Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Hepatology ; 75(2): 280-296, 2022 02.
Article in English | MEDLINE | ID: mdl-34449924

ABSTRACT

BACKGROUND AND AIMS: Methionine adenosyltransferase 1A (MAT1A) is responsible for S-adenosylmethionine (SAMe) biosynthesis in the liver. Mice lacking Mat1a have hepatic SAMe depletion and develop NASH and HCC spontaneously. Several kinases are activated in Mat1a knockout (KO) mice livers. However, characterizing the phospho-proteome and determining whether they contribute to liver pathology remain open for study. Our study aimed to provide this knowledge. APPROACH AND RESULTS: We performed phospho-proteomics in Mat1a KO mice livers with and without SAMe treatment to identify SAMe-dependent changes that may contribute to liver pathology. Our studies used Mat1a KO mice at different ages treated with and without SAMe, cell lines, in vitro translation and kinase assays, and human liver specimens. We found that the most striking change was hyperphosphorylation and increased content of La-related protein 1 (LARP1), which, in the unphosphorylated form, negatively regulates translation of 5'-terminal oligopyrimidine (TOP)-containing mRNAs. Consistently, multiple TOP proteins are induced in KO livers. Translation of TOP mRNAs ribosomal protein S3 and ribosomal protein L18 was enhanced by LARP1 overexpression in liver cancer cells. We identified LARP1-T449 as a SAMe-sensitive phospho-site of cyclin-dependent kinase 2 (CDK2). Knocking down CDK2 lowered LARP1 phosphorylation and prevented LARP1-overexpression-mediated increase in translation. LARP1-T449 phosphorylation induced global translation, cell growth, migration, invasion, and expression of oncogenic TOP-ribosomal proteins in HCC cells. LARP1 expression is increased in human NASH and HCC. CONCLUSIONS: Our results reveal a SAMe-sensitive mechanism of LARP1 phosphorylation that may be involved in the progression of NASH to HCC.


Subject(s)
Autoantigens/metabolism , Oligonucleotides/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Ribonucleoproteins/antagonists & inhibitors , Ribonucleoproteins/metabolism , S-Adenosylmethionine/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase 2/metabolism , Humans , Liver Neoplasms/metabolism , Methionine Adenosyltransferase/genetics , Mice , Mice, Knockout , Mutation , Non-alcoholic Fatty Liver Disease/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Proteomics , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , S-Adenosylmethionine/pharmacology , TOR Serine-Threonine Kinases/metabolism , SS-B Antigen
2.
Med Oncol ; 38(6): 69, 2021 May 17.
Article in English | MEDLINE | ID: mdl-34002296

ABSTRACT

Transcription factor CCAAT/enhancer-binding protein-alpha (C/EBPα) regulates myelopoiesis by coupling growth arrest with differentiation of myeloid progenitors. Mutations in one or both alleles are observed in 10-14% AML cases that render C/EBPα functionally inactive. Besides, antagonistic protein-protein interactions also impair C/EBPα expression and function. In recent independent studies, we showed that CDK2 and SKP2 downregulated C/EBPα expression in an ubiquitin-dependent proteasome degradation manner leading to differentiation block in AML. Here, we demonstrate that CDK2-instigated C/EBPα downregulation is actually mediated by SKP2. Mechanistically, we show that CDK2 stabilizes SKP2 by phosphorylating it at Ser64 and thereby potentiates C/EBPα ubiquitination and subsequent degradation in AML cells. Immunoblot experiments showed that CDK2 inhibition downregulated SKP2 levels and concomitantly enhanced C/EBPα levels in myeloid cells. We further show that while CDK2 promoted C/EBPα ubiquitination and inhibited its protein levels, negatively affected its transactivation potential and DNA binding ability, simultaneous SKP2 depletion abrogated CDK2-promoted ubiquitination and restored C/EBPα expression and function. Taken together, these findings consolidate that CDK2 potentiates SKP2-mediated C/EBPα degradation in AML and targeting CDK2-SKP2 axis can be harnessed for therapeutic benefit in AML. Hypothetical model depicts that SKP2-mediated C/EBPα proteasomal degradation is reinforced by CDK2. CDK2 phopshorylates SKP2 leading to its enhanced stabilization which in turn exaggerates C/EBPα degradation leading to differentiation arrest in AML.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , Leukemia, Myeloid, Acute/metabolism , S-Phase Kinase-Associated Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation , Cell Line, Tumor , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Down-Regulation , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Multiprotein Complexes/immunology , Phosphorylation , Protein Stability , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/immunology , Serine/metabolism
3.
Proc Natl Acad Sci U S A ; 116(37): 18561-18570, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31451657

ABSTRACT

Neutrophil migration is essential for inflammatory responses to kill pathogens; however, excessive neutrophilic inflammation also leads to tissue injury and adverse effects. To discover novel therapeutic targets that modulate neutrophil migration, we performed a neutrophil-specific microRNA (miRNA) overexpression screen in zebrafish and identified 8 miRNAs as potent suppressors of neutrophil migration. Among those, miR-199 decreases neutrophil chemotaxis in zebrafish and human neutrophil-like cells. Intriguingly, in terminally differentiated neutrophils, miR-199 alters the cell cycle-related pathways and directly suppresses cyclin-dependent kinase 2 (Cdk2), whose known activity is restricted to cell cycle progression and cell differentiation. Inhibiting Cdk2, but not DNA replication, disrupts cell polarity and chemotaxis of zebrafish neutrophils without inducing cell death. Human neutrophil-like cells deficient in CDK2 fail to polarize and display altered signaling downstream of the formyl peptide receptor. Chemotaxis of primary human neutrophils is also reduced upon CDK2 inhibition. Furthermore, miR-199 overexpression or CDK2 inhibition significantly improves the outcome of lethal systemic inflammation challenges in zebrafish. Our results therefore reveal previously unknown functions of miR-199 and CDK2 in regulating neutrophil migration and provide directions in alleviating systemic inflammation.


Subject(s)
Chemotaxis, Leukocyte/genetics , Cyclin-Dependent Kinase 2/genetics , MicroRNAs/metabolism , Neutrophils/immunology , Systemic Inflammatory Response Syndrome/immunology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/immunology , Disease Models, Animal , Down-Regulation/immunology , Gene Knockdown Techniques , Humans , Larva , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Signal Transduction/genetics , Signal Transduction/immunology , Systemic Inflammatory Response Syndrome/genetics , Zebrafish
4.
Front Immunol ; 9: 395, 2018.
Article in English | MEDLINE | ID: mdl-29568296

ABSTRACT

Because of its great potential for diversity, the immunoglobulin heavy-chain complementarity-determining region 3 (HCDR3) is taken as an antibody molecule's most important component in conferring binding activity and specificity. For this reason, HCDR3s have been used as unique identifiers to investigate adaptive immune responses in vivo and to characterize in vitro selection outputs where display systems were employed. Here, we show that many different HCDR3s can be identified within a target-specific antibody population after in vitro selection. For each identified HCDR3, a number of different antibodies bearing differences elsewhere can be found. In such selected populations, all antibodies with the same HCDR3 recognize the target, albeit at different affinities. In contrast, within unselected populations, the majority of antibodies with the same HCDR3 sequence do not bind the target. In one HCDR3 examined in depth, all target-specific antibodies were derived from the same VDJ rearrangement, while non-binding antibodies with the same HCDR3 were derived from many different V and D gene rearrangements. Careful examination of previously published in vivo datasets reveals that HCDR3s shared between, and within, different individuals can also originate from rearrangements of different V and D genes, with up to 26 different rearrangements yielding the same identical HCDR3 sequence. On the basis of these observations, we conclude that the same HCDR3 can be generated by many different rearrangements, but that specific target binding is an outcome of unique rearrangements and VL pairing: the HCDR3 is necessary, albeit insufficient, for specific antibody binding.


Subject(s)
Antibody Diversity , Complementarity Determining Regions/genetics , Immunoglobulin Heavy Chains/chemistry , Single-Chain Antibodies/genetics , Amino Acid Sequence , Antigens/immunology , Clonal Selection, Antigen-Mediated , Cyclin-Dependent Kinase 2/immunology , Datasets as Topic , Gene Rearrangement , Humans , Immunoglobulin Heavy Chains/genetics , Peptide Library , Protein Binding , Structure-Activity Relationship
5.
J Immunol ; 192(2): 649-57, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337749

ABSTRACT

Coinfection of hepatitis B virus (HBV) with hepatitis C virus (HCV) is quite common, leading to an increase in morbidity and mortality. As such, HBV vaccination is recommended in HCV-infected individuals. However, HBV vaccine responses in HCV-infected individuals are often blunted compared with uninfected populations. The mechanism for this failure of vaccine response in HCV-infected subjects remains unclear. In this study, we investigated the expression and function of an inhibitory receptor, killer cell lectin-like receptor subfamily G member 1 (KLRG1), in the regulation of CD4(+) T cells and HBV vaccine responses during HCV infection. We demonstrated that KLRG1 was overexpressed on CD4(+) T cells from HCV-infected, HBV vaccine nonresponders compared with HBV vaccine responders. The capacity of CD4(+) T cells to proliferate and secrete IL-2 cytokine was inversely associated with the level of KLRG1 expression. Importantly, blocking KLRG1 signaling resulted in a significant improvement in CD4(+) T cell proliferation and IL-2 production in HCV-infected, HBV vaccine nonresponders in response to TCR stimulation. Moreover, blockade of KLRG1 increased the phosphorylation of Akt (Ser(473)) and decreased the expression of cell cycle inhibitors p16(ink4a) and p27(kip1), which subsequently enhanced the expression of cyclin-dependent kinase 2 and cyclin E. These results suggest that the KLRG1 pathway impairs CD4(+) T cell responses to neoantigen and induces a state of immune senescence in individuals with HCV infection, raising the possibility that blocking this negative-signaling pathway might improve HBV vaccine responses in the setting of chronic viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Hepatitis B Vaccines/immunology , Hepatitis B/immunology , Hepatitis C/immunology , Lectins, C-Type/genetics , Trans-Activators/genetics , Aging/genetics , Aging/immunology , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Coinfection/genetics , Coinfection/immunology , Cyclin E/genetics , Cyclin E/immunology , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase Inhibitor p16/immunology , Cyclin-Dependent Kinase Inhibitor p27/immunology , Hepacivirus/immunology , Hepatitis B/genetics , Hepatitis B/prevention & control , Hepatitis B virus/immunology , Hepatitis C/genetics , Hepatitis C/virology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Lectins, C-Type/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Immunologic , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/immunology
6.
PLoS One ; 8(11): e81154, 2013.
Article in English | MEDLINE | ID: mdl-24260551

ABSTRACT

BACKGROUND: T cells are essential for the development of uveitis and other autoimmune diseases. After initial activation, CD4+ lymphocytes express the co-stimulatory molecule OX40 that plays an important role in T cell proliferation. Cyclin dependent kinase 2 (CdK2) plays a pivotal role in the cell cycle transition from G1 to S phase. In addition, recent research has implicated CdK2 in T cell activation. Thus, we sought to test the immunosuppressive effect of roscovitine, a potent CdK2 inhibitor, on CD4+ T cell activation, proliferation, and function. DESIGN AND METHODS: Mouse CD4+ T cells were activated by anti-CD3 and anti-CD28 antibodies. The expression of OX40, CD44, and CdK2 were analyzed by flow cytometry. In addition, cell cycle progression and apoptosis of control and roscovitine-treated T lymphocytes were measured by BrdU incorporation and annexin V assay, respectively. Furthermore, the immunoregulatory effect of roscovitine was evaluated in both ovalbumin-induced uveitis and experimental autoimmune uveitis (EAU) models. RESULTS: In this study, we found that T cell activation induced OX40 expression. Cell cycle analysis showed that more CD4+OX40+ cells entered S phase than OX40- T cells. Concurrently, CD4+OX40+ cells had a higher level of CdK2 expression. Roscovitine treatment blocked activated CD4+ cells from entering S phase. In addition, roscovitine not only reduced the viability of CD4+ lymphocytes but also suppressed T cell activation and cytokine production. Finally, roscovitine significantly attenuated the severity of T cell-dependent, OX40-enhanced uveitis. CONCLUSION: These results implicate CdK2 in OX40-augmented T cell response and expansion. Furthermore, this study suggests that roscovitine is a novel, promising, therapeutic agent for treating T cell-mediated diseases such as uveitis.


Subject(s)
Autoimmune Diseases/drug therapy , CD4-Positive T-Lymphocytes/drug effects , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Immunologic Factors/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Uveitis/drug therapy , Animals , Antibodies/pharmacology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , G1 Phase/drug effects , Gene Expression , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/genetics , Receptors, OX40/immunology , Roscovitine , S Phase/drug effects , Uveitis/chemically induced , Uveitis/immunology , Uveitis/pathology
7.
J Biol Chem ; 288(34): 24494-502, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23853094

ABSTRACT

Foxp3 is a transcription factor required for the development of regulatory T cells (Treg). Mice and humans with a loss of Foxp3 function suffer from uncontrolled autoimmunity and inflammatory disease. Expression of Foxp3 is necessary for the anti-inflammatory capacity of Treg, but whether Foxp3 activity is further subject to regulation by extracellular signals is unclear. The primary structure of Foxp3 contains four cyclin-dependent kinase (CDK) motifs (Ser/Thr-Pro) within the N-terminal repressor domain, and we show that CDK2 can partner with cyclin E to phosphorylate Foxp3 at these sites. Consistent with our previous demonstration that CDK2 negatively regulates Treg function, we find that mutation of the serine or threonine at each CDK motif to alanine (S/T→A) results in enhanced Foxp3 protein stability in CD4(+) T cells. T cells expressing the S/T→A mutant of Foxp3 showed enhanced induction (e.g. CD25) and repression (e.g. IL2) of canonical Foxp3-responsive genes, exhibited an increased capacity to suppress conventional T cell proliferation in vitro, and were highly effective at ameliorating colitis in an in vivo model of inflammatory bowel disease. These results indicate that CDK2 negatively regulates the stability and activity of Foxp3 and implicate CDK-coupled receptor signal transduction in the control of regulatory T cell function and stability.


Subject(s)
Cyclin-Dependent Kinase 2/immunology , Forkhead Transcription Factors/immunology , Inflammatory Bowel Diseases/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Amino Acid Motifs , Animals , Cyclin-Dependent Kinase 2/genetics , Disease Models, Animal , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Mutant Strains , Phosphorylation/genetics , Phosphorylation/immunology , Protein Stability , Protein Structure, Tertiary , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology
8.
Immunol Res ; 51(1): 45-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21850539

ABSTRACT

Complement system activation plays an important role in both innate and acquired immunity. Activation of the complement and the subsequent formation of C5b-9 channels (the membrane attack complex) on the cell membranes lead to cell death. However, when the number of channels assembled on the surface of nucleated cells is limited, sublytic C5b-9 can induce cell cycle progression by activating signal transduction pathways and transcription factors and inhibiting apoptosis. This induction by C5b-9 is dependent upon the activation of the phosphatidylinositol 3-kinase/Akt/FOXO1 and ERK1 pathways in a Gi protein-dependent manner. C5b-9 induces sequential activation of CDK4 and CDK2, enabling the G1/S-phase transition and cellular proliferation. In addition, it induces RGC-32, a novel gene that plays a role in cell cycle activation by interacting with Akt and the cyclin B1-CDC2 complex. C5b-9 also inhibits apoptosis by inducing the phosphorylation of Bad and blocking the activation of FLIP, caspase-8, and Bid cleavage. Thus, sublytic C5b-9 plays an important role in cell activation, proliferation, and differentiation, thereby contributing to the maintenance of cell and tissue homeostasis.


Subject(s)
Apoptosis/immunology , Cell Membrane/immunology , Complement Membrane Attack Complex/immunology , MAP Kinase Signaling System/immunology , Animals , BH3 Interacting Domain Death Agonist Protein/immunology , BH3 Interacting Domain Death Agonist Protein/metabolism , CDC2 Protein Kinase , Caspase 8/immunology , Caspase 8/metabolism , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Complement Membrane Attack Complex/metabolism , Cyclin B/immunology , Cyclin B/metabolism , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinases , Forkhead Box Protein O1 , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , G1 Phase/immunology , Humans , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Proteins/immunology , Muscle Proteins/metabolism , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , S Phase/immunology , bcl-Associated Death Protein/immunology , bcl-Associated Death Protein/metabolism
9.
Immunity ; 34(1): 24-38, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21194982

ABSTRACT

RAPL (an alternative spliced form of Rassf5) is a critical Ras-related protein1 (Rap1) effector that regulates lymphocyte adhesion. Here, we have shown that in addition to this previously described function, RAPL also negatively controls lymphocyte proliferation and prevents autoimmunity and lymphoma. RAPL-deficient mice experienced age-related lupus-like glomerulonephritis and developed B cell lymphomas. RAPL-deficient lymphocytes showed hyperproliferation by enhanced S phase entry after antigen receptor ligation. Compared to wild-type cells, RAPL-deficient naive lymphocytes had a 2- to 3-fold increase in Cdk2 kinase activity with a cytoplasmic mislocalization of the cyclin-dependent kinase inhibitor p27(kip1). RAPL was found to suppress the phosphorylation of p27(kip1) on serine 10 (S10) and promoted p27(kip1) nuclear translocation. An S10A mutation in p27(kip1) corrected its cytoplasmic accumulation, reduced hyperproliferation in RAPL-deficient lymphocytes, and suppressed glomerulonephritis and development of B cell lymphoma. Thus, RAPL serves as a checkpoint for S phase entry to prevent lymphoproliferative disorders through the spatial regulation of p27(kip1).


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , rap1 GTP-Binding Proteins/genetics , Animals , Autoimmunity/genetics , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/immunology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lymphocytes/immunology , Mice , Mice, Knockout , Mutation/genetics , Phosphorylation/genetics , Protein Transport/genetics , Protein Transport/immunology , rap1 GTP-Binding Proteins/immunology , rap1 GTP-Binding Proteins/metabolism
10.
Immunol Lett ; 127(2): 93-9, 2010 Jan 04.
Article in English | MEDLINE | ID: mdl-19854217

ABSTRACT

Accumulating data suggested that functional TLR9 was expressed in various tumor cells and TLR9 signaling could enhance the progression of tumor cells. However, the underlying mechanism of TLR9 signaling on the progression of tumors cells remains largely undefined. Our previous study demonstrated that the TLR9 agonist CpG ODNs could significantly enhance the progression of human lung cancer cells in vivo. Here we further evaluated the direct effect of CpG ODNs on the proliferation and cell cycle of human lung cancer cells. Our data showed that TLR9 agonist CpG ODNs could robustly elevate the proliferation and stimulate cell cycle entry of 95D cells in vitro, accompanied by the selectively up-regulated expression of CDK2. Furthermore, we found that down-regulation of CDK2 expression using siRNA against CDK2 could significantly inhibit the enhanced proliferation of 95D cells induced by CpG ODNs. Finally, we investigated that the CpG ODNs could selectively enhance the promoter activity of CDK2. Our findings indicated that TLR9 signaling could selectively up-regulate the expression of CDK2, which was critical for the enhanced proliferation of human lung cancer cells. Our results might provide novel insight into the understanding of functional expression of TLR9 on the progression of tumor cells.


Subject(s)
Cyclin-Dependent Kinase 2/biosynthesis , Lung Neoplasms/immunology , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/agonists , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 9/immunology
11.
J Proteome Res ; 7(4): 1445-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345606

ABSTRACT

Cellular immune mechanisms detect and destroy cancerous and infected cells via the human leukocyte antigen (HLA) class I molecules that present peptides of intracellular origin on the surface of all nucleated cells. The identification of novel, tumor-specific epitopes is a critical step in the development of immunotherapeutics for breast cancer. To directly identify peptide epitopes unique to cancerous cells, secreted human class I HLA molecules (sHLA) were constructed by deletion of the transmembrane and cytoplasmic domain of HLA A*0201. The resulting sHLA-A*0201 was transferred and expressed in breast cancer cell lines MCF-7, MDA-MB-231, and BT-20 as well as in the immortal, nontumorigenic cell line MCF10A. Stable transfectants were seeded into bioreactors for production of > 25 mg of sHLA-A*0201. Peptides eluted from affinity purified sHLA were analyzed by mass spectroscopy. Comparative analysis of HLA-A*0201 peptides revealed 5 previously uncharacterized epitopes uniquely presented on breast cancer cells. These peptides were derived from intracellular proteins with either well-defined or putative roles in breast cancer development and progression: Cyclin Dependent Kinase 2 (Cdk2), Ornithine Decarboxylase (ODC1), Kinetochore Associated 2 (KNTC2 or HEC1), Macrophage Migration Inhibitory Factor (MIF), and Exosome Component 6 (EXOSC6). Cellular recognition of the MIF, KNTC2, EXOSC6, and Cdk2 peptides by circulating CD8+ cells was demonstrated by tetramer staining and IFN-gamma ELISPOT. The identification and characterization of peptides unique to the class I of breast cancer cells provide putative targets for the development of immune diagnostic tools and therapeutics.


Subject(s)
Breast Neoplasms/immunology , Epitopes, T-Lymphocyte/analysis , HLA-A Antigens/immunology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase 2/metabolism , Cytoskeletal Proteins , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/isolation & purification , Exoribonucleases/immunology , Exoribonucleases/metabolism , Female , HLA-A Antigens/chemistry , HLA-A2 Antigen , Humans , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Mass Spectrometry , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Ornithine Decarboxylase/immunology , Ornithine Decarboxylase/metabolism
12.
Biochemistry ; 44(46): 15287-95, 2005 Nov 22.
Article in English | MEDLINE | ID: mdl-16285732

ABSTRACT

Hsp90 and its cochaperone Cdc37 cooperate to provide requisite support to numerous protein kinases involved in cellular signal transduction. In this report, we studied the interactions of Hsp90 and Cdc37 with the cyclin-dependent kinase, Cdk2. Treatment of K562 cells with the Hsp90 inhibitor, geldanamycin, caused a 75% reduction in Cdk2 levels and reduced the levels of its activating kinase, Cdk7, by more than 60%, suggesting that both of these kinases may be Hsp90 clients. Using classical pull-down assays and the Hsp90 inhibitory agents geldanamycin and molybdate, Cdk2 is shown to be a genuine client of the Hsp90 chaperone complex. Subsequently, pull-down assays directed at helix alphaC of Cdk2 are shown to disrupt Hsp90 and Cdc37 binding and explain the initial difficulties in demonstrating these interactions. Mutant constructs containing deletions of secondary structural elements from the N- and C-termini of Cdk2 were prepared and assayed for their ability to coadsorb Hsp90 and Cdc37 in a salt-stable high-affinity manner with and without the addition of molybdate. Consistent with similar work done with the cyclin-dependent kinase relative Cdk4, the presence of the G-box motif of Cdk2 was shown to be critical for Cdc37 binding, whereas consistent with work done with the Src-family tyrosine kinase Lck, the presence of helix alphaC and the stabilization of helix alphaE were shown to be needed for Hsp90 binding.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Animals , Benzoquinones , Chaperonins , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/immunology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , K562 Cells , Lactams, Macrocyclic , Quinones/pharmacology , Rabbits , Reticulocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL