Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Immunity ; 54(2): 308-323.e6, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33421362

ABSTRACT

Th17 cells are known to exert pathogenic and non-pathogenic functions. Although the cytokine transforming growth factor ß1 (TGF-ß1) is instrumental for Th17 cell differentiation, it is dispensable for generation of pathogenic Th17 cells. Here, we examined the T cell-intrinsic role of Activin-A, a TGF-ß superfamily member closely related to TGF-ß1, in pathogenic Th17 cell differentiation. Activin-A expression was increased in individuals with relapsing-remitting multiple sclerosis and in mice with experimental autoimmune encephalomyelitis. Stimulation with interleukin-6 and Activin-A induced a molecular program that mirrored that of pathogenic Th17 cells and was inhibited by blocking Activin-A signaling. Genetic disruption of Activin-A and its receptor ALK4 in T cells impaired pathogenic Th17 cell differentiation in vitro and in vivo. Mechanistically, extracellular-signal-regulated kinase (ERK) phosphorylation, which was essential for pathogenic Th17 cell differentiation, was suppressed by TGF-ß1-ALK5 but not Activin-A-ALK4 signaling. Thus, Activin-A drives pathogenic Th17 cell differentiation, implicating the Activin-A-ALK4-ERK axis as a therapeutic target for Th17 cell-related diseases.


Subject(s)
Activins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neurogenic Inflammation/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activins/genetics , Animals , Cell Differentiation , Cells, Cultured , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy , Signal Transduction
2.
Immunity ; 49(5): 886-898.e5, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30446383

ABSTRACT

Pathogenic Th17 (pTh17) cells drive inflammation and immune-pathology, but whether pTh17 cells are a Th17 cell subset whose generation is under specific molecular control remains unaddressed. We found that Ras p21 protein activator 3 (RASA3) was highly expressed by pTh17 cells relative to non-pTh17 cells and was required specifically for pTh17 generation in vitro and in vivo. Mice conditionally deficient for Rasa3 in T cells showed less pathology during experimental autoimmune encephalomyelitis. Rasa3-deficient T cells acquired a Th2 cell-biased program that dominantly trans-suppressed pTh17 cell generation via interleukin 4 production. The Th2 cell bias of Rasa3-deficient T cells was due to aberrantly elevated transcription factor IRF4 expression. RASA3 promoted proteasome-mediated IRF4 protein degradation by facilitating interaction of IRF4 with E3-ubiquitin ligase Cbl-b. Therefore, a RASA3-IRF4-Cbl-b pathway specifically directs pTh17 cell generation by balancing reciprocal Th17-Th2 cell programs. These findings indicate that a distinct molecular program directs pTh17 cell generation and reveals targets for treating pTh17 cell-related pathology and diseases.


Subject(s)
Cell Differentiation/genetics , GTPase-Activating Proteins/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Animals , Autoimmunity , Biomarkers , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , GTPase-Activating Proteins/metabolism , Gene Expression , Immunophenotyping , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice , Proteolysis , RNA, Messenger , Th17 Cells/immunology , Th2 Cells/immunology
3.
Nature ; 591(7849): 300-305, 2021 03.
Article in English | MEDLINE | ID: mdl-33505023

ABSTRACT

The inflammasome initiates innate defence and inflammatory responses by activating caspase-1 and pyroptotic cell death in myeloid cells1,2. It consists of an innate immune receptor/sensor, pro-caspase-1, and a common adaptor molecule, ASC. Consistent with their pro-inflammatory function, caspase-1, ASC and the inflammasome component NLRP3 exacerbate autoimmunity during experimental autoimmune encephalomyelitis by enhancing the secretion of IL-1ß and IL-18 in myeloid cells3-6. Here we show that the DNA-binding inflammasome receptor AIM27-10 has a T cell-intrinsic and inflammasome-independent role in the function of T regulatory (Treg) cells. AIM2 is highly expressed by both human and mouse Treg cells, is induced by TGFß, and its promoter is occupied by transcription factors that are associated with Treg cells such as RUNX1, ETS1, BCL11B and CREB. RNA sequencing, biochemical and metabolic analyses demonstrated that AIM2 attenuates AKT phosphorylation, mTOR and MYC signalling, and glycolysis, but promotes oxidative phosphorylation of lipids in Treg cells. Mechanistically, AIM2 interacts with the RACK1-PP2A phosphatase complex to restrain AKT phosphorylation. Lineage-tracing analysis demonstrates that AIM2 promotes the stability of Treg cells during inflammation. Although AIM2 is generally accepted as an inflammasome effector in myeloid cells, our results demonstrate a T cell-intrinsic role of AIM2 in restraining autoimmunity by reducing AKT-mTOR signalling and altering immune metabolism to enhance the stability of Treg cells.


Subject(s)
Autoimmunity/immunology , DNA-Binding Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , CARD Signaling Adaptor Proteins/deficiency , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Glycolysis , Humans , Inflammasomes , Inflammation/immunology , Mice , Oxidative Phosphorylation , Phosphorylation , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors for Activated C Kinase/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta
4.
J Proteome Res ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38417049

ABSTRACT

Fluorescence-activated cell sorting (FACS) is a specialized technique to isolate specific cell subpopulations with a high level of recovery and accuracy. However, the cell sorting procedure can impact the viability and metabolic state of cells. Here, we performed a comparative study and evaluated the impact of traditional high-pressure charged droplet-based and microfluidic chip-based sorting on the metabolic and phosphoproteomic profile of different cell types. While microfluidic chip-based sorted cells more closely resembled the unsorted control group for most cell types tested, the droplet-based sorted cells showed significant metabolic and phosphoproteomic alterations. In particular, greater changes in redox and energy status were present in cells sorted with the droplet-based cell sorter along with larger shifts in proteostasis. 13C-isotope tracing analysis on cells recovering postsorting revealed that the sorter-induced suppression of mitochondrial TCA cycle activity recovered faster in the microfluidic chip-based sorted group. Apart from this, amino acid and lipid biosynthesis pathways were suppressed in sorted cells, with minimum impact and faster recovery in the microfluidic chip-based sorted group. These results indicate microfluidic chip-based sorting has a minimum impact on metabolism and is less disruptive compared to droplet-based sorting.

5.
J Biol Chem ; 299(1): 102739, 2023 01.
Article in English | MEDLINE | ID: mdl-36435197

ABSTRACT

Recent discovery of the ribosomal protein (RP) RPL11 interacting with and inhibiting the E3 ubiquitin ligase function of MDM2 established the RP-MDM2-p53 signaling pathway, which is linked to biological events, including ribosomal biogenesis, nutrient availability, and metabolic homeostasis. Mutations in RPs lead to a diverse array of phenotypes known as ribosomopathies in which the role of p53 is implicated. Here, we generated conditional RPL11-deletion mice to investigate in vivo effects of impaired RP expression and its functional connection with p53. While deletion of one Rpl11 allele in germ cells results in embryonic lethality, deletion of one Rpl11 allele in adult mice does not affect viability but leads to acute anemia. Mechanistically, we found RPL11 haploinsufficiency activates p53 in hematopoietic tissues and impedes erythroid precursor differentiation, resulting in insufficient red blood cell development. We demonstrated that reducing p53 dosage by deleting one p53 allele rescues RPL11 haploinsufficiency-induced inhibition of erythropoietic precursor differentiation and restores normal red blood cell levels in mice. Furthermore, blocking the RP-MDM2-p53 pathway by introducing an RP-binding mutation in MDM2 prevents RPL11 haploinsufficiency-caused p53 activation and rescues the anemia in mice. Together, these findings demonstrate that the RP-MDM2-p53 pathway is a critical checkpoint for RP homeostasis and that p53-dependent cell cycle arrest of erythroid precursors is the molecular basis for the anemia phenotype commonly associated with RP deficiency.


Subject(s)
Anemia , Tumor Suppressor Protein p53 , Animals , Mice , Anemia/genetics , Haploinsufficiency , Mutation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Cancer Immunol Immunother ; 72(7): 2151-2168, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36828964

ABSTRACT

The metabolic stress present in the tumor microenvironment of many cancers can attenuate T cell antitumor activity, which is intrinsically controlled by the mitochondrial plasticity, dynamics, metabolism, and biogenesis within these T cells. Previous studies have reported that the complement C1q binding protein (C1QBP), a mitochondrial protein, is responsible for maintenance of mitochondrial fitness in tumor cells; however, its role in T cell mitochondrial function, particularly in the context of an antitumor response, remains unclear. Here, we show that C1QBP is indispensable for T cell antitumor immunity by maintaining mitochondrial integrity and homeostasis. This effect holds even when only one allele of C1qbp is functional. Further analysis of C1QBP in the context of chimeric antigen receptor (CAR) T cell therapy against the murine B16 melanoma model confirmed the cell-intrinsic role of C1QBP in regulating the antitumor functions of CAR T cells. Mechanistically, we found that C1qbp knocking down impacted mitochondrial biogenesis via the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha signaling pathway, as well as mitochondrial morphology via the phosphorylation of mitochondrial dynamics protein dynamin-related protein 1. In summary, our study provides a novel mitochondrial target to potentiate the plasticity and metabolic fitness of mitochondria within T cells, thus improving the immunotherapeutic potential of these T cells against tumors.


Subject(s)
Mitochondria , Mitochondrial Proteins , T-Lymphocytes , Tumor Microenvironment , Animals , Mice , Humans , Heterografts , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , T-Lymphocytes/metabolism , Gene Knockdown Techniques , Mitochondria/metabolism , Signal Transduction , Immunotherapy, Adoptive
8.
J Immunol ; 189(9): 4417-25, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22993204

ABSTRACT

Dihydroartemisinin (DHA) is an important derivative of the herb medicine Artemisia annua L., used in ancient China. DHA is currently used worldwide to treat malaria by killing malaria-causing parasites. In addition to this prominent effect, DHA is thought to regulate cellular functions, such as angiogenesis, tumor cell growth, and immunity. Nonetheless, how DHA affects T cell function remains poorly understood. We found that DHA potently suppressed Th cell differentiation in vitro. Unexpectedly, however, DHA greatly promoted regulatory T cell (Treg) generation in a manner dependent on the TGF-ßR:Smad signal. In addition, DHA treatment effectively reduced onset of experimental autoimmune encephalomyelitis (EAE) and ameliorated ongoing EAE in mice. Administration of DHA significantly decreased Th but increased Tregs in EAE-inflicted mice, without apparent global immune suppression. Moreover, DHA modulated the mammalian target of rapamycin (mTOR) pathway, because mTOR signal was attenuated in T cells upon DHA treatment. Importantly, enhanced Akt activity neutralized DHA-mediated effects on T cells in an mTOR-dependent fashion. This study therefore reveals a novel immune regulatory function of DHA in reciprocally regulating Th and Treg cell generation through the modulating mTOR pathway. It addresses how DHA regulates immune function and suggests a new type of drug for treating diseases in which mTOR activity is to be tempered.


Subject(s)
Artemisinins/pharmacology , Inflammation/prevention & control , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/physiology
9.
Cancer Biol Ther ; 23(1): 150-162, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35130111

ABSTRACT

Transforming growth factor-ß (TGF-ß) plays a critical role in regulating cell growth and differentiation. Epithelial to mesenchymal transition (EMT) induced by TGF-ß promotes cancer cell migration, invasion, and proliferation. Pirfenidone (5-methyl-1-phenyl-2(1 H)-pyridone, PFD), an approved drug for treating pulmonary and renal fibrosis, is a potent TGF-ß inhibitor and found reduced incidence of lung cancer and alleviated renal function decline. However, whether PFD plays a role in controlling renal cancer progression is largely unknown. In the present study, we demonstrated that high TGF-ß1 expression was negatively associated with ten-year overall survival of patients with renal cancer. Functionally, blockade of TGF-ß signaling with PFD significantly suppressed the progression of renal cancer in a murine model. Mechanistically, we revealed that PFD significantly decreased the expression and secretion of TGF-ß both in vitro and in vivo tumor mouse model, which further prevented TGF-ß-induced EMT and thus cell proliferation, migration, and invasion. Importantly, the downregulation of TGF-ß upon PFD treatment shaped the immunosuppressive tumor microenvironment by limiting the recruitment of tumor-infiltrating MDSCs. Therefore, our study demonstrated that PFD prevents renal cancer progression by inhibiting TGF-ß production of cancer cells and downstream signaling pathway, which might be presented as a therapeutic adjuvant for renal cancer.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Animals , Carcinoma, Renal Cell/drug therapy , Epithelial-Mesenchymal Transition , Female , Fibrosis , Humans , Kidney Neoplasms/drug therapy , Male , Mice , Pyridones , Transforming Growth Factor beta , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment
10.
Front Immunol ; 12: 707899, 2021.
Article in English | MEDLINE | ID: mdl-34335622

ABSTRACT

Pathogenic Th17 cells are critically involved in many autoimmune diseases, while non-pathogenic Th17 cells are more immune regulatory. Understanding the mechanisms of the induction and maintenance of pathogenic Th17 cells will benefit the development of therapeutic treatments of related diseases. We have shown that the transforming growth factor-ß (TGFß) induced SKI degradation and dissociation from Smad4 complex is a prerequisite for TGFß-induced Th17 cell differentiation. However, it is unclear whether and how SKI regulates pathogenic Th17 differentiation, which does not require TGFß cytokine. Here we showed that SKI expression was downregulated during pathogenic Th17 cell differentiation and the ectopic expression of SKI abrogated the differentiation of pathogenic Th17 cells. Functionally, using a knock-in mouse model, we found ectopic SKI expression specifically in T cells prevented myelin oligodendrocyte glycoprotein peptide (MOG33-55) induced experimental autoimmune encephalomyelitis (EAE), an animal model of human multiple sclerosis. We further revealed that induced SKI expression in already differentiated pathogenic Th17 cells reduced the maintenance of Th17 program and ameliorated EAE in an adoptive T cell transfer model. Therefore, our study provides valuable insights of targeting SKI to modulate pathogenic Th17 cell function and treat Th17-related diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Proto-Oncogene Proteins/metabolism , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/immunology , Th17 Cells/pathology , Transforming Growth Factor beta/metabolism
11.
Cell Mol Immunol ; 18(10): 2410-2421, 2021 10.
Article in English | MEDLINE | ID: mdl-32612153

ABSTRACT

Acute viral infection causes illness and death. In addition, an infection often results in increased susceptibility to a secondary infection, but the mechanisms behind this susceptibility are poorly understood. Since its initial identification as a marker for resident memory CD8+ T cells in barrier tissues, the function and regulation of CD103 integrin (encoded by ITGAE gene) have been extensively investigated. Nonetheless, the function and regulation of the resident CD103+CD8+ T cell response to acute viral infection remain unclear. Although TGFß signaling is essential for CD103 expression, the precise molecular mechanism behind this regulation is elusive. Here, we reveal a TGFß-SKI-Smad4 pathway that critically and specifically directs resident CD103+CD8+ T cell generation for protective immunity against primary and secondary viral infection. We found that resident CD103+CD8+ T cells are abundant in both lymphoid and nonlymphoid tissues from uninfected mice. CD103 acts as a costimulation signal to produce an optimal antigenic CD8+ T cell response to acute viral infection. There is a reduction in resident CD103+CD8+ T cells following primary infection that results in increased susceptibility of the host to secondary infection. Intriguingly, CD103 expression inversely and specifically correlates with SKI proto-oncogene (SKI) expression but not R-Smad2/3 activation. Ectopic expression of SKI restricts CD103 expression in CD8+ T cells in vitro and in vivo to hamper viral clearance. Mechanistically, SKI is recruited to the Itgae loci to directly suppress CD103 transcription by regulating histone acetylation in a Smad4-dependent manner. Our study therefore reveals that resident CD103+CD8+ T cells dictate protective immunity during primary and secondary infection. Interfering with SKI function may amplify the resident CD103+CD8+ T cell response to promote protective immunity.


Subject(s)
CD8-Positive T-Lymphocytes , DNA-Binding Proteins/genetics , Immunologic Memory , Proto-Oncogene Proteins/genetics , Virus Diseases/immunology , Animals , Mice , Proto-Oncogenes , Transforming Growth Factor beta/metabolism
12.
Mol Immunol ; 139: 184-192, 2021 11.
Article in English | MEDLINE | ID: mdl-34560414

ABSTRACT

Metastatic renal carcinoma is a kind of tumor with high degree of malignancy, but there are no effective treatment methods and strategies at present. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of high mobility group box 1 protein (HMGB1) and a tumor-specific antigen of B7H3 (CD276) for renal carcinoma therapy. Mice bearing subcutaneous human B7H3 (hB7H3)-Renca tumor were immunized with H1-pHMGB1/pB7H3, H1-pB7H3, H1-pHMGB1, or Mock vaccine. Compared to other control groups, the growth of the tumor was significantly inhibited in H1-pHMGB1/pB7H3 vaccine group. The increased proportion and mature of CD11c+ DCs were observed in the spleen of H1-pHMGB1/pB7H3 treated mice. Likewise, HMGB1 promoted B7H3 vaccine to induce tumor-specific CD8+ T cell proliferation and CTL responses. Beyond that, H1-pHMGB1/pB7H3 vaccine strengthened the induction of functional CD8+ T cells. With the depletion of CD8+ T cells, the anti-tumor effect of H1-pHMGB1/pB7H3 also disappeared, indicating that CD8+ T cells are the key factor of the anti-tumor activity of the vaccine. So, to sum up, H1-pHMGB1/pB7H3 vaccine could achieve the desired anti-tumor effect by enhancing the response of tumor-specific functional CD8+ T cell responses. H1 nanoparticle-based vaccines may have great potential and prospect in the treatment of primary solid tumors.


Subject(s)
B7 Antigens/immunology , Cancer Vaccines/pharmacology , Carcinoma, Renal Cell/immunology , HMGB1 Protein/immunology , Kidney Neoplasms/immunology , Adjuvants, Immunologic/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Nanoparticles , Vaccines, DNA/immunology
13.
J Clin Invest ; 130(11): 5893-5908, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32730228

ABSTRACT

As a hallmark of immunological aging, low-grade, chronic inflammation with accumulation of effector memory T cells contributes to increased susceptibility to many aging-related diseases. While the proinflammatory state of aged T cells indicates a dysregulation of immune homeostasis, whether and how aging drives regulatory T cell (Treg) aging and alters Treg function are not fully understood owing to a lack of specific aging markers. Here, by a combination of cellular, molecular, and bioinformatic approaches, we discovered that Tregs senesce more severely than conventional T (Tconv) cells during aging. We found that Tregs from aged mice were less efficient than young Tregs in suppressing Tconv cell function in an inflammatory bowel disease model and in preventing Tconv cell aging in an irradiation-induced aging model. Furthermore, we revealed that DDB1- and CUL4-associated factor 1 (DCAF1) was downregulated in aged Tregs and was critical to restrain Treg aging via reactive oxygen species (ROS) regulated by glutathione-S-transferase P (GSTP1). Importantly, interfering with GSTP1 and ROS pathways reinvigorated the proliferation and function of aged Tregs. Therefore, our studies uncover an important role of the DCAF1/GSTP1/ROS axis in Treg senescence, which leads to uncontrolled inflammation and immunological aging.


Subject(s)
Aging/immunology , Cellular Senescence/immunology , Protein Serine-Threonine Kinases/immunology , Reactive Oxygen Species/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Aging/genetics , Aging/pathology , Animals , Cellular Senescence/genetics , Disease Models, Animal , Down-Regulation/genetics , Down-Regulation/immunology , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology
14.
Front Immunol ; 10: 1988, 2019.
Article in English | MEDLINE | ID: mdl-31552016

ABSTRACT

Foxp3 expressing regulatory T (Treg) cells, as the central negative regulator of adaptive immune system, are essential to suppress immune response and maintain immune homeostasis. However, the function of Treg cells is frequently compromised in autoimmunity and hyper-activated in infections and tumor microenvironments. Thus, manipulating Treg cells becomes a promising therapeutic strategy for treating various diseases. Here we reported that inhibition of Cdk8/Cdk19 activity by small molecule inhibitors CCT251921 or Senexin A greatly promoted the differentiation of Treg cells and the expression of Treg signature genes, such as Foxp3, CTLA4, PD-1, and GITR. Mechanistically, we found that the augmented Treg cell differentiation was due to sensitized TGF-ß signaling by Cdk8/Cdk19 inhibition, which was associated with attenuation of IFN-γ-Stat1 signaling and enhancement of phosphorylated Smad2/3. Importantly, treatment with Cdk8/Cdk19 inhibitor CCT251921 significantly increased Treg population and ameliorated autoimmune symptoms in an experimental autoimmune encephalomyelitis (EAE) model. Taken together, our study reveals a novel role of Cdk8/Cdk19 in Treg cell differentiation and provides a potential target for Treg cell based therapeutics.


Subject(s)
Cyclin-Dependent Kinase 8/immunology , Cyclin-Dependent Kinases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/metabolism
15.
Int J Biol Sci ; 14(1): 10-20, 2018.
Article in English | MEDLINE | ID: mdl-29483821

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) are among the most common developmental defects in humans. Despite of several known CAKUT-related loci (HNF1B, PAX2, EYA1, etc.), the genetic etiology of CAKUT remains to be elucidated for most patients. In this study, we report that disruption of the Holliday Junction resolvase gene Gen1 leads to renal agenesis, duplex kidney, hydronephrosis, and vesicoureteral reflux (VUR) in mice. GEN1 interacts with SIX1 and enhances the transcriptional activity of SIX1/EYA1, a key regulatory complex of the GDNF morphogen. Gen1 mutation impairs Grem1 and Gdnf expression, resulting in excessive ureteric bud formation and defective ureteric bud branching during early kidney development. These results revealed an unidentified role of GEN1 in kidney development and suggested its contribution to CAKUT.


Subject(s)
Holliday Junction Resolvases/metabolism , Kidney/abnormalities , Kidney/metabolism , Urinary Tract/abnormalities , Urinary Tract/metabolism , Animals , HEK293 Cells , Holliday Junction Resolvases/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunoprecipitation , In Situ Hybridization , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Protein Binding , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism
16.
Article in English | MEDLINE | ID: mdl-27827866

ABSTRACT

This research aims at combined and relative effect levels on anxiety of: (1) perceived risk, knowledge, optimism, pessimism, and social trust; and (2) four sub-variables of social trust among inhabitants concerning living on heavy metal contaminated soil. On the basis of survey data from 499 Chinese respondents, results suggest that perceived risk, pessimism, optimism, and social trust have individual, significant, and direct effects on anxiety, while knowledge does not. Knowledge has significant, combined, and interactive effects on anxiety together with social trust and pessimism, respectively, but does not with perceived risk and optimism. Social trust, perceived risk, pessimism, knowledge, and optimism have significantly combined effects on anxiety; the five variables as a whole have stronger predictive values than each one individually. Anxiety is influenced firstly by social trust and secondly by perceived risk, pessimism, knowledge, and optimism. Each of four sub-variables of social trust has an individual, significant, and negative effect on anxiety. When introducing four sub-variables into one model, trust in social organizations and in the government have significantly combined effects on anxiety, while trust in experts and in friends and relatives do not; anxiety is influenced firstly by trust in social organization, and secondly by trust in the government.


Subject(s)
Anxiety/etiology , Environmental Pollution/adverse effects , Metals, Heavy/toxicity , Residence Characteristics , Soil Pollutants/toxicity , Adolescent , Adult , Aged , Aged, 80 and over , Anxiety/psychology , China , Female , Health Knowledge, Attitudes, Practice , Humans , Male , Middle Aged , Models, Psychological , Optimism , Pessimism , Trust , Young Adult
17.
Nat Commun ; 7: 10307, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728942

ABSTRACT

On activation, naive T cells grow in size and enter cell cycle to mount immune response. How the fundamental processes of T-cell growth and cell cycle entry are regulated is poorly understood. Here we report that DCAF1 (Ddb1-cullin4-associated-factor 1) is essential for these processes. The deletion of DCAF1 in T cells impairs their peripheral homeostasis. DCAF1 is upregulated on T-cell receptor activation and critical for activation-induced T-cell growth, cell cycle entry and proliferation. In addition, DCAF1 is required for T-cell expansion and function during anti-viral and autoimmune responses in vivo. DCAF1 deletion leads to a drastic stabilization of p53 protein, which can be attributed to a requirement of DCAF1 for MDM2-mediated p53 poly-ubiquitination. Importantly, p53 deletion rescues the cell cycle entry defect but not the growth defect of DCAF1-deficient cells. Therefore, DCAF1 is vital for T-cell function through p53-dependent and -independent mechanisms.


Subject(s)
Carrier Proteins/immunology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/immunology , Animals , Carrier Proteins/genetics , Cell Proliferation , Female , Gene Deletion , Lymphocyte Activation , Male , Mice , Mice, Knockout , Protein Serine-Threonine Kinases , T-Lymphocytes/cytology , Tumor Suppressor Protein p53/genetics
18.
Immunol Lett ; 135(1-2): 34-42, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-20932862

ABSTRACT

The Id3 gene has been shown to play important roles in the development and function of broad tissue types including B and T cells. Id3 deficient mice develop autoimmune disease similar to human Sjögren's syndrome. Both B and T lymphocytes have been implicated to contribute to the disease phenotype in this disease model. In order to gain a better understanding of individual cell types in this disease model, we generated an Id3 conditional allele. An LckCre transgene was used to induce Id3 deletion in developing T cells. We showed that the Id3 gene was efficiently disrupted in early thymocyte development prior to T cell receptor (TCR)-mediated positive selection. Consequently, thymocyte maturation was impaired in the conditional knockout mice. These mice developed exocrinopathy starting at two months of age and subsequently exhibited high incidence of lymphocyte infiltration to salivary glands between eight and 12 months of age. This progressive feature of disease development is very similar to those observed in Id3 germline knockout mice. This study establishes a new model for investigating the relationship between T cell development and autoimmune disease. Our observation provides an experimental case that autoimmune disease may be induced by acquired mutation in developing T cells.


Subject(s)
Disease Models, Animal , Inhibitor of Differentiation Proteins/immunology , Sjogren's Syndrome/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Gene Deletion , Humans , Inhibitor of Differentiation Proteins/genetics , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL