Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960622

ABSTRACT

A pleiotropic immunoregulatory cytokine, TGF-ß, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.


Subject(s)
Cell Differentiation , Dendritic Cells , Interferon Regulatory Factors , STAT3 Transcription Factor , Smad3 Protein , Animals , Cell Differentiation/genetics , Dendritic Cells/metabolism , Dendritic Cells/cytology , Smad3 Protein/metabolism , Smad3 Protein/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Mice , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Mice, Knockout , Mice, Inbred C57BL , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Phosphorylation , Signal Transduction
2.
Biochem Biophys Res Commun ; 484(2): 269-277, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28115165

ABSTRACT

Cancer-associated inflammation develops resistance to the epidermal growth-factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in non-small cell lung cancers (NSCLCs) harboring oncogenic EGFR mutations. Stat3-mediated interleukin (IL)-6 signaling and Smad-mediated transforming growth factor-ß (TGF-ß) signaling pathways play crucial regulatory roles in cancer-associated inflammation. However, mechanisms how these pathways regulate sensitivity and resistance to EGFR-TKI in NSCLCs remain largely undetermined. Here we show that signal transducer and activator of transcription (Stat)3 represses Smad3 in synergy with the potent negative regulators of TGF-ß signaling, c-Ski and SnoN, whereby renders gefitinib-sensitive HCC827 cells resistant. We found that IL-6 signaling via phosphorylated Stat3 induced gefitinib resistance as repressing transcription of Smad3, whereas TGF-ß enhanced gefitinib sensitivity as activating transcription of Smad3 in HCC827 cells with gefitinib-sensitizing EGFR mutation. Promoter analyses showed that Stat3 synergized with c-Ski/SnoN to repress Smad2/3/4-induced transcription of the Smad3 gene. Smad3 was found to be an apoptosis inducer, which upregulated pro-apoptotic genes such as caspase-3 and downregulated anti-apoptotic genes such as Bcl-2. Our results suggest that derepression of Smad3 can be a therapeutic strategy to prevent gefitinib-resistance in NSCLCs with gefitinib-sensitizing EGFR mutation.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Intracellular Signaling Peptides and Proteins/physiology , Lung Neoplasms/metabolism , Proto-Oncogene Proteins/physiology , Quinazolines/pharmacology , STAT3 Transcription Factor/physiology , Smad3 Protein/antagonists & inhibitors , Adenocarcinoma/pathology , Apoptosis/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Gefitinib , Humans , Interleukin-6/metabolism , Lung Neoplasms/pathology , Mutation , Signal Transduction
3.
Nat Commun ; 6: 7600, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26194464

ABSTRACT

Transforming growth factor-ß (TGF-ß) and interleukin-6 (IL-6) are the pivotal cytokines to induce IL-17-producing CD4(+) T helper cells (TH17); yet their signalling network remains largely unknown. Here we show that the highly homologous TGF-ß receptor-regulated Smads (R-Smads): Smad2 and Smad3 oppositely modify STAT3-induced transcription of IL-17A and retinoic acid receptor-related orphan nuclear receptor, RORγt encoded by Rorc, by acting as a co-activator and co-repressor of STAT3, respectively. Smad2 linker phosphorylated by extracellular signal-regulated kinase (ERK) at the serine 255 residue interacts with STAT3 and p300 to transactivate, whereas carboxy-terminal unphosphorylated Smad3 interacts with STAT3 and protein inhibitor of activated STAT3 (PIAS3) to repress the Rorc and Il17a genes. Our work uncovers carboxy-terminal phosphorylation-independent noncanonical R-Smad-STAT3 signalling network in TH17 differentiation.


Subject(s)
Cell Differentiation , STAT3 Transcription Factor/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Th17 Cells , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-17/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation
4.
EMBO Mol Med ; 7(5): 577-92, 2015 May.
Article in English | MEDLINE | ID: mdl-25766838

ABSTRACT

We have previously reported that Smad6, one of the inhibitory Smads of transforming growth factor-ß (TGF-ß)/bone morphogenetic protein (BMP) signaling, inhibits Toll-like receptor (TLR) 4 signaling by disrupting the Pellino-1-mediated TLR4 signaling complex. Here, we developed Smaducin-6, a novel membrane-tethered palmitic acid-conjugated Smad6-derived peptide composed of amino acids 422-441 of Smad6. Smaducin-6 interacted with Pellino-1, located in the inner membrane, thereby disrupting the formation of IRAK1-, RIP1-, IKKε-mediated TLR4 signaling complexes. Systemic administration of Smaducin-6 showed a significant therapeutic effect on mouse TLR4-mediated inflammatory disease models, cecal-ligation-puncture (CLP)-induced sepsis, and lipopolysaccharide-induced endotoxemia, by inhibiting pro-inflammatory cytokine production and apoptosis while enhancing neutrophil migration and bacterial clearance. Our findings provide clues to develop new peptide-based drugs to target Pellino-1 protein in TLR4 signaling pathway for the treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Endotoxemia/drug therapy , Immunosuppressive Agents/administration & dosage , Oligopeptides/administration & dosage , Sepsis/drug therapy , Smad6 Protein/administration & dosage , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/metabolism , Disease Models, Animal , Immunosuppressive Agents/metabolism , Mice , Nuclear Proteins/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Binding , Signal Transduction , Smad6 Protein/genetics , Smad6 Protein/metabolism , Treatment Outcome , Ubiquitin-Protein Ligases/metabolism
6.
EMBO Mol Med ; 5(11): 1720-39, 2013 11.
Article in English | MEDLINE | ID: mdl-24127404

ABSTRACT

Varieties of transforming growth factor-ß (TGF-ß) antagonists have been developed to intervene with excessive TGF-ß signalling activity in cancer. Activin receptor-like kinase5 (ALK5) inhibitors antagonize TGF-ß signalling by blocking TGF-ß receptor-activated Smad (R-Smad) phosphorylation. Here we report the novel mechanisms how ALK5 inhibitors exert a therapeutic effect on a mouse B16 melanoma model. Oral treatment with a novel ALK5 inhibitor, EW-7197 (2.5 mg/kg daily) or a representative ALK5 inhibitor, LY-2157299 (75 mg/kg bid) suppressed the progression of melanoma with enhanced cytotoxic T-lymphocyte (CTL) responses. Notably, ALK5 inhibitors not only blocked R-Smad phosphorylation, but also induced ubiquitin-mediated degradation of the common Smad, Smad4 mainly in CD8(+) T cells in melanoma-bearing mice. Accordingly, T-cell-specific deletion of Smad4 was sufficient to suppress the progression of melanoma. We further identified eomesodermin (Eomes), the T-box transcription factor regulating CTL functions, as a specific target repressed by TGF-ß via Smad4 and Smad3 in CD8(+) T cells. Thus, ALK5 inhibition enhances anti-melanoma CTL responses through ubiquitin-mediated degradation of Smad4 in addition to the direct inhibitory effect on R-Smad phosphorylation.


Subject(s)
Melanoma/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Smad4 Protein/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Ubiquitin/metabolism , Animals , Female , Humans , Male , Melanoma/genetics , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proteolysis , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad4 Protein/genetics , T-Box Domain Proteins/genetics , Ubiquitination , Up-Regulation
7.
Mol Cells ; 36(5): 432-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24158612

ABSTRACT

Tumor-associated macrophages (TAMs) accumulate in various cancers and promote tumor angiogenesis and metastasis, and thus may be ideal targets for the clinical diagnosis of tumor metastasis with high specificity. However, there are few specific markers to distinguish between TAMs and normal or inflammatory macrophages. Here, we show that TAMs localize in green fluorescent protein-labeled tumors of metastatic lymph nodes (MLNs) from B16F1 melanoma cells but not in necrotic tumor regions, suggesting that TAMs may promote the growth of tumor cells and the progression of tumor metastasis. Furthermore, we isolated pure populations of TAMs from MLNs and characterized their gene expression signatures compared to peritoneal macrophages (PMs), and found that TAMs significantly overexpress immunosuppressive cytokines such as IL-4, IL-10, and TGF-ß as well as proangiogenic factors such as VEGF, TIE2, and CD31. Notably, immunological analysis revealed that TIE2(+)/CD31(+) macrophages constitute the predominant population of TAMs that infiltrate MLNs, distinct from tissue or inflammatory macrophages. Importantly, these TIE2(+)/CD31(+) macrophages also heavily infiltrated MLNs from human breast cancer biopsies but not reactive hyperplastic LNs. Thus, TIE2(+)/ CD31(+) macrophages may be a unique histopathological biomarker for detecting metastasis in clinical diagnosis, and a novel and promising target for TAM-specific cancer therapy.


Subject(s)
Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Macrophages/physiology , Melanoma, Experimental/secondary , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Receptor, TIE-2/analysis , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/immunology , Lymphatic Metastasis/immunology , Macrophages/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Receptor, TIE-2/immunology , Tumor Microenvironment
8.
Yonsei Med J ; 54(2): 352-7, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23364967

ABSTRACT

PURPOSE: Patients with gestational diabetes mellitus (GDM) have been reported to exhibit the same genetic susceptibility as that observed in those with type 2 diabetes mellitus (T2DM). Recent polymorphism studies have shown that several genes are related to T2DM and GDM. The aim of this study was to examine whether certain candidate genes, previously shown to be associated with T2DM, also offer a specific genetic predisposition to GDM. MATERIALS AND METHODS: The current study was conducted in 136 Korean pregnant women, who gave birth at Gil Hospital, from October 2008 to May 2011. These study subjects included 95 subjects with GDM and 41 non-diabetic controls. We selected the specific genes of PPARγ², IGF2BP2, and KCNQ1 for study and amplified them using the polymerase chain reaction. This was followed by genotyping for single nucleotide polymorphisms. We then compared the genotype frequencies between patients with GDM and non-diabetic controls using the χ² test. We obtained and analyzed clinical information using Student's t-test, and statistical analyses were conducted using logistic regression with SPSS Statistics software, version 19.0. RESULTS: Significant differences were observed in maternal age, body mass index, weight gain and weight at time of delivery between the groups compared. Among pregnant women, polymorphisms in PPARγ² and IGF2BP2 were shown to be highly correlated with GDM occurrence, whereas no correlation was found for KCNQ1 polymorphisms. CONCLUSION: Our results indicated that genetic polymorphisms could also be of value in predicting the occurrence and diagnosis of GDM.


Subject(s)
Diabetes, Gestational/genetics , KCNQ1 Potassium Channel/genetics , PPAR gamma/genetics , RNA-Binding Proteins/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Logistic Models , Polymorphism, Single Nucleotide , Pregnancy , Republic of Korea
9.
Biochem Biophys Res Commun ; 423(4): 750-6, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22705548

ABSTRACT

Bone morphogenetic proteins (BMPs) that belong to the transforming growth factor-ß (TGF-ß) superfamily cytokines, play crucial roles in hematopoiesis. However, roles of Smad6 in hematopoiesis remained unknown in contrast to the other inhibitory Smad (I-Smad), Smad7. Here we show that Smad6 inhibits erythropoiesis in human CD34(+) cord blood hematopoietic stem cells (HSCs). Smad6 was specifically expressed in CD34(+) cord blood HSCs, which was correlated with the expression of BMP2/4/6/7 and BMP type I receptor (BMPRI). BMP-specific receptor-regulated Smads (R-Smads), Smad1 and Smad5 in cooperation with Smad4 induced transcription of the Smad6 gene. Instead of affecting cell cycle, apoptosis, self-renewal, and stemness of CD34(+) cells, Smad6 knockdown enhanced, whereas Smad6 overexpression suppressed erythropoiesis in stem cell culture and colony formation assay. Consistently, Smad6 suppressed the expression of the genes essential for erythropoiesis, such as Kruppel-like factor 1 (erythroid) (KLF1/EKLF) and GATA binding protein 2 (GATA-2). Promoter analyses showed that Smad6 repressed Smad5/4-induced transcription of the Klf1 gene. Thus, our data suggest that Smad6 indirectly maintains stemness by preventing spontaneous erythropoiesis in HSCs.


Subject(s)
Erythropoiesis/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Smad6 Protein/metabolism , Antigens, CD34/analysis , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Cells, Cultured , Fetal Blood/cytology , GATA2 Transcription Factor/genetics , Gene Knockdown Techniques , Hematopoietic Stem Cells/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Promoter Regions, Genetic , Smad6 Protein/genetics , Transcription, Genetic
10.
J Sex Med ; 6(5): 1284-96, 2009 May.
Article in English | MEDLINE | ID: mdl-19473283

ABSTRACT

INTRODUCTION: Transforming growth factor-beta1 (TGF-beta1) has been known to play a crucial role in the pathogenesis of Peyronie's disease (PD). AIM: The aim of this paper was to investigate the therapeutic effect of IN-1130, a novel small molecule inhibitor of activin receptor-like kinase (ALK)5, a type I receptor of TGF-beta, in an animal model of PD. METHODS: PD was induced in rats through repeated injections of adenovirus expressing TGF-beta1 (days 0, 3, and 6; 1 x 10(10) particles/0.1 mL, respectively) into the tunica albuginea. The rats were divided into five groups (N = 10 per group): group 1, age-matched controls without treatment; group 2, age-matched controls receiving repeated injections of IN-1130 (days 30 and 37; 5 mg/kg in 0.1 mL saline, respectively); group 3, PD rats without treatment; group 4, PD rats receiving repeated injections of saline (days 30 and 37; 0.1 mL, respectively); group 5, PD rats receiving repeated injections of IN-1130 (days 30 and 37; 5 mg/kg in 0.1 mL saline, respectively) into the lesion. MAIN OUTCOME MEASURES: Penile curvature was evaluated by use of an artificial erection test at day 45, and the penis was then harvested for histologic examination. Collagen in the plaque was quantitatively assessed by hydroxyproline determination. RESULTS: IN-1130 induced significant regression of fibrotic plaque through reduced infiltration of inflammatory cells, reduced transnuclear expression of phospho-Smad2/phospho-Smad3, reduced hydroxyproline content, and reduced cartilage content and restoration of elastin fibers in the fibrotic plaque of PD rats, which was accompanied by the correction of penile curvature. CONCLUSION: Antagonizing TGF-beta signaling through the use of ALK5 inhibitors may represent an exciting new therapeutic strategy for the future treatment of PD.


Subject(s)
Imidazoles/administration & dosage , Penile Induration/drug therapy , Penis/drug effects , Protein Kinase Inhibitors/administration & dosage , Quinoxalines/administration & dosage , Animals , Disease Models, Animal , Fibrosis/drug therapy , Humans , Male , Penile Induration/pathology , Rats
11.
Immune Netw ; 9(4): 122-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-20157598

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a highly pleiotropic cytokine playing pivotal roles in immune regulation. TGF-beta facilitates tumor cell survival and metastasis by targeting multiple cellular components. Focusing on its immunosuppressive functions, TGF-beta antagonists have been employed for cancer treatment to enhance tumor immunity. TGF-beta antagonists exert anti-tumor effects through #1 activating effector cells such as NK cells and cytotoxic CD8(+) T cells (CTLs), #2 inhibiting regulatory/suppressor cell populations, #3 making tumor cells visible to immune cells, #4 inhibiting the production of tumor growth factors. This review focuses on the effect of TGF-beta on T cells, which are differentiated into effector T cells or newly identified tumor-supporting T cells.

12.
J Sex Med ; 5(10): 2318-29, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18778311

ABSTRACT

INTRODUCTION: It has been suggested that transforming growth factor-beta1 (TGF-beta1) plays an important role in the pathogenesis of diabetes-induced erectile dysfunction. AIM: To investigate the expression and activity of Smad transcriptional factors, the key molecules for the initiation of TGF-beta-mediated fibrosis, in the penis of streptozotocin (STZ)-induced diabetic rats. METHODS: Fifty-two 8-week-old Sprague-Dawley rats were used and divided into control and diabetic groups. Diabetes was induced by an intravenous injection of STZ. MAIN OUTCOME MEASURES: Eight weeks later, erectile function was measured by electrical stimulation of the cavernous nerve (N = 12 per group). The penis was harvested and stained with Masson trichrome or antibody to TGF-beta1, phospho-Smad2 (P-Smad2), smooth muscle alpha-actin, and factor VIII (N = 12 per group). Penis specimens from a separate group of animals were used for TGF-beta1 enzyme-linked immunosorbent assay (ELISA), P-Smad2/Smad2, phospho-Smad3 (P-Smad3)/Smad3, fibronectin, collagen I, and collagen IV western blot, or hydroxyproline determination. RESULTS: Erectile function was significantly reduced in diabetic rats compared with that in controls. The expression of TGF-beta1, P-Smad2, and P-Smad3 protein evaluated by ELISA or western blot was higher in diabetic rats than in controls. Compared with that in control rats, P-Smad2 expression was higher mainly in smooth muscle cells and fibroblasts of diabetic rats, whereas no significant differences were noted in endothelial cells or in the dorsal nerve bundle. Cavernous smooth muscle and endothelial cell contents were lower in diabetic rats than in controls. Cavernous fibronectin, collagen IV, and hydroxyproline content was significantly higher in diabetic rats than in controls. CONCLUSION: Upregulation of TGF-beta1 and activation of the Smad signaling pathway in the penis of diabetic rats might play important roles in diabetes-induced structural changes and deterioration of erectile function.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Erectile Dysfunction/metabolism , Penis , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Endothelium, Vascular/metabolism , Erectile Dysfunction/blood , Fibrosis/metabolism , Male , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/blood
13.
Cancer Res ; 68(10): 3835-43, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483268

ABSTRACT

Overexpression of transforming growth factor beta (TGF-beta) is frequently associated with metastasis and poor prognosis, and TGF-beta antagonism has been shown to prevent metastasis in preclinical models with surprisingly little toxicity. Here, we have used the transplantable 4T1 model of metastatic breast cancer to address underlying mechanisms. We showed that efficacy of the anti-TGF-beta antibody 1D11 in suppressing metastasis was dependent on a synergistic combination of effects on both the tumor parenchyma and microenvironment. The main outcome was a highly significant enhancement of the CD8+ T-cell-mediated antitumor immune response, but effects on the innate immune response and on angiogenesis also contributed to efficacy. Treatment with 1D11 increased infiltration of natural killer cells and T cells at the metastatic site, and enhanced expression of coactivators (NKG2D) and cytotoxic effectors (perforin and granzyme B) on CD8+ T cells. On the tumor cells, increased expression of an NKG2D ligand (Rae1gamma) and of a death receptor (TNFRSF1A) contributed to enhanced immune cell-mediated recognition and lysis. The data suggest that elevated TGF-beta expression in the tumor microenvironment modulates a complex web of intercellular interactions that aggregately promote metastasis and progression. TGF-beta antibodies reverse this effect, and the absence of a major effect of TGF-beta antagonism on any one cell compartment may be critical for a good therapeutic window and the avoidance of autoimmune complications.


Subject(s)
Immunotherapy/methods , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/therapy , Transforming Growth Factor beta/chemistry , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Culture Media, Conditioned/pharmacology , Killer Cells, Natural/metabolism , Ligands , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Prognosis , Transfection
14.
Cancer Res ; 68(10): 3915-23, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483277

ABSTRACT

Overexpression of the immunosuppressive cytokine transforming growth factor beta (TGF-beta) is one strategy that tumors have developed to evade effective immunesurveillance. Using transplantable models of breast and colon cancer, we made the unexpected finding that CD8+ cells in tumor-bearing animals can directly promote tumorigenesis, by a mechanism that is dependent on TGF-beta. We showed that CD8+ splenocytes from tumor-bearing mice expressed elevated interleukin (IL)-17 when compared with naive mice, and that CD8+ T cells could be induced to make IL-17 on addition of TGF-beta and IL-6 in vitro. Treatment of mice with anti-TGF-beta antibodies in vivo reduced IL-17 expression both in the tumor and the locoregional lymph nodes. Although IL-17 has not previously been shown to act as a survival factor for epithelial cells, we found that IL-17 suppressed apoptosis of several tumor cell lines in vitro, suggesting that this altered T-cell polarization has the potential to promote tumorigenesis directly, rather than indirectly through inflammatory sequelae. Consistent with this hypothesis, knockdown of the IL-17 receptor in 4T1 mouse mammary cancer cells enhanced apoptosis and decreased tumor growth in vivo. Thus, in addition to suppressing immune surveillance, tumor-induced TGF-beta may actively subvert the CD8+ arm of the immune system into directly promoting tumor growth by an IL-17-dependent mechanism.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-17/physiology , Neoplasms/immunology , Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Humans , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Rats
15.
Am J Pathol ; 173(1): 68-76, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18502822

ABSTRACT

Our previous work showed that 6 weeks after cutaneous irradiation, mice null (knockout, KO) for Smad3, a cytoplasmic downstream mediator of transforming growth factor-beta, demonstrate less epidermal acanthosis and dermal inflammation than wild-type (WT) Smad3 mice. Analysis of the kinetics of inflammation showed that 6 to 8 hours after skin irradiation, there was a transient sevenfold increase in neutrophil influx in Smad3 KO mice compared with WT. Herein we describe bone marrow transplantation and skin grafting between WT and KO mice to assess the contribution of the neutrophil genotype compared with that of irradiated skin to the induction of neutrophil migration after irradiation. Results from bone marrow transplantation showed that WT marrow transplanted into KO mice enhanced neutrophil migration 6 to 8 hours after irradiation by 3.2-fold compared with KO marrow in WT mice. KO skin grafted onto either WT or KO animals showed a sixfold elevation of neutrophils after irradiation compared with grafted WT skin. These results suggest that the genotype of the irradiated skin, rather than the inflammatory cell, controls neutrophil influx. Circulating neutrophils, increased in WT mice after injection of granulocyte colony-stimulating factor, resulted in increased neutrophil migration to the skin 6 to 8 hours after irradiation and less skin damage 6 weeks after irradiation compared with untreated WT mice. Thus, early responses, including enhanced neutrophil influx, appear to contribute to subsequent cutaneous radioprotection.


Subject(s)
Neutrophil Infiltration/radiation effects , Skin/radiation effects , Smad3 Protein/deficiency , Smad3 Protein/genetics , Animals , Bone Marrow Transplantation , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Genotype , Mice , Mice, Knockout , Neutrophil Infiltration/immunology , Skin/immunology , Skin Transplantation
17.
Arthritis Rheum ; 58(3): 754-63, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18311788

ABSTRACT

OBJECTIVE: To clarify the glucose-6-phosphate isomerase (GPI)-specific CD4+ T cell lineage involved in GPI-induced arthritis and to investigate their pathologic and regulatory roles in the induction of the disease. METHODS: DBA/1 mice were immunized with GPI to induce arthritis. CD4+ T cells and antigen-presenting cells were cocultured with GPI, and cytokines in the supernatant were analyzed by enzyme-linked immunosorbent assay. Anti-interferon-gamma (anti-IFNgamma) monoclonal antibody (mAb), anti-interleukin-17 (anti-IL-17) mAb, or the murine IL-6 receptor (IL-6R) mAb MR16-1 was injected at different time points, and arthritis development was monitored visually. After MR16-1 was injected, percentages of Th1, Th2, Th17, and Treg cells were analyzed by flow cytometry, and CD4+ T cell proliferation was analyzed using carboxyfluorescein diacetate succinimidyl ester. RESULTS: GPI-specific CD4+ T cells were found to be differentiated to Th1 and Th17 cells, but not Th2 cells. Administration of anti-IL-17 mAb on day 7 significantly ameliorated arthritis (P < 0.01), whereas administration of anti-IFNgamma mAb exacerbated arthritis. Neither anti-IL-17 mAb nor anti-IFNgamma mAb administration on day 14 ameliorated arthritis. Administration of MR16-1 on day 0 or day 3 protected against arthritis induction, and MR16-1 administration on day 8 significantly ameliorated existing arthritis (P < 0.05). After administration of MR16-1, there was marked suppression of Th17 differentiation, without an increase in Th1, Th2, or Treg cells, and CD4+ T cell proliferation was also suppressed. CONCLUSION: IL-6 and Th17 play an essential role in GPI-induced arthritis. Since it has previously been shown that treatment with a humanized anti-IL-6R mAb has excellent effects in patients with rheumatoid arthritis (RA), we propose that the IL-6/IL-17 axis might also be involved in the generation of RA, especially in the early effector phase.


Subject(s)
Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/metabolism , Glucose-6-Phosphate Isomerase/adverse effects , Interleukin-17/metabolism , Interleukin-6/metabolism , Animals , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/pharmacology , Arthritis, Rheumatoid/pathology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-6/immunology , Male , Mice , Mice, Inbred DBA , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/drug effects , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/drug effects , Th2 Cells/metabolism , Th2 Cells/pathology
18.
Mod Rheumatol ; 18(1): 15-22, 2008.
Article in English | MEDLINE | ID: mdl-18092129

ABSTRACT

Interleukin-17 is a proinflammatory cytokine. Recent animal studies have shown that IL-17 plays a role in the initiation and progression of arthritis. However, whether IL-17 has a prominent role in human rheumatoid arthritis (RA) or not remains unclear. Here we investigated the role of IL-17 in patients with RA. cDNA was prepared from knee joint synovial tissues of RA (n = 11) and osteoarthritic (OA, n = 10) patients and PBMC of RA (n = 52) and healthy subjects (n = 34). IL-17 gene expression level was measured by real-time PCR, and was compared with various clinical parameters. IL-17 gene expression in synovial tissues of RA was similar to that in OA. IL-17 gene expression level in PBMC of RA patients was significantly higher than in the control. The response (changes in DAS) to two-week treatment with anti-TNF-alpha blockers (infliximab or etanercept) did not correlate with changes in IL-17 gene expression levels. The IL-17/TNF-alpha gene expression ratio at baseline (before treatment) tended to be lower in responders to the treatment. Expression of IL-17 gene in PBMC may be associated with the inflammatory process of RA. IL-17/TNF-alpha expression ratio is a potentially suitable marker of response to anti-TNF-alpha therapy.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Interleukin-17/metabolism , Knee Joint/immunology , Synovial Membrane/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacology , Arthritis, Rheumatoid/genetics , Biomarkers , Case-Control Studies , Etanercept , Female , Gene Expression , Humans , Immunoglobulin G/pharmacology , Immunologic Factors/pharmacology , Infliximab , Interleukin-17/blood , Interleukin-17/genetics , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Osteoarthritis, Knee/immunology , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/drug effects
19.
Cancer Res ; 67(18): 8643-52, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875704

ABSTRACT

The transforming growth factor-beta (TGF-beta) pathway has tumor-suppressor activity in many epithelial tissues. Because TGF-beta is a potent inhibitor of epithelial cell proliferation, it has been widely assumed that this property underlies the tumor-suppressor effect. Here, we have used a xenograft model of breast cancer to show that endogenous TGF-beta has the potential to suppress tumorigenesis through a novel mechanism, involving effects at two distinct levels in the hierarchy of cellular progeny that make up the epithelial component of the tumor. First, TGF-beta reduces the size of the putative cancer stem or early progenitor cell population, and second it promotes differentiation of a more committed, but highly proliferative, progenitor cell population to an intrinsically less proliferative state. We further show that reduced expression of the type II TGF-beta receptor correlates with loss of luminal differentiation in a clinical breast cancer cohort, suggesting that this mechanism may be clinically relevant. At a molecular level, the induction of differentiation by TGF-beta involves down-regulation of Id1, and forced overexpression of Id1 can promote tumorigenesis despite persistence of the antiproliferative effect of TGF-beta. These data suggest new roles for the TGF-beta pathway in regulating tumor cell dynamics that are independent of direct effects on proliferation.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Transforming Growth Factor beta/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Profiling , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/deficiency , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/deficiency , Transforming Growth Factor beta/deficiency , Transplantation, Heterologous
20.
Intern Med ; 46(13): 1033-7, 2007.
Article in English | MEDLINE | ID: mdl-17603247

ABSTRACT

A 46-year-old woman with systemic lupus erythematosus was hospitalized for purpura, hematochezia and hematuria. One week after admission, she developed grand mal seizures and coma and was diagnosed with thrombotic thrombocytopenic purpura (TTP) when fragmented red cells were found on the peripheral blood smear. Laboratory findings showed severe ADAMTS13 (a disintegrin-like and metalloprotease with thrombospondin type 1 repeats) deficiency and anti-ADAMTS13 antibodies, which in recent reports have indicated a poor prognosis. She was refractory to methylprednisolone pulse therapy and plasma exchange, but administration of cyclosporine induced remission without adverse effects. We propose that cyclosporine may be an effective treatment for cases of refractory TTP.


Subject(s)
ADAM Proteins/immunology , Antibody Formation/immunology , Cyclosporine/therapeutic use , Lupus Erythematosus, Systemic/diagnosis , Methylprednisolone/therapeutic use , Purpura, Thrombotic Thrombocytopenic/drug therapy , Blood Chemical Analysis , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Middle Aged , Purpura, Thrombotic Thrombocytopenic/complications , Purpura, Thrombotic Thrombocytopenic/diagnosis , Recurrence , Risk Assessment , Severity of Illness Index , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...