Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(4): 112364, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37043352

ABSTRACT

The clinical response to immune checkpoint blockade (ICB) correlates with tumor-infiltrating cytolytic T lymphocytes (CTLs) prior to treatment. However, many of these inflamed tumors resist ICB through unknown mechanisms. We show that tumors with transcription elongation deficiencies (TEdef+), which we previously reported as being resistant to ICB in mouse models and the clinic, have high baseline CTLs. We show that high baseline CTLs in TEdef+ tumors result from aberrant activation of the nucleic acid sensing-TBK1-CCL5/CXCL9 signaling cascade, which results in an immunosuppressive microenvironment with elevated regulatory T cells and exhausted CTLs. ICB therapy of TEdef+ tumors fail to increase CTL infiltration and suppress tumor growth in both experimental and clinical settings, suggesting that TEdef+, along with surrogate markers of tumor immunogenicity such as tumor mutational burden and CTLs, should be considered in the decision process for patient immunotherapy indication.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Animals , Mice , Humans , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/pathology , Immunotherapy/methods , Signal Transduction , Inflammation/drug therapy , Tumor Microenvironment
2.
J Immunother Cancer ; 10(11)2022 11.
Article in English | MEDLINE | ID: mdl-36427906

ABSTRACT

BACKGROUND: Cancer immunotherapy has taken center stage in cancer treatment. However, the current immunotherapies only benefit a small proportion of patients with cancer, necessitating better understanding of the mechanisms of tumor immune evasion and improved cancer immunotherapy strategies. Regulatory T (Treg) cells play an important role in maintaining immune tolerance through inhibiting effector T-cell function. In the tumor microenvironment, Treg cells are used by tumor cells to counteract effector T cell-mediated tumor suppression. Targeting Treg cells may thus unleash the antitumor activity of effector T cells. While systemic depletion of Treg cells can cause excessive effector T-cell responses and subsequent autoimmune diseases, controlled targeting of Treg cells may benefit patients with cancer. METHODS: Treg cells from Treg cell-specific heterozygous Cdc42 knockout mice, C57BL/6 mice treated with a Cdc42 inhibitor CASIN, and control mice were examined for their homeostasis and stability by flow cytometry. The autoimmune responses in Treg cell-specific heterozygous Cdc42 knockout mice, CASIN-treated C57BL/6 mice, and control mice were assessed by H&E staining and ELISA. Antitumor T-cell immunity in Treg cell-specific heterozygous Cdc42 knockout mice, CASIN-treated C57BL/6 mice, humanized NSGS mice, and control mice was assessed by challenging the mice with MC38 mouse colon cancer cells, KPC mouse pancreatic cancer cells, or HCT116 human colon cancer cells. RESULTS: Treg cell-specific heterozygous deletion or pharmacological targeting of Cdc42 with CASIN does not affect Treg cell numbers but induces Treg cell instability, leading to antitumor T-cell immunity without detectable autoimmune reactions. Cdc42 targeting causes an additive effect on immune checkpoint inhibitor anti-programmed cell death protein-1 antibody-induced T-cell response against mouse and human tumors. Mechanistically, Cdc42 targeting induces Treg cell instability and unleashes antitumor T-cell immunity through carbonic anhydrase I-mediated pH changes. CONCLUSIONS: Rational targeting of Cdc42 in Treg cells holds therapeutic promises in cancer immunotherapy.


Subject(s)
Colonic Neoplasms , T-Lymphocytes, Regulatory , Humans , Mice , Animals , Mice, Inbred C57BL , Immunotherapy , Mice, Knockout , Tumor Microenvironment
3.
Front Immunol ; 12: 726393, 2021.
Article in English | MEDLINE | ID: mdl-34721389

ABSTRACT

RhoA of the Rho GTPase family is prenylated at its C-terminus. Prenylation of RhoA has been shown to control T helper 17 (Th17) cell-mediated colitis. By characterizing T cell-specific RhoA conditional knockout mice, we have recently shown that RhoA is required for Th2 and Th17 cell differentiation and Th2/Th17 cell-mediated allergic airway inflammation. It remains unclear whether RhoA plays a cell-intrinsic role in regulatory T (Treg) cells that suppress effector T cells such as Th2/Th17 cells to maintain immune tolerance and to promote tumor immune evasion. Here we have generated Treg cell-specific RhoA-deficient mice. We found that homozygous RhoA deletion in Treg cells led to early, fatal systemic inflammatory disorders. The autoimmune responses came from an increase in activated CD4+ and CD8+ T cells and in effector T cells including Th17, Th1 and Th2 cells. The immune activation was due to impaired Treg cell homeostasis and increased Treg cell plasticity. Interestingly, heterozygous RhoA deletion in Treg cells did not affect Treg cell homeostasis nor cause systemic autoimmunity but induced Treg cell plasticity and an increase in effector T cells. Importantly, heterozygous RhoA deletion significantly inhibited tumor growth, which was associated with tumor-infiltrating Treg cell plasticity and increased tumor-infiltrating effector T cells. Collectively, our findings suggest that graded RhoA expression in Treg cells distinguishes tumor immunity from autoimmunity and that rational targeting of RhoA in Treg cells may trigger anti-tumor T cell immunity without causing autoimmune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , rhoA GTP-Binding Protein/deficiency , Animals , Autoimmunity , Cell Line, Tumor , Female , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Tumor Escape , rhoA GTP-Binding Protein/immunology
4.
Cells ; 8(10)2019 10 16.
Article in English | MEDLINE | ID: mdl-31623153

ABSTRACT

BACKGROUND: Schistosomiasis is an immunopathogenic disease in which Th17 cells play vital roles. Hepatic granuloma formation and subsequent fibrosis are its main pathologic manifestations and the leading causes of hepatic cirrhosis, and effective therapeutic interventions are lacking. In this study, we explored the effects of fasudil, a selective RhoA-Rho-associated kinase (ROCK) inhibitor, on Th17 cells and the pathogenesis of schistosomiasis. METHODS: Mice were infected with Schistosoma japonicum and treated with fasudil. The worm burden, hepatic granuloma formation, and fibrosis were evaluated. The roles of fasudil on Th17, Treg, and hepatic stellate cells were analyzed. RESULTS: Fasudil therapy markedly reduced the granuloma size and collagen deposit in livers from mice infected with S. japonicum. However, fasudil therapy did not affect the worm burden in infected mice. The underlying cellular and molecular mechanisms were investigated. Fasudil suppressed the activation and induced the apoptosis of CD4+ T cells. Fasudil inhibited the differentiation and effector cytokine secretion of Th17 cells, whereas it upregulated Treg cells in vitro. It also restrained the in vivo interleukin (IL)-4 and IL-17 levels in infected mice. Fasudil directly induced the apoptosis of hepatic stellate cells and downregulated the expressions of hepatic fibrogenic genes, such as collagen type I (Col-I), Col-III, and transforming growth factor-1 (TGF-ß1). These effects may contribute to its anti-pathogenic roles in schistosomiasis. CONCLUSIONS: Fasudil inhibits hepatic granuloma formation and fibrosis with downregulation of Th17 cells. Fasudil might serve as a novel therapeutic agent for hepatic fibrosis due to schistosome infections and perhaps other disorders.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Protein Kinase Inhibitors/therapeutic use , Schistosoma japonicum/pathogenicity , Th17 Cells/drug effects , Th17 Cells/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Cell Differentiation/drug effects , Collagen Type I/metabolism , Female , Flow Cytometry , Granuloma/drug therapy , Granuloma/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
5.
J Leukoc Biol ; 106(5): 1139-1151, 2019 11.
Article in English | MEDLINE | ID: mdl-31260596

ABSTRACT

Asthma is a heterogeneous chronic airway inflammation in which Th2 and Th17 cells are key players in its pathogenesis. We have reported that RhoA of Rho GTPases orchestrated glycolysis for Th2 cell differentiation and allergic airway inflammation by the use of a conditional RhoA-deficient mouse line. However, the role of RhoA in Th17 cells remains to be elucidated. In this study, we investigated the effects of RhoA deficiency on Th17 cells in the context of ex vivo cell culture systems and an in vivo house dust mites (HDM)-induced allergic airway inflammation. We found that RhoA deficiency inhibited Th17 differentiation and effector cytokine secretion, which was associated with the downregulations of Stat3 and Rorγt, key Th17 transcription factors. Furthermore, loss of RhoA markedly suppressed Th17 and neutrophil-involved airway inflammation induced by HDM in mice. The infiltrating inflammatory cells in the lungs and bronchoalveolar lavage (BAL) fluids were dramatically reduced in conditional RhoA-deficient mice. Th17 as well as Th2 effector cytokines were suppressed in the airways at both protein and mRNA levels. Interestingly, Y16, a specific RhoA inhibitor, was able to recapitulate the most phenotypes of RhoA genetic deletion in Th17 differentiation and allergic airway inflammation. Our data demonstrate that RhoA is a key regulator of Th17 cell differentiation and function. RhoA might serve as a potential novel therapeutic target for asthma and other inflammatory disorders.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Pyroglyphidae/immunology , Th17 Cells/immunology , rhoA GTP-Binding Protein/deficiency , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cell Differentiation/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , rhoA GTP-Binding Protein/immunology
6.
Exp Ther Med ; 17(5): 3859-3866, 2019 May.
Article in English | MEDLINE | ID: mdl-30988771

ABSTRACT

Ginsenoside Rb1 (GRb1) is a major ingredient of ginseng, a traditional medicine that has been used for thousands of years. Previous studies have reported that GRb1 had anti-inflammatory, antioxidant and neuroprotective effects. The current study aimed to evaluate the antinociceptive effects of GRb1 in a rat model of cancer-induced bone pain (CIBP) established by intratibial injection of Walker 256 cells. Intraperitoneal injection (i.p.) of GRb1 (5 and 10 mg/kg, but not 1 mg/kg) partially and transiently reversed the mechanical allodynia and thermal hyperalgesia in CIBP rats at 14 days following surgery when the pain behavior is established. Furthermore, repeated administration of GRb1 demonstrated persistent analgesic effect. Additionally, the protein expression and immunoreactivity of iba1, which is the maker of microglia, was significantly suppressed in CIBP rats treated with GRb1 (i.p., 10 mg/kg) from day 12 for three consecutive days compared with CIBP rats treated with a vehicle. Furthermore, upregulation of spinal interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α were also significantly inhibited by the treatment of GRb1 (i.p., 10 mg/kg) from day 12 for three consecutive days. Together, these results indicated that GRb1 may attenuate CIBP via inhibiting the activation of microglia and glial-derived proinflammatory cytokines.

7.
Clin Exp Allergy ; 49(1): 92-107, 2019 01.
Article in English | MEDLINE | ID: mdl-30307073

ABSTRACT

BACKGROUND: Asthma is an allergic airway inflammation-driven disease that affects more than 300 million people world-wide. Targeted therapies for asthma are largely lacking. Although asthma symptoms can be prevented from worsening, asthma development cannot be prevented. Cdc42 GTPase has been shown to regulate actin cytoskeleton, cell proliferation and survival. OBJECTIVES: To investigate the role and targeting of Cdc42 in Th2 cell differentiation and Th2-mediated allergic airway inflammation. METHODS: Post-thymic Cdc42-deficient mice were generated by crossing Cdc42flox/flox mice with dLckicre transgenic mice in which Cre expression is driven by distal Lck promoter. Effects of post-thymic Cdc42 deletion and pharmacological targeting Cdc42 on Th2 cell differentiation were evaluated in vitro under Th2-polarized culture conditions. Effects of post-thymic Cdc42 deletion and pharmacological targeting Cdc42 on allergic airway inflammation were evaluated in ovalbumin- and/or house dust mite-induced mouse models of asthma. RESULTS: Post-thymic deletion of Cdc42 led to reduced peripheral CD8+ T cells and attenuated Th2 cell differentiation, with no effect on closely related Th1, Th17 and induced regulatory T (iTreg) cells. Post-thymic Cdc42 deficiency ameliorated allergic airway inflammation. The selective inhibition of Th2 cell differentiation by post-thymic deletion of Cdc42 was recapitulated by pharmacological targeting of Cdc42 with CASIN, a Cdc42 activity-specific chemical inhibitor. CASIN also alleviated allergic airway inflammation. CASIN-treated Cdc42-deficient mice showed comparable allergic airway inflammation to vehicle-treated Cdc42-deficient mice, indicative of negligible off-target effect of CASIN. CASIN had no effect on established allergic airway inflammation. CONCLUSION AND CLINICAL RELEVANCE: Cdc42 is required for Th2 cell differentiation and allergic airway inflammation, and rational targeting Cdc42 may serve as a preventive but not therapeutic approach for asthma control.


Subject(s)
Asthma , Cell Differentiation , Th2 Cells/immunology , cdc42 GTP-Binding Protein , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Deletion , Mice , Mice, Transgenic , Th2 Cells/pathology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/immunology
8.
J Immunol ; 200(7): 2313-2326, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29440353

ABSTRACT

A balance between Th17 cells and regulatory T cells (Tregs) is important for host immunity and immune tolerance. The underlying molecular mechanisms remain poorly understood. Here we have identified Cdc42 as a central regulator of Th17/Treg balance. Deletion of Cdc42 in T cells enhanced Th17 differentiation but diminished induced Treg differentiation and suppressive function. Treg-specific deletion of Cdc42 decreased natural Tregs but increased effector T cells including Th17 cells. Notably, Cdc42-deficient Th17 cells became pathogenic associated with enhanced glycolysis and Cdc42-deficient Tregs became unstable associated with weakened glycolytic signaling. Inhibition of glycolysis in Cdc42-deficient Th17 cells diminished their pathogenicity and restoration of glycolysis in Cdc42-deficient Tregs rescued their instability. Intriguingly, Cdc42 deficiency in T cells led to exacerbated wasting disease in mouse models of colitis and Treg-specific deletion of Cdc42 caused early, fatal lymphoproliferative diseases. In summary, we show that Cdc42 is a bona fide regulator of peripheral tolerance through suppression of Th17 aberrant differentiation/pathogenicity and promotion of Treg differentiation/stability/function involving metabolic signaling and thus Cdc42 pathway might be harnessed in autoimmune disease therapy.


Subject(s)
Glycolysis/genetics , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , cdc42 GTP-Binding Protein/metabolism , Animals , Cell Differentiation/immunology , Cell Proliferation/genetics , Colitis/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphoproliferative Disorders/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , cdc42 GTP-Binding Protein/genetics
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 934-941, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29305916

ABSTRACT

Asthma is a chronic airway inflammation in which Th2 and Th17 cells play critical roles in its pathogenesis. We have reported that atypical protein kinase (PKC) λ/ι is a new regulator for Th2 differentiation and function. However, the role of PKCλ/ι for Th17 cells remains elusive. In this study, we explored the effect of PKCλ/ι on Th17 cells in the context of ex vivo cell culture systems and an in vivo murine model of allergic airway inflammation with the use of activated T cell-specific conditional PKCλ/ι-deficient mice. Our findings indicate that PKCλ/ι regulates Th17 cells. The secretion of Th17 effector cytokines, including IL-17, IL-21 and IL-22, were inhibited from PKCλ/ι-deficient T cells under non-skewing or Th17-skewing culture conditions. Moreover, the impaired Th17 differentiation and function by the PKCλ/ι-deficiency was associated with the downregulation of Stat3 and Rorγt, key Th17 transcription factors. We developed a model of Th17 and neutrophil-involved allergic airway inflammation by intratracheal inoculation of house dust mites. PKCλ/ι-deficiency significantly inhibited airway inflammations. The infiltrating cells in the lungs and bronchoalveolar lavage fluids were significantly reduced in conditional PKCλ/ι-deficient mice. Th17 effector cytokines were reduced in the bronchoalveolar lavage fluids and lungs at protein and mRNA levels. Thus, PKCλ/ι emerges as a critical regulator of Th17 differentiation and allergic airway hyperresponsiveness.


Subject(s)
Cell Differentiation/genetics , Inflammation , Isoenzymes/physiology , Protein Kinase C/physiology , Pyroglyphidae/immunology , Respiratory Hypersensitivity , Th17 Cells/physiology , Animals , Dermatophagoides pteronyssinus/immunology , Embryo, Mammalian , Female , Inflammation/genetics , Inflammation/immunology , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Protein Kinase C/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology
10.
PLoS One ; 13(1): e0190996, 2018.
Article in English | MEDLINE | ID: mdl-29304132

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0179565.].

11.
PLoS One ; 12(8): e0183266, 2017.
Article in English | MEDLINE | ID: mdl-28813526

ABSTRACT

The mammalian target of rapamycin (mTOR), present in mTOR complex 1 (mTORC1) and mTORC2, is a serine/threonine kinase that integrates nutrients, growth factors, and cellular energy status to control protein synthesis, cell growth, survival and metabolism. However, it remains elusive whether mTOR plays a developmental stage-specific role in tissue development and whether mTOR can function independent of its complexes and kinase activity. In this study, by inducible genetic manipulation approach, we investigated the role of mTOR and its dependence on mTOR complexes and kinase activity in mitochondrial fitness of early, progenitor stage (lineage-negative; Lin-) versus later, lineage-committed stage (lineage-positive; Lin+) of hematopoietic cells. We found that oxidative phosphorylation (OXPHOS), ATP production and mitochondrial DNA synthesis were decreased in mTOR-/- Lin- cells but increased in mTOR-/- Lin+ cells, suggesting that mTOR plays a developmental stage-specific role in OXPHOS, ATP production and mitochondrial DNA synthesis. In contrast to mTOR deletion, simultaneous deletion of Raptor, a key component of mTORC1, and Rictor, a key component of mTORC2, led to increased mitochondrial DNA in Lin- cells and decreased mitochondrial DNA and ATP production in Lin+ cells, suggesting that mTOR regulates mitochondrial DNA synthesis in Lin- and Lin+ cells and ATP production in Lin+ cells independent of mTORC1 and mTORC2. Similar to mTOR deletion, deletion of Raptor alone attenuated glycolysis and increased mitochondrial mass and mitochondrial membrane potential in Lin- cells and increased mitochondrial mass and OXPHOS in Lin+ cells, whereas deletion of Rictor alone had no effect on these mitochondrial parameters in Lin- and Lin+ cells, suggesting that mTOR regulates glycolysis and mitochondrial membrane potential in Lin- cells, OXPHOS in Lin+ cells, and mitochondrial mass in both Lin- and Lin+ cells dependent on mTORC1, but not mTORC2. Either Raptor deficiency or Rictor deficiency recapitulated mTOR deletion in decreasing OXPHOS in Lin- cells and glycolysis in Lin+ cells, suggesting that mTOR regulates OXPHOS in Lin- cells and glycolysis in Lin+ cells dependent on both mTORC1 and mTORC2. Finally, mice harboring a mTOR kinase dead D2338A knock-in mutant showed decreased glycolysis in Lin+ cells, as seen in mTOR-/- Lin+ cells, but no change in glycolysis in Lin- cells, in contrast to the decreased glycolysis in mTOR-/- Lin- cells, suggesting that mTOR regulates glycolysis in Lin+ cells dependent on its kinase activity, whereas mTOR regulates glycolysis in Lin- cells independent of its kinase activity.


Subject(s)
Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA, Mitochondrial/genetics , Glycolysis/genetics , Glycolysis/physiology , Immunoblotting , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Mutant Strains , Multiprotein Complexes/genetics , Oxidative Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics
12.
PLoS One ; 12(6): e0179565, 2017.
Article in English | MEDLINE | ID: mdl-28614408

ABSTRACT

The "hygiene hypothesis" is a theory try to explain the dramatic increases in the prevalence of autoimmune and allergic diseases over the past two to three decades in developed countries. According to this theory, reduced exposure to parasites and microorganisms in childhood is the main cause for the increased incidences of both T helper 1 (Th1)-mediated autoimmunity and Th2-mediated allergy. In this study, we investigated the impact of Schistosoma japonicum infection on the allergic airway inflammation induced by repeated intracheal inoculations of house dust mites (HDM), which is a Th17 and neutrophils dominant murine asthma model, mimicking severe asthma. We found that S. japonicum infection downregulated airway hyperresponsiveness. The infiltrating cells, Th17 and Th2 effector cytokines in the bronchoalveolar lavage (BAL) fluids and lungs were significantly reduced in the infected mice. Our findings indicated that S. japonicum infection was able to effectively inhibit host's allergic airway inflammation, which may be related to the upregulated Treg cells upon infection. To our knowledge, it is the first study to reveal the impact of S. japonicum infection on house dust mite induced severe asthma. More in depth investigation is need to elucidate the underlying mechanisms.


Subject(s)
Inflammation/immunology , Pyroglyphidae/immunology , Respiratory Hypersensitivity/immunology , Schistosoma japonicum/immunology , Schistosomiasis japonica/immunology , Animals , Asthma/blood , Asthma/immunology , Asthma/parasitology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Down-Regulation/immunology , Enzyme-Linked Immunosorbent Assay , Female , Host-Parasite Interactions/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Inflammation/blood , Inflammation/parasitology , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Pyroglyphidae/physiology , Respiratory Hypersensitivity/blood , Respiratory Hypersensitivity/parasitology , Reverse Transcriptase Polymerase Chain Reaction , Schistosoma japonicum/physiology , Schistosomiasis japonica/parasitology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
13.
J Immunol Res ; 2016: 2395645, 2016.
Article in English | MEDLINE | ID: mdl-27563682

ABSTRACT

Invariant natural killer T (iNKT) cells are unique subset of innate-like T cells recognizing glycolipids. iNKT cells can rapidly produce copious amounts of cytokines upon antigen stimulation and exert potent immunomodulatory activities for a wide variety of immune responses and diseases. We have revealed the regulatory effect of iNKT cells on autoimmunity with a serial of publications. On the other hand, the role of iNKT cells in parasitic infections, especially in recently attractive topic "hygiene hypothesis," has not been clearly defined yet. Bacterial and parasitic cell wall is a cellular structure highly enriched in a variety of glycolipids and lipoproteins, some of which may serve as natural ligands of iNKT cells. In this review, we mainly summarized the recent findings on the roles and underlying mechanisms of iNKT cells in parasite infections and their cross-talk with Th1, Th2, Th17, Treg, and innate lymphoid cells. In most cases, iNKT cells exert regulatory or direct cytotoxic roles to protect hosts against parasite infections. We put particular emphasis as well on the identification of the natural ligands from parasites and the involvement of iNKT cells in the hygiene hypothesis.


Subject(s)
Host-Parasite Interactions/immunology , Hygiene Hypothesis , Natural Killer T-Cells/immunology , Parasitic Diseases/immunology , Parasitic Diseases/parasitology , T-Lymphocyte Subsets/immunology , Animals , Cytokines/metabolism , Humans , Lymphocyte Activation/immunology , Natural Killer T-Cells/metabolism , T-Lymphocyte Subsets/metabolism
14.
J Allergy Clin Immunol ; 137(1): 231-245.e4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26100081

ABSTRACT

BACKGROUND: Mitochondrial metabolism is known to be important for T-cell activation. However, its involvement in effector T-cell differentiation has just begun to gain attention. Importantly, how metabolic pathways are integrated with T-cell activation and effector cell differentiation and function remains largely unknown. OBJECTIVE: We sought to test our hypothesis that RhoA GTPase orchestrates glycolysis for TH2 cell differentiation and TH2-mediated allergic airway inflammation. METHODS: Conditional RhoA-deficient mice were generated by crossing RhoA(flox/flox) mice with CD2-Cre transgenic mice. Effects of RhoA on TH2 differentiation were evaluated based on in vitro TH2-polarized culture conditions and in vivo in ovalbumin-induced allergic airway inflammation. Cytokine levels were measured by using intracellular staining and ELISA. T-cell metabolism was measured by using the Seahorse XF24 Analyzer and flow cytometry. RESULTS: Disruption of RhoA inhibited T-cell activation and TH2 differentiation in vitro and prevented the development of allergic airway inflammation in vivo, with no effect on TH1 cells. RhoA deficiency in activated T cells led to multiple defects in metabolic pathways, such as glycolysis and oxidative phosphorylation. Importantly, RhoA couples glycolysis to TH2 cell differentiation and allergic airway inflammation through regulating IL-4 receptor mRNA expression and TH2-specific signaling events. Finally, inhibition of Rho-associated protein kinase, an immediate downstream effector of RhoA, blocked TH2 differentiation and allergic airway inflammation. CONCLUSION: RhoA is a key component of the signaling cascades leading to TH2 differentiation and allergic airway inflammation at least in part through control of T-cell metabolism and the Rho-associated protein kinase pathway.


Subject(s)
Glycolysis , Respiratory Hypersensitivity/metabolism , Th2 Cells/metabolism , rhoA GTP-Binding Protein/metabolism , Allergens/immunology , Animals , Cell Differentiation , Inflammation/immunology , Inflammation/metabolism , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology , Th2 Cells/cytology , Th2 Cells/immunology , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/immunology
15.
Article in Chinese | MEDLINE | ID: mdl-27097497

ABSTRACT

Schistosomiasis is a chronic helminthic disease that affects about two hundred millions of people in the world. The pathogenesis of schistosome infection is primarily due to hepatic and intestinal granuloma formation around deposited eggs and subsequent fibrosis. It is known that CD4⁺ T cell subsets play critical roles in the host immunity and immunopathogenesis to schistosome infections, in which T helper 1 (Th1) and Th2 cells are major effector T cell subsets, whereas T regulatory (Treg) cells exert immunosuppressive roles in general. The recently discovered Th17 cells are also actively involved in the immune responses to the infection. During the infection, these T cell subsets cross-talk and exhibit different kinetics and roles in the control and regulation of infection progress and fibrosis. This review summarizes current findings of Th1, Th2, Th17 and Treg cells and their effector cytokines in schistosome infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Schistosoma/physiology , Schistosomiasis/immunology , Animals , Humans , Schistosoma/immunology , Schistosomiasis/parasitology , T-Lymphocyte Subsets/immunology
16.
J Immunol ; 193(12): 5973-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25398325

ABSTRACT

Thymocyte development is regulated by complex signaling pathways. How these signaling cascades are coordinated remains elusive. RhoA of the Rho family small GTPases plays an important role in actin cytoskeleton organization, cell adhesion, migration, proliferation, and survival. Nonetheless, the physiological function of RhoA in thymocyte development is not clear. By characterizing a conditional gene targeting mouse model bearing T cell deletion of RhoA, we show that RhoA critically regulates thymocyte development by coordinating multiple developmental events. RhoA gene disruption caused a strong developmental block at the pre-TCR checkpoint and during positive selection. Ablation of RhoA led to reduced DNA synthesis in CD4(-)CD8(-), CD4(+)CD8(-), and CD4(-)CD8(+) thymocytes but not in CD4(+)CD8(+) thymocytes. Instead, RhoA-deficient CD4(+)CD8(+) thymocytes showed an impaired mitosis. Furthermore, we found that abrogation of RhoA led to an increased apoptosis in all thymocyte subpopulations. Importantly, we show that the increased apoptosis was resulted from reduced pre-TCR expression and increased production of reactive oxygen species (ROS), which may be because of an enhanced mitochondrial function, as manifested by increased oxidative phosphorylation, glycolysis, mitochondrial membrane potential, and mitochondrial biogenesis in RhoA-deficient thymocytes. Restoration of pre-TCR expression or treatment of RhoA-deficient mice with a ROS scavenger N-acetylcysteine partially restored thymocyte development. These results suggest that RhoA is required for thymocyte development and indicate, to our knowledge, for the first time that fine-tuning of ROS production by RhoA, through a delicate control of metabolic circuit, may contribute to thymopoiesis.


Subject(s)
Gene Targeting , Mitochondria/genetics , Mitochondria/metabolism , Thymocytes/cytology , Thymocytes/metabolism , rhoA GTP-Binding Protein/genetics , Animals , Antigens, Surface , Apoptosis/genetics , Apoptosis/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival/genetics , Gene Expression Profiling , Immunophenotyping , Mice , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , V(D)J Recombination , rhoA GTP-Binding Protein/deficiency
17.
Haematologica ; 98(9): 1353-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23716557

ABSTRACT

mTOR integrates signals from nutrients and growth factors to control protein synthesis, cell growth, and survival. Although mTOR has been established as a therapeutic target in hematologic malignancies, its physiological role in regulating hematopoiesis remains unclear. Here we show that conditional gene targeting of mTOR causes bone marrow failure and defects in multi-lineage hematopoiesis including myelopoiesis, erythropoiesis, thrombopoiesis, and lymphopoiesis. mTOR deficiency results in loss of quiescence of hematopoietic stem cells, leading to a transient increase but long-term exhaustion and defective engraftment of hematopoietic stem cells in lethally irradiated recipient mice. Furthermore, ablation of mTOR causes increased apoptosis in lineage-committed blood cells but not hematopoietic stem cells, indicating a differentiation stage-specific function. These results demonstrate that mTOR is essential for hematopoietic stem cell engraftment and multi-lineage hematopoiesis.


Subject(s)
Gene Targeting/methods , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , TOR Serine-Threonine Kinases/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Mice , Mice, Knockout , Mice, SCID
18.
Arthritis Res Ther ; 15(2): R47, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23531237

ABSTRACT

INTRODUCTION: ß2-microglobulin (ß2m) is required for the surface expression of MHC class I and class I-like proteins such as CD1d, Qa1 and neonatal Fc receptor (FcRn), all of which may impact the development of autoimmunity. Since CD1d is known to bind and present phospholipid antigens to T cells, we asked if the deficiency of ß2m or CD1d will impact the development of anti-phospholipid antibodies as compared to other aspects of lupus autoimmunity. METHODS: We introgressed the ß2m-null genotype onto the NZB and NZW backgrounds for 12 to 14 generations to generate genetically lupus-susceptible (NZB/NZW)F1 (BWF1) mice that are ß2m-deficient (ß2m°). Circulating immunoglobulins (Ig), rheumatoid factor (RF), anti-DNA and anti-cardiolipin (anti-CL) antibodies, and renal disease were analyzed in these and CD1d-deficient (CD1d°) BWF1 mice that we had previously generated. RESULTS: Whereas ß2m° BWF1 mice had reduced serum IgG, they had increased mortality, nephritis, serum IgG anti-DNA antibody and RF as compared to heterozygous and wild-type littermates. These effects were recapitulated in CD1d° BWF1 mice, except that they also had increased serum IgG as compared to control littermates. Intriguingly, both ß2m° and CD1d° mice had lower serum anti-CL antibody levels than in control littermates. Such CD1d dependence of anti-CL antibody production is not mediated by CD1d/glycolipid-reactive iNKT cells, as these cells reduced the production of RF and anti-DNA antibodies but had no effect on anti-CL antibodies. CONCLUSIONS: We report a novel dichotomous role of ß2m and CD1d, whereby these molecules differently regulate autoimmunity against phospholipid versus non-phospholipid autoantigens.


Subject(s)
Antigens, CD1d/genetics , Autoantibodies/immunology , Autoantigens/immunology , Histocompatibility Antigens Class I/genetics , Lupus Erythematosus, Systemic/immunology , Animals , Antibodies, Antiphospholipid/immunology , Antigens, CD1d/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Germ-Line Mutation , Histocompatibility Antigens Class I/immunology , Lupus Erythematosus, Systemic/genetics , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mice, SCID
19.
J Clin Immunol ; 32(1): 106-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22002593

ABSTRACT

CD1d presents glycolipid antigens such as α-galactosylceramide (αGalCer) to invariant natural killer T cells (iNKT). We have reported that activated iNKTs inhibit IL-10-producing autoreactive B cells, while promoting or leaving intact the normal B cell responses, making iNKT modulation an attractive therapeutic modality. Here, we report that a brief treatment of young lupus-prone (NZB/NZW)F1 (BWF1) mice with two injections of αGalCer conferred a long-term protection against lupus. Long-term repeated administrations of αGalCer, however, afforded no clinical benefit. These disparate clinical effects correlated with iNKT responsiveness. While a brief treatment with αGalCer enhanced iNKT responses upon in vitro recall, the long-term αGalCer treatment resulted in reduced iNKT responses in BWF1 mice. The improvement in disease with αGalCer treatment was associated with the reduced IL-10 production. Furthermore, iNKTs directly inhibited IL-10-secreting cells in vivo in reconstituted SCID mice and inhibited IL-10-secreting B cells in vitro in co-cultures. Thus, a brief treatment with a CD1d-binding glycolipid enhances iNKT responses, reduces IL-10 production, and delays the onset of lupus, whereas long-term repeated treatments induce marked iNKT hyporesponsiveness and do not affect disease outcome in BWF1 mice. Identifying glycolipid regimens that can modulate iNKT responsiveness will have important implications for developing iNKT-based therapies for autoimmune diseases.


Subject(s)
Galactosylceramides/therapeutic use , Lupus Nephritis/prevention & control , Natural Killer T-Cells/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Galactosylceramides/administration & dosage , Interleukin-10/biosynthesis , Ligands , Lupus Nephritis/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Mice, Knockout , Mice, SCID , Proteinuria/metabolism
20.
PLoS One ; 6(10): e26536, 2011.
Article in English | MEDLINE | ID: mdl-22046304

ABSTRACT

Marginal zone B cells (MZB) mount a rapid antibody response, potently activate naïve T cells, and are enriched in autoreactive B cells. MZBs express high levels of CD1d, the restriction element for invariant natural killer T cells (iNKT). Here, we examined the effect of iNKT cells on MZB cell activation and numbers in vitro and in vivo in normal and autoimmune mice. Results show that iNKT cells activate MZBs, but restrict their numbers in vitro and in vivo in normal BALB/c and C57/BL6 mice. iNKT cells do so by increasing the activation-induced cell death and curtailing proliferation of MZB cells, whereas they promote the proliferation of follicular B cells. Sorted iNKT cells can directly execute this function, without help from other immune cells. Such MZB regulation by iNKTs is mediated, at least in part, via CD1d on B cells in a contact-dependent manner, whereas iNKT-induced proliferation of follicular B cells occurs in a contact- and CD1d-independent manner. Finally, we show that iNKT cells reduce 'autoreactive' MZB cells in an anti-DNA transgenic model, and limit MZB cell numbers in autoimmune-prone (NZB×NZW)F1 and non-obese diabetic mice, suggesting a potentially new mechanism whereby iNKT cells might regulate pathologic autoimmunity. Differential regulation of follicular B cells versus potentially autoreactive MZBs by iNKT cells has important implications for autoimmune diseases as well as for conditions that require a rapid innate B cell response.


Subject(s)
B-Lymphocyte Subsets/cytology , Homeostasis/immunology , Natural Killer T-Cells/physiology , Animals , Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , Cell Death , Cell Proliferation , Immunity, Innate , Lymphocyte Activation , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...