Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Exp Med ; 214(5): 1453-1469, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28400474

ABSTRACT

T helper type 17 cells (Th17 cells) are major contributors to many autoimmune diseases. In this study, we demonstrate that the germinal center kinase family member MINK1 (misshapen/NIK-related kinase 1) negatively regulates Th17 cell differentiation. The suppressive effect of MINK1 on induction of Th17 cells is mediated by the inhibition of SMAD2 activation through direct phosphorylation of SMAD2 at the T324 residue. The importance of MINK1 to Th17 cell differentiation was strengthened in the animal model of experimental autoimmune encephalomyelitis (EAE). Moreover, we show that the reactive oxygen species (ROS) scavenger N-acetyl cysteine boosts Th17 cell differentiation in a MINK1-dependent manner and exacerbates the severity of EAE. Thus, we have not only established MINK1 as a critical regulator of Th17 cell differentiation, but also clarified that accumulation of ROS may limit the generation of Th17 cells. The contribution of MINK1 to ROS-regulated Th17 cell differentiation may suggest an important mechanism for the development of autoimmune diseases influenced by antioxidant dietary supplements.


Subject(s)
Protein Serine-Threonine Kinases/physiology , Th17 Cells/physiology , Animals , Cell Differentiation/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Male , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Smad2 Protein/physiology
2.
Nat Commun ; 7: 11121, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27079169

ABSTRACT

The anterior pituitary harbours five distinct hormone-producing cell types, and their cellular differentiation is a highly regulated and coordinated process. Here we show that ZBTB20 is essential for anterior pituitary development and lactotrope specification in mice. In anterior pituitary, ZBTB20 is highly expressed by all the mature endocrine cell types, and to some less extent by somatolactotropes, the precursors of prolactin (PRL)-producing lactotropes. Disruption of Zbtb20 leads to anterior pituitary hypoplasia, hypopituitary dwarfism and a complete loss of mature lactotropes. In ZBTB20-null mice, although lactotrope lineage commitment is normally initiated, somatolactotropes exhibit profound defects in lineage specification and expansion. Furthermore, endogenous ZBTB20 protein binds to Prl promoter, and its knockdown decreases PRL expression and secretion in a lactotrope cell line MMQ. In addition, ZBTB20 overexpression enhances the transcriptional activity of Prl promoter in vitro. In conclusion, our findings point to ZBTB20 as a critical regulator of anterior pituitary development and lactotrope specification.


Subject(s)
Cell Lineage/genetics , Lactotrophs/metabolism , Pituitary Gland, Anterior/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Hypopituitarism/genetics , Hypopituitarism/metabolism , Hypothalamus/embryology , Hypothalamus/growth & development , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Lactotrophs/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pituitary Gland, Anterior/embryology , Pituitary Gland, Anterior/growth & development , Prolactin/genetics , Prolactin/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
3.
Cell Mol Immunol ; 11(2): 150-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24362470

ABSTRACT

The combination of immunotherapy and chemotherapy is regarded as a promising approach for the treatment of certain types of cancer. However, the underlying mechanisms need to be fully investigated to guide the design of more efficient protocols for cancer chemoimmunotherapy. It is well known that danger-associated molecular patterns (DAMPs) can activate immune cells, including dendritic cells (DCs), via Toll-like receptors (TLRs); however, the role of DAMPs released from chemical drug-treated tumor cells in the activation of the immune response needs to be further elucidated. Here, we found that colorectal cancer (CRC) cells treated with oxaliplatin (OXA) and/or 5-fluorouracil (5-Fu) released high levels of high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). After OXA/5-Fu therapy, the sera of CRC patients also exhibited increased levels of HMGB1 and HSP70, both of which are well-known DAMPs. The supernatants of dying CRC cells treated with OXA/5-Fu promoted mouse and human DC maturation, with upregulation of HLA-DR, CD80 and CD86 expression and enhancement of IL-1ß, TNF-α, MIP-1α, MIP-1ß, RANTES and IP-10 production. Vaccines composed of DCs pulsed with the supernatants of chemically stressed CRC cells induced a more significant IFN-γ-producing Th1 response both in vitro and in vivo. However, the supernatants of chemically stressed CRC cells failed to induce phenotypic maturation and cytokine production in TLR4-deficient DCs, indicating an essential role of TLR4 in DAMP-induced DC maturation and activation. Furthermore, pulsing with the supernatants of chemically stressed CRC cells did not efficiently induce an IFN-γ-producing Th1 response in TLR4-deficient DCs. Collectively, these results demonstrate that DAMPs released from chemically stressed cancer cells can activate DCs via TLR4 and enhance the induction of an anti-tumor T-cell immune response, delineating a clinically relevant immuno-adjuvant pathway triggered by DAMPs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cancer Vaccines , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Dendritic Cells/immunology , Immunotherapy/methods , T-Lymphocytes/immunology , Toll-Like Receptor 4/metabolism , Animals , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cytokines/metabolism , DNA Damage/immunology , Dendritic Cells/drug effects , Fluorouracil/administration & dosage , HMGB1 Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Staging , Neoplasm Transplantation , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Toll-Like Receptor 4/genetics , Tumor Burden , Vaccination
4.
J Biol Chem ; 288(34): 24600-8, 2013 08 23.
Article in English | MEDLINE | ID: mdl-23857586

ABSTRACT

The molecular mechanisms that fine-tune the Toll-like receptor (TLR)-triggered innate immune response need further investigation. As an important transcription factor, zinc finger proteins (ZFPs) play important roles in many cell functions, including development, differentiation, tumorigenesis, and functions of the immune system. However, the role of ZFP members in the innate immune responses remains unclear. Here we showed that the expression of C2H2-type ZFP, ZFP64, was significantly up-regulated in macrophages upon stimulation with TLR ligands, including LPS, CpG oligodeoxynucleotides, or poly(I:C). ZFP64 overexpression promoted TLR-triggered TNF-α, IL-6, and IFN-ß production in macrophages. Coincidently, knockdown of ZFP64 expression significantly inhibited the production of the above cytokines. However, activation of MAPK and IRF3 was not responsible for the ZFP64-mediated promotion of cytokine production. Interestingly, ZFP64 significantly up-regulated TLR-induced NF-κB activation. ZFP64 could bind to the promoter of the TNF-α, IL-6, and IFN-ß genes in macrophages only after TLR ligation. Furthermore, ZFP64 associated with the NF-κB p65 subunit upon LPS stimulation, and TLR-ligated macrophages showed a lower level of p65 recruitment to the TNF-α, IL-6, and IFN-ß gene promoter in the absence of ZFP64. The data identify ZFP64 as a downstream positive regulator of TLR-initiated innate immune responses by associating with the NF-κB p65 subunit, enhancing p65 recruitment to the target gene promoters and increasing p65 activation and, thus, leading to the promotion of TLR-triggered proinflammatory cytokine and type I interferon production. Our findings add mechanistic insight into the efficient activation of the TLR innate response against invading pathogens.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-beta/biosynthesis , Macrophages, Peritoneal/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Adjuvants, Immunologic/pharmacology , Animals , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interferon Inducers/pharmacology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Oligodeoxyribonucleotides/pharmacology , Poly I-C/pharmacology , Promoter Regions, Genetic , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Transcription Factor RelA/genetics , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
5.
J Biol Chem ; 288(23): 16225-16234, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23609450

ABSTRACT

Sepsis-associated immunosuppression (SAIS) is regarded as one of main causes for the death of septic patients at the late stage because of the decreased innate immunity with a more opportunistic infection. LPS-tolerized macrophages, which are re-challenged by LPS after prior exposure to LPS, are regarded as the common model of hypo-responsiveness for SAIS. However, the molecular mechanisms of endotoxin tolerance and SAIS remain to be fully elucidated. In addition, negative regulation of the Toll-like receptor (TLR)-triggered innate inflammatory response needs further investigation. Here we show that expression of immune responsive gene 1 (IRG1) was highly up-regulated in the peripheral blood mononuclear cells of septic patients and in LPS-tolerized mouse macrophages. IRG1 significantly suppressed TLR-triggered production of proinflammatory cytokines TNF-α, IL-6, and IFN-ß in LPS-tolerized macrophages, with the elevated expression of reactive oxygen species (ROS) and A20. Moreover, ROS enhanced A20 expression by increasing the H3K4me3 modification of histone on the A20 promoter domain, and supplement of the ROS abrogated the IRG1 knockdown function in breaking endotoxin tolerance by increasing A20 expression. Our results demonstrate that inducible IRG1 promotes endotoxin tolerance by increasing A20 expression through ROS, indicating a new molecular mechanism regulating hypoinflammation of sepsis and endotoxin tolerance.


Subject(s)
DNA-Binding Proteins/immunology , Gene Expression Regulation/drug effects , Hydro-Lyases/immunology , Immune Tolerance/drug effects , Immunity, Innate/drug effects , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/toxicity , Macrophages/immunology , Nuclear Proteins/immunology , Proteins/immunology , Reactive Oxygen Species/immunology , Sepsis/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Carboxy-Lyases , Cysteine Endopeptidases , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Histones/genetics , Histones/immunology , Humans , Hydro-Lyases/genetics , Interferon-beta/genetics , Interferon-beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/immunology , Macrophages/pathology , Male , Mice , Monocytes/immunology , Monocytes/pathology , Nuclear Proteins/genetics , Proteins/genetics , Sepsis/genetics , Sepsis/pathology , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Ubiquitin-Protein Ligases/genetics
6.
Am J Pathol ; 180(6): 2375-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22538089

ABSTRACT

IL-10-producing CD1d(hi)CD5(+) B cells, also known as B10 cells, have been shown to possess a regulatory function in the inhibition of immune responses, but whether and how B10 cells suppress the development of autoimmune arthritis remain largely unclear. In this study, we detected significantly decreased numbers of IL-10-producing B cells, but increased IL-17-producing CD4(+) T (Th17) cells in both spleen and draining lymph nodes of mice during the acute stage of collagen-induced arthritis (CIA) when compared with adjuvant-treated control mice. On adoptive transfer of in vitro expanded B10 cells, collagen-immunized mice showed a marked delay of arthritis onset with reduced severity of both clinical symptoms and joint damage, accompanied by a substantial reduction in the number of Th17 cells. To determine whether B10 cells directly inhibit the generation of Th17 cells in culture, naive CD4(+) T cells labeled with carboxyfluorescein succinimidyl ester (CFSE) were co-cultured with B10 cells. These B10 cells suppressed Th17 cell differentiation via the reduction of STAT3 phosphorylation and retinoid-related orphan receptor γt (RORγt) expression. Moreover, Th17 cells showed significantly decreased proliferation when co-cultured with B10 cells. Although adoptive transfer of Th17 cells triggered the development of collagen-induced arthritis in IL-17(-/-)DBA/1J mice, co-transfer of B10 cells with Th17 cells profoundly delayed the onset of arthritis. Thus, our findings suggest a novel regulatory role of B10 cells in arthritic progression via the suppression of Th17 cell generation.


Subject(s)
Arthritis, Experimental/prevention & control , B-Lymphocyte Subsets/immunology , Interleukin-10/biosynthesis , Th17 Cells/immunology , Adoptive Transfer/methods , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocyte Subsets/transplantation , Cell Differentiation/immunology , Cells, Cultured , Interleukin-17/biosynthesis , Interleukin-17/deficiency , Lymph Nodes/immunology , Lymphocyte Transfusion/methods , Mice , Mice, Inbred DBA , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation/immunology , STAT3 Transcription Factor/metabolism , Spleen/immunology
7.
Cell Mol Immunol ; 7(4): 296-305, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20418899

ABSTRACT

Here, we investigated the antitumor effect of adenovirus-mediated gene transfer of LIGHT, the tumor-necrosis factor (TNF) superfamily member also known as TNFSF14, in the murine A20 B-cell lymphoma. LIGHT gene modification resulted in upregulated expression of Fas and the accessory molecule--intercellular adhesion molecule-1 (ICAM-1) on A20 cells and led to enhanced A20 cell apoptosis. LIGHT-modified A20 cells effectively stimulated the proliferation of T lymphocytes and interferon (IFN)-gamma production in vitro. Immunization of BALB/c mice with a LIGHT-modified A20 cell vaccine efficiently elicited protective immunity against challenge with the parental tumor cell line. Adenovirus-mediated gene transfer of LIGHT by intratumoral injection exerted a very potent antitumor effect against pre-existing A20 cell lymphoma in BALB/c mice. This adenovirus-mediated LIGHT therapy induced substantial splenic natural killer (NK) and cytotoxic T lymphocyte (CTL) activity, enhanced tumor infiltration by inflammatory cells and increased chemokine expression of CC chemokine ligand 21 (CCL21), IFN-inducible protein-10 (IP-10) and monokine induced by IFN-gamma (Mig) from tumor tissues. Thus, adenovirus-mediated LIGHT therapy might have potential utility for the prevention and treatment of B-cell lymphoma.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Immunity/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokines/metabolism , Female , Gene Transfer Techniques , Humans , Immunization , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/prevention & control , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/immunology
9.
Mol Immunol ; 44(10): 2686-96, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17223196

ABSTRACT

Inhibition of dendritic cell (DC) migration into tissues and secondary lymphoid organs is an efficient way to induce immunosuppression and tolerance. CCR7 and PGE(2) are critical for DC migration to secondary lymphoid organs where DC initiate immune response. Triptolide, an active component purified from the medicinal plant Tripterygium Wilfordii Hook F., is a potent immunosuppressive drug capable of prolonging allograft survival in organ transplantation by inhibiting T cell activation and proliferation. Considering the essential role in T cell tolerance of DC migration to secondary lymphoid organs, here we demonstrate that triptolide can significantly inhibit LPS-triggered upregulation of CCR7 expression and PGE(2) production by inhibiting cyclooxygenase-2 (COX-2) expression in DC, thus impairing DC migration towards CCR7 ligand CCL19/MIP-3betain vitro. Moreover, triptolide-treated DC display impaired migration into secondary lymphoid organs and in vivo administration of triptolide also inhibits DC migration. Further studies show that the triptolide-mediated inhibitory effects of LPS-induced activation of phosphatidylinositol-3 kinase (PI3-K)/Akt and nuclear NF-kappaB activation are involved in down-regulation of COX-2 and CCR7 expression resulting in impaired migration to secondary lymphoid organs of DC. Therefore, inhibition of DC migration through decreasing COX-2 and CCR7 expression via PI3-K/Akt and NF-kappaB signal pathways provides additional mechanistic explanation for triptolide's immunosuppressive effect.


Subject(s)
Cell Movement/drug effects , Cyclooxygenase 2/metabolism , Dendritic Cells/drug effects , Diterpenes/pharmacology , Immunosuppressive Agents/pharmacology , Phenanthrenes/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Animals , Chemokine CCL19 , Chemokines, CC/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Epoxy Compounds/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, CCR7 , Receptors, Chemokine/metabolism
10.
Biochem Biophys Res Commun ; 345(3): 1122-30, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-16713992

ABSTRACT

Triptolide, an active component purified from the medicinal plant Tripterygium wilfordii Hook F., is potent in anti-inflammation and immunosuppression. Dendritic cells (DC), one of important targets of immunosuppressants, play crucial roles in linking the innate immunity and adaptive immunity. However, the effects of triptolide on DC have not been fully elucidated. Chemoattraction of neutrophils and T cells by DC may favor their interactions and initiation of immune response. Here we demonstrate that triptolide significantly impairs DC-mediated chemoattraction of neutrophils and T cells both in vitro and in vivo by suppressing DC production of CC and CXC chemokines including MIP-1alpha, MIP-1beta, MCP-1, RANTES, TARC, and IP-10 in response to LPS. Furthermore, triptolide-mediated inhibition of NF-kappaB activation, Stat3 phosphorylation and increase of SOCS1 expression in DC may be involved in the inhibitory effect of triptolide. Our study provides a novel mechanistic explanation for the anti-inflammatory and immunosuppressive activities of triptolide.


Subject(s)
Dendritic Cells/cytology , Diterpenes/pharmacology , Immunosuppressive Agents/pharmacology , NF-kappa B/metabolism , Neutrophils/cytology , Phenanthrenes/pharmacology , STAT3 Transcription Factor/metabolism , T-Lymphocytes/cytology , Animals , Anti-Inflammatory Agents/pharmacology , Bone Marrow Cells/cytology , Dendritic Cells/metabolism , Epoxy Compounds , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Repressor Proteins/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/metabolism
11.
Immunol Lett ; 98(1): 91-6, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15790513

ABSTRACT

Sinomenine (SN), an immunnosuppressive compound derived from the Chinese medicinal plant Sinomenium acutum, has been used to treat autoimmune diseases effectively. Previous studies show SN can inhibit lymphocytes proliferation and macrophage production of pro-inflammatory factors. However, little is known about the mechanisms by which SN inhibits macrophage functions. In this study, we demonstrated that SN could inhibit the proliferation of murine macrophages RAW264.7 by inducing apoptosis in a dose- and time-dependent manner. We found activation of extracellular signal-regulated protein kinase (ERK) in SN-treated macrophages, and requirement for ERK activation in SN-induced apoptosis of macrophages. Contemporarily, the expression of p27/KIP1, proapoptotic factor Bax increased, and expression of Bcl-2 decreased, which might cooperate to induce apoptosis. Inhibiting ERK activation reduced the increased expression of p27 and Bax, but had no effect on the decreased expression of Bcl-2, suggesting the involvement of ERK activation in the SN-induced increased expression of p27 and Bax. These results demonstrated that SN could induce apoptosis of macrophages through activation of ERK, and ERK activation might partially involve in the increased expression of p27 and Bax in apoptotic macrophages. Therefore, induction of macrophage apoptosis through ERK activation may be one of mechanisms by which SN exhibits its immunosuppressive function.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunosuppressive Agents/pharmacology , Macrophages/enzymology , Morphinans/pharmacology , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Macrophages/drug effects , Mice
12.
Biochem Biophys Res Commun ; 319(3): 980-6, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15184078

ABSTRACT

Dendritic cells (DCs) are the most potent antigen-presenting cells that play crucial roles in the regulation of immune response. Triptolide, an active component purified from the medicinal plant Tripterygium wilfordii Hook F., has been demonstrated to act as a potent immunosuppressive drug capable of inhibiting T cell activation and proliferation. However, little is known about the effects of triptolide on DCs. The present study shows that triptolide does not affect phenotypic differentiation and LPS-induced maturation of murine DCs. But triptolide can dramatically reduce cell recovery by inducing apoptosis of DCs at concentration as low as 10ng/ml, as demonstrated by phosphatidylserine exposure, mitochondria potential decrease, and nuclear DNA condensation. Triptolide induces activation of p38 in DCs, which precedes the activation of caspase 3. SB203580, a specific kinase inhibitor for p38, can block the activation of caspase 3 and inhibit the resultant apoptosis of DCs. Our results suggest that the anti-inflammatory and immunosuppressive activities of triptolide may be due, in part, to its apoptosis-inducing effects on DCs.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Dendritic Cells/drug effects , Diterpenes/pharmacology , Immunosuppressive Agents/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Phenanthrenes/pharmacology , Animals , Caspase 3 , Cell Differentiation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Enzyme Activation , Epoxy Compounds , Male , Mice , Mice, Inbred C57BL , Phenotype , Phosphorylation , p38 Mitogen-Activated Protein Kinases
13.
J Cell Biochem ; 88(5): 932-40, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12616532

ABSTRACT

The enzyme, glucosamine-6-phosphate isomerase (GNPI) or deaminase (GNPDA) (EC 5.3.1.10), catalyzes the conversion of GNP to fructose-6-phosphate and ammonia, with an aldo/keto isomerization and an amination/deamination. A hamster sperm-derived protein (Oscillin) with high similarity to bacterial GNPI has been proved to be capable of inducing calcium oscillation in eggs at fertilization. GNPI/Oscillin was supposed to be an important factor in starting embryonic development. From the cDNA library of human dendritic cells (DC), we isolated a novel full-length cDNA encoding a 276-amino acid-residue protein that shares high homology with human GNPI/Oscillin. So, the novel molecule is named as GNPI2. The GNPI2 gene consists of seven exons and six introns. It is mapped to chromosome 4. Northern blot analysis indicated that the tissue distribution of GNPI2 mRNA is different from that of human GNPI or Oscillin mRNA. GNPI2 is ubiquitously expressed in most of human tissues with high expression in testis, ovary, placenta, and heart. Like GNPI, the recombinant GNPI2 has been proved to have the enzymatic activity to catalyze the conversion of GNP to fructose-6-phosphate. Our results indicated that GNPI2 is a novel protein with definite function as a GNPI.


Subject(s)
Aldose-Ketose Isomerases/biosynthesis , Aldose-Ketose Isomerases/chemistry , Aldose-Ketose Isomerases/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Calcium-Binding Proteins , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/chemistry , Fructosephosphates/biosynthesis , Humans , Molecular Sequence Data , Proteins/chemistry , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Sequence Homology
SELECTION OF CITATIONS
SEARCH DETAIL