Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Curr Pharm Des ; 29(42): 3408-3420, 2023.
Article in English | MEDLINE | ID: mdl-37936452

ABSTRACT

INTRODUCTION: Breast cancer has emerged as the most widespread cancer globally surpassing lung cancer, and has become a primary cause of mortality among women. While MFHAS1 has been implicated in the pathophysiology of various diseases, its precise involvement in breast cancer remains unclear. METHODS: This study endeavors to elucidate the regulatory function of MFHAS1 in breast cancer cell pyroptosis and the associated molecular mechanisms. Our findings indicate that the inhibition of MFHAS1 can impede the proliferation and invasion of breast cancer cells, while also inducing cell pyroptosis via caspase1-dependent activation of GSDMD. RESULTS: This process results in the cleavage of cell membranes, leading to the release of inflammatory factors and LDH. Subsequent investigations revealed that the silencing of MFHAS1 can promote JNK phosphorylation, thereby activating the JNK signaling cascade. Notably, this effect can be counteracted by the JNK-specific inhibitor sp600125. Ultimately, our investigation substantiated the identical function of MFHAS1 in breast cancer tissue derived from animal models. CONCLUSION: To summarize, our findings demonstrate that the inhibition of MFHAS1 elicits pyroptosis in human breast cancer cells through the facilitation of JNK phosphorylation and the activation of the downstream NF-κB/caspase-1/GSDMD signaling cascade, thereby proposing the prospect of MFHAS1 as a viable therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Pyroptosis , Animals , Female , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Gasdermins , MAP Kinase Signaling System , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Oncogene Proteins/pharmacology , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/pharmacology , Pyroptosis/genetics , Pyroptosis/physiology , Signal Transduction , Caspase 1/metabolism
2.
Physiol Res ; 72(S3): S277-S286, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37888971

ABSTRACT

Neuroblastoma represents 8-10 % of all malignant tumors in childhood and is responsible for 15 % of cancer deaths in the pediatric population. Aggressive neuroblastomas are often resistant to chemotherapy. Canonically, neuroblastomas can be classified according to the MYCN (N-myc proto-oncogene protein) gene amplification, a common marker of tumor aggressiveness and poor prognosis. It has been found that certain compounds with chelating properties may show anticancer activity, but there is little evidence for the effect of chelators on neuroblastoma. The effect of new chelators characterized by the same functional group, designated as HLZ (1-hydrazino phthalazine), on proliferation (WST-1 and methylene blue assay), cell cycle (flow cytometry), apoptosis (proliferation assay after use of specific pharmacological inhibitors and western blot analysis) and ROS production (fluorometric assay based on dichlorofluorescein diacetate metabolism) was studied in three neuroblastoma cell lines with different levels of MYCN amplification. The molecules were effective only on MYCN-non-amplified cells in which they arrested the cell cycle in the G0/G1 phase. We investigated the mechanism of action and identified the activation of cell signaling that involves protein kinase C.


Subject(s)
Neuroblastoma , Oncogene Proteins , Child , Humans , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/therapeutic use , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Oncogene Proteins/pharmacology , Nuclear Proteins/genetics , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Neuroblastoma/drug therapy , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Apoptosis , Cell Proliferation
3.
Article in English | MEDLINE | ID: mdl-36231664

ABSTRACT

The G-protein-coupled receptor for estrogen (GPER1) is a transmembrane receptor involved in the progression and development of various neoplasms whose ligand is estradiol (E2). 17ß-aminoestrogens (17ß-AEs) compounds, analogs to E2, are possible candidates for use in hormone replacement therapy (HRT), but our knowledge of their pharmacological profile is limited. Thus, we explored the molecular recognition of GPER1 with different synthetic 17ß-AEs: prolame, butolame, and pentolame. We compared the structure and ligand recognition sites previously reported for a specific agonist (G1), antagonists (G15 and G36), and the natural ligand (E2). Then, the biological effects of 17ß-AEs were analyzed through cell viability and cell-cycle assays in two types of female cancer. In addition, the effect of 17ß-AEs on the phosphorylation of the oncoprotein c-fos was evaluated, because this molecule is modulated by GPER1. Molecular docking analysis showed that 17ß-AEs interacted with GPER1, suggesting that prolame joins GPER1 in a hydrophobic cavity, similarly to G1, G15, and E2. Prolame induced cell proliferation in breast (MCF-7) and cervical cancer (SIHA) cells; meanwhile, butolame and pentolame did not affect cell proliferation. Neither 17ß-AEs nor E2 changed the activation of c-fos in MCF-7 cells. Meanwhile, in SIHA cells, E2 and 17ß-AEs reduced c-fos phosphorylation. Thus, our data suggest that butolame and pentolame, but not prolame, could be used for HRT without presenting a potential risk of inducing breast- or cervical-cancer-cell proliferation. The novelty of this work lies in its study of compound analogs to E2 that may represent important therapeutic strategies for women in menopause, with non-significant effects on the cell viability of cancer cells. The research focused on the interactions of GPER1, a molecule recently associated with promoting and maintaining various neoplasms.


Subject(s)
Breast Neoplasms , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Alcohols , Breast Neoplasms/drug therapy , Cell Proliferation , Estradiol/pharmacology , Estrenes , Estrogens/pharmacology , Female , Humans , Ligands , Molecular Docking Simulation , Oncogene Proteins/pharmacology
4.
Int J Biol Sci ; 18(9): 3859-3873, 2022.
Article in English | MEDLINE | ID: mdl-35813469

ABSTRACT

Chemotherapy has been widely used as a clinical treatment for cancer over the years. However, its effectiveness is limited because of resistance of cancer cells to programmed cell death (PCD) after treatment with anticancer drugs. To elucidate the resistance mechanism, we initially focused on cancer cell-specific mitophagy, an autophagic degradation of damaged mitochondria. This is because mitophagy has been reported to provide cancer cells with high resistance to anticancer drugs. Our data showed that TRIP-Br1 oncoprotein level was greatly increased in the mitochondria of breast cancer cells after treatment with various anticancer drugs including staurosporine (STS), the main focus of this study. STS treatment increased cellular ROS generation in cancer cells, which triggered mitochondrial translocation of TRIP-Br1 from the cytosol via dephosphorylation of TRIP-Br1 by protein phosphatase 2A (PP2A). Up-regulated mitochondrial TRIP-Br1 suppressed cellular ROS levels. In addition, TRIP-Br1 rapidly removed STS-mediated damaged mitochondria by activating mitophagy. It eventually suppressed STS-mediated PCD via degradation of VDACI, TOMM20, and TIMM23 mitochondrial membrane proteins. TRIP-Br1 enhanced mitophagy by increasing expression levels of two crucial lysosomal proteases, cathepsins B and D. In conclusion, TRIP-Br1 can suppress the sensitivity of breast cancer cells to anticancer drugs by activating autophagy/mitophagy, eventually promoting cancer cell survival.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Antineoplastic Agents/pharmacology , Apoptosis , Autophagy , Breast Neoplasms/drug therapy , Female , Humans , Mitophagy , Oncogene Proteins/pharmacology , Reactive Oxygen Species/metabolism
5.
J Cell Mol Med ; 24(8): 4736-4747, 2020 04.
Article in English | MEDLINE | ID: mdl-32160403

ABSTRACT

Maresin Conjugates in Tissue Regeneration 1 (MCTR1) is a newly identified macrophage-derived sulfido-conjugated mediator that stimulates the resolution of inflammation. This study assessed the role of MCTR1 in alveolar fluid clearance (AFC) in a rat model of acute lung injury (ALI) induced by lipopolysaccharide (LPS). Rats were intravenously injected with MCTR1 at a dose of 200 ng/rat, 8 hours after administration of 14 mg/kg LPS. The level of AFC was then determined in live rats. Primary rat ATII (Alveolar Type II) epithelial cells were also treated with MCTR1 (100 nmol/L) in a culture medium containing LPS for 8 hours. MCTR1 treatment improved AFC (18.85 ± 2.07 vs 10.11 ± 1.08, P < .0001) and ameliorated ALI in rats. MCTR1 also significantly promoted AFC by up-regulating epithelial sodium channel (ENaC) and Na+ -K+ -adenosine triphosphatase (Na, K-ATPase) expressions in vivo. MCTR1 also activated Na, K-ATPase and elevated phosphorylated-Akt (P-Akt) by up-regulating the expression of phosphorylated Nedd4-2 (P-Nedd4-2) in vivo and in vitro. However, BOC-2 (ALX inhibitor), KH7 (cAMP inhibitor) and LY294002 (PI3K inhibitor) abrogated the improved AFC induced by MCTR1. Based on the findings of this study, MCTR1 may be a novel therapeutic approach to improve reabsorption of pulmonary oedema during ALI/acute respiratory distress syndrome (ARDS).


Subject(s)
Acute Lung Injury/therapy , Alveolar Epithelial Cells/drug effects , Cell Cycle Proteins/pharmacology , Oncogene Proteins/pharmacology , Pulmonary Alveoli/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Alveolar Epithelial Cells/metabolism , Animals , Cell Cycle Proteins/genetics , Epithelial Sodium Channels/genetics , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Oncogene Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Pulmonary Alveoli/metabolism , Rats , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/genetics
6.
Inflammation ; 41(2): 400-408, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29168081

ABSTRACT

Hyperglycemia is a highly dangerous factor to various diseases, even resulting in death of people. Inflammation plays a key role in this process. The aim of this study was to explore the role of malignant fibrous histiocytoma amplified sequence 1 (MFHAS1) in high-glucose induced inflammation. Our research showed that high glucose stimulated the expression of MFHAS1, and overexpression of MFHAS1 can attenuate high-glucose induced inflammation in endothelial cells by decreasing the secretion of cytokines interleukin-1ß (IL-1ß), interleukin-1α (IL-1α), adhesion molecule intercellular adhesion molecule-1 (ICAM), interleukin-6 (IL-6), interleukin-8 (IL-8), and chemokine ligand 1 (CXCL-1). Furthermore, we found that MFHAS1 promoted the phosphorylation of Akt and the expression of heme oxygenase-1 (HO-1). Our results indicated that MFHAS1 deadened high-glucose induced inflammation by activating AKT/HO-1 pathway, suggesting that MFHAS1 may act as a new therapeutic target of diabetes mellitus.


Subject(s)
Cell Cycle Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Glucose/physiology , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation/drug therapy , Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Humans , Hyperglycemia/drug therapy , Hyperglycemia/pathology , Inflammation/prevention & control , Oncogene Proteins/metabolism
7.
Microsc Microanal ; 23(1): 69-76, 2017 02.
Article in English | MEDLINE | ID: mdl-28162122

ABSTRACT

Cyclin E1 (CCNE1) is a core component of cell cycle regulation that drives the transition into the S phase. CCNE1 plays critical roles in cell cycle, cell proliferation, and cellular functions. However, the function of CCNE1 in early embryonic development is limited. In the present study, the function and expression of Ccne1 in porcine early parthenotes were examined. Immunostaining experiments showed that CCNE1 localized in the nucleus, starting at the four-cell stage. Knockdown of Ccne1 by double-stranded RNA resulted in the failure of blastocyst formation and induced blastocyst apoptosis. Ccne1 depletion increased expression of the pro-apoptotic gene Bax, and decreased the expression of Oct4 and the rate of inner cell mass (ICM)/trophectoderm formation. The results indicated that CCNE1 affects blastocyst formation by inducing cell apoptosis and ICM formation during porcine embryonic development.


Subject(s)
Cyclin E/pharmacology , Cyclin E/physiology , Embryonic Development/drug effects , Embryonic Development/physiology , Microscopy, Fluorescence/methods , Animals , Apoptosis/drug effects , Blastocyst/drug effects , Blastocyst Inner Cell Mass/drug effects , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation/drug effects , Cyclin E/genetics , Embryonic Stem Cells/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Octamer Transcription Factor-3/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Oncogene Proteins/physiology , Oocytes , RNA, Double-Stranded/analysis , Swine , bcl-2-Associated X Protein/metabolism
8.
J Gastroenterol Hepatol ; 31(1): 145-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26189649

ABSTRACT

BACKGROUND AND AIM: Because neutrophil gelatinase-associated lipocalin (NGAL) is known to provide significant bacteriostatic effects during infectious conditions, we tested the hypothesis that this protein is up-regulated and secreted into the intraluminal cavity of the gut under critically ill conditions and is thus responsible for the regulation of bacterial overgrowth. METHODS: With our institutional approval, male C57BL/6J mouse (6-7 weeks) were enrolled and applied for lipopolysaccharide or peritonitis model compared with naïve control. We assessed NGAL protein concentrations in intestinal lumen and up-regulation of NGAL expression in intestinal tissues in in vivo as well as ex vivo settings. Simultaneously, we examined the effects of NGAL protein administration on the growth of Escherichia coli (E. coli) in in vivo and in vitro experimental settings. The localization of NGAL in intestinal tissues and lumen was also assessed by immunohistological approach using NGAL antibody. RESULTS: Both lipopolysaccharide and peritonitis insults evoked the marked up-regulation of NGAL mRNA and protein levels in gut tissues such as crypt cells. In addition, the administration of NGAL protein significantly inhibited the outgrowth of enteric E. coli under both in vitro and in vivo conditions, accompanied by histological evidence. CONCLUSION: Neutrophil gelatinase-associated lipocalin protein accompanied by apparent bacteriostatic action accumulated in the intestinal wall and streamed into the mucosal layer during critically ill state, thereby possibly shaping microbiota homeostasis in the gut.


Subject(s)
Acute-Phase Proteins/pharmacology , Acute-Phase Proteins/physiology , Intestines/microbiology , Lipocalins/pharmacology , Lipocalins/physiology , Microbiota/drug effects , Oncogene Proteins/pharmacology , Oncogene Proteins/physiology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Critical Illness , Disease Models, Animal , Escherichia coli/growth & development , Gene Expression , Homeostasis/drug effects , Intestinal Mucosa/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Microbiota/physiology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peritonitis/microbiology , Up-Regulation
9.
Neurodegener Dis ; 15(1): 13-23, 2015.
Article in English | MEDLINE | ID: mdl-25500798

ABSTRACT

BACKGROUND: Dopaminergic degeneration is a major finding in brains of patients with Parkinson's disease (PD), together with Lewy bodies, intraneuronal inclusions mainly composed of the fibrillogenic protein α-synuclein (α-syn). The familial-PD-related protein DJ-1 was reported to reduce dopaminergic degeneration triggered by α-syn or by the dopaminergic-selective neurotoxin 6-hydroxydopamine (6-OHDA). OBJECTIVE: The aim was to further investigate the role of DJ-1 in dopaminergic degeneration and to see whether a cell-permeable recombinant form of DJ-1 (TAT-DJ-1) could restore dopamine depletion in vivo, thus representing an innovative therapeutic approach. METHODS: We developed in vitro (PC12/TetOn cells and mouse primary mesencephalic neurons) and in vivo models [including DJ-1 knockout (-/-) mice] to investigate DJ-1 in dopaminergic degeneration. RESULTS: We found that in PC12/TetOn cells overexpressing α-syn with the familial-PD linked mutation A30P, DJ-1 silencing increased α-syn (A30P) toxicity. Primary mesencephalic neurons from DJ-1 (-/-) mice were more vulnerable to a cell-permeable form of α-syn (TAT-α-syn) and to 6-OHDA. Intrastriatally administered TAT-DJ-1 reduced 6-OHDA toxicity in vivo in C57BL/6 mice. Finally, when we injected TAT-α-syn (A30P) in the striatum of DJ-1 (-/-) animals, dopamine was depleted more than in the control strain. CONCLUSION: DJ-1 appears to have a protective role against dopaminergic degeneration triggered by α-syn or 6-OHDA, reinforcing the possible therapeutic importance of this protein in PD.


Subject(s)
Dopaminergic Neurons/drug effects , Nerve Degeneration/prevention & control , Oncogene Proteins/pharmacology , Oxidopamine/pharmacology , Peroxiredoxins/pharmacology , alpha-Synuclein/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Oxidopamine/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Protein Deglycase DJ-1 , Up-Regulation , alpha-Synuclein/metabolism
10.
Exp Neurol ; 254: 41-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440229

ABSTRACT

Lipocalin-2 (LCN2) is an acute phase protein induced in response to injury, infection or other inflammatory stimuli. Based on the previously reported involvement of LCN2 in chemokine induction and in the recruitment of neutrophils at the sites of infection or tissue injury, we investigated the role of LCN2 in the pathogenesis of chronic/persistent inflammatory pain hypersensitivity. In the complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model, LCN2 expression was strongly induced in the ipsilateral hindpaws, peaking at 12h after CFA injection and then gradually subsiding. In CFA-injected hindpaw tissues, LCN2 and its receptor 24p3R were mainly expressed in infiltrating neutrophils and macrophages. CFA-induced thermal hyperalgesia and mechanical allodynia were significantly diminished in Lcn2-deficient mice compared to wild-type animals. Furthermore, neutrophil infiltration, myeloperoxidase activity, expression of TNF-α, IL-1ß and MIP-2 in CFA-injected hindpaws, and spinal glial activation were markedly reduced by Lcn2 deficiency. An intraplantar injection of recombinant LCN2 protein induced thermal and mechanical hypersensitivities in naïve mice, and this was accompanied by neutrophil and macrophage infiltration into the hindpaws and glial activation in the dorsal horn of the spinal cord. Taken together, our results show that inflammatory cell-derived LCN2 at the sites of inflammation plays important roles in central sensitization and the subsequent nociceptive behavior in the rodent model of chronic inflammatory pain.


Subject(s)
Acute-Phase Proteins/immunology , Chronic Pain/immunology , Hyperalgesia/immunology , Inflammation/immunology , Lipocalins/immunology , Oncogene Proteins/immunology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Cells, Cultured , Chronic Pain/genetics , Cytokines/immunology , Disease Models, Animal , Female , Freund's Adjuvant/pharmacology , Hyperalgesia/genetics , Inflammation/chemically induced , Inflammation/genetics , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Nociceptors/immunology , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Recombinant Proteins/pharmacology , Spinal Cord/immunology
11.
J Biol Chem ; 288(33): 24116-27, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23836894

ABSTRACT

Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.


Subject(s)
Acute-Phase Proteins/metabolism , Chemokines/metabolism , Lipocalins/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Oncogene Proteins/metabolism , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Signal Transduction , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/pathology , Chemokines/genetics , Gene Expression Regulation/drug effects , Hyperalgesia/complications , Hyperalgesia/metabolism , Hyperalgesia/pathology , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Models, Biological , Neuralgia/enzymology , Neuralgia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Orthop Res ; 31(7): 1046-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23483583

ABSTRACT

Lipocalin 2 (LCN2) has recently emerged as a novel adipokine involved in different processes including arthritis and chondrocyte inflammatory response. However, little is known about its activity on chondrocyte homeostasis and its regulation by nitric oxide (NO) Hence, we performed a set of experiments aimed to achieve a better understanding of this relationship. Cell vitality was tested in the ATDC5 cell line by the MTT colorimetric assay. Protein expression and gene expression was evaluated by Western blot and real time RT-PCR, respectively. NO production (determined as nitrite accumulation) was assayed by the Griess reaction. First, we demonstrated that LCN2 decreased murine chondrocytes vitality. Next, LCN2 co-stimulation with LPS enhanced NOS2 protein expression by murine chondrocytes. In addition, inhibition of LPS-induced nitric oxide production by aminoguanidine, a selective NOS2 inhibitor, significantly reduced LPS-mediated LCN2 expression. In contrast, treatment of murine chondrocytes with sodium nitroprussiate (SNP), a classic NO donor, scarcely induced LCN2 expression. Intriguingly, SNP addition to LPS-challenged chondrocytes, treated with aminoguanidine, provoked a strong induction of LCN2 expression. Finally, murine ATDC5 cells, co-cultured with LPS pre-challenged macrophages, had higher LCN2 expression in comparison with murine chondrocytes co-cultured with non pre-challenged macrophages. In this work we have described for the first time that NO is able to exert a control on LCN2 expression, suggesting the existence of a feedback loop regulating its expression.


Subject(s)
Acute-Phase Proteins/metabolism , Chondrocytes/metabolism , Lipocalins/metabolism , Nitric Oxide/metabolism , Oncogene Proteins/metabolism , Toll-Like Receptor 4/metabolism , Acute-Phase Proteins/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Guanidines/pharmacology , Lipocalin-2 , Lipocalins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Oncogene Proteins/pharmacology
13.
Eur J Immunol ; 42(12): 3346-57, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22965758

ABSTRACT

Attraction of neutrophils to sites of infection or tissue injury is an essential prerequisite for an efficient innate immune response. Herein, we provide novel evidence that the antimicrobial protein, neutrophil gelatinase associated lipocalin (24p3 or lipocalin-2, Lcn2) is a central regulator of this process. Lcn2 is produced by several cell types but high amounts are released by neutrophils. Using human and murine neutrophils, we found that the addition of recombinant Lcn2 significantly stimulated their migration, which was independent of IL-8/keratinocyte chemokine formation. Mechanistically, this could be traced back to Lcn2-mediated changes of Erk1/2 signaling. Accordingly, the i.p. injection of Lcn2 into C57BL/6 mice stimulated the mobilization of neutrophils while we found a significantly reduced neutrophil chemotactic activity of cells obtained from Lcn2 KO mice. This observation transmitted to a reduced accumulation of neutrophils in intra-dermal lesions infected with Salmonella typhimurium in Lcn2 KO mice as compared to WT mice. This was not only due to a reduced chemotaxis but also to an impaired cellular adhesion of neutrophils in the absence of Lcn2. We herein describe a novel role of Lcn2 as an important paracrine chemoattractant and an indispensable factor for neutrophil function in inflammation.


Subject(s)
Acute-Phase Proteins/immunology , Chemotaxis/immunology , Lipocalins/immunology , Neutrophils/immunology , Oncogene Proteins/immunology , Paracrine Communication/immunology , Proto-Oncogene Proteins/immunology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Chemotaxis/drug effects , Chemotaxis/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Male , Mice , Mice, Knockout , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Paracrine Communication/drug effects , Paracrine Communication/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella typhimurium/immunology
14.
Vitam Horm ; 88: 333-54, 2012.
Article in English | MEDLINE | ID: mdl-22391311

ABSTRACT

Arsenic has been used for ages as a therapeutic agent. Currently, it is an FDA approved drug to treat acute promyelocytic leukemia where it leads to degradation of the PML-RAR fusion protein. It has been shown to have various other targets in cells such as JNK, NFκB, thioredoxin reductase, and MAPK pathways. Most of its effects in cells have been through arsenic's ability to bind to thiol groups in cysteine residues. Recent evidence has shown that arsenic can inhibit the Hedgehog pathway by inhibiting GLI proteins. The proposed mechanism of action is through direct binding. Potential binding sites include the critical cysteine residues in GLI zinc finger domains. The role of the Hedgehog pathway has been implicated in many cancers such as basal cell carcinoma, medulloblastoma, Ewing sarcoma, and rhabdoid tumors. Current Hedgehog pathway inhibitors have been fraught with resistance issues and so arsenic trioxide may provide an alternative therapy when combined with these other inhibitors or after acquired resistance.


Subject(s)
Arsenicals/therapeutic use , Bone Neoplasms/drug therapy , Cerebellar Neoplasms/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Medulloblastoma/drug therapy , Oncogene Proteins/pharmacology , Oxides/therapeutic use , Sarcoma, Ewing/drug therapy , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Arsenicals/pharmacology , Cerebellar Neoplasms/pathology , Hedgehog Proteins/pharmacology , Humans , Leukemia, Promyelocytic, Acute/pathology , Medulloblastoma/pathology , Oxides/pharmacology
15.
Arthritis Rheum ; 64(5): 1620-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22083497

ABSTRACT

OBJECTIVE: The mechanism by which anti-DNA antibodies mediate lupus nephritis has yet to be conclusively determined. Previously, we found that treatment of mesangial cells with anti-DNA antibodies induced high expression of neutrophil gelatinase-associated lipocalin (NGAL), an iron-binding protein up-regulated in response to kidney injury. We undertook this study to determine whether NGAL is instrumental in the pathogenesis of nephritis, is induced as part of repair, or is irrelevant to damage/repair pathways. METHODS: To investigate the role of NGAL in antibody-mediated nephritis, we induced nephrotoxic nephritis by passive antibody transfer to 129/SyJ and C57BL/6 mice. To determine if NGAL up-regulation is instrumental, we compared the severity of renal damage in NGAL wild-type mice and NGAL-knockout mice following induction of nephrotoxic nephritis. RESULTS: We found that kidney NGAL expression, as well as urine NGAL levels, were significantly increased in mice with nephrotoxic nephritis as compared to control-injected mice. Tight correlations were observed between NGAL expression, renal histopathology, and urine NGAL excretion. NGAL-knockout mice had attenuated proteinuria and improved renal histopathology compared to wild-type mice. Similarly, following nephritis induction, NGAL injection significantly exacerbated nephritis and decreased survival. NGAL induced apoptosis via caspase 3 activation and up-regulated inflammatory gene expression in kidney cells in vitro and when injected in vivo. CONCLUSION: We conclude that kidney binding of pathogenic antibodies stimulates local expression of NGAL, which plays a crucial role in the pathogenesis of nephritis via promotion of inflammation and apoptosis. NGAL blockade may be a novel therapeutic approach for the treatment of nephritis mediated by pathogenic antibodies, including anti-glomerular basement membrane disease and lupus nephritis.


Subject(s)
Acute-Phase Proteins/metabolism , Lipocalins/metabolism , Nephritis/metabolism , Oncogene Proteins/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression , Gene Silencing , Glomerular Mesangium/drug effects , Glomerular Mesangium/immunology , Glomerular Mesangium/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Longevity , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephritis/genetics , Nephritis/pathology , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , RNA, Small Interfering/genetics , Up-Regulation
16.
J Biol Chem ; 287(7): 4808-17, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22117066

ABSTRACT

Our objective was to determine whether lipocalin-2 (Lcn2) regulates cardiomyocyte apoptosis, the mechanisms involved, and the functional significance. Emerging evidence suggests that Lcn2 is a proinflammatory adipokine associated with insulin resistance and obesity-related complications, such as heart failure. Here, we used both primary neonatal rat cardiomyocytes and H9c2 cells and demonstrated for the first time that Lcn2 directly induced cardiomyocyte apoptosis, an important component of cardiac remodeling leading to heart failure. This was shown by detection of DNA fragmentation using TUNEL assay, phosphatidylserine exposure using flow cytometry to detect annexin V-positive cells, caspase-3 activity using enzymatic assay and immunofluorescence, and Western blotting for the detection of cleaved caspase-3. We also observed that Lcn2 caused translocation of the proapoptotic protein Bax to mitochondria and disruption of mitochondrial membrane potential. Using transient transfection of GFP-Bax, we confirmed that Lcn2 induced co-localization of Bax with MitoTracker® dye. Importantly, we used the fluorescent probe Phen Green SK to demonstrate an increase in intracellular iron in response to Lcn2, and depleting intracellular iron using an iron chelator prevented Lcn2-induced cardiomyocyte apoptosis. Administration of recombinant Lcn2 to mice for 14 days increased cardiomyocyte apoptosis as well as an acute inflammatory response with compensatory changes in cardiac functional parameters. In conclusion, Lcn2-induced cardiomyocyte apoptosis is of physiological significance and occurs via a mechanism involving elevated intracellular iron levels and Bax translocation.


Subject(s)
Acute-Phase Proteins/metabolism , Apoptosis/physiology , Iron/metabolism , Lipocalins/metabolism , Myocytes, Cardiac/metabolism , Oncogene Proteins/metabolism , Acute-Phase Proteins/pharmacology , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Line , DNA Fragmentation/drug effects , In Situ Nick-End Labeling , Lipocalin-2 , Lipocalins/pharmacology , Mice , Myocytes, Cardiac/cytology , Oncogene Proteins/pharmacology , Phosphatidylserines/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Rats , bcl-2-Associated X Protein/metabolism
17.
Oncotarget ; 1(1): 9-21, 2010 May.
Article in English | MEDLINE | ID: mdl-20634973

ABSTRACT

Changes in cell morphology and rearrangements of the actin cytoskeleton are common features accompanying cell transformation induced by various oncogenes. In this study, we show that promyelocytic leukemia zinc finger protein (PLZF) binds to the promoter of smooth muscle α-actin, reducing mRNA and protein levels encoded by this gene and resulting in a reorganization of the actin cytoskeleton. In cultures of chicken embryo fibroblasts (CEF), this effect on α-actin expression is correlated with a change in cellular phenotype from spindle shaped to polygonal and flattened. This morphological change is dependent on Ras function. The polygonal, flattened CEF show a high degree of resistance to the transforming activity of several oncoproteins. Our results support the conclusion that reorganization of the actin cytoskeleton plays an important role in tumor suppression by PLZF.


Subject(s)
Actins/metabolism , Cell Shape/physiology , Cell Transformation, Neoplastic , Cytoskeleton/metabolism , Fibroblasts/cytology , Gene Expression Regulation/physiology , Kruppel-Like Transcription Factors/genetics , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Chick Embryo , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Fibroblasts/metabolism , Humans , Oncogene Proteins/pharmacology , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Exp Cell Res ; 316(4): 649-56, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19769964

ABSTRACT

LRRK2 is an autosomal dominant gene whose mutations cause familial Parkinson's disease (PD). The LRRK2 protein contains a functional kinase and a GTPase domain. PD phenotypes caused by LRRK2 mutations are similar to those of idiopathic PD, implying that LRRK2 is an important participant in PD pathogenesis. Of LRRK2's PD-specific mutations, the G2019S is the most frequently observed one. Its over-expression is known to increase kinase activity and neurotoxicity compared to wild type (WT) LRRK2. Here, using a simple colorimetric cell viability assay, we analyzed LRRK2's neurotoxicity in dopaminergic SN4741 cells following treatment with hydrogen peroxide. When WT, G2019S, or empty vector was expressed in SN4741 cells, cell death was modestly and significantly increased in the order of G2019S>WT>vector. When these transfected cells were treated with hydrogen peroxide to mimic oxidative stress, cellular neurotoxicity was enhanced in the same order (i.e. G2019S>WT>vector). Moreover, incubation of SN4741 cells with conditioned medium from cells expressing G2019S and subjected to hydrogen peroxide treatment exhibited 10-15% more cell death than conditioned medium from cells transfected with vector or WT, suggesting that G2019S-expressing cells secrete a factor(s) affecting viability of neighboring cells. The kinase domain was mapped to be responsible for oxidative stress-induced neurotoxicity. In addition, over-expression of WT and G2019S LRRK2 lead to a weak, but significant, increase in intracellular reactive oxygen species (ROS) in the order of G2019S>WT as measured by DCFH-DA assay in both the presence and absence of H(2)O(2) treatment. Furthermore, in G2019S-expressing cells, co-expression of the anti-oxidant protein DJ-1 or ERK inhibitor treatment restored survival rate to a level similar to that of cells transfected with control vector under H(2)O(2) treatment. Taken together, our data suggest that the LRRK2 kinase domain increases the generation of ROS and causes enhanced neurotoxicity under H(2)O(2) treatment, which can be at least partially rescued by DJ-1 or the ERK inhibitor.


Subject(s)
Neurons/pathology , Oxidative Stress/genetics , Phosphotransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , Biological Assay/methods , Blotting, Western , Cell Survival , Cells, Cultured , Cloning, Molecular , Enzyme Inhibitors/pharmacology , Genetic Vectors , Humans , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mutation/genetics , Oncogene Proteins/pharmacology , Oxidative Stress/drug effects , Plasmids , Protein Deglycase DJ-1 , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/analysis , Signal Transduction
19.
Oncogene ; 29(9): 1394-404, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-19946337

ABSTRACT

Increased activity of MYC protein-family members is a common feature in many cancers. Using neuroblastoma as a tumor model, we established a microRNA (miRNA) signature for activated MYCN/c-MYC signaling in two independent primary neuroblastoma tumor cohorts and provide evidence that c-MYC and MYCN have overlapping functions. On the basis of an integrated approach including miRNA and messenger RNA (mRNA) gene expression data we show that miRNA activation contributes to widespread mRNA repression, both in c-MYC- and MYCN-activated tumors. c-MYC/MYCN-induced miRNA activation was shown to be dependent on c-MYC/MYCN promoter binding as evidenced by chromatin immunoprecipitation. Finally, we show that pathways, repressed through c-MYC/MYCN miRNA activation, are highly correlated to tumor aggressiveness and are conserved across different tumor entities suggesting that c-MYC/MYCN activate a core set of miRNAs for cooperative repression of common transcriptional programs related to disease aggressiveness. Our results uncover a widespread correlation between miRNA activation and c-MYC/MYCN-mediated coding gene expression modulation and further substantiate the overlapping functions of c-MYC and MYCN in the process of tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Gene Regulatory Networks/drug effects , Genes, myc/physiology , MicroRNAs/pharmacology , Neuroblastoma/genetics , Nuclear Proteins/pharmacology , Oncogene Proteins/pharmacology , Promoter Regions, Genetic/drug effects , Cell Line, Tumor , Gene Regulatory Networks/physiology , Gene Silencing/physiology , Genes, myc/genetics , Humans , MicroRNAs/biosynthesis , N-Myc Proto-Oncogene Protein , Neuroblastoma/therapy , RNA, Small Interfering/pharmacology , Transcription Factors/physiology , Treatment Outcome
20.
Exp Cell Res ; 315(18): 3140-51, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19732769

ABSTRACT

Environmental temperature variations are the most common stresses experienced by a wide range of organisms. Lipocalin 2 (Lcn2/NGAL) is expressed in various normal and pathologic conditions. However, its precise functions have not been fully determined. Here we report the induction of Lcn2 by thermal stresses in vivo, and its role following exposure to cold and heat stresses in vitro. Induction of Lcn2 in liver, heart and kidney was detected by RT-PCR, Western blot and immunohistochemistry following exposure of mice to heat and cold stresses. When CHO and HEK293T cells overexpressing NGAL were exposed to cold stress, cell proliferation was higher compared to controls. Down-regulatrion of NGAL by siRNA in A549 cells resulted in less proliferation when exposed to cold stress compared to control cells. The number of apoptotic cells and expression of pro-apoptotic proteins were lower in the NGAL overexpressing CHO and HEK293T cells, but were higher in the siRNA-transfected A549 cells compared to controls, indicating that NGAL protects cells against cold stress. Following exposure of the cells to heat stress, ectopic expression of NGAL protected cells while addition of exogenous recombinant NGAL to the cell culture medium exacerbated the toxicity of heat stress specially when there was low or no endogenous expression of NGAL. It had a dual effect on apoptosis following heat stress. NGAL also increased the expression of HO-1. Lcn2/NGAL may have the potential to improve cell proliferation and preservation particularly to prevent cold ischemia injury of transplanted organs or for treatment of some cancers by hyperthermia.


Subject(s)
Acute-Phase Proteins/metabolism , Heat Stress Disorders/metabolism , Heme Oxygenase-1/metabolism , Hypothermia/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , CHO Cells , Cell Line, Tumor , Cold Temperature , Cricetinae , Cricetulus , Down-Regulation/genetics , Down-Regulation/physiology , Gene Knockdown Techniques , Heme Oxygenase-1/drug effects , Humans , Kidney/cytology , Kidney/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Myocardium/cytology , Myocardium/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , RNA, Small Interfering/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL