokine receptor subunit, gp130, caused autoimmune disease, and this was also influenced by the genetic background. CIA in D/J.gp130F759 is useful for evaluating drugs in a relatively short period because sustained activation of STAT3 may enhance the disease symptoms.


Subject(s)
Arthritis, Experimental/genetics , Cytokine Receptor gp130/genetics , Genetic Predisposition to Disease , Point Mutation , Animals , Antirheumatic Agents/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Hyperplasia/genetics , Hyperplasia/pathology , Joints/pathology , Lymphoid Tissue/pathology , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Organ Size , Species Specificity , Spleen/pathology , Stifle/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...