Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Front Immunol ; 14: 1133111, 2023.
Article in English | MEDLINE | ID: mdl-37234153

ABSTRACT

Modulation of surface T cell antigen receptor (TCR) expression is crucial for proper T cell development and maintenance of mature T cell function at steady state and upon stimulation. We previously determined that CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule that served as a potential member of the γc cytokine family, contributes to antitumor responses by augmenting CD8+ T cell-mediated immunity. Here we show that T cell-specific deletion of Ccdc134 decreased peripheral mature CD4+ and CD8+ T cells, which resulted in impaired T cell homeostasis. Moreover, Ccdc134-deficient T cells exhibited an attenuated response to TCR stimulation in vitro, showing lower activation and proliferative capacity. This was further reflected in vivo, rendering mice refractory to T cell-mediated inflammatory and antitumor responses. More importantly, CCDC134 is associated with TCR signaling components, including CD3ϵ, and attenuated TCR signaling in Ccdc134-deficient T cells via altered CD3ϵ ubiquitination and degradation. Taken together, these findings suggest a role for CCDC134 as a positive regulator of TCR-proximal signaling and provide insight into the cell-intrinsic functional consequences of Ccdc134 deficiency in the attenuation of T cell-mediated inflammatory and antitumor responses.


Subject(s)
CD8-Positive T-Lymphocytes , Signal Transduction , Mice , Animals , Receptors, Antigen, T-Cell/metabolism , Lymphocyte Activation , Cytokines/metabolism
2.
Environ Sci Pollut Res Int ; 29(39): 59317-59327, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35384541

ABSTRACT

Most studies have shown that improper disposal of e-waste can accelerate the release of high concentrations of polybrominated diphenyl ethers (PBDEs), and this situation causes environmental pollution and human health risks. The recycling technology of waste electronic plastics based on solvent processes can reduce environmental pollution and health risks from PBDEs. In this study, high impact polystyrene (HIPS) from waste TV sets was taken as the research object, and d-limonene and n-propanol were used as solvent and precipitant, respectively. We studied the relationship between the precipitation conditions and the size of precipitate particles, and the effect laws of precipitation conditions on the removal percentage of PBDEs were discussed. Transferring behavior of PBDEs during precipitation was investigated, and the parameters suitable for removing PBDEs from HIPS solution were confirmed. Results showed that lower HIPS concentration in d-limonene, lower precipitation temperature, higher mass ratio of n-propanol to HIPS solution, and greater stirring speed were conducive to form smaller and more uniform precipitate particles. All conditions (concentration, temperature, mass ratio, and stirring rate) that could increase the solubility of PBDEs in the mixed solvent of limonene and n-propanol or decrease the swelling degree of HIPS precipitate particles, or reduce the size of particles could improve the removal percentage of PBDEs. The investigated results indicated that insoluble PBDEs (e.g., decabromodiphenyl ether) transferred into the HIPS precipitate mainly through the generated crystals and then precipitated together with the HIPS particles, and soluble PBDEs (e.g., octabromodiphenyl ether) migrated into the precipitate by the solution entrained. The precipitate particles, which measured approximately 1.0 mm (on average), were obtained when the solution containing 10% of HIPS from waste TV shell was precipitated by adding n-propanol equivalent to twice the mass of the solution at 40 °C and 3000 r/min stirring speed. The total concentration of PBDEs in the precipitate particles (dried) was reduced to 2369 mg/kg, and 88.06% of the PBDEs in the original plastic solution was successfully removed by this process.


Subject(s)
Electronic Waste , Halogenated Diphenyl Ethers , 1-Propanol , Electronic Waste/analysis , Environmental Monitoring , Halogenated Diphenyl Ethers/analysis , Humans , Limonene , Plastics/chemistry , Polystyrenes , Solvents
3.
Cancer Cell Int ; 21(1): 510, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34560882

ABSTRACT

BACKGROUND: Chordomas are rare, slow-growing and locally aggressive bone sarcomas. At present, chordomas are difficult to manage due to their high recurrence rate, metastasis tendency and poor prognosis. The underlying mechanisms of chordoma tumorigenesis and progression urgently need to be explored to find the effective therapeutic targets. Our previous data demonstrates that EGFR plays important roles in chordoma development and CKLF-like MARVEL transmembrane domain containing (CMTM)3 suppresses gastric cancer metastasis by inhibiting the EGFR/STAT3/EMT signaling pathway. However, the roles and mechanism of CMTM3 in chordomas remain unknown. METHODS: Primary chordoma tissues and the paired adjacent non-tumor tissues were collected to examine the expression of CMTM3 by western blot. The expression of CMTM3 in chordoma cell lines was tested by Real-time PCR and western blot. CCK-8 and colony forming unit assay were performed to delineate the roles of CMTM3 in cell proliferation. Wound healing and Transwell assays were performed to assess cell migration and invasion abilities. A xenograft model in NSG mice was used to elucidate the function of CMTM3 in vivo. Signaling pathways were analyzed by western blot and IHC. RNA-seq was performed to further explore the mechanism regulated by CMTM3 in chordoma cells. RESULTS: CMTM3 expression was downregulated in chordoma tissues compared with paired normal tissues. CMTM3 suppressed proliferation, migration and invasion of chordoma cells in vitro and inhibited tumor growth in vivo. CMTM3 accelerated EGFR degradation, suppressed EGFR/STAT3/EMT signaling pathway, upregulated TP53 expression and enriched the TP53 signaling pathway in chordoma cells. CONCLUSIONS: CMTM3 inhibited tumorigenesis and development of chordomas through activating the TP53 signaling pathway and suppressing the EGFR/STAT3 signaling pathway, which suppressed EMT progression. CMTM3 might be a potential therapeutic target for chordomas.

4.
Cell Immunol ; 352: 104100, 2020 06.
Article in English | MEDLINE | ID: mdl-32305130

ABSTRACT

B-1a cells represent a distinct B cell population with unique phenotype, self-renewing capacity and restricted Igµ repertoire. They primarily locate in body cavity and also exist in spleen. The different subpopulations of B-1a cells are heavily affected by local environment. Our previous studies revealed that MARVEL-domain-containing membrane protein, CMTM7, was involved in B-1a cell development. Here, we focused its influence on peritoneal and splenic B-1a cells. Unlike peritoneal B-1a cells, we found that splenic Cmtm7-/- B-1a cells expressed higher level of CD5, CD80 and CD86 compared with WT counterparts. They also exhibited an enhanced tonic BCR signals in steady state. Though the cell viability was unaffected in vitro, Cmtm7 knockout markedly promoted splenic B-1a cell apoptosis in situ, which was likely associated with down-regulation of Il-5rα. With regard to Igµ repertoire, peritoneal and splenic Cmtm7-/- B-1a cells exhibit similar changes exemplified by the loss of VH11 and gain of VH12, whereas an increase in VH1 usage and skewed J segments from JH1 to JH2 and JH4 families could only be detected within splenic Cmtm7-/- B-1a cells. Overall, these data indicate that Cmtm7 functions differently in peritoneal and splenic B-1a cells and plays a more important role in splenic cells.


Subject(s)
B-Lymphocyte Subsets/metabolism , Chemokines/metabolism , MARVEL Domain-Containing Proteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Apoptosis/immunology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Cell Membrane/metabolism , Cell Proliferation , Chemokines/immunology , Female , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Spleen/immunology , Spleen/pathology
5.
Eur J Immunol ; 50(6): 809-821, 2020 06.
Article in English | MEDLINE | ID: mdl-32022930

ABSTRACT

Terminal differentiation of B cells into antibody-secreting cells is the foundation of humoral immune response. B-1 cells, which are different from B-2 cells, preferentially differentiate into plasma cells. CMTM7 is a MARVEL-domain-containing membrane protein predominantly expressed in B cells that plays an important role in B-1a cell development. The present study assessed CMTM7 function in response to antigen stimulation. Following immunization with T cell-dependent and T cell-independent antigens, Cmtm7-deficient mice exhibited decreased IgM but normal IgG responses in vivo. In vitro stimulation with LPSs induced Cmtm7-/- B-1 cell activation, whereas proliferation was marginally reduced. Notably, Cmtm7 deficiency markedly suppressed plasma cell differentiation in response to TLR agonists, accompanied by a decrease in IgM and IL-10 production. At the molecular level, loss of Cmtm7 repressed the downregulation of Pax5 and the upregulation of Xbp1, Irf4, and Prdm1. Furthermore, p38 phosphorylation was inhibited in Cmtm7-/- B-1 cells. Experiments using a p38 inhibitor revealed that p38 activation was essential for the terminal differentiation of B-1 cells, suggesting that Cmtm7 contributes to B-1 cell differentiation by maintaining p38 activation. Overall, the data reveal the crucial functions of CMTM7 in TLR-induced terminal differentiation and p38 activation in B-1 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Chemokines/immunology , MAP Kinase Signaling System/immunology , MARVEL Domain-Containing Proteins/immunology , Plasma Cells/immunology , Toll-Like Receptors/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Chemokines/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-10/genetics , Interleukin-10/immunology , MAP Kinase Signaling System/genetics , MARVEL Domain-Containing Proteins/genetics , Mice , Mice, Knockout , Plasma Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Toll-Like Receptors/genetics , p38 Mitogen-Activated Protein Kinases/genetics
6.
Cell Discov ; 5: 64, 2019.
Article in English | MEDLINE | ID: mdl-31839985

ABSTRACT

Antibodies have a common structure consisting of two identical heavy (H) and two identical light (L) chains. It is widely accepted that a single mature B cell produces a single antibody through restricted synthesis of only one VHDJH (encoding the H-chain variable region) and one VLJL (encoding the L-chain variable region) via recombination. Naive B cells undergo class-switch recombination (CSR) from initially producing membrane-bound IgM and IgD to expressing more effective membrane-bound IgG, IgA, or IgE when encountering antigens. To ensure the "one cell - one antibody" paradigm, only the constant region of the H chain is replaced during CSR, while the rearranged VHDJH pattern and the L chain are kept unchanged. To define those long-standing classical concepts at the single-cell transcriptome level, we applied the Chromium Single-Cell Immune Profiling Solution and Sanger sequencing to evaluate the Ig transcriptome repertoires of single B cells. Consistent with the "one cell - one antibody" rule, most of the B cells showed one V(D)J recombination pattern. Intriguingly, however, two or more VHDJH or VLJL recombination patterns of IgH chain or IgL chain were also observed in hundreds to thousands of single B cells. Moreover, each Ig class showed unique VHDJH recombination pattern in a single B-cell expressing multiple Ig classes. Together, our findings reveal an unprecedented presence of multi-Ig specificity in some single B cells, implying regulation of Ig gene rearrangement and class switching that differs from the classical mechanisms of both the "one cell - one antibody" rule and CSR.

7.
Acta Biochim Biophys Sin (Shanghai) ; 51(9): 915-924, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31435638

ABSTRACT

CMTM4 (CKLF-like MARVEL transmembrane domain containing 4), a potential tumor suppressor gene, is involved in several types of malignancies. It has been reported to be downregulated and exhibit anti-tumorigenic activities by regulating cell growth and cell cycle in clear cell renal cell carcinoma. It has also been identified as a tumor suppressor in hepatocellular carcinoma (HCC), and its negative expression is a risk factor for poor prognosis of HCC patients. In the present study, an integrated bioinformatics analysis based on The Cancer Genome Atlas (TCGA) database showed that CMTM4 was frequently reduced in colorectal cancer (CRC) and high expression of CMTM4 was associated with increased overall survival rates. Based on these findings, we adopted gain-of-function and lost-of-function strategies using SW480 and HT29 CRC cell lines which have relatively low and high endogenous CMTM4 levels, respectively. We observed impeded cell proliferation and migration upon overexpression of CMTM4 in SW480 cells, and the opposite effects were observed upon knockdown of CMTM4 in HT-29 cells. Cell signaling pathways essential for CRC progression were then examined, and the phosphorylation levels of AKT, ERK1/2, and STAT3 were found to be decreased by CMTM4 overexpression in SW480 cells and elevated by CMTM4 silencing in HT29 cells. Their inhibitors were used to validate that the three signaling pathways contributed to the inhibitory effects of CMTM4 on CRC cells. Taken together, our results suggest that CMTM4 plays a tumor suppressive role in CRC.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , MARVEL Domain-Containing Proteins/physiology , Adenocarcinoma/pathology , Cell Movement , Cell Proliferation , HT29 Cells , Humans , MAP Kinase Signaling System , MARVEL Domain-Containing Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
8.
Int Immunol ; 31(11): 715-728, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31081901

ABSTRACT

Innate-like B-1a cells are an important cell population for production of natural IgM and interleukin-10 (IL-10), and act as the first line against pathogens. We determined that CMTM7 is essential for B-1a cell development. Following Cmtm7 (CKLF-like MARVEL transmembrane domain-containing 7) knockout, B-1a cell numbers decreased markedly in all investigated tissues. Using a bone marrow and fetal liver adoptive transfer model and conditional knockout mice, we showed that the reduction of B-1a cells resulted from B-cell-intrinsic defects. Because of B-1a cell loss, Cmtm7-deficient mice produced less IgM and IL-10, and were more susceptible to microbial sepsis. Self-renewal and homeostasis of mature B-1a cells in Cmtm7-/- mice were not impaired, suggesting the effect of Cmtm7 on B-1a cell development. Further investigations demonstrated that the function of Cmtm7 in B-1a cell development occurred at the specific transitional B-1a (TrB-1a) stage. Cmtm7 deficiency resulted in a slow proliferation and high cell death rate of TrB-1a cells. Thus, Cmtm7 controls B-1a cell development at the transitional stage.


Subject(s)
Chemokines/immunology , MARVEL Domain-Containing Proteins/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , B-Lymphocyte Subsets/immunology , Cell Death , Cell Proliferation , Chemokines/deficiency , MARVEL Domain-Containing Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/immunology
9.
FASEB J ; : fj201800020RRR, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29799787

ABSTRACT

FAM19A1 is a member of the family with sequence similarity 19 with unknown function. FAM19A1 mRNA expression is restricted to the CNS. Here, we report that FAM19A1 is a classic secretory protein, and expression levels correlate with brain development, increasing from embryonic d 12.5, peaking between postnatal d (P)1 and P7 and decreasing at wk 8. The adult hippocampus is a region of FAM19A1 high expression. Recombinant FAM19A1 suppressed the proliferation and self-renewal of neural stem cells (NSCs) and altered the lineage progression of NSCs with promoted neuron differentiation and suppressed astrocyte differentiation. Although GPCR 1 (GPR1) has been reported to be expressed in the CNS, its functions in the brain remain unclear. We identified GPR1 to be a functional receptor for FAM19A1. FAM19A1 interacted with GPR1 via the N-terminal domain (GPR1-ND), and its NSC modulatory functions required the Rho-associated protein kinase (ROCK) /ERK1/2 and ROCK/signal transducer and activator of transcription 3 signaling pathways. GPR1-ND that selectively bound to FAM19A1 neutralized the effects of FAM19A1 on NSC functions. Taken together, our results show, for the first time to our knowledge, that FAM19A1 is a novel regulatory factor of the proliferation and differentiation of NSCs, and identified a novel mechanism by which GPCR mediates the effects of FAM19A1 on NSC functions that may be important for brain development and neurogenesis. Additional exploration of the functions of FAM19A1 and GPR1 in the CNS may broaden the range of therapeutic options available for major brain disorders.-Zheng, C., Chen, D., Zhang, Y., Bai, Y., Huang, S., Zheng, D., Liang, W., She, S., Peng, X., Wang, P., Mo, X., Song, Q., Lv, P., Huang, J., Ye, R. D., Wang, Y. FAM19A1 is a new ligand for GPR1 that modulates neural stem-cell proliferation and differentiation.

10.
Brain Behav Immun ; 71: 158-168, 2018 07.
Article in English | MEDLINE | ID: mdl-29548993

ABSTRACT

CCDC134 (coiled-coil domain containing 134), a cytokine-like molecule, was previously reported to exert antitumor effects by augmenting CD8+ T-cell mediated immunity. However, the dynamic changes in CCDC134 expression patterns in the spinal cord that may be involved in the progression of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, remains unclear. In this study, we found that CCDC134 expression was markedly increased in the spinal cord during the progression of EAE. Furthermore, we demonstrated that CCDC134 significantly reduced the severity and slowed the progression of EAE, which correlated with reduced spinal cord inflammation and demyelination. The underlying mechanism of CCDC134-induced effects involved inhibition of T helper (Th)-1 and Th17 cell differentiation and secretion of its key effector molecules IFN-γ and IL-17A via regulation of JAK/STAT signaling. These findings indicate that CCDC134 exerts potent anti-inflammatory effects through the selective modulation of pathogenic Th1 and Th17 cells by targeting critical signaling pathways. The study provides insights into the role of CCDC134 as a unique therapeutic agent for the treatment of autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Membrane Proteins/pharmacology , Animals , Cell Differentiation/drug effects , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Signal Transduction/drug effects , Spinal Cord/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/physiology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/physiology
11.
Circulation ; 138(1): 48-63, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29453251

ABSTRACT

BACKGROUND: Obesity plays crucial roles in the development of cardiovascular diseases. However, the mechanisms that link obesity and cardiovascular diseases remain elusive. Compelling evidence indicates that adipokines play an important role in obesity-related cardiovascular diseases. Here, we found a new adipokine-named family with sequence similarity 19, member A5 (FAM19A5), a protein with unknown function that was predicted to be distantly related to the CC-chemokine family. We aimed to test whether adipose-derived FAM19A5 regulates vascular pathology on injury. METHODS: DNA cloning, protein expression, purification, and N-terminal sequencing were applied to characterize FAM19A5. Adenovirus infection and siRNA transfection were performed to regulate FAM19A5 expression. Balloon and wire injury were performed in vivo on the rat carotid arteries and mouse femoral arteries, respectively. Bioinformatics analysis, radioactive ligand-receptor binding assays, receptor internalization, and calcium mobilization assays were used to identify the functional receptor for FAM19A5. RESULTS: We first characterized FAM19A5 as a secreted protein, and the first 43 N-terminal amino acids were the signal peptides. Both FAM19A5 mRNA and protein were abundantly expressed in the adipose tissue but were downregulated in obese mice. Overexpression of FAM19A5 markedly inhibited vascular smooth muscle cell proliferation and migration and neointima formation in the carotid arteries of balloon-injured rats. Accordingly, FAM19A5 silencing in adipocytes significantly promoted vascular smooth muscle cell activation. Adipose-specific FAM19A5 transgenic mice showed greater attenuation of neointima formation compared with wild-type littermates fed with or without Western-style diet. We further revealed that sphingosine-1-phosphate receptor 2 was the functional receptor for FAM19A5, with a dissociation constant (Kd) of 0.634 nmol/L. Inhibition of sphingosine-1-phosphate receptor 2 or its downstream G12/13-RhoA signaling circumvented the suppressive effects of FAM19A5 on vascular smooth muscle cell proliferation and migration. CONCLUSIONS: We revealed that a novel adipokine, FAM19A5, was capable of inhibiting postinjury neointima formation via sphingosine-1-phosphate receptor 2-G12/13-RhoA signaling. Downregulation of FAM19A5 during obesity may trigger cardiometabolic diseases.


Subject(s)
Adipose Tissue/metabolism , Cytokines/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima , Receptors, Lysosphingolipid/metabolism , Vascular System Injuries/metabolism , Adipocytes/metabolism , Animals , Calcium Signaling , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cytokines/genetics , Disease Models, Animal , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Obesity/genetics , Obesity/metabolism , Paracrine Communication , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors , Tissue Culture Techniques , Vascular System Injuries/genetics , Vascular System Injuries/pathology , rhoA GTP-Binding Protein/metabolism
12.
Protein Cell ; 9(9): 785-798, 2018 09.
Article in English | MEDLINE | ID: mdl-28536942

ABSTRACT

Leukocyte differentiation antigens (LDAs) play important roles in the immune system, by serving as surface markers and participating in multiple biological activities, such as recognizing pathogens, mediating membrane signals, interacting with other cells or systems, and regulating cell differentiation and activation. Data mining is a powerful tool used to identify novel LDAs from whole genome. LRRC25 (leucine rich repeat-containing 25) was predicted to have a role in the function of myeloid cells by a large-scale "omics" data analysis. Further experimental validation showed that LRRC25 is highly expressed in primary myeloid cells, such as granulocytes and monocytes, and lowly/intermediately expressed in B cells, but not in T cells and almost all NK cells. It was down-regulated in multiple acute myeloid leukemia (AML) cell lines and bone marrow cells of AML patients and up-regulated after all-trans retinoic acid (ATRA)-mediated granulocytic differentiation in AML cell lines and acute promyelocytic leukemia (APL; AML-M3, FAB classification) cells. Localization analysis showed that LRRC25 is a type I transmembrane molecule. Although ectopic LRRC25 did not promote spontaneous differentiation of NB4 cells, knockdown of LRRC25 by siRNA or shRNA and knockout of LRRC25 by the CRISPR-Cas9 system attenuated ATRA-induced terminal granulocytic differentiation, and restoration of LRRC25 in knockout cells could rescue ATRA-induced granulocytic differentiation. Therefore, LRRC25, a potential leukocyte differentiation antigen, is a key regulator of ATRA-induced granulocytic differentiation.


Subject(s)
Antigens, Differentiation/metabolism , Cell Differentiation/drug effects , Granulocytes/cytology , Granulocytes/drug effects , Membrane Proteins/metabolism , Tretinoin/pharmacology , Antigens, Differentiation/immunology , Cell Line, Tumor , Granulocytes/immunology , Granulocytes/metabolism , Humans , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , RNA, Small Interfering/pharmacology
13.
Sci Rep ; 7(1): 5107, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698550

ABSTRACT

Monocytes/macrophages have been found to be an important component of colitis. However, the key chemokine that initiates the CCR2+ monocytes migration from circulation to colitis tissue remains to be undiscovered. PC3-secreted microprotein (PSMP) is a novel chemokine whose receptor is CCR2. The physiological and pathological functions of PSMP have not yet been reported. In this study, PSMP was found to be expressed in colitis and colonic tumor tissues from patients and significantly up-regulated in mouse DSS-induced colitis tissues. PSMP overexpression in the colon aggravated the DSS-induced colitis and the anti-PSMP neutralizing antibody mollified the colitis by reducing macrophage infiltration and inhibiting the expression of IL-6, TNF-α and CCL2. Furthermore, we demonstrated that lipopolysaccharide and muramyl dipeptide induced PSMP expression in the colonic epithelial cells. PSMP was up-regulated in the initial stage prior to IL-6, TNF-α and CCL2 up-regulated expression in DSS colitis and promoted the M1 macrophages to produce CCL2. PSMP chemo-attracted Ly6Chi monocytes in a CCR2 dependent manner via in situ chemotaxis and adoptive transfer assays. Our data identify PSMP as a key molecule in ulcerative colitis, which provides a novel mechanism of monocyte/macrophage migration that affects gut innate immunity and makes PSMP a potential target for controlling colitis.


Subject(s)
Colitis/metabolism , Macrophages/metabolism , Monocytes/metabolism , Neoplasm Proteins/metabolism , Prostatic Secretory Proteins/metabolism , Receptors, CCR2/metabolism , Animals , Cell Movement , Chemokine CCL2/metabolism , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Humans , Interleukin-6/metabolism , Male , Mice , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
14.
Oncoimmunology ; 6(4): e1292195, 2017.
Article in English | MEDLINE | ID: mdl-28507796

ABSTRACT

Identification of novel stimulatory cytokines with antitumor function would have great value in tumor immunotherapy investigations. Here, we report LYG1 (Lysozyme G-like 1) identified through the strategy of Immunogenomics as a novel classical secretory protein with tumor-inhibiting function. LYG1 recombinant protein (rhLYG1) could significantly suppress the growth of B16 tumors in WT B6 mice, but not in SCID-beige mice, Rag1-/- mice, CD4+- or CD8+ T cell-deleted mice. It could increase the number of CD4+ and CD8+ T cells in tumor-infiltrating lymphocytes, tumor-draining lymph nodes, and spleens, and promote IFNγ production by T cells in tumor-bearing mice. In vitro experiments demonstrated that rhLYG1 could directly enhance IFNγ secretion by CD4+ T cells, but not CD8+ T cells. Moreover, it could promote the activation, proliferation, and IFNγ production of tumor antigen-specific CD4+ T cells. The tumor-inhibiting effect of LYG1 was eliminated in Ifng-/- mice. Furthermore, LYG1 deficiency accelerated B16 and LLC1 tumor growth and inhibited the function of T cells. In summary, our findings reveal a tumor-inhibiting role for LYG1 through promoting the activation, proliferation, and function of CD4+ T cells in antitumor immune responses, offering implications for novel tumor immunotherapy.

15.
Appl Immunohistochem Mol Morphol ; 25(2): 122-128, 2017 02.
Article in English | MEDLINE | ID: mdl-26574634

ABSTRACT

Previous studies have demonstrated that overexpression of CMTM8 inhibits cell growth and induces apoptosis in multiple types of cancer cells, whereas the downregulation of CMTM8 induces the epithelial-to-mesenchymal (EMT)-like phenotype in hepatocyte carcinoma cells, implying its important roles in tumorigenesis and tumor metastasis. No extensive studies on the expression of CMTM8 in either normal or tumorous human tissues have been reported to date. Here, using real-time quantitative polymerase chain reaction, we analyzed CMTM8 expression in multiple normal human tissue samples. Moreover, by applying high-throughput immunohistochemical staining of tissue microarrays with homemade anti-CMTM8 antibodies, we studied CMTM8 expression in carcinoma samples and adjacent normal samples of 6 types of human tissues. CMTM8 is widely expressed in many normal human tissues and is frequently downregulated or absent in multiple solid tumors (liver, lung, colon, rectum, esophagus, stomach). χ tests revealed a significant negative correlation between CMTM8 expression and tumorigenesis: liver, lung (squamous carcinoma), colon, rectum, P<0.0001; esophagus, P<0.001; stomach, P<0.01. Real-time quantitative polymerase chain reaction analysis of samples from esophageal carcinomas and the adjacent normal tissues revealed that CMTM8 mRNA levels are reduced in carcinomas compared with normal tissues, indicating that CMTM8 is potentially downregulated at the mRNA level (P<0.01). This is the first extensive study of CMTM8 expression in both normal and tumorous human tissues. Our findings strongly supported the potential role of CMTM8 as a novel tumor suppressor and may shape further functional studies on this gene.


Subject(s)
Chemokines/metabolism , Down-Regulation , MARVEL Domain-Containing Proteins/metabolism , Neoplasms/metabolism , Chemokines/genetics , HeLa Cells , Hep G2 Cells , Humans , Immunohistochemistry , MARVEL Domain-Containing Proteins/genetics , Neoplasms/pathology , RNA, Messenger/genetics
16.
Cancer Lett ; 386: 77-86, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27867015

ABSTRACT

CMTM3 (CKLF-like MARVEL transmembrane domain containing 3), a tumor suppressor gene, is involved in multiple types of malignancies. CMTM3 knockdown promotes metastasis of gastric cancer via the STAT3/Twist1/EMT signaling pathway. Strong epidermal growth factor receptor1 (EGFR) expression is significantly associated with tumor metastasis and poor outcomes of gastric cancer patients. In this paper, we show that CMTM3 suppresses epidermal growth factor (EGF)-mediated migration and STAT3 signaling, downregulates EGFR expression via accelerating EGFR degradation in gastric cancer cells. CMTM3 colocalizes with early endosome markers Rab5 and EEA1. Co-immunoprecipitation (Co-IP) assay further confirms that CMTM3 interacts with Rab5. More importantly, CMTM3 markedly increases Rab5 activity. The suppressive effects of CMTM3 on EGFR expression and EGF-mediated migration can be abrogated by the siRNA against Rab5. Finally, we found that the C-terminal region of CMTM3 plays more important roles in the tumor suppressive effects of CMTM3. Overall, this study demonstrates that CMTM3 decreases EGFR expression, facilitates EGFR degradation, and inhibits the EGF-mediated tumorigenicity of gastric cancer cells via enhancing Rab5 activity.


Subject(s)
Cell Movement/drug effects , Chemokines/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/agonists , MARVEL Domain-Containing Proteins/metabolism , Stomach Neoplasms/enzymology , rab5 GTP-Binding Proteins/metabolism , Chemokines/genetics , Down-Regulation , Endosomes/drug effects , Endosomes/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , MARVEL Domain-Containing Proteins/genetics , Neoplasm Invasiveness , Protein Interaction Domains and Motifs , Proteolysis , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection , Vesicular Transport Proteins/metabolism , rab5 GTP-Binding Proteins/genetics
18.
Oncotarget ; 7(20): 29507-19, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27121055

ABSTRACT

CMTM3 (CKLF-like MARVEL transmembrane domain containing 3) possesses tumor suppressor properties in multiple types of malignancies. Restoration of CMTM3 significantly inhibits the metastasis of gastric cancer, and its expression level is correlated with prognosis. However, the physiological effects and the mechanism of CMTM3 remain unknown. Here, we suppress CMTM3 expression by shRNA to explore its endogenous effects and its mechanism of action in gastric cancer. Stable knockdown of CMTM3 promotes cell migration, invasion and tumor metastasis, increases MMP2 expression and enhances MMP2 activity. CMTM3 inhibits EMT along with the upregulation of E-cadherin and the downregulation of N-cadherin, Vimentin and Twist1. It has no obvious effects on Zeb1 and Snail. CMTM3 suppresses the phosphorylation of STAT3 but not Akt. More importantly, the EMT phenotype and cell migration induced by CMTM3 knockdown can be reversed by the Jak2/STAT3 inhibitor JSI-124 or by siRNA against STAT3 or Twist1. Overall, this study demonstrates that knockdown of CMTM3 promotes the metastasis of gastric cancer through the STAT3/Twist1/EMT pathway.


Subject(s)
Neoplasm Invasiveness/genetics , Nuclear Proteins/metabolism , Signal Transduction/physiology , Stomach Neoplasms/pathology , Twist-Related Protein 1/metabolism , Animals , Cell Line, Tumor , Chemokines/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Heterografts , Humans , MARVEL Domain-Containing Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
19.
J Cell Sci ; 129(9): 1831-42, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26966188

ABSTRACT

The family with sequence similarity 3 (FAM3) gene family is a cytokine-like gene family with four members FAM3A, FAM3B, FAM3C and FAM3D. In this study, we found that FAM3D strongly chemoattracted human peripheral blood neutrophils and monocytes. To identify the FAM3D receptor, we used chemotaxis, receptor internalization, Ca(2+) flux and radioligand-binding assays in FAM3D-stimulated HEK293 cells that transiently expressed formyl peptide receptor (FPR)1 or FPR2 to show that FAM3D was a high affinity ligand of these receptors, both of which were highly expressed on the surface of neutrophils, and monocytes and macrophages. After being injected into the mouse peritoneal cavity, FAM3D chemoattracted CD11b+ Ly6G+ neutrophils in a short time. In response to FAM3D stimulation, phosphorylated ERK1/2 and phosphorylated p38 MAPK family proteins were upregulated in the mouse neutrophils, and this increase was inhibited upon treatment with an inhibitor of FPR1 or FPR2. FAM3D has been reported to be constitutively expressed in the gastrointestinal tract. We found that FAM3D expression increased significantly during colitis induced by dextran sulfate sodium. Taken together, we propose that FAM3D plays a role in gastrointestinal homeostasis and inflammation through its receptors FPR1 and FPR2.


Subject(s)
Chemotaxis , Colitis/metabolism , Cytokines/metabolism , MAP Kinase Signaling System , Monocytes/metabolism , Neutrophils/metabolism , Receptors, Formyl Peptide , Receptors, Lipoxin , Animals , Colitis/genetics , Colitis/pathology , Cytokines/genetics , Dextran Sulfate/toxicity , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Mice , Monocytes/pathology , Neutrophils/pathology , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/agonists , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism
20.
Tumour Biol ; 37(5): 6217-25, 2016 May.
Article in English | MEDLINE | ID: mdl-26615421

ABSTRACT

Previous research revealed that CMTM8 acts as a tumor suppressor gene in variety cancers. However, the role of CMTM8 in bladder cancer has never been reported. In this study, the expression profile of CMTM8 was examined in bladder cancer tissues and bladder cancer cell lines. The effects of CMTM8 on bladder cancer cell proliferation, apoptosis, migration, and invasion were examined. Bladder tumor tissues from 84 cases were examined for CMTM8 expression by immunohistochemistry. Disease-specific survival was investigated using a Kaplan-Meier analysis, and Cox proportional hazards analysis was assessed. Our results showed that upregulation of CMTM8 in the T24 cell line could suppress T24 cells proliferation, migration and invasion and enhance the sensitivity to Epirubicin. Kaplan-Meier analysis revealed that the expression of CMTM8 was correlated with the survival time of bladder cancer patients. Altogether, our data suggested that CMTM8 is an important tumor suppressor gene in human bladder cancer and qualified as a useful prognostic indicator for patients with bladder cancer.


Subject(s)
Biomarkers, Tumor/genetics , Chemokines/genetics , MARVEL Domain-Containing Proteins/genetics , Prognosis , Urinary Bladder Neoplasms/genetics , Adult , Aged , Apoptosis/genetics , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Chemokines/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , MARVEL Domain-Containing Proteins/biosynthesis , Male , Middle Aged , Tumor Suppressor Proteins/genetics , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL