Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
J Immunother ; 41(3): 141-150, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29528991

ABSTRACT

We are developing whole, heat-killed, recombinant Saccharomyces cerevisiae yeast, engineered to encode target proteins, which stimulate immune responses against malignant cells expressing those targets. This phase 1 trial, enrolling patients with advanced colorectal or pancreas cancer, was designed to evaluate safety, immunogenicity, response, and overall survival of ascending doses of the GI-4000 series of products, which express 3 different forms of mutated Ras proteins. The study enrolled 33 heavily pretreated subjects (14 with pancreas and 19 with colorectal cancer), whose tumors were genotyped before enrollment to identify the specific ras mutation and thereby to identify which GI-4000 product to administer. No dose limiting toxicities were observed and no subject discontinued treatment due to a GI-4000 related adverse event (AE). The majority of AEs and all fatal events were due to underlying disease progression and AE frequencies were not significantly different among dose groups. GI-4000 was immunogenic, as Ras mutation-specific immune responses were detected on treatment in ∼60% of subjects. No objective tumor responses were observed but based on imaging, clinical status and/or biochemical markers, stable disease was observed in 6 subjects (18%) on day 29, while 1 subject had stable disease at days 57 and 85 follow-up visits. The median overall survival was 3.3 months (95% confidence interval, 2.3-5.3 mo), and 5 subjects survived past the 48-week follow-up period. No significant dose-dependent trends for survival were observed. This first clinical trial in humans with GI-4000 demonstrated a favorable safety profile and immunogenicity in the majority of subjects.


Subject(s)
Biological Therapy , Gene Expression , Mutation , Neoplasms/genetics , Neoplasms/therapy , Saccharomyces cerevisiae/genetics , ras Proteins/genetics , Adult , Aged , Biological Therapy/methods , Biomarkers, Tumor , Complement Activation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunity , Male , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Saccharomyces cerevisiae/immunology
3.
Clin Vaccine Immunol ; 24(12)2017 Dec.
Article in English | MEDLINE | ID: mdl-29046306

ABSTRACT

As yet, very few vaccine candidates with activity in animals against Mycobacterium tuberculosis infection have been tested as therapeutic postexposure vaccines. We recently described two pools of mycobacterial proteins with this activity, and here we describe further studies in which four of these proteins (Rv1738, Rv2032, Rv3130, and Rv3841) were generated as a fusion polypeptide and then delivered in a novel yeast-based platform (Tarmogen) which itself has immunostimulatory properties, including activation of Toll-like receptors. This platform can deliver antigens into both the class I and class II antigen presentation pathways and stimulate strong Th1 and Th17 responses. In mice this fusion vaccine, designated GI-19007, was immunogenic and elicited strong gamma interferon (IFN-γ) and interleukin-17 (IL-17) responses; despite this, they displayed minimal prophylactic activity in mice that were subsequently infected with a virulent clinical strain. In contrast, in a therapeutic model in the guinea pig, GI-19007 significantly reduced the lung bacterial load and reduced lung pathology, particularly in terms of secondary lesion development, while significantly improving survival in one-third of these animals. In further studies in which guinea pigs were vaccinated with BCG before challenge, therapeutic vaccination with GI-19007 initially improved survival versus that of animals given BCG alone, although this protective effect was gradually lost at around 400 days after challenge. Given its apparent ability to substantially limit bacterial dissemination within and from the lungs, GI-19007 potentially can be used to limit lung damage as well as facilitating chemotherapeutic regimens in infected individuals.


Subject(s)
Gene Transfer Techniques , Mycobacterium tuberculosis/immunology , Saccharomyces cerevisiae/genetics , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Female , Guinea Pigs , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Post-Exposure Prophylaxis/methods , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , Treatment Outcome , Tuberculosis/immunology , Tuberculosis/pathology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
4.
Methods Mol Biol ; 1404: 529-545, 2016.
Article in English | MEDLINE | ID: mdl-27076321

ABSTRACT

GlobeImmune's Tarmogen(®) immunotherapy platform utilizes recombinant Saccharomyces cerevisiae yeast as a vaccine vector to deliver heterologous antigens for activation of disease-specific, targeted cellular immunity. The vaccines elicit immune-mediated killing of target cells expressing viral and cancer antigens in vivo via a CD8(+) CTL-mediated mechanism. Tarmogens are not neutralized by host immune responses and can be administered repeatedly to boost antigen-specific immunity. Production of the vaccines yields stable off-the-shelf products that avoid the need for patient-specific manufacturing found with other immunotherapeutic approaches. Tarmogens for the treatment of chronic hepatitis B and C and various cancers were well tolerated and immunogenic in phase 1 and 2 clinical trials encompassing >600 subjects. The platform is being widely utilized in basic vaccine research and the most rapid path to success in these endeavors follows from optimal immunoassay selection and execution. This chapter provides detailed methods for the construction and preclinical immunogenicity testing of yeast-based immunotherapeutic products to support the rapid and efficient use of this versatile technology.


Subject(s)
Saccharomyces cerevisiae/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Animals , Flow Cytometry , Immunization , Mice , Spleen/immunology , T-Lymphocytes/cytology , Vaccines, Synthetic/genetics
5.
Discov Med ; 21(114): 109-16, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27011046

ABSTRACT

Fas ligand (FasL, CD95L) is a 40-kDa type II transmembrane protein that binds to Fas (CD95) receptors and promotes programmed cell death. Fas receptors are expressed at higher levels in many tumors than in normal cells; however, systemic administration of FasL or agonistic anti-Fas antibodies to mice with tumors caused lethal hepatitis. Somewhat paradoxically, elimination of Fas or FasL from tumors also leads to death induced by CD95 receptor/ligand elimination (DICE). At face value, this suggests that Fas signaling not only kills normal cells, but that it also is essential for tumor cell survival. Targeting this pathway may not only fail to kill tumors, but instead may even enhance their growth, leading some to report the demise of Fas ligand in cancer immunotherapy. But, to paraphrase Mark Twain, is this death an exaggeration? Here, we provide a careful examination of the literature exploring the merits of FasL as a novel form of cancer immunotherapy. With local administration using delivery vectors that achieve high levels of expression in the tumor environment, our results indicate that the potential for systemic toxicity is eliminated in higher mammals, and that a systemic anti-tumor response ensues, which delays or prevents progression and simultaneously attacks distant metastases.


Subject(s)
Fas Ligand Protein/toxicity , Fas Ligand Protein/therapeutic use , Immunotherapy , Neoadjuvant Therapy , Neoplasms/pathology , Animals , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Graft Rejection/immunology , Humans , Neoplasms/immunology
6.
Cancer Immunol Immunother ; 64(11): 1449-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26250807

ABSTRACT

The potential of mesenchymal stromal cells (MSCs) to inhibit anti-tumor immunity is becoming increasingly well recognized, but the precise steps affected by these cells during the development of an anti-tumor immune response remain incompletely understood. Here, we examined how MSCs affect the steps required to mount an effective anti-tumor immune response following administration of adenovirus Fas ligand (Ad-FasL) in the Lewis lung carcinoma (LL3) model. Administration of bone marrow-derived MSCs with LL3 cells accelerated tumor growth significantly. MSCs inhibited the inflammation induced by Ad-FasL in the primary tumors, precluding their rejection; MSCs also reduced the consequent expansion of tumor-specific T cells in the treated hosts. When immune T cells were transferred to adoptive recipients, MSCs impaired, but did not completely abrogate the ability of these T cells to promote elimination of secondary tumors. This impairment was associated with a modest reduction in tumor-infiltrating T cells, with a significant reduction in tumor-infiltrating macrophages, and with a reorganization of the stromal environment. Our data indicate that MSCs in the tumor environment reduce the efficacy of immunotherapy by creating a functional and anatomic barrier that impairs inflammation, T cell priming and expansion, and T cell function-including recruitment of effector cells.


Subject(s)
Carcinoma, Lewis Lung/immunology , Inflammation/prevention & control , Mesenchymal Stem Cells/physiology , T-Lymphocytes/immunology , Tumor Microenvironment , Adenoviridae/genetics , Animals , Cytotoxicity, Immunologic , Fas Ligand Protein/genetics , Fas Ligand Protein/physiology , Mice , T-Lymphocytes/physiology
7.
PLoS One ; 9(7): e101904, 2014.
Article in English | MEDLINE | ID: mdl-25051027

ABSTRACT

Chronic hepatitis B infection (CHB) is characterized by sub-optimal T cell responses to viral antigens. A therapeutic vaccine capable of restoring these immune responses could potentially improve HBsAg seroconversion rates in the setting of direct acting antiviral therapies. A yeast-based immunotherapy (Tarmogen) platform was used to make a vaccine candidate expressing hepatitis B virus (HBV) X, surface (S), and Core antigens (X-S-Core). Murine and human immunogenicity models were used to evaluate the type and magnitude of HBV-Ag specific T cell responses elicited by the vaccine. C57BL/6J, BALB/c, and HLA-A*0201 transgenic mice immunized with yeast expressing X-S-Core showed T cell responses to X, S and Core when evaluated by lymphocyte proliferation assay, ELISpot, intracellular cytokine staining (ICS), or tumor challenge assays. Both CD4+ and CD8+ T cell responses were observed. Human T cells transduced with HBc18-27 and HBs183-91 specific T cell receptors (TCRs) produced interferon gamma (IFNγ following incubation with X-S-Core-pulsed dendritic cells (DCs). Furthermore, stimulation of peripheral blood mononuclear cells (PBMCs) isolated from CHB patients or from HBV vaccine recipients with autologous DCs pulsed with X-S-Core or a related product (S-Core) resulted in pronounced expansions of HBV Ag-specific T cells possessing a cytolytic phenotype. These data indicate that X-S-Core-expressing yeast elicit functional adaptive immune responses and supports the ongoing evaluation of this therapeutic vaccine in patients with CHB to enhance the induction of HBV-specific T cell responses.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B, Chronic/prevention & control , T-Lymphocytes/immunology , Vaccination , Animals , Cell Proliferation , Cells, Cultured , Cross-Priming , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Hepatitis B Vaccines/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Liver Neoplasms/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Saccharomyces cerevisiae/genetics , T-Lymphocytes/virology , Trans-Activators/genetics , Trans-Activators/immunology , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Regulatory and Accessory Proteins
8.
PLoS One ; 9(6): e100629, 2014.
Article in English | MEDLINE | ID: mdl-24945807

ABSTRACT

Nfatc2 and Tob1 are intrinsic negative regulators of T cell activation. Nfatc2-deficient and Tob1-deficient T cells show reduced thresholds of activation; however, whether these factors have independent or overlapping roles in negative regulation of T cell responses has not been previously examined. Here, we show that Nfatc2 knockout (KO) but not Tob1 KO mice have age-associated accumulation of persistently activated T cells in vivo and expansion of the CD44+ memory cell compartment and age-associated lymphocytic infiltrates in visceral organs, without significant changes in numbers of CD4+CD25+Foxp3+ regulatory T cells (Treg). In vitro, CD4+CD25- "conventional" T cells (Tconvs) from both KO strains showed greater proliferation than wild type (WT) Tconvs. However, while Tregs from Nfatc2 KO mice retained normal suppressive function, Tregs from Tob1 KOs had enhanced suppressive activity. Nfatc2 KO Tconvs expanded somewhat more rapidly than WT Tconvs under conditions of homeostatic proliferation, but their accelerated growth capacity was negated, at least acutely, in a lymphoreplete environment. Finally, Nfatc2 KO mice developed a previously uncharacterized increase in B-cell malignancies, which was not accelerated by the absence of Tob1. The data thus support the prevailing hypothesis that Nfatc2 and Tob1 are non-redundant regulators of lymphocyte homeostasis.


Subject(s)
B-Lymphocytes/metabolism , Carcinogenesis/genetics , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , NFATC Transcription Factors/genetics , T-Lymphocytes, Regulatory/metabolism , Animals , B-Lymphocytes/pathology , CD4 Antigens/genetics , CD4 Antigens/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carrier Proteins/metabolism , Cell Transformation, Neoplastic , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Deletion , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins , Lymphoma/metabolism , Lymphoma/pathology , Male , Mice , Mice, Knockout , NFATC Transcription Factors/deficiency , Signal Transduction , T-Lymphocytes, Regulatory/pathology
9.
Mol Ther ; 20(12): 2234-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22850679

ABSTRACT

Fas ligand (FasL) gene therapy for cancer has shown promise in rodents; however, its efficacy in higher mammals remains unknown. Here, we used intratumoral FasL gene therapy delivered in an adenovirus vector (Ad-FasL) as neoadjuvant to standard of care in 56 dogs with osteosarcoma. Tumors from treated dogs had greater inflammation, necrosis, apoptosis, and fibrosis at day 10 (amputation) compared to pretreatment biopsies or to tumors from dogs that did not receive Ad-FasL. Survival improvement was apparent in dogs with inflammation or lymphocyte-infiltration scores >1 (in a 3-point scale), as well as in dogs that had apoptosis scores in the top 50th percentile (determined by cleaved caspase-3). Survival was no different than that expected from standard of care alone in dogs with inflammation scores ≤1 or apoptosis scores in the bottom 50th percentile. Reduced Fas expression by tumor cells was associated with prognostically advantageous inflammation, and this was seen only in dogs that received Ad-FasL. Together, the data suggest that Ad-FasL gene therapy improves survival in a subset of large animals with naturally occurring tumors, and that at least in some tumor types like osteosarcoma, it is most effective when tumor cells fail to express Fas.


Subject(s)
Bone Neoplasms/therapy , Fas Ligand Protein/genetics , Genetic Therapy/methods , Animals , Apoptosis/genetics , Apoptosis/physiology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Dogs , Necrosis , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/therapy
10.
J Immunother ; 35(1): 14-22, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22130158

ABSTRACT

In current clinical trails, whole yeast-based immunotherapy expressing hepatitis C viral antigens demonstrated statistically significant improvement in end of treatment responses when combined with type I interferon based standard of care, even in standard of care resistant patients. Although preclinical data suggest yeast vaccination, such as type I interferon, facilitates CD8 T-cell immunity, the capacity of yeast to generate immunity in patients resistant to type I interferon calls into question the mechanism(s) underpinning the efficacy of this approach. We show yeast and a Toll-like receptor exclusive agonist, Pam3Cys, differ in CD8 T-cell generation when combined with an agonistic CD40 antibody. Although both yeast and PamCys were largely Toll-like receptor dependent, the primary CD8 response generated by yeast was significantly less than Pam3Cys in wild-type hosts even in a CD4 T-cell-deficient setting. In addition, immunization of IL6 mice with yeast produced a 3-fold to 6-fold increased CD8 response while the Pam3Cys response was unaffected. The yeast but not Pam3Cys-driven CD8 response was inhibited in both wild-type and IL-6 hosts by blocking interleukin (IL)-12. In addition, IL6 mice had increased CD86 expression on their dendritic cells after yeast immunization also inhibited by IL-12 blockade. Collectively, our results indicate the CD8 T-cell response to yeast but not Pam3Cys is influenced by IL-6-mediated control of IL-12 critical for dendritic cell activation. To our knowledge this is the first demonstration that yeast directly influence IL-12-associated CD8 T-cell immunity providing an additional route whereby recombinant yeast may provide efficacy independent of type I interferon.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Immunotherapy/methods , Interleukin-6/metabolism , Saccharomyces cerevisiae/immunology , Animals , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Interferon Type I/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Lipoproteins/administration & dosage , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology , Vaccination
11.
BMC Cancer ; 10: 619, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21062482

ABSTRACT

BACKGROUND: The etiology of hemangiosarcoma remains incompletely understood. Its common occurrence in dogs suggests predisposing factors favor its development in this species. These factors could represent a constellation of heritable characteristics that promote transformation events and/or facilitate the establishment of a microenvironment that is conducive for survival of malignant blood vessel-forming cells. The hypothesis for this study was that characteristic molecular features distinguish hemangiosarcoma from non-malignant endothelial cells, and that such features are informative for the etiology of this disease. METHODS: We first investigated mutations of VHL and Ras family genes that might drive hemangiosarcoma by sequencing tumor DNA and mRNA (cDNA). Protein expression was examined using immunostaining. Next, we evaluated genome-wide gene expression profiling using the Affymetrix Canine 2.0 platform as a global approach to test the hypothesis. Data were evaluated using routine bioinformatics and validation was done using quantitative real time RT-PCR. RESULTS: Each of 10 tumor and four non-tumor samples analyzed had wild type sequences for these genes. At the genome wide level, hemangiosarcoma cells clustered separately from non-malignant endothelial cells based on a robust signature that included genes involved in inflammation, angiogenesis, adhesion, invasion, metabolism, cell cycle, signaling, and patterning. This signature did not simply reflect a cancer-associated angiogenic phenotype, as it also distinguished hemangiosarcoma from non-endothelial, moderately to highly angiogenic bone marrow-derived tumors (lymphoma, leukemia, osteosarcoma). CONCLUSIONS: The data show that inflammation and angiogenesis are important processes in the pathogenesis of vascular tumors, but a definitive ontogeny of the cells that give rise to these tumors remains to be established. The data do not yet distinguish whether functional or ontogenetic plasticity creates this phenotype, although they suggest that cells which give rise to hemangiosarcoma modulate their microenvironment to promote tumor growth and survival. We propose that the frequent occurrence of canine hemangiosarcoma in defined dog breeds, as well as its similarity to homologous tumors in humans, offers unique models to solve the dilemma of stem cell plasticity and whether angiogenic endothelial cells and hematopoietic cells originate from a single cell or from distinct progenitor cells.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hemangiosarcoma/metabolism , Neovascularization, Pathologic , Animals , Cell Line, Tumor , Dogs , Female , Hemangiosarcoma/veterinary , Humans , Inflammation , Male , Mutation , Oligonucleotide Array Sequence Analysis , Von Hippel-Lindau Tumor Suppressor Protein/genetics , ras Proteins/genetics
12.
Cancer Immunol Immunother ; 58(2): 171-85, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18523772

ABSTRACT

The survival of naive T cells is compromised in the absence of molecules encoded by the major histocompatibility complex (MHC) while antigen-experienced T cells survive. We hypothesized that survival pressures in an in vivo, MHC-deficient environment would permit enrichment of less frequent antigen-experienced autoreactive cells at the expense of the majority of antigen naive T cells. To test this hypothesis, we generated MHC class I- and class II-deficient mice in NOD and C57Bl/6 (B6) backgrounds, and examined the capacity of adoptively transferred autoimmune-prone NOD T cells, or non-autoimmune prone naive B6 T cells, respectively, to reject transplanted wild-type pancreatic islets or transplantable tumors in the MHC-deficient mice. In the MHC-deficient environment, CD4 T cells acquired self-hostile properties (islet rejection and tumor invasion) that were independent from their genetic propensity for autoreactivity, while CD8 T cells required appropriate prior exposure to antigen in order to survive and function (reject tumor) in this environment; however, disengagement of Tob1, a negative regulator of proliferation, led to a reverse phenotype with regard to persistence of CD4 and CD8 T cells in the MHC-deficient environment. Our data suggest that self-peptide/MHC interactions have dual roles to facilitate survival and restrain autoreactivity, thus acting as integral components of an intrinsic network of negative regulation that maintains tolerance.


Subject(s)
Autoimmunity , Desensitization, Immunologic , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Animals , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Survival , Immune Tolerance , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
13.
Immunol Res ; 41(2): 137-53, 2008.
Article in English | MEDLINE | ID: mdl-18415030

ABSTRACT

It is now apparent that naïve peripheral T cells are a dynamic population where active processes prevent inappropriate activation while supporting survival. The process of thymic education makes naïve peripheral T cells dependent on interactions with self-MHC for survival. However, as these signals can potentially result in inappropriate activation, various non-redundant, intrinsic negative regulatory molecules including Tob, Nfatc2, and Smad3 actively enforce T cell quiescence. Interactions among these pathways are only now coming to light and may include positive or negative crosstalk. In the case of positive crosstalk, self-MHC initiated signals and intrinsic negative regulatory factors may cooperate to dampen T cell activation and sustain peripheral tolerance in a binary fashion (on-off). In the case of negative crosstalk, self-MHC signals may promote survival through partial activation while intrinsic negative regulatory factors act as rheostats to restrain cell cycle entry and prevent T cells from crossing a threshold that would break tolerance.


Subject(s)
Major Histocompatibility Complex/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Proliferation , GTP-Binding Proteins/metabolism , Homeostasis , Humans , Lymphocyte Activation , NFATC Transcription Factors/metabolism , Protein Kinases/metabolism , Signal Transduction , Smad3 Protein/metabolism , T-Lymphocyte Subsets/metabolism , Tumor Suppressor Proteins/metabolism
14.
Mol Immunol ; 44(4): 479-87, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16584774

ABSTRACT

The diabetes-prone biobreeding (BB-DP) rat contains the lyp mutation which results in lymphopenia and promotes the progression of a T cell-mediated autoimmune attack of the pancreas in certain rat strains. This mutation has been mapped to a gene which bears homology to human Gimap5/Ian5 and results in the truncation and loss of activity of this protein. The lymphopenic state induced by the loss of this protein has led to the proposal that Gimap5 has an anti-apoptotic function. Previously we described an additional phenotype of incomplete activation mediated by the loss of Gimap5 function. Here we further characterize this incomplete activation phenotype and map a potential signal transduction pathway leading to activation. We show that CD5 expression on peripheral T cells is elevated in Gimap5 animals, while thymocyte expression remains similar between the two strains. Additionally, we show that NF-kappaB but not NFAT is activated in unstimulated Gimap5 mutant T cells as compared to unstimulated wild type T cells. Mapping this activation to its upstream source we show that activation of NF-kappaB is correlated with an activation of IKK. Using a variety of kinase inhibitors we further map this increase in IKK to an increase in MEK activation. Finally, to counter the possibility that activation is an indirect consequence of the lymphopenic environment, we created bone marrow chimeras in which Gimap5 mutant T cells developed in a normal environment and show that these cells retain their activated phenotype. Together, we interpret these data as demonstrating that the activation caused by loss of Gimap5 is a cell intrinsic phenomenon caused, in part, by a MEK-dependent activation of IKK. This, in turn, would suggest that Gimap5 functions to promote both T cell survival and quiescence and that these pathways are biochemically linked.


Subject(s)
GTP-Binding Proteins/genetics , NF-kappa B/genetics , Signal Transduction/genetics , Animals , CD5 Antigens/biosynthesis , CD5 Antigens/immunology , Cells, Cultured , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Enzyme Activation/genetics , Gene Deletion , MAP Kinase Signaling System/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Rats , Rats, Mutant Strains , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Eur J Immunol ; 34(9): 2396-406, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15307172

ABSTRACT

The Biobreeding diabetes-prone rat suffers from a profound peripheral lymphopenia and yet succumbs to a T cell-dependent autoimmune disease. Lymphopenia segregates with a mutated chromosomal locus, termed lyp, recently identified as a frameshift mutation in IAN4. Others have correlated loss of IAN4 function with decreased mitochondrial integrity resulting in T cell apoptosis. Here we report that IAN4-/- T cells enter a state similar to that of partial activation wherein they down-regulate CD62L and undergo incomplete blasting yet do not progress through mitosis. When given a strong stimulus, this partial activation phenotype can be overcome. This phenotype can be recapitulated in wild-type T cells through suboptimal stimulation. The phenotype is not simply a reaction to the lymphopenic environment, as spontaneous CD62L down-regulation occurs in mature single-positive medullary thymocytes that develop within a non-lymphopenic environment, and normal T cells do not undergo similar blasting when parked in a lymphopenic environment. Finally, we show that IAN4-/- T cells are more readily triggered via TCR stimulation. Thus, in addition to their role in apoptosis, IAN family members may also play a role in regulating the T cell activation state through modulation of TCR signaling strength.


Subject(s)
Apoptosis , Histocompatibility Antigens Class II/physiology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cells, Cultured , Down-Regulation , Histocompatibility Antigens Class II/genetics , Homeostasis , L-Selectin/analysis , Rats , Rats, Inbred F344 , Receptors, Antigen, T-Cell/physiology
18.
Cancer Res ; 64(15): 5084-8, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15289309

ABSTRACT

Activating mutations in Ras oncoproteins represent attractive targets for cancer immunotherapy, but few vectors capable of generating immune responses required for tumor killing without vector neutralization have been described. Whole recombinant yeast heterologously expressing mammalian mutant Ras proteins were used to immunize mice in a carcinogen-induced lung tumor model. Therapeutic immunization with the whole recombinant yeast caused complete regression of established Ras mutation-bearing lung tumors in a dose-dependent, antigen-specific manner. In combination with the genomic sequencing of tumors in patients, the yeast-based immunotherapeutic approach could be applied to treat Ras mutation-bearing human cancers.


Subject(s)
Immunotherapy/methods , Lung Neoplasms/prevention & control , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Adenoma/chemically induced , Adenoma/immunology , Adenoma/prevention & control , Animals , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred Strains , Mutation , Neoplasms, Experimental/chemically induced , Proto-Oncogene Proteins p21(ras)/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/physiology , Urethane
19.
J Pharmacol Exp Ther ; 311(2): 758-69, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15231866

ABSTRACT

We used primary peripheral blood T cells, a population that exists in G(0) and can be stimulated to enter the cell cycle synchronously, to define more precisely the effects of nicotine on pathways that control cell cycle entry and progression. Our data show that nicotine decreased the ability of T cells to transit through the G(0)/G(1) boundary (acquire competence) and respond to progression signals. These effects were due to nuclear factor of activated T cells c2 (NFATc2)-dependent repression of cyclin-dependent kinase 4 (CDK4) expression. Growth arrest at the G(0)/G(1) boundary was further enforced by inhibition of cyclin D2 expression and by increased expression and stabilization of p27Kip1. Intriguingly, T cells from habitual users of tobacco products and from NFATc2-deficient mice constitutively expressed CDK4 and were resistant to the antiproliferative effects of nicotine. These results indicate that nicotine impairs T cell cycle entry through NFATc2-dependent mechanisms and suggest that, in the face of chronic nicotine exposure, selection may favor cells that can evade these effects. We postulate that cross talk between nicotinic acetylcholine receptors and growth factor receptor-activated pathways offers a novel mechanism by which nicotine may directly impinge on cell cycle progression. This offers insight into possible reasons that underlie the unique effects of nicotine on distinct cell types and identifies new targets that may be useful control tobacco-related diseases.


Subject(s)
Cell Cycle/drug effects , DNA-Binding Proteins/metabolism , Nicotine/pharmacology , Nuclear Proteins/metabolism , T-Lymphocytes/drug effects , Transcription Factors/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cytokines/pharmacology , DNA-Binding Proteins/physiology , Humans , Mice , NFATC Transcription Factors , Nuclear Proteins/physiology , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , T-Lymphocytes/cytology , Nicotiana/chemistry , Transcription Factors/physiology
20.
Clin Immunol ; 112(1): 54-65, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15207782

ABSTRACT

Signals transmitted by binding of Fas ligand (FasL) to the Fas receptor (CD95/Apo-1) have pleiotropic effects on cellular function that present opportunities for therapeutic applications. For example, depending on the circumstances, overexpression of FasL can enhance, prevent, or reverse growth of spontaneous or transplantable tumors. Furthermore, local administration of FasL into a single paw in susceptible mice protects from or reduces the severity of collagen-induced arthritis (CIA) in all paws. Here, we define mechanisms that mediate systemic protection induced by locally delivered FasL. Protection is not solely dependent on local interactions between Fas and FasL, but rather requires induction of a paradoxical inflammatory response that not only destroys Fas-resistant tumors, but also recruits motile, activated, Fas-bearing T cells that are Fas sensitive. We demonstrate by following the antigen-specific recruitment and subsequent termination of transgenic T cells that activated T cells, including autoreactive cells responsible for CIA, are eliminated within this inflammatory environment through the overexpressed FasL. The nature of the inflammatory response, which depends on the Fas ligand being cell bound and not soluble, and the magnitude of FasL expression within the inflammatory milieu are essential for this effect, as arthritogenic inflammation alone resulting from CIA induction is insufficient to ameliorate the disease or eliminate antigen-specific T cells, even upon systemic delivery of soluble FasL. These data show that gene delivery of membrane-bound FasL can effectively recruit and eliminate autoreactive T cells.


Subject(s)
Arthritis, Experimental/prevention & control , Autoimmunity/drug effects , Membrane Glycoproteins/pharmacology , T-Lymphocytes/drug effects , Adenoviridae/genetics , Adoptive Transfer , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoimmunity/immunology , Cell Death/immunology , Cytotoxicity, Immunologic , Fas Ligand Protein , Female , Lymphocyte Activation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...