Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Free Radic Biol Med ; 208: 458-467, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37678654

ABSTRACT

Ferroptosis is a regulated form of cell death, the mechanism of which is still to be understood. 15-lipoxygenase (15LOX) complex with phosphatidylethanolamine (PE)-binding protein 1 (PEBP1) catalyzes the generation of pro-ferroptotic cell death signals, hydroperoxy-polyunsaturated PE. We focused on gaining new insights into the molecular basis of these pro-ferroptotic interactions using computational modeling and liquid chromatography-mass spectrometry experiments. Simulations of 15LOX-1/PEBP1 complex dynamics and interactions with lipids revealed that association with the membrane triggers a conformational change in the complex. This conformational change facilitates the access of stearoyl/arachidonoyl-PE (SAPE) substrates to the catalytic site. Furthermore, the binding of SAPE promotes tight interactions within the complex and induces further conformational changes that facilitate the oxidation reaction. The reaction yields two hydroperoxides as products, 15-HpETE-PE and 12-HpETE-PE, at a ratio of 5:1. A significant effect of PEBP1 is observed only on the predominant product. Moreover, combined experiments and simulations consistently demonstrate the significance of PEBP1 P112E mutation in generating ferroptotic cell death signals.


Subject(s)
Arachidonate 15-Lipoxygenase , Ferroptosis , Phosphatidylethanolamine Binding Protein , Cell Death , Ferroptosis/physiology , Oxidation-Reduction , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/physiology , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/physiology , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/metabolism , Humans , Animals , Swine
2.
Cell Mol Immunol ; 18(4): 992-1004, 2021 04.
Article in English | MEDLINE | ID: mdl-32901127

ABSTRACT

Aberrant inflammasome activation contributes to the pathogenesis of various human diseases, including atherosclerosis, gout, and metabolic disorders. Elucidation of the underlying mechanism involved in the negative regulation of the inflammasome is important for developing new therapeutic targets for these diseases. Here, we showed that Raf kinase inhibitor protein (RKIP) negatively regulates the activation of the NLRP1, NLRP3, and NLRC4 inflammasomes. RKIP deficiency enhanced caspase-1 activation and IL-1ß secretion via NLRP1, NLRP3, and NLRC4 inflammasome activation in primary macrophages. The overexpression of RKIP in THP-1 cells inhibited NLRP1, NLRP3, and NLRC4 inflammasome activation. RKIP-deficient mice showed increased sensitivity to Alum-induced peritonitis and Salmonella typhimurium-induced inflammation, indicating that RKIP inhibits NLRP3 and NLRC4 inflammasome activation in vivo. Mechanistically, RKIP directly binds to apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and competes with NLRP1, NLRP3, or NLRC4 to interact with ASC, thus interrupting inflammasome assembly and activation. The depletion of RKIP aggravated inflammasome-related diseases such as monosodium urate (MSU)-induced gouty arthritis and high-fat diet (HFD)-induced metabolic disorders. Furthermore, the expression of RKIP was substantially downregulated in patients with gouty arthritis or type 2 diabetes (T2D) compared to healthy controls. Collectively, our findings suggest that RKIP negatively regulates NLRP1, NLRP3, and NLRC4 inflammasome activation and is a potential therapeutic target for the treatment of inflammasome-related diseases.


Subject(s)
Arthritis, Gouty/immunology , Diabetes Mellitus, Type 2/immunology , Inflammasomes/immunology , Macrophages/immunology , Peritonitis/immunology , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Arthritis, Gouty/metabolism , Arthritis, Gouty/pathology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Mice , Mice, Inbred C57BL , Peritonitis/metabolism , Peritonitis/pathology , Phosphatidylethanolamine Binding Protein/genetics , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
3.
Cells ; 8(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083461

ABSTRACT

Lung cancer is the most deadly neoplasm with the highest incidence in both genders, with non-small cell lung cancer (NSCLC) being the most frequent subtype. Somatic mutations within the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene are key drivers of NSCLC progression, with EGFR inhibitors being particularly beneficial for patients carrying the so-called "EGFR-sensitizing mutations". However, patients eventually acquire resistance to these EGFR inhibitors, and a better knowledge of other driven and targetable proteins will allow the design of increasingly accurate drugs against patients' specific molecular aberrations. Raf kinase inhibitory protein (RKIP) is an important modulator of relevant intracellular signaling pathways, including those controlled by EGFR, such as MAPK. It has been reported that it has metastasis suppressor activity and a prognostic role in several solid tumors, including lung cancer. In the present review, the potential use of RKIP in the clinic as a prognostic biomarker and predictor of therapy response in lung cancer is addressed.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Phosphatidylethanolamine Binding Protein/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Mutation/drug effects , Prognosis , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 115(42): E9859-E9868, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30282734

ABSTRACT

The signaling cascades triggered by the cross-linkage of immunoglobulin E (IgE) with its high-affinity receptor (FcεRI) on mast cells contribute to multiple allergic disorders, such as asthma, rhinitis, and atopic dermatitis. Restraint of intracellular signals for mast cell activation is essential to restore homeostasis. In this study, we found that Raf kinase inhibitor protein (RKIP) negatively regulated mast cell activation. RKIP-deficient mast cells showed greater IgE-FcεRI-mediated activation than wild-type mast cells. Consistently, RKIP deficiency in mast cells rendered mice more sensitive to IgE-FcεRI-mediated allergic responses and ovalbumin-induced airway inflammation. Mechanistically, RKIP interacts with the p85 subunit of PI3K, prevents it from binding to GRB2-associated binding protein 2 (Gab2), and eventually inhibits the activation of the PI3K/Akt/NF-κB complex and its downstream signaling. Furthermore, the expression of RKIP was significantly down-regulated in the peripheral blood of asthma patients and in the IgE-FcεRI-stimulated mast cells. Collectively, our findings not only suggest that RKIP plays an important role in controlling mast cell-mediated allergic responses but also provide insight into therapeutic targets for mast cell-related allergic diseases.


Subject(s)
Anaphylaxis/immunology , Asthma/immunology , Mast Cells/immunology , Phosphatidylethanolamine Binding Protein/physiology , Receptors, IgE/metabolism , Anaphylaxis/metabolism , Animals , Asthma/metabolism , Cell Degranulation , Cells, Cultured , Child , Humans , Mast Cells/metabolism , Mice , Mice, Knockout , Signal Transduction
5.
EMBO Rep ; 19(6)2018 06.
Article in English | MEDLINE | ID: mdl-29674348

ABSTRACT

Th17 cells contribute to the development of autoimmune diseases by secreting interleukin-17 (IL-17), which activates its receptor (IL-17R) that is expressed on epithelial cells, macrophages, microglia, and resident neuroectodermal cells. However, the mechanisms through which IL-17R-mediated signaling contributes to the development of autoimmune disease have not been completely elucidated. Here, we demonstrate that Raf-1 kinase inhibitor protein (RKIP) deficiency in mice ameliorates the symptoms of experimental autoimmune encephalomyelitis (EAE). Adoptive T-cell-transfer experiments demonstrate that RKIP plays a predominant role in Th17-mediated, but not in Th1-mediated immune responses. RKIP deficiency has no effect on Th17-cell differentiation ex vivo, nor does it affect Th17-cell differentiation in EAE mice. However, RKIP significantly promotes IL-17R-induced proinflammatory cytokine and chemokine production. Mechanistically, RKIP directly interacts with IL-17RA and Act1 to promote the formation of an IL-17R-Act1 complex, resulting in enhanced MAPK- and P65-mediated NF-κB activation and downstream cytokine production. Together, these findings indicate that RKIP functions as an essential modulator of the IL-17R-Act1 axis in IL-17R signaling, which promotes IL-17-induced inflammation and autoimmune neuroinflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Phosphatidylethanolamine Binding Protein/physiology , Receptors, Interleukin-17/metabolism , Th17 Cells/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylethanolamine Binding Protein/genetics , Signal Transduction
6.
Andrology ; 5(5): 1032-1051, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28859251

ABSTRACT

This study aimed to identify sperm proteomic signatures regulating sperm functions and fertility by: (i) comparing the sperm electrophoretic protein profiles and identifying the differentially abundant proteins among breeding bulls differing in fertility status and (ii) elucidating the possible role of one of the identified novel proteins, PEBP4 on sperm function and fertility. The grouping of bulls as fertile (n = 6) and low fertile (n = 6) was performed based on bull fertility index and infertile (n = 6) based on semen rejection rate (>33%). The sperm motility, fructolysis index, acrosomal reaction, intracellular calcium levels, and seminal plasma fructose and calcium levels were studied among fertility groups. The differentially expressed sperm proteins observed in single- and two-dimensional gel electrophoresis (2DE) were identified using Nano-LC-MS/MS. In the fertile bulls, the expression levels of calmodulin (CALM1), spermadhesinZ13 (SPADH2), and phosphatidylethanolamine-binding protein 4 (PEBP4) were significantly (p < 0.05) higher than in other fertility groups. In bovine, expression of PEBP4 a novel seminal protein was not observed in spermatozoa of infertile bulls. When the bulls were grouped based on the presence (n = 8) or absence (n = 10) of PEBP4 protein in spermatozoa, a positive significant (p < 0.05) association of this protein with the percentage of motile, type-A spermatozoa, and sperm fructose uptake was observed. Further, PEBP4 was localized in elongated spermatids, Leydig cells, excurrent duct system, and principal piece of spermatozoa. These findings suggest a crucial role for the PEBP4 protein in spermiogenesis, epididymal sperm maturation, and sperm motility. This first study in bovine indicates the positive association of PEBP4 in regulating sperm maturation, functions, and fertility and could be a potential marker for predicting semen quality and fertility.


Subject(s)
Cattle/physiology , Fertility , Phosphatidylethanolamine Binding Protein/physiology , Spermatozoa/physiology , Acrosome Reaction , Animals , Biomarkers/metabolism , Calcium Signaling , Cattle Diseases/metabolism , Epididymis/metabolism , Fructose/metabolism , Infertility, Male/metabolism , Infertility, Male/veterinary , Male , Proteome , Semen/metabolism , Sperm Maturation , Sperm Motility
7.
Cancer Res ; 76(12): 3644-54, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27197200

ABSTRACT

RAF kinase inhibitor protein (RKIP) is a seminal regulator of intracellular signaling and exhibits both antimetastatic and antitumorigenic properties. Decreased expression of RKIP has been described in several human malignancies, including acute myelogenous leukemia (AML). As the mechanisms leading to RKIP loss in AML are still unclear, we aimed to analyze the potential involvement of miRNAs within this study. miRNA microarray and qPCR data of more than 400 AML patient specimens revealed correlation between decreased expression of RKIP and increased expression of miR-23a, a member of the miR-23a/27a/24-2 cluster. In functional experiments, overexpression of miR-23a decreased RKIP mRNA and protein expression, whereas miR-23a inhibition caused the opposite effect. By using an RKIP 3'-untranslated region luciferase reporter construct with and without mutation or deletion of the putative miR-23a-binding site, we could show that RKIP modulation by miR-23a is mediated via direct binding to this region. Importantly, miR-23a overexpression induced a significant increase of proliferation in hematopoietic cells. Simultaneous transfection of an RKIP expression construct lacking the miR-23a-binding sites reversed this phenotype, indicating that this effect is truly mediated via downregulation of RKIP. Finally, by analyzing more than 4,300 primary patient specimens via database retrieval from The Cancer Genome Atlas, we could highlight the importance of the miR-23a/RKIP axis in a broad range of human cancer entities. In conclusion, we have identified miR-23a as a negative regulator of RKIP expression in AML and have provided data that suggest the importance of our observation beyond this tumor entity. Cancer Res; 76(12); 3644-54. ©2016 AACR.


Subject(s)
Leukemia, Myeloid, Acute/pathology , MicroRNAs/physiology , Phosphatidylethanolamine Binding Protein/physiology , Cell Line, Tumor , Cell Proliferation , Humans , Leukemia, Myeloid, Acute/genetics , Phosphatidylethanolamine Binding Protein/analysis , Phosphatidylethanolamine Binding Protein/genetics
8.
Biochim Biophys Acta ; 1860(2): 384-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26116914

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common type of liver cancer with high mortality and poor prognosis. Mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways have been implicated in promoting tumor cell proliferation and invasion of HCC cells. METHODS: As a potential inhibitor of tumor metastasis, the role of Raf kinase inhibitor protein (RKIP) in HCC development and the functional relevance with MAPK and NF-κB signaling pathways were investigated. The levels of RKIP expression were examined in human HCC tissues and correlated with tumor stages and metastatic status. Function of RKIP in cellular proliferation, migration, invasion and apoptosis was investigated in HCC cell lines by either overexpressing or knocking down RKIP expression. Mouse xenograft model was established to assess the effect of RKIP expression on tumor growth. RESULTS: Our results demonstrated decreased RKIP expression in HCC tissues and a strong correlation with tumor grade and distant metastasis. Manipulation of RKIP expression in HCCLM3 and HepG2 cells indicated that RKIP functioned to inhibit HCC cell motility and invasiveness, and contributed to tumor growth inhibition in vivo. Mechanistic studies showed that the function of RKIP was mediated through MAPK and NF-κB signaling pathways. However, cell type-dependent RKIP regulation on these two pathways was also suggested, indicating the complex nature of signaling network. CONCLUSION: Our study provides a better understanding on the molecular mechanisms of HCC metastasis and sets the foundation for the development of targeted therapeutics for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Phosphatidylethanolamine Binding Protein/physiology , Signal Transduction/physiology , Animals , Carcinoma, Hepatocellular/secondary , Cell Movement , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Humans , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Neoplasm Invasiveness
9.
Plant J ; 84(5): 949-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26466761

ABSTRACT

Distinct molecular mechanisms integrate changes in ambient temperature into the genetic pathways that govern flowering time in Arabidopsis thaliana. Temperature-dependent eviction of the histone variant H2A.Z from nucleosomes has been suggested to facilitate the expression of FT by PIF4 at elevated ambient temperatures. Here we show that, in addition to PIF4, PIF3 and PIF5, but not PIF1 and PIF6, can promote flowering when expressed specifically in phloem companion cells (PCC), where they can induce FT and its close paralog, TSF. However, despite their strong potential to promote flowering, genetic analyses suggest that the PIF genes seem to have only a minor role in adjusting flowering in response to photoperiod or high ambient temperature. In addition, loss of PIF function only partially suppressed the early flowering phenotype and FT expression of the arp6 mutant, which is defective in H2A.Z deposition. In contrast, the chemical inhibition of gibberellic acid (GA) biosynthesis resulted in a strong attenuation of early flowering and FT expression in arp6. Furthermore, GA was able to induce flowering at low temperature (15°C) independently of FT, TSF, and the PIF genes, probably directly at the shoot apical meristem. Together, our results suggest that the timing of the floral transition in response to ambient temperature is more complex than previously thought and that GA signaling might play a crucial role in this process.


Subject(s)
Arabidopsis/growth & development , Gibberellins/metabolism , Plant Growth Regulators/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Flowers/genetics , Flowers/growth & development , Flowers/metabolism , Gibberellins/pharmacology , Histones/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Nucleosomes/metabolism , Phosphatidylethanolamine Binding Protein/genetics , Phosphatidylethanolamine Binding Protein/metabolism , Phosphatidylethanolamine Binding Protein/physiology , Photoperiod , Plant Growth Regulators/pharmacology , Signal Transduction , Temperature
10.
Cancer Res ; 75(19): 4063-73, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26238785

ABSTRACT

Triple-negative breast cancer (TNBC) patients have the highest risk of recurrence and metastasis. Because they cannot be treated with targeted therapies, and many do not respond to chemotherapy, they represent a clinically underserved group. TNBC is characterized by reduced expression of metastasis suppressors such as Raf kinase inhibitory protein (RKIP), which inhibits tumor invasiveness. Mechanisms by which metastasis suppressors alter tumor cells are well characterized; however, their ability to regulate the tumor microenvironment and the importance of such regulation to metastasis suppression are incompletely understood. Here, we use species-specific RNA sequencing to show that RKIP expression in tumors markedly reduces the number and metastatic potential of infiltrating tumor-associated macrophages (TAM). TAMs isolated from nonmetastatic RKIP(+) tumors, relative to metastatic RKIP(-) tumors, exhibit a reduced ability to drive tumor cell invasion and decreased secretion of prometastatic factors, including PRGN, and shed TNFR2. RKIP regulates TAM recruitment by blocking HMGA2, resulting in reduced expression of numerous macrophage chemotactic factors, including CCL5. CCL5 overexpression in RKIP(+) tumors restores recruitment of prometastatic TAMs and intravasation, whereas treatment with the CCL5 receptor antagonist Maraviroc reduces TAM infiltration. These results highlight the importance of RKIP as a regulator of TAM recruitment through chemokines such as CCL5. The clinical significance of these interactions is underscored by our demonstration that a signature comprised of RKIP signaling and prometastatic TAM factors strikingly separates TNBC patients based on survival outcome. Collectively, our findings identify TAMs as a previously unsuspected mechanism by which the metastasis-suppressor RKIP regulates tumor invasiveness, and further suggest that TNBC patients with decreased RKIP activity and increased TAM infiltration may respond to macrophage-based therapeutics.


Subject(s)
Chemokines/physiology , Chemotaxis , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Neoplasm Metastasis/immunology , Neoplasm Proteins/physiology , Phosphatidylethanolamine Binding Protein/physiology , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor/transplantation , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CCL5/physiology , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , Disease-Free Survival , Female , Gene Expression Profiling , Gene Knockdown Techniques , HMGA2 Protein/physiology , Heterografts/immunology , Humans , Mammary Neoplasms, Experimental/drug therapy , Maraviroc , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, CCR5/drug effects , Sequence Analysis, RNA , Triazoles/pharmacology , Triazoles/therapeutic use , Triple Negative Breast Neoplasms/mortality
11.
Neurobiol Aging ; 35(12): 2870-2880, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25104559

ABSTRACT

Raf kinase inhibitor protein (RKIP) is a major negative mediator of the extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. The downregulation of RKIP is correlated with many cancers, but the mechanisms that underlie this downregulation and its roles in the nervous system remain unclear. Here, we demonstrate that RKIP is a substrate of cyclin-dependent kinase 5 (CDK5) in neurons and that the phosphorylation of RKIP at T42 causes the release of Raf-1. Moreover, T42 phosphorylation promotes the exposure and recognition of the target motif "KLYEQ" in the C-terminus of RKIP by chaperone Hsc70 and the subsequent degradation of RKIP via chaperone-mediated autophagy (CMA). Furthermore, in the brain sample of Parkinson's disease (PD) patients and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride-induced and transgenic PD models, we demonstrate that CDK5-mediated phosphorylation and autophagy of RKIP are involved in the overactivation of the ERK/MAPK cascade, leading to S-phase reentry and neuronal loss. These findings provide evidence for the role of the CDK5/RKIP/ERK pathway in PD pathogenesis and suggest that this pathway may be a suitable therapeutic target in PD.


Subject(s)
Autophagy/genetics , Cyclin-Dependent Kinase 5/physiology , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Phosphatidylethanolamine Binding Protein/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Humans , MAP Kinase Signaling System/physiology , Molecular Targeted Therapy , Parkinson Disease/therapy , Phosphatidylethanolamine Binding Protein/physiology , Phosphorylation/genetics , Rats
12.
Invest Ophthalmol Vis Sci ; 55(9): 5788-94, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25125607

ABSTRACT

PURPOSE: Mutations in the cilia-centrosomal protein of centrosomal protein of 290 kDa (CEP290) result in severe ciliopathies, including autosomal recessive early onset childhood blindness disorder Leber congenital amaurosis (LCA). The Cep290(rd16) (retinal degeneration 16) mouse model of CEP290-LCA exhibits accumulation of CEP290-interacting protein Raf-1 kinase inhibitory protein (RKIP) prior to onset of retinal degeneration (by postnatal day P14). We hypothesized that reducing RKIP levels in the Cep290(rd16) mouse will delay or improve retinal phenotype. METHODS: We generated double mutant mice by combining the Cep290(rd16) and Rkip(ko) alleles (Cep290(rd16):Rkip(+/ko) and Cep290(rd16):Rkip(ko/ko)). Retinal function was assessed by ERG and retinal morphology and protein trafficking were assessed by histology, transmission electron microscopy (TEM), and immunofluorescence analysis. Cell death was examined by apoptosis. RESULTS: Prior to testing our hypothesis, we examined ERG and retinal morphology of Rkip(ko/ko) mice and did not find any detectable differences compared with wild-type mice. The Cep290(rd16):Rkip(+/ko) mice exhibited similar retinopathy as Cep290(rd16); however, Cep290(rd16): Rkip(ko/ko) double knockout mice demonstrated a substantial improvement (>9-fold) in photoreceptor function and structure at P18 as of Cep290(rd16) mice. We consistently detected transient preservation of photoreceptors at P18 and polarized trafficking of opsins to sensory cilia in the double mutant mice; however, retinal degeneration ensued by P30. CONCLUSIONS: Our studies implicate CEP290-RKIP pathway in CEP290-retinal degeneration and suggest that targeting RKIP levels can delay photoreceptor degeneration, assisting in extending the time-window for treating such rapidly progressing blindness disorder.


Subject(s)
Phosphatidylethanolamine Binding Protein/physiology , Retinal Degeneration/physiopathology , Analysis of Variance , Animals , Antigens, Neoplasm , Apoptosis/physiology , Cell Cycle Proteins , Ciliary Body/metabolism , Cytoskeletal Proteins , Disease Models, Animal , Electroretinography , Mice , Mice, Knockout , Nuclear Proteins , Opsins/metabolism , Phosphatidylethanolamine Binding Protein/deficiency , Photoreceptor Cells, Vertebrate/physiology , Retinal Degeneration/pathology
13.
PLoS One ; 9(3): e90062, 2014.
Article in English | MEDLINE | ID: mdl-24594691

ABSTRACT

Human phosphatidylethanolamine-binding protein 4(hPEBP4) is a novel anti-apoptosis molecule associated with the resistance of tumors to apoptotic agents. Here we sought to investigate the role of hPEBP4 in the radioresistance of rectal cancer. Immunohistochemistry analysis showed hPEBP4 was expressed in 27/33 of rectal cancer specimens, but only in 2/33 of neighboring normal mucosa. Silencing the expression of hPEBP4 with siRNA significantly reduced the clonogenic survival and enhanced the apoptosis of rectal cancer cells on irradiation. Instead, forced overexpression of hPEBP4 promoted its survival and decreased the apoptosis. Western blot showed hPEBP4 could increase the radiation-induced Akt activation, for which reactive oxygen specimen(ROS) was required. The radioresistance effect of hPEBP4 was reversed after given LY-294002 to inhibit Akt activation or antioxidant to abolish the ROS production. We also confirmed that effect of hPEBP4 in vivo with nude mice. Thus we concluded that hPEBP4, specifically expressed in rectal cancer cells, is associated with radioresistance of rectal cancer, implying that modulation of hPEBP4 may have important therapeutic implications in radiotherapy of rectal cancer.


Subject(s)
Phosphatidylethanolamine Binding Protein/physiology , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance/physiology , Reactive Oxygen Species/metabolism , Rectal Neoplasms/radiotherapy , Animals , Base Sequence , DNA Primers , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Phosphatidylethanolamine Binding Protein/metabolism , Polymerase Chain Reaction , Rectal Neoplasms/enzymology , Rectal Neoplasms/metabolism
14.
Crit Rev Oncog ; 19(6): 405-15, 2014.
Article in English | MEDLINE | ID: mdl-25597351

ABSTRACT

The Raf kinase inhibitor protein 1 (RKIP-1) was the first reported endogenous inhibitor of Raf-1-MEK-ERK/MAPK cascade, by interfering with the phosphorylation of MEK by Raf-1. However, RKIP's functions related to the MAPK signaling are far more complex. Newer data indicate that by modulating different protein-protein interactions, RKIP is involved in fine-tuning cell signaling, modulating ERK dynamics, and regulating cross talk between different pathways. Here, we describe the molecular mechanisms by which RKIP controls MAPK signaling at different levels and vice versa and its regulation via feedback phosphorylation. We also focus on several discrepancies and questions that remain, such as the RKIP binding regulation by Raf-1 N-region phosphorylation, the possible B-Raf inhibition, and the effects of RKIP-lipid binding. We also describe how RKIP's role as key signaling modulator of many cell fate decisions leads to the fact that fine control of RKIP activity and regulation is crucial to avoid pathological processes, such as metastasis, pulmonary arterial hypertension, and heart failure.


Subject(s)
MAP Kinase Signaling System , Phosphatidylethanolamine Binding Protein/physiology , Animals , Feedback, Physiological , Humans , Lipid Metabolism , Phosphatidylethanolamine Binding Protein/chemistry , Phosphorylation , Protein Binding , Protein Structure, Tertiary
15.
Crit Rev Oncog ; 19(6): 447-54, 2014.
Article in English | MEDLINE | ID: mdl-25597354

ABSTRACT

Cancer is one of the deadliest diseases worldwide, accounting for about 8 million deaths a year. For solid tumors, cancer patients die as a result of the metastatic spread of the tumor to the rest of the body. Therefore, there is a clinical need for understanding the molecular and cellular basis of metastasis, identifying patients whose tumors are more likely to metastasize, and developing effective therapies against metastatic progression. Over the years, Raf kinase inhibitory protein (RKIP) has emerged as a natural suppressor of the metastatic process, constituting a tool for studying metastasis and its clinical outcomes. Here, we review RKIP's role as a metastasis suppressor and the signaling networks and genes regulated by RKIP in metastatic, triple-negative breast cancer. We also highlight the clinical implications and power of building gene signatures based on RKIP-regulated signaling modules in identifying cancer patients that are at higher risk for metastases. Finally, we highlight the potential of RKIP as a tool for developing new therapeutic strategies in cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, Tumor Suppressor , Neoplasm Metastasis/genetics , Phosphatidylethanolamine Binding Protein/physiology , Humans , MAP Kinase Signaling System/physiology , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
16.
Crit Rev Oncog ; 19(6): 431-45, 2014.
Article in English | MEDLINE | ID: mdl-25597353

ABSTRACT

Cancer remains one of the most dreadful diseases. Whereas most treatment regimens for various cancers have resulted in improved clinical responses and sometimes cures, unfortunately, subsets of cancer patients are either pretreatment resistant or develop resistance following therapy. These subsets of patients develop cross-resistance to unrelated therapeutics and usually succumb to death. Thus, delineating the underlying molecular mechanisms of resistance of various cancers and identifying molecular targets for intervention are the current main focus of research investigations. One approach to investigate cancer resistance has been to identify pathways that regulate resistance and develop means to disrupt these pathways in order to override resistance and sensitize the resistant cells to cell death. Hence, we have identified one pathway that is dysregulated in cancer, namely, the NF-κB/Snail/YY1/RKIP loop, that has been shown to regulate, in large part, tumor cell resistance to apoptosis by chemotherapeutic and immunotherapeutic cytotoxic drugs. The dysregulated resistant loop is manifested by the overexpression of NF-κB, Snail, and YY1 activities and the underexpression of RKIP. The induction of RKIP expression results in the downregulation of NF-κB, Snail, and YY1 and the sensitization of resistant cells to drug-induced apoptosis. These findings identified RKIP, in addition to its antiproliferative and metastatic suppressor functions, as an anti-resistance factor. This brief review describes the role of RKIP in the regulation of drug sensitivity via disruption of the NF-κB/Snail/ YY1/RKIP loop that regulates resistance in cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Immunotherapy , Neoplasms/therapy , Phosphatidylethanolamine Binding Protein/physiology , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , YY1 Transcription Factor/genetics , YY1 Transcription Factor/metabolism
17.
Crit Rev Oncog ; 19(6): 489-96, 2014.
Article in English | MEDLINE | ID: mdl-25597358

ABSTRACT

The Raf kinase inhibitor protein (RKIP) increasingly evolves as an important regulator of intracellular signaling networks and thus participates in diverse physiological functions ranging from growth and differentiation processes to muscle contraction. Several molecular events contribute to the ability of RKIP to tightly coordinate kinase signaling. The elucidation of the molecular mechanisms leading to substrate specificity of RKIP and substrate binding efficacy is of great interest for a better understanding of the overall role of RKIP in the organism but also for the design of specific and potent kinase inhibitors. In this work, we will review mechanistic details of the regulation of RKIP as inhibitor of Raf-1 and G protein-coupled receptor kinase 2 (GRK2) that enable RKIP to coordinate the cell's balance between inhibition and potentiation of mitogenic ERK1/2 signaling--a prominent example of RKIP's function as a regulator of intracellular signaling.


Subject(s)
Phosphatidylethanolamine Binding Protein/physiology , Signal Transduction , Animals , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/metabolism , Humans , Phosphatidylethanolamine Binding Protein/metabolism , Protein Binding , Signal Transduction/genetics
18.
Crit Rev Oncog ; 19(6): 483-8, 2014.
Article in English | MEDLINE | ID: mdl-25597357

ABSTRACT

Raf kinase inhibitory protein (RKIP) is a highly conserved regulator of many signaling networks whose loss or inactivation can lead to a variety of disease states. The multifaceted roles played by RKIP are enabled by an allosteric structure that is controlled through phosphorylation of RKIP and dynamics in the RKIP pocket loop. Perhaps the most striking feature of RKIP is that it can assume multiple functional states. Specifically, phosphorylation redirects RKIP from a state that binds and inhibits Raf-1 to a state that binds and inhibits GRK2. Recent evidence suggests the presence of a third functional state that facilitates RKIP phosphorylation. Here, we present a three-state model to explain the RKIP functional switch and discuss the role of the pocket loop in regulating RKIP activity.


Subject(s)
Models, Biological , Phosphatidylethanolamine Binding Protein/chemistry , Phosphatidylethanolamine Binding Protein/physiology , Humans , Models, Molecular , Phospholipids/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-raf/metabolism , Structure-Activity Relationship
19.
Crit Rev Oncog ; 19(6): 469-81, 2014.
Article in English | MEDLINE | ID: mdl-25597356

ABSTRACT

Helicobacter pylori is a helical bacterium that colonizes the stomach in over half of the world's population. Infection with this bacterium has been linked to peptic ulcer disease and gastric cancer. The bacterium has been shown to affect regulatory pathways in its host cells through specific virulence factors that control gene expression. Infection with H. pylori increases levels of phosphorylation of Raf kinase inhibitor protein (pRKIP) in gastric adenocarcinoma (AGS) cells in vitro and in vivo. We investigated the role of H. pylori in the phosphorylation of RKIP as a possible mechanism to downregulate pro-survival signals in gastric adenocarcinoma. pRKIP induces RKIP transcriptional activity, which serves to induce apoptosis of damaged cells to prevent further tumorigenesis. Infection of wild type and RKIP knockout mice with H. pylori for 2 months further confirmed roles of RKIP and pRKIP in the prevention of gastric cancer progression. Loss of RKIP in AGS cells results in increased expression of the Cag A virulence factor after H. pylori infection and RKIP overexpression inhibits H. pylori-mediated STAT3 phosphorylation and STAT3 and NF-κB transcriptional activity. We examined the role of mTOR (mammalian target of rapamycin) after H. pylori infection on the phosphorylation of RKIP. Cells treated with rapamycin, an inhibitor of mTOR, displayed less expression of pRKIP after H. pylori infection. Microarray antibody analysis was conducted on wild-type and RKIP-knockdown AGS cells and showed that in the absence of RKIP, there was increased expression of pro-tumorigenic proteins such as EGFR, Raf-1, and MAPKs. Although further work is needed to confirm the interaction of RKIP and mTOR in AGS cells as a result of H. pylori infection, we hypothesize that H. pylori-mediated induction of pro-survival signaling in gastric epithelial cells induces a feedback response through the activation of RKIP. The phosphorylated, or active, form of RKIP is important in protecting gastric epithelial cells from tumorigenesis after H. pylori infection.


Subject(s)
Helicobacter Infections/complications , Helicobacter pylori/physiology , Phosphatidylethanolamine Binding Protein/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Animals , Carcinogenesis/genetics , Helicobacter Infections/genetics , Humans , Mice , Mice, Knockout , Phosphatidylethanolamine Binding Protein/genetics , Signal Transduction/genetics
20.
Crit Rev Oncog ; 19(6): 505-16, 2014.
Article in English | MEDLINE | ID: mdl-25597360

ABSTRACT

In 1984, a cytosolic protein was isolated from bovine brain and coined phosphatidylethanolamine binding protein (PEBP) to describe its phospholipid-binding potential. Its cellular function remained elusive for more than a decade until it was discovered that PEBP had the ability to suppress the Raf1-mitogen activated protein kinase (MAPK) pathway, earning it the new name of Raf1 kinase inhibitory protein (RKIP). This milestone discovery has paved the way for numerous studies that have now extended the reach of RKIP's function to other signaling cascades, within the context of various physiological and pathophysiological systems. This review will summarize our current knowledge of the neurophysiological roles of RKIP in the mammalian brain, including its function in the circadian clock and synaptic plasticity. It will also discuss evidence for an involvement of RKIP and its derived neuropeptide, hippocampal cholinergic neurostimulating peptide (HCNP), in neural development and differentiation. Implications in certain pathologies such as Alzheimer's disease and brain cancer will be highlighted. By chronicling the diverse functions of RKIP in the brain, we hope that this review will serve as a timely resource that ignites future studies on this versatile, multifaceted protein in the nervous system.


Subject(s)
Brain/metabolism , Mammals , Phosphatidylethanolamine Binding Protein/physiology , Animals , Cattle , Gene Expression , Humans , Mammals/physiology , Phosphatidylethanolamine Binding Protein/chemistry , Protein Conformation , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...