Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
Chem Biol Interact ; 390: 110871, 2024 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-38228243

RESUMO

The phenolic antioxidant 3,5-dihydroxy-4-methoxybenzyl alcohol (DHMBA), found in the Pacific oyster Crassostrea gigas, is a superior peroxyl radical scavenger compared to other materials, including Trolox. DHMBA may play an important role in the prevention of health disorders. This study elucidates whether DHMBA prevents the impairment of mineralization of mouse osteoblastic MC3T3-E1 cells under inflammatory conditions by using mouse macrophage RAW264.7 cells in vitro. Culturing with DHMBA (1-100 µM) did not affect the proliferation and death of MC3T3-E1 cells. DHMBA stimulated osteoblastic mineralization. DHMBA blocked the decrease in mineralization of MC3T3-E1 cells caused by culture with the inflammatory cytokine TNF-α. DHMBA inhibited the production of TNF-α by stimulation with lipopolysaccharide (LPS) in RAW264.7 cells. The growth of MC3T3-E1 cells was suppressed by coculture with macrophages under LPS stimulation through the crosstalk of both cells. Interestingly, the growth of MC3T3-E1 cells was suppressed by culturing with the conditioned medium obtained by culturing macrophages with LPS. The effect of the conditioned medium was blocked by the presence of DHMBA or Bay 11-7082, an inhibitor of the TNF-α pathway. The blocking effect of DHMBA was not further enhanced in the presence of Bay 11-7082. Mechanistically, DHMBA was found to decrease the levels of NF-κB p65 and the activity of NF-κB reporter expression in MC3T3-E1 cells. DHMBA was shown to prevent the impairment of osteoblastic mineralization via TNF-α signaling involved in macrophage activation in the bone marrow microenvironment. This study may provide a novel strategy for the therapy of osteoblastic impairment.


Assuntos
Álcoois Benzílicos , NF-kappa B , Nitrilas , Sulfonas , Fator de Necrose Tumoral alfa , Camundongos , Animais , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/metabolismo , Ativação de Macrófagos , Meios de Cultivo Condicionados/farmacologia , Osteoblastos/metabolismo , Diferenciação Celular
2.
Life Sci ; 340: 122448, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38246519

RESUMO

Regucalcin is a unique calcium-binding protein first discovered in rat liver in 1978. Regucalcin has multiple functions as an inhibitor of various cellular signaling pathways that regulate cell activity. The expression of the regucalcin gene can be altered by various physiological and pathological factors such as diet (nutrients), hormones, diabetes, alcohol and drugs. Several transcription factors have been identified on the regucalcin gene, including AP-1, NF1-A1, RGPR-p117, ß-catenin, NF-κB, STAT3 and hypoxia-inducible factor-1α (HIF-1α). Notably, regucalcin plays an important role in the development of several cancers by controlling cell growth. Clinically, many studies have reported that the expression of the regucalcin gene is downregulated in various human cancers. In addition, higher expression of regucalcin in tumor tissue has been associated with longer patient survival, suggesting that regucalcin may act as a potential suppressor of various types of human cancer. Regucalcin may offer a novel therapeutic strategy and diagnostic tool for cancer treatment. However, the underlying mechanism by which regucalcin expression is reduced in human cancer is still unclear. A deeper understanding of regucalcin reduction and function in cancer is needed to discover potential resistance mechanisms and biomarkers, and to improve regucalcin-targeting agents. We review recent findings on regucalcin gene expression in cancer. We discuss the possible mechanisms by which regucalcin expression is downregulated in cancer cells to facilitate understanding of how regucalcin regulates cell growth function. This mini-review may lead to better therapeutic targets with regucalcin.


Assuntos
Neoplasias , Transdução de Sinais , Ratos , Animais , Humanos , Regulação para Baixo , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Neoplasias/genética , Regiões Promotoras Genéticas , Proteínas de Ligação ao Cálcio/metabolismo
3.
Cancers (Basel) ; 15(22)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38001749

RESUMO

Regucalcin, a calcium-binding protein lacking the EF-hand motif, was initially discovered in 1978. Its name is indicative of its function in calcium signaling regulation. The rgn gene encodes for regucalcin and is situated on the X chromosome in both humans and vertebrates. Regucalcin regulates pivotal enzymes involved in signal transduction and has an inhibitory function, which includes protein kinases, protein phosphatases, cysteinyl protease, nitric oxide dynthetase, aminoacyl-transfer ribonucleic acid (tRNA) synthetase, and protein synthesis. This cytoplasmic protein is transported to the nucleus where it regulates deoxyribonucleic acid and RNA synthesis as well as gene expression. Overexpression of regucalcin inhibits proliferation in both normal and cancer cells in vitro, independent of apoptosis. During liver regeneration in vivo, endogenous regucalcin suppresses cell growth when overexpressed. Regucalcin mRNA and protein expressions are significantly downregulated in tumor tissues of patients with various types of cancers. Patients exhibiting upregulated regucalcin in tumor tissue have shown prolonged survival. The decrease of regucalcin expression is linked to the advancement of cancer. Overexpression of regucalcin carries the potential for preventing and treating carcinogenesis. Additionally, extracellular regucalcin has displayed control over various types of human cancer cells. Regucalcin may hold a prominent role as a regulatory factor in cancer development. Supplying the regucalcin gene could prove to be a valuable asset in cancer treatment. The therapeutic value of regucalcin suggests its potential significance in treating cancer patients. This review delves into the most recent research on the regulatory role of regucalcin in human cancer development, providing a novel approach for treatment.

4.
Curr Mol Med ; 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37365791

RESUMO

BACKGROUND AND OBJECTIVE: The novel marine factor 3,5-dihydroxy-4-methoxybenzyl alcohol (DHMBA) was originally identified in the Pacific oyster Crassostrea Gigas. DHMBA has been shown to prevent oxidative stress by scavenging radicals and enhance the production of antioxidant proteins. However, the pharmacologic role of DHMBA has been poorly understood. Inflammation is implicated in the pathogenesis of many diseases. Inflammatory cytokines are produced in macrophages with stimulation of lipopolysaccharide (LPS) and are used as biomarkers that cause diverse disease conditions. Therefore, this study has been undertaken to elucidate whether DHMBA expresses anti-inflammatory effects in in vitro mouse macrophage RAW264.7 cells. METHODS: Mouse macrophage RAW264.7 cells were cultured in a medium containing 10% fetal bovine serum (FBS) with or without DHMBA (1-1000 µM). RESULTS: Culturing with DHMBA (1-1000 µM) suppressed the growth and stimulated the death of RAW264.7 cells in vitro, leading to a decrease in cell number. Treatment with DHMBA reduced the levels of Ras, PI3K, Akt, MAPK, phospho-MAPK, and mTOR, which are signalling factors to promote cell proliferation, and it raised the levels of p53, p21, Rb, and regucalcin, which are cell growth suppressors. DHMBA treatment elevated caspase-3 and cleaved caspase-3 levels. Interestingly, DHMBA treatment repressed the production of inflammatory cytokines, including tumor necrosis factor-α, interleukin-6, interleukin-1ß, or prostaglandin E2, which were enhanced by LPS stimulation. Notably, the levels of NF-κB p65 increased by LPS treatment, and this augmentation was repressed by DHMBA treatment. Moreover, LPS treatment stimulated osteoclastogenesis of RAW264.7 cells. This stimulation was blocked by DHMBA treatment, and this effect was not caused by the presence of an NF-κB signalling inhibitor. CONCLUSION: DHMBA was found to potentially suppress the activity of inflammatory macrophages in vitro, suggesting its therapeutic usefulness in inflammatory conditions.

5.
Cell Signal ; 107: 110663, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37001596

RESUMO

Macrophages in the cancer microenvironments may play a regulatory role in the progression and metastasis of prostate cancer cells. However, the crosstalk between macrophages and prostate cancer cells is poorly understood. This study elucidates whether inflammatory macrophages regulate the proliferation and death of human prostate cancer cells in vitro. The RAW264.7 mouse macrophages were cocultured with PC-3 or DU-145 wild-type cells by using a Transwell chamber in vitro. RAW264.7 cells were cocultured with PC-3 or DU-145 cells in the presence of lipopolysaccharide (LPS). This coculturing blocked the proliferation and accelerated the death of cancer cells. Interestingly, cancer cell proliferation was repressed and death was promoted by the addition of the conditioned medium obtained from RAW264.7 cells treated with LPS. Culturing with LPS mostly augmented the production of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the culture medium of RAW264.7 cells. The effects of the conditioned medium on the proliferation and death of PC-3 or DU-145 cells were blocked by NF-κB or STAT3 signaling inhibitors. Moreover, the effects of the conditioned medium on the proliferation and death of prostate cancer cells were not expressed in regucalcin-overexpressing cancer cells that diminish the levels of NF-κB p65 and STAT3. Culturing with extracellular TNF-α, IL-6, or regucalcin triggered inhibition of the proliferation of PC-3 wild-type cells. The levels of regucalcin in PC-3 cells were elevated by TNF-α or IL-6 stimulation. This study demonstrates that inflammatory macrophages triggered the loss of prostate cancer cells via the signaling process of NF-κB, STAT3, or regucalcin.


Assuntos
Neoplasias da Próstata , Fator de Necrose Tumoral alfa , Camundongos , Masculino , Animais , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Meios de Cultivo Condicionados/farmacologia , Lipopolissacarídeos/farmacologia , Transdução de Sinais , Macrófagos/metabolismo , Microambiente Tumoral
6.
Life Sci ; 314: 121328, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36584916

RESUMO

AIMS: Regucalcin, which plays a multifunctional role in cell regulation, contributes as a suppressor in carcinogenesis. Survival of cancer patients is prolonged with high expression of regucalcin in tumor tissues. Ovarian cancer is the most lethal in gynecologic malignancies. This study elucidates the repressive role of regucalcin on the growth of human ovarian cancer SK-OV-3 cells that are resistant to cytotoxic cancer drugs. MATERIALS AND METHODS: SK-OV-3 wild type-cells and regucalcin-overexpressing cells (transfectants) were cultured in Dulbecco's Modification of Eagle's Medium containing 10 % fetal bovine serum. KEY FINDINGS: Colony formation and proliferation of SK-OV-3 cells were repressed by regucalcin overexpression. The suppressive effects of regucalcin on proliferation were independent of cell death. The proliferation of SK-OV-3 wild-type cells was repressed by various inhibitors, including cell cycle, signaling processes, and transcriptional activity. The effects of all inhibitors were not revealed in transfectants, suggesting the involvement of multiple signaling pathways in regucalcin effects. Of note, the overexpressed regucalcin declined the levels of Ras, Akt, mitogen-activating protein kinase, NF-κB p65, ß-catenin, and STAT3, while it raised the levels of tumor suppressors p53 and Rb, and cell cycle inhibitor p21. Interestingly, the stimulatory effects of epidermal growth factor (EGF) on cell proliferation were blocked in regucalcin-overexpressing cells. Extracellular regucalcin repressed the proliferation independent of the death of SK-OV-3 cells and blocked EGF-enhanced cell proliferation. SIGNIFICANCES: The overexpressed regucalcin may repress cell proliferation by targeting diverse signal pathways, including EGF signaling. This study offers a novel approach to the treatment of ovarian cancer with regucalcin.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Humanos , Feminino , Fator de Crescimento Epidérmico/farmacologia , Proliferação de Células , Transdução de Sinais , Linhagem Celular Tumoral , Antineoplásicos/farmacologia
7.
Life Sci ; 306: 120795, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-35835253

RESUMO

AIMS: RGPR-p117 was originally discovered as a novel transcription factor, which specifically binds to a nuclear factor I (NFI) consensus motif TTGGC(N)6CC in the promoter region of the regucalcin gene. RGPR-p117 is also called as Lztr2 and SEC16B. The role of RGPR-p117 in cell regulation is poorly understood. This study was undertaken to determine whether the overexpression of RGPR-p117 impacts the proliferation of normal rat kidney proximal tubular epithelial NRK-52E cells in vitro. MAIN METHODS: The NRK-52E wild-type cells and RGPR-p117-overexpressing NRK-52E cells were cultured in DMEM containing fetal bovine serum. KEY FINDINGS: The overexpression of RGPR-p117 repressed colony formation and proliferation of NRK-52E cells. Interestingly, RGPR-p117 overexpression blocked cell proliferation promoted by culturing with Bay K 8644, a calcium-entry agonist, and phorbol 12-myristate 13-acetate, an activator of protein kinase C. The depressive effects of RGPR-p117 overexpression on cell proliferation were not occurred by culturing with various inhibitors of cell cycle and intracellular signaling processes. RGPR-p117 overexpression increased the translocation of RGPR-p117 into the nucleus of NRK-52E cells. Mechanistically, RGPR-p117 overexpression diminished the levels of Ras, PI3 kinase, Akt, mitogen-activated protein kinase, and mTOR, while it raised the levels of p53, Rb, p21, and regucalcin. Furthermore, RGPR-p117 overexpression protected cell death caused by apoptosis-inducing factors, suggesting that the suppressive effects of RGPR-p117 on cell growth are independent of cell death. SIGNIFICANCE: The present study demonstrates that the overexpressed transcription factor RGPR-p117 suppresses cell proliferation via targeting diverse signaling processes, suggesting a role of RGPR-p117 in cell regulation.


Assuntos
Proteínas de Ligação ao Cálcio , Proteínas de Ligação a DNA/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Rim/metabolismo , Fatores de Transcrição NFI/genética , Regiões Promotoras Genéticas , Ratos , Transdução de Sinais
8.
Prostate ; 82(10): 1025-1039, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35365850

RESUMO

BACKGROUND: Prostate cancer is a bone metastatic cancer and is the second leading cause of cancer-related death in men. Prolonged progression-free survival of prostate cancer patients is associated with high regucalcin expression in the tumor tissues. This study investigates the underlying mechanism by which regucalcin prevents bone metastatic activity of prostate cancer cells. METHODS: Human prostate cancer PC-3 or DU-145 wild-type cells or regucalcin-overexpressing PC-3 or DU-145 cells (transfectants) were cultured in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum. RESULTS: Overexpressed regucalcin suppressed the migration and invasion of bone metastatic human prostate cancer cells in vitro, and it reduced the levels of key proteins in metastasis including Ras, Akt, MAPK, RSK-2, mTOR, caveolin-1, and integrin ß1. Invasion of prostate cancer cells was promoted by coculturing with preosteoblastic MC3T3-E1 or preosteoclastic RAW264.7 cells. Coculturing with cancer cells and bone cells repressed the growth of preosteoblastic cells and enhanced osteoclastogenesis of preosteoclastic cells, and these alterations were caused by a conditioned medium from cancer cell culture. Disordered differentiation of bone cells was prevented by regucalcin overexpression. Production of tumor necrosis factor-α (TNF-α) in cancer cells was blocked by overexpressed regucalcin. Of note, the effects of conditioned medium on bone cells were prevented by NF-κB inhibitor. TNF-α may be important as a mediator in the crosstalk between cancer cells and bone cells. CONCLUSION: Overexpression of regucalcin suppressed the migration, invasion, and bone metastatic activity of human prostate cancer cells. This study may provide a new strategy for therapy with the regucalcin gene transfer.


Assuntos
Neoplasias Ósseas , Proteínas de Ligação ao Cálcio , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias da Próstata , Neoplasias Ósseas/secundário , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Meios de Cultivo Condicionados , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Células PC-3 , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Anticancer Drugs ; 33(5): 424-436, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35324521

RESUMO

Prostate cancer is metastatic cancer and is the second leading cause of cancer-related death in men. It is needed to develop more effective treatment for metastatic prostate cancer. The present study investigates whether the novel factor 3,5-dihydroxy-4-methoxybenzyl alcohol (DHMBA), which was isolated from marine oyster, suppresses the activity of metastatic human prostate cancer PC-3 or DU-145 cells. Culture of DHMBA (1 or 10 µM) suppressed colony formation and growth of PC-3 or DU-145 cells in vitro. Suppressive effects of DHMBA on cell proliferation were not occurred by culturing with intracellular signaling inhibitors. Mechanistically, DHMBA (10 µM) reduced the levels of key proteins linked to promotion of cell growth, including Ras, PI3K, Akt, MAPK, and mTOR in PC-3 cells. Interestingly, DHMBA increased the levels of cancer suppressor p53, p21, Rb, and regucalcin. Moreover, culture of DHMBA simulated the death of PC-3 and DU-145 cells. This effect was implicated to caspase-3 activation in cells. Interestingly, the effects of DHMBA on cell proliferation and death were blocked by culturing with an inhibitor of aryl hydrocarbon receptor linked to transcriptional regulation. Furthermore, culture of DHMBA inhibited production of reactive oxygen species in PC-3 or DU-145 cells. Of note, DHMBA blocked migration and invasion by diminishing their related protein levels, including NF-κB 65, caveolin-1 and integrin ß1. The novel marine factor DHMBA was demonstrated to suppress metastatic prostate cancer cells via targeting diverse signaling pathways. This study may provide a new strategy for prostate cancer therapy with DHMBA.


Assuntos
Neoplasias da Próstata , Álcoois Benzílicos , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Masculino , Células PC-3 , Neoplasias da Próstata/patologia , Transdução de Sinais
10.
Oncology ; 100(7): 399-412, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35340010

RESUMO

INTRODUCTION: Regucalcin plays a multifunctional role in the regulation of cellular function including metabolism, signaling process, and transcriptional activity in maintaining cell homeostasis. Downregulated expression or activity of regucalcin contributes to the development of malignancies in various types of human cancer. Survival of cancer patients, including metastatic prostate cancer, is prolonged with high expression of regucalcin in the tumor tissues. METHODS: We elucidate whether extracellular regucalcin conquers the growth, migration, invasion, and adhesion of metastatic human prostate cancer PC-3 and DU-145 cells. RESULTS: Extracellular regucalcin (0.1, 1, and 10 nM) of physiologic levels (1 nM at human serum) inhibited colony formation and growth of PC-3 and DU-145 cells, while it did not have an effect on cell death. Repressive effects of extracellular regucalcin on the proliferation were not exhibited by the presence of inhibitors of the cell cycle, intracellular signaling process, and transcriptional activity, suggesting that the signals of extracellular regucalcin are transmitted to block cell growth. Furthermore, extracellular regucalcin (0.1, 1, or 10 nM) inhibited migration, invasion, and adhesion of PC-3 and DU-145 cells. Mechanistically, extracellular regucalcin (10 nM) decreased the levels of various signaling proteins including Ras, posphatidylinositol-3 kinase, mitogen-activated protein kinase, mechanistic target of rapamycin, RSK-2, caveolin-1, and integrin ß1 in PC-3 cells. DISCUSSION AND CONCLUSION: Thus, extracellular regucalcin may play a suppressive role in growth, migration, invasion, and adhesion, which are involved in the metastatic activity of human prostate cancer cells, via affecting diverse signaling processes. This study may provide a new strategy in preventing metastatic prostate cancer with exogenous regucalcin.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias da Próstata , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino
11.
FEBS Open Bio ; 12(1): 175-191, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34709731

RESUMO

We previously isolated derrisfolin A, a novel rotenoid derivative, from the stems of Derris trifoliata Lour. (Leguminosae). Here, we report that derrisfolin A induces the expression of endogenous regucalcin (RGN) protein in both pancreatic MIN6 ß-cells and RAW264.7 macrophages. Induction of RGN expression by derrisfolin A or retrovirus-mediated gene transfer in MIN6 cells and RAW264.7 macrophages significantly decreased lipopolysaccharide (LPS)-induced mRNA expression of Nos2, Il1b, and Tnf via nuclear factor-κB activation; reduced LPS-induced apoptosis in MIN6 cells, accompanied by decreased production of nitric oxide, interleukin-1ß, and tumor necrosis factor-α; and attenuated generation of LPS-induced reactive oxygen species, malondialdehyde, and 3-nitrotyrosine in MIN6 cells. Additionally, in co-cultures of MIN6 cells with RAW264.7 macrophages in the presence of LPS, induction of RGN expression by derrisfolin A or retrovirus-mediated gene transfer in RAW264.7 macrophages attenuated apoptosis and oxidative/nitrosative stress in MIN6 cells. These results suggest that the induction of RGN expression in MIN6 cells was effective in suppressing LPS-induced inflammatory cytotoxicity and that in co-culture conditions, the induction of RGN expression in RAW264.7 macrophages blocked LPS-induced paracrine effects of RAW264.7 macrophages on inflammatory cytotoxicity in MIN6 cells. Our findings suggest that derrisfolin A, a chemical inducer of RGN, might be useful for developing a new drug against macrophage-associated ß-cell inflammation in type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Lipopolissacarídeos , Animais , Diabetes Mellitus Tipo 2/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Células RAW 264.7
12.
Anticancer Drugs ; 32(5): 558-566, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33595948

RESUMO

Hepatocellular carcinoma (HCC) is one of the most prevalent malignant diseases and causes a third of cancer-related death. The prognosis and effective treatment of advanced HCC remains poor in spite of the development of novel therapeutic strategies. In the present study, we investigate anticancer effects of the botanical molecule p-hydroxycinnamic acid (HCA) in the HepG2 liver cancer model in vitro. Culturing with HCA (10-1000 nM) suppressed colony formation and growth of HepG2 cells. Mechanistically, culturing with HCA decreased levels of Ras, PI3K, Akt, MAPK, NF-κB p65 and ß-catenin, which are linked to processes of cell signaling and transcription, and increased levels of retinoblastoma and regucalcin, which are suppressors for carcinogenesis. These alterations may lead to the suppression of cell growth. Furthermore, culturing with HCA (10-1000 nM) stimulated cell death due to increased caspase-3 levels. Interestingly, the effects of HCA on the growth and death of HepG2 cells were inhibited by culturing with CH223191, an antagonist of aryl hydrocarbon receptor (AHR), suggesting that the flavonoid effects are, at least partly, mediated by activation of AHR signaling. Notably, HCA blocked stimulatory effects of Bay K 8644, an agonist of L-type calcium channel, on the growth of HepG2 cells. Thus, our study demonstrates that HCA suppresses the growth and stimulates the death of human liver cancer HepG2 cells in vitro. The botanical molecule HCA may therefore be a useful tool in the treatment of HCC, providing a novel strategy for the therapy of human liver cancers.


Assuntos
Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Cumáricos/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Compostos Azo/farmacologia , Células Hep G2 , Humanos , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Transdução de Sinais
13.
J Cancer Res Clin Oncol ; 147(2): 339-350, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33001270

RESUMO

Bone metastatic prostate cancer is one of the most common malignancies in developed countries and the second leading cause of cancer-related death in men. There remains no effective treatment for metastatic prostate cancer. We investigate here the anticancer effects of botanical component p-hydroxycinnamic acid (HCA) on the PC-3 cells in vitro model of bone metastatic human prostate cancer. Culturing with HCA (10-1000 nM) suppressed colony formation and growth of PC-3 cells. Mechanistically, culturing with HCA decreased protein levels of Ras, PI3K, Akt, MAPK, NF-κB p65 and ß-catenin related to processes of cell signaling and transcription, and it increased levels of p21, p53, retinoblastoma and regucalcin, which are suppressors in carcinogenesis. These alterations can lead to suppression of cell growth. Furthermore, culturing with HCA increased cell death and caspase-3 levels. The effects of HCA on the growth and death of PC-3 cells were blocked by culturing with CH223191, an antagonist of aryl hydrocarbon receptor (AHR), suggesting that HCA effects are partly involved in AHR signaling. Interestingly, HCA suppressed the stimulatory effects of Bay K 8644, an agonist of L-type calcium channel, on the growth of PC-3 cells. Coculturing of PC-3 cells and preosteoblastic MC-3T3 E1 cells increased osteoblastic mineralization. This increase was not attenuated by treatment of HCA that stimulated mineralization. Notably, osteoclastogenesis from preosteoclastic RAW264.7 cells was enhanced by coculturing with PC-3 cells, and this enhancement was suppressed by treatment with HCA (10-1000 nM). Thus, HCA has anticancer effects on bone metastatic human prostate cancer, potentially providing a novel therapeutic tool.


Assuntos
Neoplasias Ósseas/secundário , Ácidos Cumáricos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Apoptose/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Neoplasias Ósseas/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Camundongos , Células PC-3 , Neoplasias da Próstata/patologia , Células RAW 264.7 , Receptores de Hidrocarboneto Arílico/fisiologia
14.
Transl Oncol ; 14(1): 100955, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33232921

RESUMO

Prostate cancer, which is a bone metastatic cancer, is the second leading cause of cancer-related death in men. There is no effective treatment for metastatic prostate cancer. Regucalcin has been shown to contribute as a suppressor in various types of human cancers. In the present study, furthermore, we investigate an involvement of regucalcin in suppression of prostate cancer. Regucalcin expression was compared in 131 primary tumor tissues and 19 metastatic tumor tissues in prostate cancer patients. Regucalcin expression in the metastatic tumor was found to be reduced as compared with that in primary tumor. The progression-free survival rate was prolonged in patients with a higher regucalcin expression. Translationally, overexpression of regucalcin in bone metastatic human prostate cancer PC-3 and DU-145 cells suppressed colony formation and cell growth in vitro. Mechanistically, overexpressed regucalcin enhanced the levels of p53, Rb, and p21, and decreased the levels of Ras, PI3 kinase, Akt, and mitogen-activated protein kinase, leading to suppression of cell growth. Furthermore, higher regucalcin expression suppressed the levels of nuclear factor-κB p65, ß-catenin, and signal transducer and activator of transcription 3, which regulate a transcription activity. Cell growth was promoted by culturing with the calcium agonist Bay K 8644. This effect was blocked by overexpression of regucalcin. Notably, overexpressed regucalcin suppressed bone metastatic activity of PC-3 and DU-145 cells when cocultured with preosteoblastic or preosteoclastic cells. Regucalcin may suppress the development of human prostate cancer, suggesting that gene delivery systems in which its expression is forced may be a novel therapeutic strategy.

15.
Tissue Cell ; 67: 101447, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33137709

RESUMO

Regucalcin plays a multifunctional role in cell regulation as a suppressor in the processes of intracellular signaling and transcription, leading to inhibition of cell growth. The downregulated expression or activity of regucalcin has been shown to contribute to the development of carcinogenesis in various types of human cancer. The wild-type tumor suppressor TP53 gene encodes for a transcriptional factor p53. This protein may play a role in cell proliferation. Loss of p53 function may induce cell transformation during carcinogenesis and tumor progression of human cancer. We investigate whether or not extracellular regucalcin suppresses the proliferation of non-tumorigenic human mammary epithelial MCF 10A cells with loss of p53 in vitro. Loss of p53 did not impact colony formation and proliferation of the cells. Interestingly, p53 loss caused decrease in the cell cycle suppressor p21, but not retinoblastoma and regucalcin, as compared with those of wild-type MCF 10A cells. Notably, extracellular regucalcin suppressed colony formation and proliferation of wild-type MCF 10A cells and p53 (-/-) cells, while it did not have an effect on cell death. Mechanistically, extracellular regucalcin decreased levels of various signaling factors including Ras, phosphatidylinositol-3 kinase, mitogen-activated protein kinase (MAPK), phospho-MAPK, and signal transducer and activator of transcription 3 in wild-type MCF 10A cells and p53 (-/-) cells. Thus, extracellular regucalcin was found to suppress the growth of MCF 10A cells with loss of p53. Extracellular regucalcin may play a role as a suppressor in the growth of human mammary epithelial cells with p53 loss, providing a novel strategy for cancer.


Assuntos
Proteínas de Ligação ao Cálcio/farmacologia , Hidrolases de Éster Carboxílico/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Espaço Extracelular/química , Glândulas Mamárias Humanas/citologia , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Epiteliais/efeitos dos fármacos , Humanos , Ratos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/metabolismo
16.
Int J Exp Pathol ; 101(6): 248-263, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32985761

RESUMO

Human umbilical vein endothelial cells (HUVECs) are a pivotal component of the hematopoietic microenvironment linked to the modulation of the immune response, inflammation and carcinogenesis. HUVEC expresses the aryl hydrocarbon receptor (AHR), which regulates gene expression by binding to the xenobiotic-responsive element. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent agonist for AHR signalling. Treatment with TCDD (0.1-100 nmol/L) was found to suppress the proliferation and to stimulate the death of HUVEC. TCDD's effects were abolished by culturing with CH223191, an inhibitor of AHR signalling. Mechanistically, TCDD treatment increased the protein levels of cell growth suppressors, including p53, Rb, p21 and regucalcin, and caspase-3 implicated in apoptotic cell death, and decreased the levels of Stat3, mitogen-activated protein kinase (MAPK/Erk1/2) and phospho-MAPK/Erk1/2. Treatment with polyunsaturated fatty acids (PUFAs), including docosahexaenoic acid, eicosapentaenoic acid and arachidonic acid, suppressed the proliferation and stimulated the death of HUVEC in vitro, and decreased the levels of Stat3, MAPK/Erk1/2 and phospho-MAPK/Erk1/2 and increased caspase-3. Notably, the effects of TCDD in suppressing proliferation and stimulating death of HUVEC were modulated by coculturing with PUFAs. These effects were reversed by treatment with CH223191, an inhibitor of AHR. Treatment with both TCDD and PUFAs collaboratively enhanced the levels of AHR, CYP1A1, p53, p21, Rb and regucalcin. Moreover, TCDD suppressed migration with wound healing of HUVEC. Notably, the combination of TCDD and PUFAs revealed potent suppressive effects on angiogenesis of HUVEC, potentially related to disorders of the stromal microenvironment.


Assuntos
Apoptose/efeitos dos fármacos , Compostos Azo/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Ácidos Graxos Insaturados/farmacologia , Dibenzodioxinas Policloradas/farmacologia , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas , Transdução de Sinais/efeitos dos fármacos , Ácido Araquidônico/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores
17.
FEBS Open Bio ; 10(10): 1967-1984, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32783343

RESUMO

Dysregulation of adipocyte differentiation and dysfunction play key roles in the pathogenesis of obesity and associated disorders such as diabetes and metabolic syndrome, and as such, a better understanding of the molecular mechanism of adipogenesis may help to elucidate the pathological condition of obesity and its associated disorders. Regucalcin (RGN) plays multiple regulatory roles in intracellular Ca2+ signaling pathways in mammalian cells. Here, we report that overexpression of RGN enhances lipid accumulation in 3T3-L1 adipocyte cells after adipogenic stimulation, accompanied by upregulation of adipocyte differentiation marker proteins. In contrast, genetic disruption of RGN inhibited adipogenic stimulation-induced differentiation of 3T3-L1 cells. Furthermore, RGN overexpression in differentiated 3T3-L1 adipocytes blocked inflammatory crosstalk between 3T3-L1 adipocytes and RAW264.7 macrophages in a transwell coculture system. Knockdown of RGN expression in cocultured 3T3-L1 adipocytes enhanced their susceptibility to RAW264.7 macrophage-mediated inflammation. These results suggest that RGN is required for 3T3-L1 adipocyte differentiation and that it exerts anti-inflammatory activity against 3T3-L1 adipocyte inflammation after coculture with RAW264.7 macrophages. Thus, RGN may be a novel regulator of adipocyte differentiation and act as a suppressor of inflammation in macrophage-infiltrated adipocyte tissue.


Assuntos
Adipócitos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/fisiologia , Adipogenia/genética , Adipogenia/fisiologia , Animais , Proteínas de Ligação ao Cálcio/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Técnicas de Cocultura , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Macrófagos/metabolismo , Camundongos , Obesidade/metabolismo , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
18.
Mol Cell Biochem ; 472(1-2): 173-185, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32591915

RESUMO

Hepatocellular carcinoma is one of the most prevalent malignant diseases and causes a third of cancer-related death. The consequences of altered calcium homeostasis in cancer cells may contribute to tumor progression. Regucalcin plays an inhibitory role in calcium signaling linked to transcription regulation. Regucalcin gene expression is downregulated in the tumor tissues of liver cancer patients, suggesting an involvement as a suppressor in hepatocarcinogenesis. We investigated whether Bay K 8644, an agonist of the L-type Ca2+ channel, promotes the growth of human liver cancer and if the effect of Bay K 8644 is suppressed by overexpressed regucalcin using the HepG2 cell model. The colony formation and growth of HepG2 cells were promoted by culturing with Bay K 8644 (0.1-10 nM). This effect was suppressed by inhibitors of signaling processes linked to cell proliferation, including PD98059 and wortmannin. Death of HepG2 cells was stimulated by Bay K 8644 with higher concentrations (25 and 100 nM). The effects of Bay K 8644 on cell growth and death were abolished by verapamil, an antagonist of calcium channel. Mechanistically, culturing with Bay K 8644 increased levels of mitogen-activated protein kinase (MAPK) and phospho-MAPK. Notably, overexpressed regucalcin suppressed Bay K 8644-promoted growth and death of HepG2 cells. Furthermore, overexpressed regucalcin prevented growth and increased death induced by thapsigargin, which induces the release of intracellular stored calcium. Thus, higher regucalcin expression suppresses calcium signaling linked to the growth of liver cancer cells, providing a novel strategy in treatment of hepatocellular carcinoma with delivery of the regucalcin gene.


Assuntos
Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/efeitos adversos , Agonistas dos Canais de Cálcio/efeitos adversos , Canais de Cálcio Tipo L/química , Proteínas de Ligação ao Cálcio/metabolismo , Carcinoma Hepatocelular/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Hepáticas/prevenção & controle , Apoptose , Proteínas de Ligação ao Cálcio/genética , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Células Tumorais Cultivadas
19.
Curr Mol Med ; 20(8): 643-652, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32065089

RESUMO

Backgrounds and Objective: Inflammation is implicated in the pathogenesis of many diseases. Inflammatory cytokines with painful conditions are well known as biomarkers in human muscle pain, and they are produced by macrophages. Metaxalone is used as a skeletal muscle relaxant, but the mechanism by which metaxalone acts is unknown. This study was undertaken to investigate whether or not metaxalone exhibits an inhibitory effect on the activity of inflammatory macrophages in vitro. METHODS: Mouse macrophage RAW264.7 cells were cultured in Dulbecco's Modification of Eagle's Medium containing 10% fetal bovine serum in the presence of metaxalone. Cell growth was assayed by counting the number of cells attached to culture dishes. Inflammatory cytokines released into the culture medium were analyzed with the ELISA kit. RESULTS: Metaxalone (1-100 µM) was found to decrease the number of macrophages by inhibiting the proliferation and stimulating the death of RAW264.7 cells in vitro. The combination of metaxalone (0.1 or 1 µM) and ß-caryophyllene (10 or 50 µM), which alone did not have a significant effect on the cell number, caused potential effects on the growth and death of RAW264.7 cells. Mechanistically, molecular levels of mitogenactivated protein kinase were decreased by treatment with metaxalone or ß- caryophyllene, and each effect was enhanced by their combination. Furthermore, levels of caspase-3 were increased by metaxalone or ß-caryophyllene and enhanced by their combination. Notably, productions of inflammatory cytokines, including tumor necrosis factor-α, interleukin-6 or prostaglandin E2, which were enhanced by lipopolysaccharide (LPS), were repressed by culturing with metaxalone. Levels of cyclooxygenase (COX)-1, COX-2 and nuclear factor kappa B, which were increased by LPS treatment, were reduced by metaxalone. CONCLUSION: Metaxalone was found to suppress the activity of inflammatory macrophages in vitro.


Assuntos
Citocinas/antagonistas & inibidores , Sinergismo Farmacológico , Mediadores da Inflamação/antagonistas & inibidores , Inflamação/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Oxazolidinonas/farmacologia , Sesquiterpenos Policíclicos/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Citocinas/metabolismo , Combinação de Medicamentos , Técnicas In Vitro , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Fármacos Neuromusculares/farmacologia , Células RAW 264.7
20.
Cancer Invest ; 38(1): 37-51, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31868021

RESUMO

Regucalcin plays a pivotal role as a suppressor of human carcinogenesis, and downregulation of regucalcin expression may contribute to the promotion of human osteosarcoma. Overexpression of regucalcin suppressed the proliferation of Saos-2 human osteosarcoma cells in vitro and decreased the protein levels of multiple signaling components, transcription factors, and tumor suppressors. Interestingly, extracellular regucalcin repressed colony formation and proliferation of Saos-2 cells, and reduced the protein levels of multiple signaling components, cell cycle inhibitor, and various transcription factors. Thus, regucalcin suppressed the growth of human osteosarcoma cells, providing a novel strategy with the gene therapy for treatment of osteosarcoma.


Assuntos
Neoplasias Ósseas/terapia , Proteínas de Ligação ao Cálcio/metabolismo , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteossarcoma/terapia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/isolamento & purificação , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/isolamento & purificação , Hidrolases de Éster Carboxílico/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fígado , Osteossarcoma/genética , Osteossarcoma/patologia , Prognóstico , Ratos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...