Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
2.
Diabetes ; 63(11): 3960-73, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24760139

ABSTRACT

Mechanisms associated with type 1 diabetes (T1D) development remain incompletely defined. Using a sensitive array-based bioassay where patient plasma is used to induce transcriptional responses in healthy leukocytes, we previously reported disease-specific, partially interleukin (IL)-1-dependent signatures associated with preonset and recent onset (RO) T1D relative to unrelated healthy control subjects (uHC). To better understand inherited susceptibility in T1D families, we conducted cross-sectional and longitudinal analyses of healthy autoantibody-negative (AA(-)) high HLA-risk siblings (HRS) (DR3 and/or DR4) and AA(-) low HLA-risk siblings (LRS) (non-DR3/non-DR4). Signatures, scored with a novel ontology-based algorithm, and confirmatory studies differentiated the RO T1D, uHC, HRS, and LRS plasma milieus. Relative to uHC, T1D family members exhibited an elevated inflammatory state, consistent with innate receptor ligation that was independent of HLA, AA, or disease status and included elevated plasma IL-1α, IL-12p40, CCL2, CCL3, and CCL4 levels. Longitudinally, signatures of T1D progressors exhibited increasing inflammatory bias. Conversely, HRS possessing decreasing AA titers revealed emergence of an IL-10/transforming growth factor-ß-mediated regulatory state that paralleled temporal increases in peripheral activated CD4(+)/CD45RA(-)/FoxP3(high) regulatory T-cell frequencies. In AA(-) HRS, the familial innate inflammatory state also was temporally supplanted by immunoregulatory processes, suggesting a mechanism underlying the decline in T1D susceptibility with age.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Chemokine CCL2/blood , Chemokine CCL3/blood , Chemokine CCL4/blood , Child , Cross-Sectional Studies , Female , Humans , Interleukin-1/blood , Interleukin-10/blood , Interleukin-12 Subunit p40/blood , Longitudinal Studies , Male , T-Lymphocytes, Regulatory/metabolism , Young Adult
3.
Biochim Biophys Acta ; 1821(12): 1501-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960380

ABSTRACT

Microparticles (MPs) are membrane-bound vesicles shed normally or as a result of various (pathological) stimuli. MPs contain a wealth of bio-active macromolecules. Aminophospholipid phosphatidylserine (PS) is present on the surface of many MPs. As PS and phosphatidylethanolamine (PE) are related, yet distinct aminophospholipids, the purpose of this study was to systematically and directly assess PE exposure on MPs. We examined MPs from various human cellular sources (human breast cancer, endothelial, red and white blood cells) by flow cytometry using a PE-specific probe, duramycin, and two PS-specific probes, annexin V and lactadherin. PS and PE exposure percentage was comparable on vascular and blood cell-derived MPs (80-90% of MP-gated events). However, the percentage of malignant breast cancer MPs exposing PE (~90%) was significantly higher than PS (~50%). Thus, while PS and PE exposure can result from a general loss of membrane asymmetry, there may also be distinct mechanisms of PE and PS exposure on MPs that vary by cellular source.


Subject(s)
Cell-Derived Microparticles/metabolism , Endothelial Cells/metabolism , Erythrocytes/metabolism , Phosphatidylethanolamines/metabolism , Bacteriocins/chemistry , Bacteriocins/metabolism , Bacteriocins/pharmacology , Biotin/chemistry , Blood Coagulation/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell-Derived Microparticles/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Microscopy, Confocal , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Phosphatidylethanolamines/chemistry
4.
J Vis Exp ; (53)2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21808226

ABSTRACT

Regulatory T cells (Tregs) are critical mediators of immune tolerance to self-antigens. In addition, they are crucial regulators of the immune response following an infection. Despite efforts to identify unique surface marker on Tregs, the only unique feature is their ability to suppress the proliferation and function of effector T cells. While it is clear that only in vitro assays can be used in assessing human Treg function, this becomes problematic when assessing the results from cross-sectional studies where healthy cells and cells isolated from subjects with autoimmune diseases (like Type 1 Diabetes-T1D) need to be compared. There is a great variability among laboratories in the number and type of responder T cells, nature and strength of stimulation, Treg:responder ratios and the number and type of antigen-presenting cells (APC) used in human in vitro suppression assays. This variability makes comparison between studies measuring Treg function difficult. The Treg field needs a standardized suppression assay that will work well with both healthy subjects and those with autoimmune diseases. We have developed an in vitro suppression assay that shows very little intra-assay variability in the stimulation of T cells isolated from healthy volunteers compared to subjects with underlying autoimmune destruction of pancreatic ß-cells. The main goal of this piece is to describe an in vitro human suppression assay that allows comparison between different subject groups. Additionally, this assay has the potential to delineate a small loss in nTreg function and anticipate further loss in the future, thus identifying subjects who could benefit from preventive immunomodulatory therapy. Below, we provide thorough description of the steps involved in this procedure. We hope to contribute to the standardization of the in vitro suppression assay used to measure Treg function. In addition, we offer this assay as a tool to recognize an early state of immune imbalance and a potential functional biomarker for T1D.


Subject(s)
Immune System Diseases/diagnosis , Immune System Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Flow Cytometry/methods , Humans , T-Lymphocytes, Regulatory/cytology
5.
Prostaglandins Other Lipid Mediat ; 94(1-2): 34-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21167293

ABSTRACT

Endocannabinoids (ECs), anandamide (AEA) and 2-arachidonoylglycerol (2-AG), inhibit proliferation of carcinoma cells. Several enzymes hydrolyze ECs to reduce endogenous EC concentrations and produce eicosanoids that promote cell growth. In this study, we determined the effects of EC hydrolysis inhibitors and a putative EC, 2-arachidonylglyceryl ether (noladin ether, NE) on proliferation of prostate carcinoma (PC-3, DU-145, and LNCaP) cells. PC-3 cells had the least specific hydrolysis activity for AEA and administration of AEA effectively inhibited cell proliferation. The proliferation inhibition was blocked by SR141716A (a selective CB1R antagonist) but not SR144528 (a selective CB2R antagonist), suggesting a CB1R-mediated inhibition mechanism. On the other hand, specific hydrolysis activity for 2-AG was high and 2-AG inhibited proliferation only in the presence of EC hydrolysis inhibitors. NE inhibited proliferation in a concentration-dependent manner; however, SR141716A, SR144528 and pertussis toxin did not block the NE-inhibited proliferation, suggesting a CBR-independent mechanism of NE. A peroxisome proliferator-activated receptor gamma (PPARγ) antagonist GW9662 did not block the NE-inhibited proliferation, suggesting that PPARγ was not involved. NE also induced cell cycle arrest in G(0)/G(1) phase in PC-3 cells. NE inhibited the nuclear translocation of nuclear factor-kappa B (NF-κB p65) and down-regulated the expression of cyclin D1 and cyclin E in PC-3 cells, suggesting the NF-κB/cyclin D and cyclin E pathways are involved in the arrest of G1 cell cycle and inhibition of cell growth. These results indicate therapeutic potentials of EC hydrolysis inhibitors and the enzymatically stable NE in prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids , Glycerides/pharmacology , Prostatic Neoplasms/metabolism , Anilides/pharmacology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Cyclin E/metabolism , Humans , Male , NF-kappa B/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology
6.
PLoS One ; 5(12): e15154, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21151941

ABSTRACT

BACKGROUND: In type 1 diabetes (T1D), a prototypic autoimmune disease, effector T cells destroy beta cells. Normally, CD4(+)CD25(+high), or natural regulatory T cells (Tregs), counter this assault. In autoimmunity, the failure to suppress CD4(+)CD25(low) T cells is important for disease development. However, both Treg dysfunction and hyperactive responder T-cell proliferation contribute to disease. METHODS/PRINCIPAL FINDINGS: We investigated human CD4(+)CD25(low) T cells and compared them to CD4(+)CD25(-) T cells in otherwise equivalent in vitro proliferative conditions. We then asked whether these differences in suppression are exacerbated in T1D. In both single and co-culture with Tregs, the CD4(+)CD25(low) T cells divided more rapidly than CD4(+)CD25(-) T cells, which manifests as increased proliferation/reduced suppression. Time-course experiments showed that this difference could be explained by higher IL-2 production from CD4+CD25(low) compared to CD4+CD25- T cells. There was also a significant increase in CD4+CD25(low) T-cell proliferation compared to CD4+CD25- T cells during suppression assays from RO T1D and at-risk subjects (n = 28, p = 0.015 and p = 0.024 respectively). CONCLUSIONS/SIGNIFICANCE: The in vitro dual suppression assays proposed here could highlight the impaired sensitivity of certain responder T cells to the suppressive effect of Tregs in human autoimmune diseases.


Subject(s)
Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/cytology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Separation , Coculture Techniques , Flow Cytometry , Humans , Insulin-Secreting Cells/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Kinetics , Leukocytes, Mononuclear/cytology , Risk , T-Lymphocytes, Regulatory/cytology
7.
PLoS One ; 5(4): e9893, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405005

ABSTRACT

BACKGROUND: Pituitary corticotroph tumors secrete excess adrenocorticotrophic hormone (ACTH) resulting in Cushing's disease (CD). Standard treatment includes surgery and, if not successful, radiotherapy, both of which have undesirable side effects and frequent recurrence of the tumor. Pharmacotherapy using PPARgamma agonists, dopamine receptor agonists, retinoic acid or somatostatin analogs is still experimental. Curcumin, a commonly used food additive in South Asian cooking, has potent growth inhibitory effects on cell proliferation. Our laboratory recently demonstrated that curcumin inhibited growth and induced apoptosis in prolactin- and growth hormone-producing tumor cells. Subsequently, Schaaf et.al. confirmed our findings and also showed the in vivo effectiveness of curcumin to suppress pituitary tumorigenesis. However the molecular mechanism that mediate this effect of curcumin are still unknown. PRINCIPAL FINDINGS: Using the mouse corticotroph tumor cells, AtT20 cells, we report that curcumin had a robust, irreversible inhibitory effect on cell proliferation and clonogenic property. The curcumin-induced growth inhibition was accompanied by decreased NFkappaB activity. Further, curcumin down-regulated the pro-survival protein Bcl-xL, depolarized the mitochondrial membrane, increased PARP cleavage, which led to apoptotic cell death. Finally, curcumin had a concentration-dependent suppressive effect on ACTH secretion from AtT20 cells. CONCLUSION: The ability of curcumin to inhibit NFkappaB and induce apoptosis in pituitary corticotroph tumor cells leads us to propose developing it as a novel therapeutic agent for the treatment of CD.


Subject(s)
ACTH-Secreting Pituitary Adenoma/drug therapy , Cell Proliferation/drug effects , Curcumin/pharmacology , Pituitary ACTH Hypersecretion/drug therapy , ACTH-Secreting Pituitary Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Animals , Antineoplastic Agents , Apoptosis/drug effects , Cell Line, Tumor , Curcumin/therapeutic use , Dose-Response Relationship, Drug , Mice , NF-kappa B/antagonists & inhibitors
8.
Anticancer Res ; 30(2): 499-504, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20332461

ABSTRACT

Medulloblastoma (MB) is the most common malignant brain tumor in children. Bcl-2 and MMP-9 promote the pathogenesis and progression of MB. The expression of both bcl-2 and MMP-9 is regulated by the transcription factor NF-kappaB. Curcumin, a natural food additive, has a potent anti-proliferative effect, presumably mediated through NF-kappaB suppression. The tumor-suppressing effects of curcumin are well documented, however, its effect on MB is unknown. Our objectives were to: a) examine the effect of curcumin on MB cell proliferation and apoptosis; b) characterize the mechanism that mediates the effect of curcumin; c) examine the effects of curcumin on MB cell migration. We report that curcumin inhibited cell proliferation and blocked clonogenicity of MB cells. Furthermore, curcumin down-regulated bcl-2 and bcl(x)l, leading to caspase-mediated cell death. Finally, curcumin blocked migration of MB cells. Thus, we propose developing curcumin as a novel therapeutic agent for MB.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cerebellar Neoplasms/pathology , Curcumin/pharmacology , Medulloblastoma/pathology , Blotting, Western , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/metabolism , Colony-Forming Units Assay , Humans , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Wound Healing/drug effects , bcl-X Protein/metabolism
9.
Endocrinology ; 149(8): 4158-67, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450960

ABSTRACT

Prolactinomas are the most prevalent functional pituitary adenomas. Dopamine D2 receptor (D2R) agonists, such as bromocriptine are the first line of therapy; however, drug intolerance/resistance to D2R agonists exists. Apart from D2R agonists, there is no established medical therapy for prolactinomas; therefore, identifying novel therapeutics is warranted. Curcumin, a commonly used food additive in South Asian cooking, inhibits proliferation of several tumor cell lines; however, its effect on pituitary tumor cell proliferation has not been determined. Our objectives were to: 1) determine whether curcumin inhibits proliferation of pituitary tumor cell lines; 2) identify the signaling intermediaries that mediate the effect of curcumin; 3) examine whether curcumin inhibited pituitary hormone production and release; and 4) examine whether curcumin could enhance the growth-inhibitory effect of bromocriptine. Using rat lactotroph cell lines, GH3 and MMQ cells, we report that curcumin had a robust dose and time-dependent inhibitory effect on GH3 and MMQ cell proliferation. Inhibitory effects of curcumin persisted, even on removal of curcumin, and curcumin also blocked colony formation ability of pituitary tumor cells. The growth-inhibitory effect of curcumin was accompanied by decreased expression of cyclin D3 and ser 780 phosphorylation of retinoblastoma protein. In addition, curcumin also induced apoptosis in both GH3 and MMQ cells. Furthermore, curcumin suppresses intracellular levels and release of both prolactin and GH. Finally, we show that low concentrations of curcumin enhanced the growth-inhibitory effect of bromocriptine on MMQ cell proliferation. Taken together we demonstrate that curcumin inhibits pituitary tumor cell proliferation, induces apoptosis, and decreases hormone production and release, and thus, we propose developing curcumin as a novel therapeutic tool in the management of prolactinomas.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Pituitary Hormones/metabolism , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Animals , Antineoplastic Agents/pharmacology , Bromocriptine/pharmacology , Clone Cells/drug effects , Cyclin D3 , Cyclins/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , Phosphorylation/drug effects , Pituitary Hormones/blood , Pituitary Neoplasms/blood , Prolactinoma/blood , Rats , Retinoblastoma Protein/metabolism , Time Factors , Tumor Cells, Cultured
10.
J Pharmacol Exp Ther ; 322(3): 1228-36, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17600139

ABSTRACT

Clinical studies have shown gallium nitrate to have significant antitumor activity against non-Hodgkin's lymphoma and bladder cancer, thus indicating that gallium-based drugs have potential for further development as antineoplastic agents. In this study, we compared the cytotoxicity of gallium maltolate, a novel gallium compound, with gallium nitrate in lymphoma cell lines, including p53 variant and unique gallium nitrate-resistant cells. We found that gallium maltolate inhibited cell proliferation and induced apoptosis through the mitochondrial pathway at lower concentrations and more rapidly than gallium nitrate. Gallium maltolate produced an increase in intracellular reactive oxygen species (ROS) within 2 h of incubation with cells; this effect could be blocked by mitoquinone, a mitochondria-targeted antioxidant. The role of the transferrin receptor (TfR) in gallium maltolate's action was examined using monoclonal antibody (MoAb) 42/6 to block TfR function. However, although MoAb 42/6 reduced gallium maltolate-induced caspase-3 activity, it had only a minor effect on cell growth inhibition. Importantly, gallium maltolate induced apoptosis in cells resistant to gallium nitrate, and, unlike gallium nitrate, its cytotoxicity was not affected by cellular p53 status. Cellular gallium uptake was greater with gallium maltolate than with gallium nitrate. We conclude that gallium maltolate inhibits cell proliferation and induces apoptosis more efficiently than gallium nitrate. Gallium maltolate is incorporated into lymphoma cells to a greater extent than gallium nitrate via both TfR-independent and -dependent pathways; it has significant activity against gallium nitrate-resistant cells and acts independently of p53. Further studies to evaluate its antineoplastic activity in vivo are warranted.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Gallium/pharmacology , Lymphoma/drug therapy , Organometallic Compounds/pharmacology , Pyrones/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation , Humans , Lymphoma/pathology , Receptors, Transferrin , Tumor Suppressor Protein p53
11.
PLoS One ; 2(1): e146, 2007 Jan 03.
Article in English | MEDLINE | ID: mdl-17206281

ABSTRACT

BACKGROUND: In experimental models, Type 1 diabetes T1D can be prevented by adoptive transfer of CD4+CD25+ (FoxP3+) suppressor or regulatory T cells. Recent studies have found a suppression defect of CD4+CD25+(high) T cells in human disease. In this study we measure apoptosis of CD4+CD25+(high) T cells to see if it could contribute to reduced suppressive activity of these cells. METHODS AND FINDINGS: T-cell apoptosis was evaluated in children and adolescent 35 females/40 males subjects comprising recent-onset and long-standing T1D subjects and their first-degree relatives, who are at variable risk to develop T1D. YOPRO1/7AAD and intracellular staining of the active form of caspase 3 were used to evaluate apoptosis. Isolated CD4+CD25+(high) and CD4+CD25- T cells were co-cultured in a suppression assay to assess the function of the former cells. We found that recent-onset T1D subjects show increased apoptosis of CD4+CD25+(high) T cells when compared to both control and long-standing T1D subjects p<0.0001 for both groups. Subjects at high risk for developing T1D 2-3Ab+ve show a similar trend p<0.02 and p<0.01, respectively. On the contrary, in long-standing T1D and T2D subjects, CD4+CD25+(high) T cell apoptosis is at the same level as in control subjects p = NS. Simultaneous intracellular staining of the active form of caspase 3 and FoxP3 confirmed recent-onset FoxP3+ve CD4+CD25+(high) T cells committed to apoptosis at a higher percentage 15.3+/-2.2 compared to FoxP3+ve CD4+CD25+(high) T cells in control subjects 6.1+/-1.7 p<0.002. Compared to control subjects, both recent-onset T1D and high at-risk subjects had significantly decreased function of CD4+CD25+(high) T cells p = 0.0007 and p = 0.007, respectively. CONCLUSIONS: There is a higher level of ongoing apoptosis in CD4+CD25+(high) T cells in recent-onset T1D subjects and in subjects at high risk for the disease. This high level of CD4+CD25+(high) T-cell apoptosis could be a contributing factor to markedly decreased suppressive potential of these cells in recent-onset T1D subjects.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Child , Diabetes Mellitus, Type 1/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Risk Factors
12.
J Immunol ; 177(10): 7275-86, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082646

ABSTRACT

Human type 1 diabetes mellitus (T1DM) arises through autoimmune destruction of pancreatic beta cells and is modeled in many respects by the lymphopenic and spontaneously diabetic BioBreeding (BB) DRlyp/lyp rat. Previously, preonset expression profiling of whole DRlyp/lyp pancreatic lymph nodes (PLN) revealed innate immune activity, specifically that of mast cells and eosinophils. Furthermore, we observed that pancreatic islets of DRlyp/lyp rats as well as those of diabetes-inducible BB DR(+/+) rats potentially recruit innate cells through eotaxin expression. Here we determine that lifelong eotaxin expression begins before 40 days of life and is localized specifically to beta cells. In this report, we find that PLN mast cells are more abundant in DRlyp/lyp compared with related BB DR(+/+) rats (2.1 +/- 0.9% vs 0.9 +/- 0.4% of total cells, p < 0.0001). DRlyp/lyp PLN mast cell gene expression profiling revealed an activated population and included significant overrepresentation of transcripts for mast cell protease 1, cationic trypsinogen, carboxypeptidase A, IL-5, and phospholipase Cgamma. In the DR(+/+) rat, which develops T1DM upon depletion of T regulator cells, mast cells displayed gene expression consistent with the negative regulation of degranulation, including significant overrepresentation of transcripts encoding tyrosine phosphatase SHP-1, lipid phosphatase SHIP, and E3 ubiquitin ligase c-Cbl. To recapitulate the negative mast cell regulation observed in the DR(+/+) rats, we treated DRlyp/lyp rats with the mast cell "stabilizer" cromolyn, which significantly (p < 0.05) delayed T1DM onset. These findings are consistent with a growing body of evidence in human and animal models, where a role for mast cells in the initiation and progression of autoimmune disease is emerging.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Genetic Predisposition to Disease , Mast Cells/physiology , Animals , Cell Count , Chemokine CCL11 , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Cromolyn Sodium/administration & dosage , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Female , Gene Expression Profiling , Hypoglycemic Agents/administration & dosage , Immunity, Innate/genetics , Immunoglobulin E/blood , Immunophenotyping , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Lymph Nodes/cytology , Lymph Nodes/enzymology , Lymph Nodes/metabolism , Lymphopenia/enzymology , Lymphopenia/genetics , Lymphopenia/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Rats , Rats, Inbred BB , Rats, Inbred WF , Reverse Transcriptase Polymerase Chain Reaction
13.
J Invest Dermatol ; 125(6): 1130-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354182

ABSTRACT

CD 200 is a widely expressed transmembrane glycoprotein that transmits an inhibitory signal after ligation of the structurally homologous CD 200-receptor-1 (CD 200 R1). Recently, we showed that CD 200 is expressed on keratinocytes and plays a role in protecting hair follicles from autoimmune attack. Here, we report the characterization of cell surface and mRNA expression of CD 200 R1 by cells of the murine epidermis. In addition, we report mRNA expression for other members of the CD 200 R-family (R2-R4) by quantitative real-time RT-PCR. Variable levels of CD 200 R1, R2, R3, and R4 mRNA were detected in bulk epidermal cell suspensions. Freshly isolated Langerhans cells (LC) preferentially expressed CD 200 R1. Consistent with an inhibitory role for CD 200:CD 200 R1 interaction, LC obtained from mice deficient in CD 200 (CD 200(-/-)) were in a heightened state of activation as compared with wild-type (CD 200(+/+)) cells. Freshly isolated dendritic epidermal T cells (DETC) expressed low levels of CD 200 R1, R2, and R3 mRNA, but they preferentially increased cell surface and mRNA expression of CD 200 R1 upon activation in vitro. In functional assays using sub-optimal CD3 signaling, immobilized CD 200 inhibited DETC proliferation and cytokine secretion. Collectively, these results suggest that CD 200:CD 200 R interactions may play a role in regulating both LC and DETC in cutaneous immune reactions.


Subject(s)
Antigens, Surface/genetics , Epidermis/physiology , Membrane Glycoproteins/genetics , Receptors, Cell Surface/genetics , Animals , Cell Line , Dendritic Cells/immunology , Ear , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Orexin Receptors , RNA, Messenger/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction
14.
J Invest Dermatol ; 123(5): 880-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15482475

ABSTRACT

CD200 (OX-2) is a transmembrane glycoprotein that transmits an immunoregulatory signal through the CD200 receptor (CD200R) to attenuate inflammatory reactions and promote immune tolerance. CD200 expression in the skin has not been described previously. We now report that freshly isolated cells of the murine epidermis contain a subpopulation of major histocompatibility complex (MHC) class II-negative, CD3-negative keratinocytes that are CD200-positive. CD200 expression was accentuated in keratinocytes comprising the outer root sheath of the murine hair follicle (HF). When syngeneic skin grafts were exchanged between gender-matched wild-type (WT) and CD200-deficient C57BL/6 mice, significant perifollicular and intrafollicular inflammation was observed, eventually leading to the destruction of virtually all HF (alopecia) without significant loss of the CD200-negative grafts. Minimal and transient inflammation was observed in WT grafts, which persisted long term with hair. There was a 2-fold increase in graft-infiltrating T cells in CD200-deficient skin at 14 d. Alopecia and skin lesions were induced in CD200-deficient hosts by adoptive transfer of splenocytes from WT mice previously grafted with CD200-negative skin, but not from mice grafted with WT skin. Collectively, these results suggest that the expression of CD200 in follicular epithelium attenuates inflammatory reactions and may play a role in maintaining immune tolerance to HF-associated autoantigens.


Subject(s)
Alopecia/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Hair Follicle/immunology , Immune Tolerance/physiology , Adoptive Transfer , Alopecia/genetics , Alopecia/physiopathology , Animals , Antigens, CD , Bone Marrow Transplantation , Cells, Cultured , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/physiopathology , Female , Hair Follicle/cytology , Keratinocytes/cytology , Keratinocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenotype , Skin Transplantation , Spleen/cytology , T-Lymphocytes/immunology , Transplantation Chimera
15.
Cell Immunol ; 227(2): 129-39, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15135295

ABSTRACT

We have been investigating whether alloantigen-specific CD4(+)25+ regulatory T cells can be identified for use in treating graft-versus-host disease. CD150, which is upregulated on the surface of all activated T lymphocytes, was identified as a candidate marker for alloantigen-activated CD4(+)25+ regulatory T cells by gene chip analysis. Freshly isolated CD4(+)25+ cells had only low cell-surface expression of CD150, comparable to that of CD4(+)25- T cells. Increased CD150 expression was observed on all T cells after coculture with allogeneic stimulator cells. When purified CD4(+)25+ cells were precultured with allogeneic stimulator cells, then sorted into CD150+ and CD150- subsets, allosuppressive activity was contained primarily in the CD150+ fraction. These cells also suppressed the proliferation of alloantigen-activated autologous T cells, and they could be expanded in vitro without loss of their suppressive capacity. These results suggest that CD150 can be used as a marker for the identification of purified alloantigen-activated CD4(+)25+ regulatory T cells.


Subject(s)
CD4 Antigens/analysis , Glycoproteins/analysis , Immunoglobulins/analysis , Isoantigens/immunology , Lymphocyte Activation , Receptors, Interleukin-2/analysis , T-Lymphocytes/immunology , Antigens, CD , Humans , Lymphocyte Culture Test, Mixed , Receptors, Antigen, T-Cell/physiology , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1
16.
Med Immunol ; 1(1): 3, 2002 Nov 19.
Article in English | MEDLINE | ID: mdl-12495444

ABSTRACT

BACKGROUND: Long term treatment with immunosuppressive agents results in nephrotoxicity in renal transplant recipients. We explored the effect of combination of Tacrolimus (TAC) and Sirolimus (SRL) on the immune system in renal transplant recipients. METHODS: 10 stable renal transplant recipients were selected to participate in a pharmacokinetic study with a combination of TAC and SRL. Blood was drawn on day zero and 14 days post treatment. Lymphocyte proliferation was quantified by 3H-thymidine uptake assay (results expressed as counts per minute). The mRNA expression was studied by RT-PCR and serum levels of cytokines were quantified by ELISA and a cytokine bead array system. RESULTS: Lymphocyte proliferative response to PHA (p < 0.05), Con A (p < 0.006) and Anti-CD3 (p <0.005) were significantly decreased in patients who received both TAC and SRL compared to TAC alone. The mRNA expression of proinflammatory cytokines TNF-alpha (p < 0.05), cyclins G (p < 0.01) and E (p < 05) were decreased, and of TGF-beta (p < 0.03) and p21 (p < 0.05) were increased in patients treated with this combination. Circulating levels of IFN-gamma (p < 0.04), IL-4 (p < 0.02), and Il-2 (p < 0.03) were significantly inhibited and elevation of TGF-beta (p < 0.04) was observed in patients treated with TAC and SRL combination. CONCLUSION: These novel findings demonstrate that addition of SRL to TAC therapy enhances immuno modulation and causes increased immunosuppression providing a rationale for this concomitant therapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...