Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
J Biol Chem ; 297(6): 101330, 2021 12.
Article in English | MEDLINE | ID: mdl-34688667

ABSTRACT

CD4+ T cells differentiate into subsets that promote immunity or minimize damage to the host. T helper 17 cells (Th17) are effector cells that function in inflammatory responses. T regulatory cells (Tregs) maintain tolerance and prevent autoimmunity by secreting immunosuppressive cytokines and expressing check point receptors. While the functions of Th17 and Treg cells are different, both cell fate trajectories require T cell receptor (TCR) and TGF-ß receptor (TGF-ßR) signals, and Th17 polarization requires an additional IL-6 receptor (IL-6R) signal. Utilizing high-resolution phosphoproteomics, we identified that both synergistic and additive interactions between TCR, TGF-ßR, and IL-6R shape kinase signaling networks to differentially regulate key pathways during the early phase of Treg versus Th17 induction. Quantitative biochemical analysis revealed that CD4+ T cells integrate receptor signals via SMAD3, which is a mediator of TGF-ßR signaling. Treg induction potentiates the formation of the canonical SMAD3/4 trimer to activate a negative feedback loop through kinases PKA and CSK to suppress TCR signaling, phosphatidylinositol metabolism, and mTOR signaling. IL-6R signaling activates STAT3 to bind SMAD3 and block formation of the SMAD3/4 trimer during the early phase of Th17 induction, which leads to elevated TCR and PI3K signaling. These data provide a biochemical mechanism by which CD4+ T cells integrate TCR, TGF-ß, and IL-6 signals via generation of alternate SMAD3 complexes that control the development of early signaling networks to potentiate the choice of Treg versus Th17 cell fate.


Subject(s)
Receptors, Antigen, T-Cell/immunology , Receptors, Interleukin-6/immunology , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Line , Cells, Cultured , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology
2.
J Immunol ; 207(10): 2456-2464, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34615735

ABSTRACT

Lactoferrin (LF) is known to possess anti-inflammatory activity, although its mechanisms of action are not well-understood. The present study asked whether LF affects the commitment of inducible regulatory T cells (Tregs). LF substantially promoted Foxp3 expression by mouse activated CD4+T cells, and this activity was further enhanced by TGF-ß1. Interestingly, blocking TGF-ß with anti-TGF-ß Ab completely abolished LF-induced Foxp3 expression. However, no significant amount of soluble TGF-ß was released by LF-stimulated T cells, suggesting that membrane TGF-ß (mTGF-ß) is associated. Subsequently, it was found that LF binds to TGF-ß receptor III, which induces reactive oxygen species production and diminishes the expression of mTGF-ß-bound latency-associated peptide, leading to the activation of mTGF-ß. It was followed by phosphorylation of Smad3 and enhanced Foxp3 expression. These results suggest that LF induces Foxp3+ Tregs through TGF-ß receptor III/reactive oxygen species-mediated mTGF-ß activation, triggering canonical Smad3-dependent signaling. Finally, we found that the suppressive activity of LF-induced Tregs is facilitated mainly by CD39/CD73-induced adenosine generation and that this suppressor activity alleviates inflammatory bowel disease.


Subject(s)
Lactoferrin/metabolism , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Colitis/immunology , Colitis/metabolism , Lactoferrin/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
3.
Fish Shellfish Immunol ; 105: 41-52, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32629101

ABSTRACT

Transforming growth factor-ß type III receptor (TßR3), as a co-receptor of TGF-ß superfamily, plays critical roles in development and growth as well as some disease pathogeneses by presenting ligands to other receptors in vertebrates. However, the identification and functional characterization of TßR3 had not been reported yet in invertebrates. In the present study, TßR3 was first identified and characterized in mud crab Scylla paramamosain. The obtained cDNA length of SpTßR3 was 2, 424 bp with a 1, 854 bp open reading frame, which encoded a putative peptide of 617 amino acids containing a typical transmembrane region and a Zona pellucida (ZP) domain. Real-time PCR results showed that SpTßR3 was predominantly expressed at early embryonic development stage and early postmolt stage, suggesting its participation in development and growth. We report, for the first time in invertebrates, the challenge of both Vibro alginolyticus and Poly (I:C) could alter the expression patterns of SpTßR3. Notably, the expression levels of SpIKK, two NF-κB members (SpRelish and SpDorsal), and five antimicrobial peptide genes (SpCrustin and SpALF1-4) were significantly suppressed when SpTßR3 was interfered in vivo. Secondly, the overexpression of SpTßR3 in vitro could activate NF-κB signaling through the dual-luciferase reporter assays. Furthermore, the bacterial clearance assay after SpTßR3 was silenced in vivo highlighted the potential of SpTßR3 in activating the innate immune responses. These results implied the involvement of SpTßR3 in the innate immune responses by regulating the NF-κB pathway. This study first indicated that TßR3 was present in invertebrate, and it participated in not only the development and growth but also the innate immunity of S. paramamosain. It also provided new insights into the origin or evolution of TGF-ß receptors in crustacean species and even in invertebrates.


Subject(s)
Brachyura/genetics , Brachyura/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction/genetics , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Base Sequence , Gene Expression Profiling , Phylogeny , Poly I-C/pharmacology , Receptors, Transforming Growth Factor beta/chemistry , Sequence Alignment , Vibrio alginolyticus/physiology
4.
Mol Ther ; 28(2): 548-560, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31870622

ABSTRACT

The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor ß (TGF-ß) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.


Subject(s)
Genital Neoplasms, Female/immunology , Immunotherapy, Adoptive , Ovarian Neoplasms/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Epitopes/genetics , Epitopes/immunology , Female , Genital Neoplasms, Female/therapy , Humans , Mice , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
5.
Cancer Immunol Res ; 7(5): 784-796, 2019 05.
Article in English | MEDLINE | ID: mdl-30872264

ABSTRACT

The chemokine CXCL13 mediates recruitment of B cells to tumors and is essential for the formation of tertiary lymphoid structures (TLSs). TLSs are thought to support antitumor immunity and are associated with improved prognosis. However, it remains unknown whether TLSs are formed in response to the general inflammatory character of the tumor microenvironment, or rather, are induced by (neo)antigen-specific adaptive immunity. We here report on the finding that the TGFß-dependent CD103+CD8+ tumor-infiltrating T-cell (TIL) subpopulation expressed and produced CXCL13. Accordingly, CD8+ T cells from peripheral blood activated in the presence of TGFß upregulated CD103 and secreted CXCL13. Conversely, inhibition of TGFß receptor signaling abrogated CXCL13 production. CXCL13+CD103+CD8+ TILs correlated with B-cell recruitment, TLSs, and neoantigen burden in six cohorts of human tumors. Altogether, our findings indicated that TGFß plays a noncanonical role in coordinating immune responses against human tumors and suggest a potential role for CXCL13+CD103+CD8+ TILs in mediating B-cell recruitment and TLS formation in human tumors.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL13/immunology , Integrin alpha Chains/immunology , Ovarian Neoplasms/immunology , Receptors, Transforming Growth Factor beta/immunology , Antigens, Neoplasm/immunology , Female , Humans
6.
Eur Cytokine Netw ; 30(3): 107-113, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31957700

ABSTRACT

Interleukin-10 (IL-10) is a pleiotropic cytokine, which has both regulatory and stimulatory effects on different immune cell types. Different studies have reported the importance of IL-10 and Transforming growth factor-beta (TGF-ß) in the regulation of B cell class switching the production of immunoglobulin A (IgA); however, the underlying mechanisms remain to be fully elucidated. The objective of this study was to investigate the TGF-ß response during B stimulation of human B cells by IL-10. Pan B cells of healthy donors were negatively purified by a magnetic cell separation technique. B cells were cultured with multimeric CD40 ligand (mCD40L) and IL-10 for two and seven days. After harvesting in specific days, TGF-ß receptor II and surface IgA expression was determined by flow cytometry, while IgA and TGF-ß secretion was assessed by enzyme-linked immunosorbent assay. B cells endogenously expressed TGF-ß receptor II and after 48 hours cultivation with mCD40L or mCD40L plus IL-10, both the expression of this receptor and the production of TGF-ß were significantly increased. Notably, TGF-ß levels following stimulation with mCD40L and IL-10 were higher than those produced by B cells stimulated with mCD40L alone. Furthermore, at day 7 and following IL-10 stimulation, there was a significant rise in the amount of IgA secretion by class-switched plasma cells, which was higher than stimulation with mCD40L alone. Our findings suggest that IL-10 can modulate TGF-ß production and TGF-ß receptor expression in mCD40-activated human B lymphocytes.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin A/immunology , Interleukin-10/immunology , Receptors, Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/immunology , Up-Regulation/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , Humans , Immunoglobulin Class Switching/immunology , Lymphocyte Activation/immunology
7.
J Nucl Med ; 59(8): 1234-1242, 2018 08.
Article in English | MEDLINE | ID: mdl-29674421

ABSTRACT

We have developed the 16F12 mouse monoclonal antibody (mAb), which targets the Müllerian-inhibiting substance receptor, type II (MISRII), expressed by ovarian tumors. Here, we assessed in preclinical models the possibility of using radiolabeled 16F12 in a theranostic approach for small-volume ovarian peritoneal carcinomatosis, such as after cytoreductive surgery. Methods: DOTA-, DTPA- or deferoxamine mesylate-conjugated 16F12 mAb was radiolabeled with ß-particle (177Lu) or α-particle (213Bi) emitters for therapeutic use and with 89Zr for PET imaging. On the 13th postxenograft day, mice bearing intraperitoneal MISRII-positive AN3CA endometrial carcinoma cell xenografts were treated by conventional intraperitoneal radioimmunotherapy (IP-RIT) with 10 MBq of 177Lu-16F12 or 12.9 MBq of 213Bi-16F12 or by brief intraperitoneal radioimmunotherapy (BIP-RIT) using 50 MBq of 177Lu-16F12 or 37 MBq of 213Bi-16F12. For BIP-RIT, 30 min after injection of the radiolabeled mAbs, the peritoneal cavity was washed to remove the unbound radioactivity. The biodistribution of 177Lu- and 213Bi-16F12 mAbs was determined and then used for dose assessment. Hematologic toxicity was also monitored. Results: The 16F12 mAb was satisfactorily radiolabeled for both therapy and imaging. IP-RIT with 177Lu-16F12 was slightly more efficient in delaying tumor growth than IP-RIT with 213Bi-16F12. Conversely, 213Bi-16F12 was more efficient than 177Lu-16F12 in BIP-RIT. The biodistribution analysis showed that the tumor-to-blood uptake ratio was significantly higher with BIP-RIT than with IP-RIT for both 213Bi- and 177Lu-16F12. Hematologic toxicity was more pronounced with 177Lu-16F12 than with 213Bi-16F12. SPECT/CT images (after BIP-RIT with 177Lu-16F12) and PET/CT images (after injection of 89Zr-16F12 in the tail vein) showed focal uptake at the tumor site. Conclusion: Radiolabeled 16F12 could represent a new theranostic tool for small-volume ovarian peritoneal carcinomatosis. Specifically, 213Bi-16F12-based BIP-RIT could be proposed to selected patients as an alternative adjuvant treatment immediately after cytoreductive surgery. An anti-MISRII mAb is currently being used in a first-in-human study, thus making radiolabeled anti-MISRII mAbs a realistic theranostic option for the clinic.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/radiotherapy , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Deferoxamine/chemistry , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Isotope Labeling , Mice , Ovarian Neoplasms/metabolism , Pentetic Acid/chemistry , Positron Emission Tomography Computed Tomography , Radiochemistry , Tissue Distribution
8.
Nat Commun ; 9(1): 741, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467463

ABSTRACT

A majority of cancers fail to respond to immunotherapy with antibodies targeting immune checkpoints, such as cytotoxic T-lymphocyte antigen-4 (CTLA-4) or programmed death-1 (PD-1)/PD-1 ligand (PD-L1). Cancers frequently express transforming growth factor-ß (TGFß), which drives immune dysfunction in the tumor microenvironment by inducing regulatory T cells (Tregs) and inhibiting CD8+ and TH1 cells. To address this therapeutic challenge, we invent bifunctional antibody-ligand traps (Y-traps) comprising an antibody targeting CTLA-4 or PD-L1 fused to a TGFß receptor II ectodomain sequence that simultaneously disables autocrine/paracrine TGFß in the target cell microenvironment (a-CTLA4-TGFßRIIecd and a-PDL1-TGFßRIIecd). a-CTLA4-TGFßRIIecd is more effective in reducing tumor-infiltrating Tregs and inhibiting tumor progression compared with CTLA-4 antibody (Ipilimumab). Likewise, a-PDL1-TGFßRIIecd exhibits superior antitumor efficacy compared with PD-L1 antibodies (Atezolizumab or Avelumab). Our data demonstrate that Y-traps counteract TGFß-mediated differentiation of Tregs and immune tolerance, thereby providing a potentially more effective immunotherapeutic strategy against cancers that are resistant to current immune checkpoint inhibitors.


Subject(s)
Antibodies/therapeutic use , Breast Neoplasms/therapy , Immunotherapy , Melanoma/therapy , Transforming Growth Factor beta/immunology , Animals , Antibodies/genetics , Antibodies/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cohort Studies , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Lymphocytes, Tumor-Infiltrating , Melanoma/genetics , Melanoma/immunology , Mice , Mice, Inbred NOD , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/genetics
9.
Nat Commun ; 9(1): 17, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29295981

ABSTRACT

Multiple sclerosis (MS) is a T cell-mediated autoimmune disease of the central nervous system. Foxp3+ regulatory T (Treg) cells are reduced in frequency and dysfunctional in patients with MS, but the underlying mechanisms of this deficiency are unclear. Here, we show that induction of human IFN-γ-IL-17A-Foxp3+CD4+ T cells is inhibited in the presence of circulating exosomes from patients with MS. The exosomal miRNA profile of patients with MS differs from that of healthy controls, and let-7i, which is markedly increased in patients with MS, suppresses induction of Treg cells by targeting insulin like growth factor 1 receptor (IGF1R) and transforming growth factor beta receptor 1 (TGFBR1). Consistently, the expression of IGF1R and TGFBR1 on circulating naive CD4+ T cells is reduced in patients with MS. Thus, our study shows that exosomal let-7i regulates MS pathogenesis by blocking the IGF1R/TGFBR1 pathway.


Subject(s)
Exosomes/immunology , MicroRNAs/immunology , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Exosomes/genetics , Exosomes/metabolism , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Male , MicroRNAs/genetics , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptor, IGF Type 1 , Receptor, Transforming Growth Factor-beta Type I , Receptors, Somatomedin/genetics , Receptors, Somatomedin/immunology , Receptors, Somatomedin/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcriptome/immunology
10.
Cancer Prev Res (Phila) ; 10(11): 612-624, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29093011

ABSTRACT

Epithelial ovarian carcinoma (EOC) is the most prevalent form of ovarian cancer in the United States, representing approximately 85% of all cases and causing more deaths than any other gynecologic malignancy. We propose that optimized control of EOC requires the incorporation of a vaccine capable of inducing safe and effective preemptive immunity in cancer-free women. In addition, we hypothesize that ovarian-specific self-proteins that are "retired" from autoimmune-inducing expression levels as ovaries age but are expressed at high levels in emerging EOC may serve as vaccine targets for mediating safe and effective primary immunoprevention. Here, we show that expression of the extracellular domain of anti-Müllerian hormone receptor II (AMHR2-ED) in normal tissues is confined exclusively to the human ovary, drops to nonautoimmune inducing levels in postmenopausal ovaries, and is at high levels in approximately 90% of human EOC. We found that AMHR2-ED vaccination significantly inhibits growth of murine EOC and enhances overall survival without inducing oophoritis in aged female mice. The observed inhibition of EOC growth was mediated substantially by induction of AMHR2-ED-specific IgG antibodies that agonize receptor signaling of a Bax/caspase-3-dependent proapoptotic cascade. Our results indicate that AMHR2-ED vaccination may be particularly useful in providing safe and effective preemptive immunity against EOC in women at high genetic or familial risk who have the greatest need for a preventive vaccine and ultimately in cancer-free postmenopausal women who account for 75% of all EOC cases. Cancer Prev Res; 10(11); 612-24. ©2017 AACRSee related editorial by Shoemaker et al., p. 607.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/therapy , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Aged , Aged, 80 and over , Cancer Vaccines/immunology , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance/immunology , Middle Aged , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Oophoritis/epidemiology , Oophoritis/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovary/immunology , Ovary/pathology , Polyendocrinopathies, Autoimmune/epidemiology , Polyendocrinopathies, Autoimmune/immunology , Postmenopause , Protein Serine-Threonine Kinases , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vaccination/methods , Xenograft Model Antitumor Assays
11.
Cell Immunol ; 319: 43-52, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28864263

ABSTRACT

Cholera toxin B subunit fusion to autoantigens such as proinsulin (CTB-INS) down regulate dendritic cell (DC) activation and stimulate synthesis of DC immunosuppressive cytokines. Recent studies of CTB-INS induction of immune tolerance in human DCs indicate that increased biosynthesis of indoleamine 2,3-dioxygenase (IDO1) may play an important role in CTB-INS vaccine suppression of DC activation. Studies in murine models suggest a role for transforming growth factor beta (TGF-ß) in the stimulation of IDO1 biosynthesis, for the induction of tolerance in DCs. Here, we investigated the contribution of TGF-ß superfamily proteins to CTB-INS induction of IDO1 biosynthesis in human monocyte-derived DCs (moDCs). We show that CTB-INS upregulates the level of TGF-ß1, activin-A and the TGF-ß activator, integrin αvß8 in human DCs. However, inhibition of endogenous TGF-ß, activin-A or addition of biologically active TGF-ß1, and activin-A, did not inhibit or stimulate IDO1 biosynthesis in human DCs treated with CTB-INS. While inhibition with the kinase inhibitor, RepSox, blocked SMAD2/3 phosphorylation and diminished IDO1 biosynthesis in a concentration dependent manner. Specific blocking of the TGF-ß type 1 kinase receptor with SB-431542 did not arrest IDO1 biosynthesis, suggesting the involvement of a different kinase pathway other than TGF-ß type 1 receptor kinase in CTB-INS induction of IDO1 in human moDCs. Together, our experimental findings identify additional immunoregulatory proteins induced by the CTB-INS fusion protein, suggesting CTB-INS may utilize multiple mechanisms in the induction of tolerance in human moDCs.


Subject(s)
Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta1/genetics , Activins/genetics , Activins/immunology , Animals , Cell Differentiation , Cholera Toxin/genetics , Cholera Toxin/immunology , Cloning, Molecular , Dendritic Cells/cytology , Dendritic Cells/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Integrins/genetics , Integrins/immunology , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Primary Cell Culture , Proinsulin/genetics , Proinsulin/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/immunology , Smad3 Protein/genetics , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology
12.
Cell Physiol Biochem ; 41(3): 1189-1198, 2017.
Article in English | MEDLINE | ID: mdl-28472799

ABSTRACT

BACKGROUND/AIMS: Immunosuppression is one of the hallmarks of cancer; however, its molecular mechanism remains unknown. In the present study, we sought to investigate the expression and activation of yes-associated protein 1 (YAP-1) and its roles in T cells within hepatocellular carcinoma (HCC). METHODS: The expression and activation of YAP-1 were accessed by real-time PCR, immunohistochemistry staining, western blot, and flow cytometry. The potential regulation effect of YAP-1 on Regulatory T cells (Tregs) differentiation was predicted using bioinformatics tools and verified by in vitro studies. RESULTS: Significant overexpression and activation of YAP-1 was detected within peripheral blood mononuclear cells and showed positive linear correlation to Treg percentage; it may serve as a valuable indicator of a bad prognosis. Using in vitro studies, we found that overexpression and activation of YAP-1 can promote naïve T cell polarization stimulation to Tregs by increasing the expression of TGFBR2. The YAP-1/TEADs DNA binding site was spotted within the promoter region of TGFBR2 and related to its transcription activity. YAP-1 acted as a co-activator of TGFBR2 transcription by binding directly to the TGFBR2 promoter through TEADs. CONCLUSION: Overexpression and activation of YAP-1 in HCC T cells can induce immunosuppression by promoting Treg differentiation via transcriptional enhancement of TGFBR2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , T-Lymphocytes, Regulatory/immunology , Adaptor Proteins, Signal Transducing/immunology , Adult , Binding Sites , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphoproteins/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction , T-Lymphocytes, Regulatory/pathology , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic , YAP-Signaling Proteins
13.
Oncotarget ; 8(23): 37061-37079, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28427157

ABSTRACT

Ovarian cancer is the leading cause of death in women with gynecological cancers and despite recent advances, new and more efficient therapies are crucially needed. Müllerian Inhibiting Substance type II Receptor (MISRII, also named AMHRII) is expressed in most ovarian cancer subtypes and is a novel potential target for ovarian cancer immunotherapy. We previously developed and tested 12G4, the first murine monoclonal antibody (MAb) against human MISRII. Here, we report the humanization, affinity maturation and glyco-engineering steps of 12G4 to generate the Fc-optimized 3C23K MAb, and the evaluation of its in vivo anti-tumor activity. The epitopes of 3C23K and 12G4 were strictly identical and 3C23K affinity for MISRII was enhanced by a factor of about 14 (KD = 5.5 × 10-11 M vs 7.9 × 10-10 M), while the use of the EMABling® platform allowed the production of a low-fucosylated 3C23K antibody with a 30-fold KD improvement of its affinity to FcγRIIIa. In COV434-MISRII tumor-bearing mice, 3C23K reduced tumor growth more efficiently than 12G4 and its combination with carboplatin was more efficient than each monotherapy with a mean tumor size of 500, 1100 and 100 mm3 at the end of treatment with 3C23K (10 mg/kg, Q3-4D12), carboplatin (60 mg/kg, Q7D4) and 3C23K+carboplatin, respectively. Conversely, 3C23K-FcKO, a 3C23K form without affinity for the FcγRIIIa receptor, did not display any anti-tumor effect in vivo. These results strongly suggested that 3C23K mechanisms of action are mainly Fc-related. In vitro, antibody-dependent cytotoxicity (ADCC) and antibody-dependent cell phagocytosis (ADCP) were induced by 3C23K, as demonstrated with human effector cells. Using human NK cells, 50% of the maximal lysis was obtained with a 46-fold lower concentration of low-fucosylated 3C23K (2.9 ng/ml) than of 3C23K expressed in CHO cells (133.35 ng/ml). As 3C23K induced strong ADCC with human PBMC but almost none with murine PBMC, antibody-dependent cell phagocytosis (ADCP) was then investigated. 3C23K-dependent (100 ng/ml) ADCP was more active with murine than human macrophages (only 10% of living target cells vs. about 25%). These in vitro results suggest that the reduced ADCC with murine effectors could be partially balanced by ADCP activity in in vivo experiments. Taken together, these preclinical data indicate that 3C23K is a new promising therapeutic candidate for ovarian cancer immunotherapy and justify its recent introduction in a phase I clinical trial.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Peptide/immunology , Receptors, Transforming Growth Factor beta/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Female , Glycosylation , Humans , Mice, Nude , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Protein Engineering
14.
Immunity ; 46(4): 660-674, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423340

ABSTRACT

Regulatory T cells (Treg cells) perform suppressive functions in disparate tissue environments and against many inflammatory insults, yet the tissue-enriched factor(s) that influence Treg cell phenotype and function remain largely unknown. We have shown a vital role for transforming growth factor-ß (TGF-ß) signals in safe-guarding specific Treg cell functions. TGF-ß signals were dispensable for steady-state Treg cell homeostasis and for Treg cell suppression of T cell proliferation and T helper-1 (Th1) cell differentiation. However, Treg cells require TGF-ß signals to appropriately dampen Th17 cells and regulate responses in the gastrointestinal tract. TGF-ß signaling maintains CD103 expression, promotes expression of the colon-specific trafficking molecule GPR15, and inhibits expression of GPR174, a receptor for lysophosphatidylserine, on Treg cells, collectively supporting the accumulation and retention of Treg cells in the colon and control of colitogenic responses. Thus, we reveal an unrecognized function for TGF-ß signaling as an upstream factor controlling Treg cell activity in specific tissue environments.


Subject(s)
Organ Specificity/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Homeostasis/immunology , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Transforming Growth Factor beta/metabolism
15.
Clin Immunol ; 178: 79-85, 2017 05.
Article in English | MEDLINE | ID: mdl-28161409

ABSTRACT

Chronic mucocutaneous candidiasis, characterized by persistent or recurrent fungal infections, represents the clinical hallmark in gain-of-function (GOF) signal transducer and activator of transcription 1 (STAT1) mutation carriers. Several cases of intracranial aneurysms have been reported in patients with GOF STAT1 mutation but the paucity of reported cases likely suggested this association still as serendipity. In order to endorse this association, we link the development of intracranial aneurysms with STAT1 GOF mutation by presenting the two different cases of a patient and her mother, and demonstrate upregulated phosphorylated STAT4 and IL-12 receptor ß1 upon stimulation in patient's blood cells. We also detected increased transforming growth factor (TGF)-ß type 2 receptor expression, particularly in CD14+ cells, and a slightly higher phosphorylation rate of SMAD3. In addition, the mother of the patient developed disseminated bacille Calmette-Guérin disease after vaccination, speculating that GOF STAT1 mutations may confer a predisposition to weakly virulent mycobacteria.


Subject(s)
Candidiasis, Chronic Mucocutaneous/genetics , Intracranial Aneurysm/genetics , STAT1 Transcription Factor/genetics , Adjuvants, Immunologic/adverse effects , Adult , Angiography, Digital Subtraction , BCG Vaccine/adverse effects , Candidiasis, Chronic Mucocutaneous/complications , Candidiasis, Chronic Mucocutaneous/immunology , Candidiasis, Chronic Mucocutaneous/metabolism , Cerebral Angiography , Female , Humans , Intracranial Aneurysm/complications , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/metabolism , Mothers , Mutation , Phosphoproteins/immunology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Interleukin-12/immunology , Receptors, Interleukin-12/metabolism , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , Smad3 Protein/immunology , Smad3 Protein/metabolism , Tuberculosis/chemically induced , Tuberculosis/immunology , Young Adult
16.
Gastroenterology ; 152(1): 36-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27773809

ABSTRACT

Transforming growth factor (TGF)-ß cytokines signal via a complex network of pathways to regulate proliferation, differentiation, adhesion, migration, and other functions in many cell types. A high percentage of colorectal tumors contain mutations that disrupt TGF-ß family member signaling. We review how TGF-ß family member signaling is altered during development of colorectal cancer, models of study, interaction of pathways, and potential therapeutic strategies.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad Proteins/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Colorectal Neoplasms/immunology , Germ-Line Mutation , Homeostasis , Humans , Mice , Mice, Knockout , Receptors, Transforming Growth Factor beta/immunology , Smad Proteins/metabolism
17.
Am J Physiol Endocrinol Metab ; 312(3): E161-E174, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27894066

ABSTRACT

Hypothalamic inflammation was recently found to mediate obesity-related hypertension, but the responsible upstream mediators remain unexplored. In this study, we show that dietary obesity is associated with extracellular release of mitochondrial DNA (mtDNA) into the cerebrospinal fluid and that central delivery of mtDNA mimics transforming growth factor-ß (TGFß) excess to activate downstream signaling pathways. Physiological study reveals that central administration of mtDNA or TGFß is sufficient to cause hypertension in mice. Knockout of the TGFß receptor in proopiomelanocortin neurons counteracts the hypertensive effect of not only TGFß but also mtDNA excess, while the hypertensive action of central mtDNA can be blocked pharmacologically by a TGFß receptor antagonist or genetically by TGFß receptor knockout. Finally, we confirm that obesity-induced hypertension can be reversed through central treatment with TGFß receptor antagonist. In conclusion, circulating mtDNA in the brain employs neural TGFß pathway to mediate a central inflammatory mechanism of obesity-related hypertension.


Subject(s)
Blood Pressure/immunology , DNA, Mitochondrial/immunology , Hypertension/immunology , Hypothalamus/immunology , Obesity/immunology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Animals , Benzamides/pharmacology , Blotting, Western , DNA, Mitochondrial/cerebrospinal fluid , DNA, Mitochondrial/metabolism , DNA, Mitochondrial/pharmacology , Diet, High-Fat , Dioxoles/pharmacology , Hypertension/etiology , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Neurons/immunology , Neurons/metabolism , Obesity/complications , Pro-Opiomelanocortin/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/immunology , Third Ventricle , Transforming Growth Factor beta1/pharmacology
18.
J Immunol ; 197(11): 4344-4350, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27793996

ABSTRACT

Immune tolerance between the fetus and mother represents an active process by which the developing fetus must not mount immune responses to noninherited Ags on chimeric maternal cells that reside in fetal tissue. This is, in part, mediated by the suppressive influence of CD4+FOXP3+CD25+ regulatory T cells (Tregs). Fetal secondary lymphoid organs have an increased frequency of Tregs and, as compared with adult T cells, fetal naive CD4+ T cells exhibit a strong predisposition to differentiate into Tregs when stimulated. This effect is mediated by the TCR and TGF-ß pathways, and fetal T cells show significantly increased Treg differentiation in response to anti-CD3 and TGF-ß stimulation. Naive fetal T cells also exhibit increased signaling through the TGF-ß pathway, with these cells demonstrating increased expression of the signaling mediators TGF-ßRI, TGF-ßRIII, and SMAD2, and higher levels of SMAD2/SMAD3 phosphorylation. Increased fetal Treg differentiation is mediated by the RNA-binding protein Lin28b, which is overexpressed in fetal T cells as compared with adult cells. When Lin28b expression is decreased in naive fetal T cells, they exhibit decreased Treg differentiation that is associated with decreased TGF-ß signaling and lowered expression of TGF-ßRI, TGF-ßRIII, and SMAD2. Lin28b regulates the maturation of let-7 microRNAs, and these TGF-ß signaling mediators are let-7 targets. We hypothesize that loss of Lin28b expression in fetal T cells leads to increased mature let-7, which causes decreased expression of TGF-ßRI, TGF-ßRIII, and SMAD2 proteins. A reduction in TGF-ß signaling leads to reduced Treg numbers.


Subject(s)
Cell Differentiation/immunology , Fetus/immunology , RNA-Binding Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , CD3 Complex/immunology , Female , Fetus/cytology , Gene Expression Regulation, Developmental/immunology , Humans , Male , MicroRNAs/immunology , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Antigen, T-Cell/immunology , Receptors, Transforming Growth Factor beta/immunology , Smad2 Protein/immunology , T-Lymphocytes, Regulatory/cytology
19.
Proc Natl Acad Sci U S A ; 113(35): 9816-21, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27540116

ABSTRACT

Clathrin, a cytosolic protein composed of heavy and light chain subunits, assembles into a vesicle coat, controlling receptor-mediated endocytosis. To establish clathrin light chain (CLC) function in vivo, we engineered mice lacking CLCa, the major CLC isoform in B lymphocytes, generating animals with CLC-deficient B cells. In CLCa-null mice, the germinal centers have fewer B cells, and they are enriched for IgA-producing cells. This enhanced switch to IgA production in the absence of CLCa was attributable to increased transforming growth factor ß receptor 2 (TGFßR2) signaling resulting from defective endocytosis. Internalization of C-X-C chemokine receptor 4 (CXCR4), but not CXCR5, was affected in CLCa-null B cells, and CLC depletion from cell lines affected endocytosis of the δ-opioid receptor, but not the ß2-adrenergic receptor, defining a role for CLCs in the uptake of a subset of signaling receptors. This instance of clathrin subunit deletion in vertebrates demonstrates that CLCs contribute to clathrin's role in vivo by influencing cargo selectivity, a function previously assigned exclusively to adaptor molecules.


Subject(s)
B-Lymphocytes/immunology , Clathrin Light Chains/genetics , Endocytosis/immunology , Gene Deletion , Immunoglobulin Class Switching , Animals , B-Lymphocytes/pathology , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Clathrin Light Chains/immunology , Gene Expression Regulation , Humans , Immunoglobulin A/biosynthesis , Immunoglobulin A/genetics , Liver/cytology , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , Myocardium/immunology , Organ Specificity , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/immunology , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/immunology , Receptors, Transforming Growth Factor beta/agonists , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
20.
Sci Signal ; 9(418): ra27, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26956486

ABSTRACT

Transforming growth factor-ß (TGF-ß) is an immunosuppressive cytokine that inhibits the proinflammatory functions of T cells, and it is a major factor in abrogating T cell activity against tumors. Canonical TGF-ß signaling results in the activation of Smad proteins, which are transcription factors that regulate target gene expression. We found that the cell surface molecule platelet endothelial cell adhesion molecule-1 (PECAM-1) facilitated noncanonical (Smad-independent) TGF-ß signaling in T cells. Subcutaneously injected tumor cells that are dependent on TGF-ß-mediated suppression of immunity for growth grew more slowly in PECAM-1(-/-) mice than in their wild-type counterparts. T cells isolated from PECAM-1(-/-) mice demonstrated relative insensitivity to the TGF-ß-dependent inhibition of interferon-γ (IFN-γ) production, granzyme B synthesis, and cellular proliferation. Similarly, human T cells lacking PECAM-1 demonstrated decreased sensitivity to TGF-ß in a manner that was partially restored by reexpression of PECAM-1. Co-incubation of T cells with TGF-ß and a T cell-activating antibody resulted in PECAM-1 phosphorylation on an immunoreceptor tyrosine-based inhibitory motif (ITIM) and the recruitment of the inhibitory Src homology 2 (SH2) domain-containing tyrosine phosphatase-2 (SHP-2). Such conditions also induced the colocalization of PECAM-1 with the TGF-ß receptor complex as identified by coimmunoprecipitation, confocal microscopy, and proximity ligation assays. These studies indicate a role for PECAM-1 in enhancing the inhibitory functions of TGF-ß in T cells and suggest that therapeutic targeting of the PECAM-1-TGF-ß inhibitory axis represents a means to overcome TGF-ß-dependent immunosuppression within the tumor microenvironment.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Amino Acid Motifs , Animals , Granzymes/genetics , Granzymes/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Smad Proteins/genetics , Smad Proteins/immunology , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL