Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 23(1): 45, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38424542

ABSTRACT

BACKGROUND: In the myeloid compartment of the tumor microenvironment, CD244 signaling has been implicated in immunosuppressive phenotype of monocytes. However, the precise molecular mechanism and contribution of CD244 to tumor immunity in monocytes/macrophages remains elusive due to the co-existing lymphoid cells expressing CD244. METHODS: To directly assess the role of CD244 in tumor-associated macrophages, monocyte-lineage-specific CD244-deficient mice were generated using cre-lox recombination and challenged with B16F10 melanoma. The phenotype and function of tumor-infiltrating macrophages along with antigen-specific CD8 T cells were analyzed by flow cytometry and single cell RNA sequencing data analysis, and the molecular mechanism underlying anti-tumorigenic macrophage differentiation, antigen presentation, phagocytosis was investigated ex vivo. Finally, the clinical feasibility of CD244-negative monocytes as a therapeutic modality in melanoma was confirmed by adoptive transfer experiments. RESULTS: CD244fl/flLysMcre mice demonstrated a significant reduction in tumor volume (61% relative to that of the CD244fl/fl control group) 14 days after tumor implantation. Within tumor mass, CD244fl/flLysMcre mice also showed higher percentages of Ly6Clow macrophages, along with elevated gp100+IFN-γ+ CD8 T cells. Flow cytometry and RNA sequencing data demonstrated that ER stress resulted in increased CD244 expression on monocytes. This, in turn, impeded the generation of anti-tumorigenic Ly6Clow macrophages, phagocytosis and MHC-I antigen presentation by suppressing autophagy pathways. Combining anti-PD-L1 antibody with CD244-/- bone marrow-derived macrophages markedly improved tumor rejection compared to the anti-PD-L1 antibody alone or in combination with wild-type macrophages. Consistent with the murine data, transcriptome analysis of human melanoma tissue single-cell RNA-sequencing dataset revealed close association between CD244 and the inhibition of macrophage maturation and function. Furthermore, the presence of CD244-negative monocytes/macrophages significantly increased patient survival in primary and metastatic tumors. CONCLUSION: Our study highlights the novel role of CD244 on monocytes/macrophages in restraining anti-tumorigenic macrophage generation and tumor antigen-specific T cell response in melanoma. Importantly, our findings suggest that CD244-deficient macrophages could potentially be used as a therapeutic agent in combination with immune checkpoint inhibitors. Furthermore, CD244 expression in monocyte-lineage cells serve as a prognostic marker in cancer patients.


Subject(s)
Melanoma , Monocytes , Humans , Animals , Mice , Monocytes/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Macrophages/metabolism , CD8-Positive T-Lymphocytes , Carcinogenesis/metabolism , Tumor Microenvironment , Signaling Lymphocytic Activation Molecule Family/metabolism
2.
Int J Nanomedicine ; 14: 8305-8320, 2019.
Article in English | MEDLINE | ID: mdl-31806959

ABSTRACT

BACKGROUND: Phosphatidylcholine (PC) and Omega-3 fatty acid (Omega-3) are promising therapeutic molecules for treating inflammatory bowel disease (IBD). PURPOSE: Based on the IBD therapeutic potential of nanoparticles, we herein sought to develop Omega-3-incorporated PC nanoparticles (liposomes) as an orally administrable vehicle for treating IBD. METHODS: Liposomes prepared with or without Omega-3 incorporation were compared in terms of colloidal stability and anitiinflammatory effects. RESULTS: The incorporation of free Omega-3 (alpha-linolenic acid, eicosapentaenoic acid or docosahexaenoic acid) into liposomes induced time-dependent membrane fusion, resulting in particle size increase from nm to µm during storage. In contrast, krill oil incorporation into liposomes (KO liposomes) did not induce the fusion and the particle size maintained <250 nm during storage. KO liposomes also maintained colloidal stability in simulated gastrointestinal conditions and exhibited a high capacity to entrap the IBD drug, budesonide (BDS). KO liposomes greatly suppressed the lipopolysaccharide-induced production of pro-inflammatory cytokines in cultured macrophages and completely restored inflammation-impaired membrane barrier function in an intestinal barrier model. In mice subjected to dextran sulfate sodium-induced colitis, oral administration of BDS-entrapped KO liposomes suppressed tumor necrosis factor-α production (by 84.1%), interleukin-6 production (by 35.3%), and the systemic level of endotoxin (by 96.8%), and slightly reduced the macroscopic signs of the disease. CONCLUSION: Taken together, KO liposomes may have great potential as a nanovehicle for oral delivery of IBD drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/drug therapy , Euphausiacea/chemistry , Liposomes/pharmacology , Oils/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Budesonide/chemistry , Budesonide/pharmacology , Caco-2 Cells , Colitis/chemically induced , Cytokines/metabolism , Dextran Sulfate/toxicity , Fatty Acids, Omega-3/chemistry , Female , Humans , Lipopolysaccharides/pharmacology , Liposomes/chemistry , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL
3.
Chem Pharm Bull (Tokyo) ; 67(9): 966-976, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31257308

ABSTRACT

Honokiol, a biphenolic neolignan isolated from Magnolia officinalis, was reported to have a promising anti-inflammatory activity for the treatment of various diseases. There are many efforts on the synthesis and structure-activity relationship of honokiol derivatives. However, regioselective O-alkylation of honokiol remains a challenge and serves as a tool to provide not only some derivatives but also chemical probes for target identification and mode of action. In this study, we examined the reaction condition for regioselective O-alkylation, in which C2 and C4'-alkylated analogs of honokiol were synthesized and evaluated for inhibitory activity on nitric oxide production and cyclooxygenase-2 expression. Furthermore, we successfully synthesized a potential photoaffinity probe consisting of biotin and benzophenone based on a C4'-alkylated derivative.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biphenyl Compounds/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Inflammation/drug therapy , Lignans/pharmacology , Alkylation , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Humans , Inflammation/metabolism , Lignans/chemical synthesis , Lignans/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Molecular Structure , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , RAW 264.7 Cells , Stereoisomerism
4.
Life Sci ; 231: 116593, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31228512

ABSTRACT

Inflammasomes are the major mechanistic complexes that include members of the NOD-like receptor (NLRs) or AIM2-like receptors (ALRs) families, which are affiliated with the innate immune system. Once NLRs or ALRs are activated by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), the caspase-1 or -11 is activated by binding with NLRs or ALRs via its own unique cytosolic domains. As a result, caspase-1 or -11 enhances the production of IL-1ß and IL-18, which results in inflammation via the recruitment of immune cells, such as macrophages, and the promotion of programmed cell death mechanisms such as pyroptosis. In addition, the consistent cascades of inflammasomes would precede both minor and severe autoimmune diseases and cancers. The clinical relevance of inflammasomes in multiple forms of cancer highlights their therapeutic promise as molecular targets. To closely analyze the physiological roles of inflammasomes in cancers, here, we describe the fundamental knowledge regarding the current issues of inflammasomes in relevant cancers, and discuss possible therapeutic values in targeting these inflammasomes for the prevention and treatment of cancer.


Subject(s)
Inflammasomes/metabolism , Inflammasomes/physiology , Neoplasms/therapy , Alarmins/metabolism , Animals , Apoptosis/physiology , Autoimmune Diseases/immunology , Carrier Proteins/metabolism , Caspase 1/metabolism , Caspases/metabolism , Humans , Immunity, Innate/immunology , Inflammation/immunology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , NLR Proteins/physiology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Signal Transduction
5.
Pharmacol Res ; 144: 73-78, 2019 06.
Article in English | MEDLINE | ID: mdl-30959160

ABSTRACT

Protein tyrosine phosphatases (PTPs), which are ubiquitously expressed in hematopoietic and non-hematopoietic cells, are critical for regulating cell proliferation as well as differentiation in the physiology of multicellular organisms. PTPs regulate the intracellular signaling mechanism of immune cells via dephosphorylation of multiple targets and are associated with the onset of various autoimmune diseases through genomic alterations. PTPs also affect disease through their role in innate and/or acquired immunity. By modulating multiple substrates, PTPN12, a member of the proline-, glutamic acid-, serine- and threonine-rich (PEST) family of PTPs, is an important regulator of cell migration and adhesion. According to its newly identified roles and functions, PTPN12 is considered a promising therapeutic target against critical diseases, including cancer, diabetes, metabolic disease and autoimmune diseases. In this review, we provide an overview of PTPs and discuss the critical roles of PTPN12/PTP-PEST in tumor progression.


Subject(s)
Neoplasms/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Animals , Disease Progression , Drug Discovery , Enzyme Inhibitors/pharmacology , Humans , Immunity/drug effects , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 12/antagonists & inhibitors
6.
Clin Exp Vaccine Res ; 7(1): 16-23, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29399576

ABSTRACT

Dendritic cells (DCs) are the most professional antigen presenting cells that play important roles in connection between innate and adaptive immune responses. Numerous studies revealed that the functions of DCs are related with the capture and processing of antigen as well as the migration to lymphoid tissues for the presenting antigens to T cells. These unique features of DCs allow them to be considered as therapeutic vaccines that can induce immune responses and anti-tumor activity. Here, we discuss and understand the immunological basis of DCs and presume the possibilities of DC-based vaccines for the promising cancer therapy.

7.
Arch Pharm Res ; 41(2): 219-228, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29147873

ABSTRACT

Adjuvants are essential vaccine components used to enhance, accelerate, and/or prolong adaptive immunity against specific vaccine antigens. In this study, we compared the adjuvanticity of two adjuvant formulations containing de-O-acylated lipooligosaccharide (dLOS), a toll-like receptor 4 agonist, on the Japanese encephalitis (JE) vaccine in mice. Mice were immunized once or twice at a two-week interval with inactivated JE vaccine in the absence or presence of adjuvant. We found that both the alum- and the liposome-based formulation induced significantly faster and higher serum IgG antibody responses as compared with the non-adjuvanted vaccine after either one or two immunizations. The antibody titers of the mouse immune sera correlated with 50% plaque reduction neutralization test (PRNT50) antibody titers. In addition, the dLOS/liposome formulation was more effective in inducing a Th1-type immune response than the dLOS/alum formulation, as suggested by a strong antigen-specific interferon (IFN)-γ response. Based on these results, we suggest that both alum- and liposome-based adjuvant formulations containing dLOS may be used for the development of JE vaccines with improved immunogenicity.


Subject(s)
Adjuvants, Immunologic , Antigens, Bacterial/immunology , Japanese Encephalitis Vaccines/immunology , Lipopolysaccharides/immunology , Acylation/immunology , Adjuvants, Immunologic/blood , Animals , Antigens, Bacterial/blood , Drug Compounding , Female , Japanese Encephalitis Vaccines/blood , Lipopolysaccharides/blood , Mice , Mice, Inbred BALB C , Protein Binding/immunology
8.
J Microbiol Biotechnol ; 28(1): 136-144, 2018 Jan 28.
Article in English | MEDLINE | ID: mdl-29081214

ABSTRACT

Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis. Bacillus Calmette-Guérin (BCG) vaccine is the only TB vaccine currently available, but it is not sufficiently effective in preventing active pulmonary TB or adult infection. With the purpose of developing an improved vaccine against TB that can overcome the limitations of the current BCG vaccine, we investigated whether adjuvant formulations containing de-O-acylated lipooligosaccharide (dLOS) are capable of enhancing the immunogenicity and protective efficacy of TB subunit vaccine. The results revealed that dLOS/dimethyl dioctadecyl ammonium bromide (DDA) adjuvant formulation significantly increased both humoral and Th1-type cellular responses to TB subunit vaccine that are composed of three antigens, Ag85A, ESAT-6, and HspX. The adjuvanted TB vaccine also effectively induced Th1-type response in a BCG-primed mouse model, suggesting a potential as a booster vaccine. Finally, dLOS/DDA-adjuvanted TB vaccine showed protective efficacy against M. tuberculosis infection in vitro and in vivo. These data indicate that dLOS/DDA adjuvant enhances the Th1-type immunity and protective efficacy of TB subunit vaccine suggesting that it would be a promising adjuvant candidate for development of a booster vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/immunology , Lipopolysaccharides/administration & dosage , Liposomes/administration & dosage , Quaternary Ammonium Compounds/administration & dosage , Th1 Cells/immunology , Tuberculosis Vaccines/immunology , Animals , Antigens, Bacterial/administration & dosage , Disease Models, Animal , Female , Mice, Inbred C57BL , Tuberculosis/prevention & control , Tuberculosis Vaccines/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
9.
Drug Deliv ; 24(1): 1587-1597, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29029595

ABSTRACT

Studies have shown that insertion of oleic acid into lipid bilayers can modulate the membrane properties of liposomes so as to improve their function as drug carriers. Considering that 2-hydroxyoleic acid (2OHOA), a potential antitumor agent currently undergoing clinical trials, is a derivative of oleic acid, we explored the possibility of developing 2OHOA-inserted liposomes as a multifunctional carrier of antitumor drugs in the present study. The insertion of 2OHOA into lipid bilayers was confirmed by surface charge determination and differential scanning calorimetry. 2OHOA insertion greatly decreased the order of dimyristoylphosphatidylcholine packing, produced a nanosized (<100 nm) dispersion, and improved the colloidal stability of liposomes during storage. Moreover, 2OHOA-inserted liposome forms exhibited greater growth inhibitory activity against cancer cells compared with free 2OHOA, and the growth-inhibitory activity of liposomal 2OHOA was selective for tumor cells. 2OHOA insertion greatly increased the liposome-incorporated concentration of hydrophobic model drugs, including mitoxantrone, paclitaxel, and all-trans retinoic acid (ATRA). The in vitro anticancer activity of ATRA-incorporated/2OHOA-inserted liposomes was significantly higher than that of ATRA-incorporated conventional liposomes. In a B16-F10 melanoma syngeneic mouse model, the tumor growth rate was significantly delayed in mice treated with ATRA-incorporated/2OHOA-inserted liposomes compared with that in the control group. Immunohistochemical analyses revealed that the enhanced antitumor activity of ATRA-incorporated/2OHOA-inserted liposomes was due, at least in part, to increased induction of apoptosis. Collectively, our findings indicate that 2OHOA-inserted liposomes exhibit multiple advantages as antitumor drug carriers, including the ability to simultaneously deliver two anticancer drugs - 2OHOA and incorporated drug - to the tumor tissue.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Liposomes/chemistry , Oleic Acids/pharmacology , Animals , Cell Line, Tumor , Dimyristoylphosphatidylcholine/chemistry , Drug Stability , Humans , Lipid Bilayers , Mice , Nanoparticles , Particle Size , Surface Properties
10.
Immune Netw ; 17(4): 214-227, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28860951

ABSTRACT

Cytokines are molecules that play critical roles in the regulation of a wide range of normal functions leading to cellular proliferation, differentiation and survival, as well as in specialized cellular functions enabling host resistance to pathogens. Cytokines released in response to infection, inflammation or immunity can also inhibit cancer development and progression. The predominant intracellular signaling pathway triggered by cytokines is the JAK-signal transducer and activator of transcription (STAT) pathway. Knockout mice and clinical human studies have provided evidence that JAK-STAT proteins regulate the immune system, and maintain immune tolerance and tumor surveillance. Moreover, aberrant activation of the JAK-STAT pathways plays an undeniable pathogenic role in several types of human cancers. Thus, in combination, these observations indicate that the JAK-STAT proteins are promising targets for cancer therapy in humans. The data supporting this view are reviewed herein.

11.
Nature ; 544(7651): 493-497, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28424516

ABSTRACT

Cancer cells elude anti-tumour immunity through multiple mechanisms, including upregulated expression of ligands for inhibitory immune checkpoint receptors. Phagocytosis by macrophages plays a critical role in cancer control. Therapeutic blockade of signal regulatory protein (SIRP)-α, an inhibitory receptor on macrophages, or of its ligand CD47 expressed on tumour cells, improves tumour cell elimination in vitro and in vivo, suggesting that blockade of the SIRPα-CD47 checkpoint could be useful in treating human cancer. However, the pro-phagocytic receptor(s) responsible for tumour cell phagocytosis is(are) largely unknown. Here we find that macrophages are much more efficient at phagocytosis of haematopoietic tumour cells, compared with non-haematopoietic tumour cells, in response to SIRPα-CD47 blockade. Using a mouse lacking the signalling lymphocytic activation molecule (SLAM) family of homotypic haematopoietic cell-specific receptors, we determined that phagocytosis of haematopoietic tumour cells during SIRPα-CD47 blockade was strictly dependent on SLAM family receptors in vitro and in vivo. In both mouse and human cells, this function required a single SLAM family member, SLAMF7 (also known as CRACC, CS1, CD319), expressed on macrophages and tumour cell targets. In contrast to most SLAM receptor functions, SLAMF7-mediated phagocytosis was independent of signalling lymphocyte activation molecule-associated protein (SAP) adaptors. Instead, it depended on the ability of SLAMF7 to interact with integrin Mac-1 (refs 18, 19, 20) and utilize signals involving immunoreceptor tyrosine-based activation motifs. These findings elucidate the mechanism by which macrophages engulf and destroy haematopoietic tumour cells. They also reveal a novel SAP adaptor-independent function for a SLAM receptor. Lastly, they suggest that patients with tumours expressing SLAMF7 are more likely to respond to SIRPα-CD47 blockade therapy.


Subject(s)
Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Macrophage-1 Antigen/metabolism , Macrophages/immunology , Phagocytosis/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Actins/metabolism , Animals , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , CD47 Antigen/immunology , CD47 Antigen/metabolism , Female , Hematologic Neoplasms/drug therapy , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family/deficiency
12.
Arch Pharm Res ; 39(11): 1588-1596, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27562774

ABSTRACT

Macrophages are traditional innate immune cells that play critical roles in the clearance of pathogens and the maintenance of tissue homeostasis. Accumulating evidence proves that macrophages affect cancer initiation and malignancy. Macrophages can be categorized into two extreme subsets, classically activated (M1) and alternatively activated (M2) macrophages based on their distinct functional abilities in response to microenvironmental stimuli. In a tumor microenvironment, tumor associated macrophages (TAMs) are considered to be of the polarized M2 phenotype that enhances tumor progression and represent a poor prognosis. Furthermore, TAMs enhance tumor angiogenesis, growth, metastasis, and immunosuppression by secreting a series of cytokines, chemokines, and proteases. The regulation of macrophage polarization is considered to be a potential future therapy for cancer management.


Subject(s)
Cell Polarity/immunology , Macrophage Activation/immunology , Macrophages/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Adaptive Immunity/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Movement/drug effects , Cell Movement/immunology , Cell Polarity/drug effects , Cytokines/genetics , Cytokines/immunology , Humans , Immunity, Innate/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Paclitaxel/therapeutic use , Tumor Microenvironment/drug effects
13.
Molecules ; 21(8)2016 Aug 13.
Article in English | MEDLINE | ID: mdl-27529212

ABSTRACT

Naturally occurring homoisoflavonoids containing either 5,7-dihydroxy-6-methoxy or 7-hydroxy-5,6-dimethoxy groups such as the antiangiogenic homoisoflavanone, cremastranone, were synthesized via three or four linear steps from the known 4-chromenone. This facile synthesis includes chemoselective 1,4-reduction of 4-chromenone and selective deprotection of 3-benzylidene-4-chromanone a containing C7-benzyloxy group.


Subject(s)
Biological Products/chemical synthesis , Isoflavones/chemical synthesis , Biological Products/chemistry , Isoflavones/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure
14.
Cell Mol Life Sci ; 71(21): 4221-41, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25037158

ABSTRACT

Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.


Subject(s)
Gene Expression Regulation , Neural Stem Cells/cytology , Animals , Brain/physiology , Cell Lineage , Cognition , Cognition Disorders/metabolism , Epigenesis, Genetic , Humans , Hypoxia , Inflammation , Lipids/chemistry , Neurodegenerative Diseases/metabolism , Neurogenesis/physiology , Regeneration
15.
Mol Cell Biol ; 34(5): 888-99, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366546

ABSTRACT

Dendritic cells (DCs) capture and process antigens in peripheral tissues, migrate to lymphoid tissues, and present the antigens to T cells. PTPN12, also known as PTP-PEST, is an intracellular protein tyrosine phosphatase (PTP) involved in cell-cell and cell-substratum interactions. Herein, we examined the role of PTPN12 in DCs, using a genetically engineered mouse lacking PTPN12 in DCs. Our data indicated that PTPN12 was not necessary for DC differentiation, DC maturation, or cytokine production in response to inflammatory stimuli. However, it was needed for full induction of T cell-dependent immune responses in vivo. This function largely correlated with the need of PTPN12 for DC migration from peripheral sites to secondary lymphoid tissues. Loss of PTPN12 in DCs resulted in hyperphosphorylation of the protein tyrosine kinase Pyk2 and its substrate, the adaptor paxillin. Pharmacological inhibition of Pyk2 or downregulation of Pyk2 expression also compromised DC migration, suggesting that Pyk2 deregulation played a pivotal role in the migration defect caused by PTPN12 deficiency. Together, these findings identified PTPN12 as a key regulator in the ability of DCs to induce antigen-induced T cell responses. This is due primarily to the role of PTPN12 in DC migration from peripheral sites to secondary lymphoid organs through regulation of Pyk2.


Subject(s)
Autoimmunity/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , T-Lymphocytes/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Autoimmunity/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/immunology , Focal Adhesion Kinase 2/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Paxillin/genetics , Paxillin/immunology , Paxillin/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , T-Lymphocytes/metabolism , Tyrosine/genetics , Tyrosine/immunology , Tyrosine/metabolism
16.
Mol Cell Biol ; 33(12): 2458-69, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23589331

ABSTRACT

Macrophages can undergo cell-cell fusion, leading to the formation of multinucleated giant cells and osteoclasts. This process is believed to promote the proteolytic activity of macrophages toward pathogens, foreign bodies, and extracellular matrices. Here, we examined the role of PTP-PEST (PTPN12), a cytoplasmic protein tyrosine phosphatase, in macrophage fusion. Using a macrophage-targeted PTP-PEST-deficient mouse, we determined that PTP-PEST was not needed for macrophage differentiation or cytokine production. However, it was necessary for interleukin-4-induced macrophage fusion into multinucleated giant cells in vitro. It was also needed for macrophage fusion following implantation of a foreign body in vivo. Moreover, in the RAW264.7 macrophage cell line, PTP-PEST was required for receptor activator of nuclear factor kappa-B ligand (RANKL)-triggered macrophage fusion into osteoclasts. PTP-PEST had no impact on expression of fusion mediators such as ß-integrins, E-cadherin, and CD47, which enable macrophages to become fusion competent. However, it was needed for polarization of macrophages, migration induced by the chemokine CC chemokine ligand 2 (CCL2), and integrin-induced spreading, three key events in the fusion process. PTP-PEST deficiency resulted in specific hyperphosphorylation of the protein tyrosine kinase Pyk2 and the adaptor paxillin. Moreover, a fusion defect was induced upon treatment of normal macrophages with a Pyk2 inhibitor. Together, these data argue that macrophage fusion is critically dependent on PTP-PEST. This function is seemingly due to the ability of PTP-PEST to control phosphorylation of Pyk2 and paxillin, thereby regulating cell polarization, migration, and spreading.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Macrophages/metabolism , Paxillin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Animals , CD47 Antigen/biosynthesis , Cadherins/biosynthesis , Cell Differentiation , Cell Fusion , Cell Line , Cell Movement , Cell Proliferation , Chemokine CCL2/metabolism , Focal Adhesion Kinase 2/antagonists & inhibitors , Giant Cells , Integrin beta Chains/biosynthesis , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/metabolism , Phosphorylation , RANK Ligand/metabolism
17.
J Biol Chem ; 287(51): 43180-90, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23105101

ABSTRACT

Protein-tyrosine phosphatase (PTP)-PEST (PTPN12) is ubiquitously expressed. It is essential for normal embryonic development and embryonic viability in mice. Herein we addressed the involvement of PTP-PEST in endothelial cell functions using a combination of genetic and biochemical approaches. By generating primary endothelial cells from an inducible PTP-PEST-deficient mouse, we found that PTP-PEST is not needed for endothelial cell differentiation and proliferation or for the control of endothelial cell permeability. Nevertheless, it is required for integrin-mediated adhesion and migration of endothelial cells. PTP-PEST-deficient endothelial cells displayed increased tyrosine phosphorylation of Cas, paxillin, and Pyk2, which were previously also implicated in integrin functions. By eliminating PTP-PEST in endothelial cells in vivo, we obtained evidence that expression of PTP-PEST in endothelial cells is required for normal vascular development and embryonic viability. Therefore, PTP-PEST is a key regulator of integrin-mediated functions in endothelial cells seemingly through its capacity to control Cas, paxillin, and Pyk2. This function explains at least in part the essential role of PTP-PEST in embryonic development and viability.


Subject(s)
Blood Vessels/embryology , Cell Membrane Permeability , Cell Movement , Embryo, Mammalian/enzymology , Endothelial Cells/cytology , Endothelial Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Animals , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cell Shape , Embryo, Mammalian/cytology , Female , Focal Adhesion Kinase 2/metabolism , Genotype , Integrins/metabolism , Intercellular Junctions/metabolism , Mice , Mice, Inbred C57BL , Paxillin/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/deficiency
18.
Nat Immunol ; 13(5): 439-47, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22513334

ABSTRACT

Lymphocyte activation must be tightly regulated to ensure sufficient immunity to pathogens and prevent autoimmunity. Protein tyrosine phosphatases (PTPs) serve critical roles in this regulation by controlling the functions of key receptors and intracellular signaling molecules in lymphocytes. In some cases, PTPs inhibit lymphocyte activation, whereas in others they promote it. Here we discuss recent progress in elucidating the roles and mechanisms of action of PTPs in lymphocyte activation. We also review the accumulating evidence that genetic alterations in PTPs are involved in human autoimmunity.


Subject(s)
Lymphocyte Activation , Lymphocytes/enzymology , Protein Tyrosine Phosphatases/metabolism , Animals , Antigens, CD/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Humans , Leukocyte Common Antigens/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily K/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 2/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Signaling Lymphocytic Activation Molecule Family Member 1
19.
Mol Cells ; 33(4): 407-14, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22395814

ABSTRACT

Lck Interacting Membrane protein (LIME) was previously characterized as a transmembrane adaptor protein mediating TCR-dependent T cell activation. Here, we show that LIME associates with Vav in response to TCR stimulation and is required for Vav guanine nucleotide exchange factor (GEF) activity for Rac1. Consistent with this finding, actin polymerization at the immunological synapse (IS) was markedly enhanced by overexpression of LIME, but was reduced by expression of a LIME shRNA. Moreover, TCR-mediated cell adhesion to ICAM-1, laminin, or fibronectin was downregulated by expression of LIME shRNA. In addition, in the IS, LIME but not LAT was found to localize at the peripheral-supramolecular activation cluster (p-SMAC) where the integrins were previously shown to be localized. Together, these results establish LIME as a transmembrane adaptor protein linking TCR stimulation to IS formation and integrin activation through activation of Vav.


Subject(s)
Actins , Adaptor Proteins, Vesicular Transport , Cell Adhesion , Gene Expression Regulation, Developmental , Immunological Synapses , Proto-Oncogene Proteins c-vav , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Apoptosis Regulatory Proteins , Binding Sites , Cell Adhesion/genetics , Cell Adhesion/immunology , Gene Expression Regulation, Developmental/immunology , Humans , Immunological Synapses/metabolism , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Transcriptional Activation , rac1 GTP-Binding Protein/metabolism
20.
Immunity ; 33(2): 167-80, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20727793

ABSTRACT

PTP-PEST (encoded by Ptpn12) is an intracellular protein tyrosine phosphatase belonging to the same family as LYP. LYP inhibits secondary T cell responses by suppressing Src family protein tyrosine kinases and is implicated in human autoimmunity. To determine the function of PTP-PEST in T cells, we generated mice with a conditionally deleted allele of Ptpn12. By removing PTP-PEST in T cells, we determined that PTP-PEST was not necessary for T cell development or primary responses. However, PTP-PEST was required for secondary T cell responses, anergy prevention, and autoimmunity induction. PTP-PEST specifically regulated the phosphorylation of Pyk2, a substrate of the Src family kinase Fyn. It also promoted the formation of T cell homoaggregates, which are known to enhance T cell activation. Thus, PTP-PEST controls Pyk2 activity and is a positive regulator of secondary T cell activation. These data illustrate the critical role of protein tyrosine phosphatases in T cell regulation.


Subject(s)
Focal Adhesion Kinase 2/immunology , Focal Adhesion Kinase 2/metabolism , Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , T-Lymphocytes/immunology , Animals , Cell Aggregation , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 12/deficiency , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...