Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Toxicol Pathol ; 51(4): 205-215, 2023 06.
Article En | MEDLINE | ID: mdl-37578155

The evaluation of changes in the immune system serves to determine the efficacy and potential immunotoxicologic effects of new products under development. Toxicologic pathologists play critical roles in identifying immune system changes that drive the immunosafety determination. Standard pathology evaluations of therapies and chemicals remain similar; however, biopharmaceutical therapies have moved from simply affecting the immune system to being specifically developed to modify the immune system, which can impact interpretation. Recent explosive growth in immunomodulatory therapies presents a challenge to the toxicologic pathologist, toxicologist, and regulatory reviewer in terms of evaluating the clinical relevance and potential adversity of immune system changes. Beyond the recognition of such changes, there is an increasing expectation to evaluate, describe, and interpret how therapies affect complex immune system pathways for both immunomodulatory therapies and non-immunomodulatory drugs with off-target immunotoxic effects. In this opinion piece, considerations regarding immune system evaluation, the current landscape of immunomodulatory therapies, a brief description of immunotoxicologic (and immunopathologic) endpoints, the importance of integrating such immunosafety data, and relevance to adversity determination are discussed. Importantly, we describe how the current paradigm of determining adversity for immune system changes may be challenging or insufficient and propose a harmonized and flexible approach for assessing adversity.

2.
Am J Pathol ; 191(2): 335-352, 2021 02.
Article En | MEDLINE | ID: mdl-33181139

Human T-lymphotropic virus type 1 (HTLV-1) causes adult T-cell leukemia, a disease commonly associated with hypercalcemia and osteolysis. There is no effective treatment for HTLV-1, and the osteolytic mechanisms are not fully understood. Mice expressing the HTLV-1 oncogene Tax, driven by the human granzyme B promoter (Tax+), develop osteolytic tumors. To investigate the progression of the bone-invasive malignancies, wild-type, Tax+, and Tax+/interferon-γ-/- mice were assessed using necropsy, histologic examination, IHC analysis, flow cytometry, and advanced imaging. Tax+ and Tax+/interferon-γ-/- malignancies of the ear, tail, and foot comprised poorly differentiated, round to spindle-shaped cells with prominent neutrophilic infiltrates. Tail tumors originated from muscle, nerve, and/or tendon sheaths, with frequent invasion into adjacent bone. F4/80+ and anti-mouse CD11b (Mac-1)+ histiocytic cells predominated within the tumors. Three Tax+/interferon-γ-/- cell lines were generated for in vivo allografts, in vitro gene expression and bone resorption assays. Two cell lines were of monocyte/macrophage origin, and tumors formed in vivo in all three. Differences in Pthrp, Il6, Il1a, Il1b, and Csf3 expression in vitro were correlated with differences in in vivo plasma calcium levels, tumor growth, metastasis, and neutrophilic inflammation. Tax+ mouse tumors were classified as bone-invasive histiocytic sarcomas. The cell lines are ideal for further examination of the role of HTLV-1 Tax in osteolytic tumor formation and the development of hypercalcemia and tumor-associated inflammation.


Cell Line, Tumor , Disease Models, Animal , Genes, pX , HTLV-I Infections/complications , Histiocytic Sarcoma , Animals , Carcinogenesis/genetics , Histiocytic Sarcoma/pathology , Histiocytic Sarcoma/virology , Human T-lymphotropic virus 1/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes , Osteolysis/pathology , Osteolysis/virology
3.
Infect Immun ; 87(8)2019 08.
Article En | MEDLINE | ID: mdl-31182615

CD4+ T helper 1 (Th1) cells producing interferon gamma (IFN-γ) are critical for the resolution of visceral leishmaniasis (VL). MicroRNA 155 (miR155) promotes CD4+ Th1 responses and IFN-γ production by targeting suppressor of cytokine signaling-1 (SOCS1) and Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP-1) and therefore could play a role in the resolution of VL. To determine the role of miR155 in VL, we monitored the course of Leishmania donovani infection in miR155 knockout (miR155KO) and wild-type (WT) C57BL/6 mice. miR155KO mice displayed significantly higher liver and spleen parasite loads than WT controls and showed impaired hepatic granuloma formation. However, parasite growth eventually declined in miR155KO mice, suggesting the induction of a compensatory miR155-independent antileishmanial pathway. Leishmania antigen-stimulated splenocytes from miR155KO mice produced significantly lower levels of Th1-associated IFN-γ than controls. Interestingly, at later time points, levels of Th2-associated interleukin-4 (IL-4) and IL-10 were also lower in miR155KO splenocyte supernatants than in WT mice. On the other hand, miR155KO mice displayed significantly higher levels of IFN-γ, iNOS, and TNF-α gene transcripts in their livers than WT mice, indicating that distinct organ-specific antiparasitic mechanisms were involved in control of L. donovani infection in miR155KO mice. Throughout the course of infection, organs of miR155KO mice showed significantly more PDL1-expressing Ly6Chi inflammatory monocytes than WT mice. Conversely, blockade of Ly6Chi inflammatory monocyte recruitment in miR155KO mice significantly reduced parasitic loads, indicating that these cells contributed to disease susceptibility. In conclusion, we found that miR155 contributes to the control of L. donovani but is not essential for infection resolution.


Leishmania donovani , Leishmaniasis, Visceral/immunology , MicroRNAs/physiology , Animals , Granuloma/etiology , Interferon-gamma/physiology , Mice , Mice, Inbred C57BL , Monocytes/physiology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/physiology
4.
J Infect Dis ; 219(4): 599-608, 2019 01 29.
Article En | MEDLINE | ID: mdl-30239895

Background: New drugs are needed for leishmaniasis because current treatments such as pentavalent antimonials are toxic and require prolonged administration, leading to poor patient compliance. Ibrutinib is an anticancer drug known to modulate T-helper type 1 (Th1)/Th2 responses and has the potential to regulate immunity against infectious disease. Methods: In this study, we evaluated the efficacy of oral ibrutinib as a host-targeted treatment for visceral leishmaniasis (VL) caused by Leishmania donovani using an experimental mouse model. Results: We found that oral ibrutinib was significantly more effective than the pentavalent antimonial sodium stibogluconate (70 mg/kg) for the treatment of VL caused by L. donovani. Ibrutinib treatment increased the number of interleukin 4- and interferon γ-producing natural killer T cells in the liver and spleen and enhanced granuloma formation in the liver. Further, ibrutinib treatment reduced the influx of Ly6Chi inflammatory monocytes, which mediate susceptibility to L. donovani. Finally, ibrutinib treatment was associated with the increased production of the cytokines interferon γ, tumor necrosis factor α, interleukin 4, and interleukin 13 in the liver and spleen, which are associated with protection against L. donovani. Conclusions: Our findings show that oral ibrutinib is highly effective for the treatment of VL caused by L. donovani and mediates its antileishmanial activity by promoting host immunity. Therefore, ibrutinib could be a novel host-targeted drug for the treatment of VL.


Immunologic Factors/administration & dosage , Leishmania donovani/growth & development , Leishmaniasis, Visceral/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Adenine/analogs & derivatives , Administration, Oral , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Immunity, Cellular , Mice , Mice, Inbred BALB C , Piperidines , Treatment Outcome
5.
Front Immunol ; 9: 1593, 2018.
Article En | MEDLINE | ID: mdl-30042766

Macrophages and their monocyte precursors mediate innate immune responses and can promote a spectrum of phenotypes from pro-inflammatory to pro-resolving. Currently, there are few markers that allow for robust dissection of macrophage phenotype. We recently identified CD38 as a marker of inflammatory macrophages in murine in vitro and in vivo models. However, it is unknown whether CD38 plays a similar marker and/or functional role in human macrophages and inflammatory diseases. Here, we establish that CD38 transcript and protein are robustly induced in human macrophages exposed to LPS (±IFN-γ) inflammatory stimuli, but not with the alternative stimulus, IL-4. Pharmacologic and/or genetic CD38 loss-of-function significantly reduced the secretion of inflammatory cytokines IL-6 and IL-12p40 and glycolytic activity in human primary macrophages. Finally, monocyte analyses in systemic lupus erythematosus patients revealed that, while all monocytes express CD38, high CD38 expression in the non-classical monocyte subpopulation is associated with disease. These data are consistent with an inflammatory marker role for CD38 in human macrophages and monocytes.

6.
Cell Transplant ; 26(1): 11-21, 2017 01 24.
Article En | MEDLINE | ID: mdl-27393133

Bilirubin has been recognized as a powerful cytoprotectant when used at physiologic doses and was recently shown to have immunomodulatory effects in islet allograft transplantation, conveying donor-specific tolerance in a murine model. We hypothesized that bilirubin, an antioxidant, acts to suppress the innate immune response to islet allografts through two mechanisms: 1) by suppressing graft release of damage-associated molecular patterns (DAMPs) and inflammatory cytokines, and 2) by producing a tolerogenic phenotype in antigen-presenting cells. Bilirubin was administered intraperitoneally before pancreatic procurement or was added to culture media after islet isolation in AJ mice. Islets were exposed to transplant-associated nutrient deprivation and hypoxia. Bilirubin significantly decreased islet cell death after isolation and hypoxic stress. Bilirubin supplementation of islet media also decreased the release of DAMPs (HMGB1), inflammatory cytokines (IL-1ß and IL-6), and chemokines (MCP-1). Cytoprotection was mediated by the antioxidant effects of bilirubin. Treatment of macrophages with bilirubin induced a regulatory phenotype, with increased expression of PD-L1. Coculture of these macrophages with splenocytes led to expansion of Foxp3+ Tregs. In conclusion, exogenous bilirubin supplementation showed cytoprotective and antioxidant effects in a relevant model of islet isolation and hypoxic stress. Suppression of DAMP release, alterations in cytokine profiles, and tolerogenic effects on macrophages suggest that the use of this natural antioxidant may provide a method of preconditioning to improve outcomes after allograft transplantation.


Bilirubin/therapeutic use , Immune Tolerance/drug effects , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/methods , Animals , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , HMGB1 Protein/metabolism , Immune Tolerance/immunology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoassay , Mice
7.
Cell Immunol ; 309: 61-68, 2016 11.
Article En | MEDLINE | ID: mdl-27614845

The outcome of visceral leishmaniasis, caused by parasite Leishmania donovani, depends on the recruitment of leishmanicidal Th1 cells. Chemokine receptor CXCR3, preferentially expressed by Th1 cells, is critical for migration of these T cells during infection. During chronic VL, there is a decrease in the presence of CXCR3-expressing CD4+ T cells in the spleen, which is associated with high parasitic burden in this organ. We therefore examined whether T cell-specific expression of CXCR3 in mice (CXCR3Tg) would promote resistance to VL. L. donovani infected CXCR3Tg mice showed increased accumulation of T cells in the spleens compared to WT littermates (CXCR3+/+). However, CXCR3+ T cells from CXCR3Tg mice showed low CD69 expression and these mice developed fewer granulomas. Additionally, both groups of mice showed similar cytokine profiles and parasitic burdens during the course of infection. In summary, although T cell-specific expression of CXCR3 promoted the accumulation of CXCR3-expressing T cells during L. donovani infection, this did not enhance resistance to VL.


Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Liver/physiology , Receptors, CXCR3/metabolism , Spleen/physiology , Th1 Cells/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Movement/genetics , Cells, Cultured , Lectins, C-Type/metabolism , Liver/parasitology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Specificity , Receptors, CXCR3/genetics , Spleen/parasitology , Th1 Cells/parasitology , Th1-Th2 Balance , Transgenes/genetics
8.
Am J Vet Res ; 77(6): 604-12, 2016 Jun.
Article En | MEDLINE | ID: mdl-27227498

OBJECTIVE To evaluate the effects of damage-associated molecular patterns (DAMPs) derived from disrupted mitochondria on canine splenocytes and other immune cells. SAMPLES Liver, spleen, and bone marrow samples obtained from 8 cadavers of healthy research Beagles that had been euthanized for other purposes. PROCEDURES Mitochondria were obtained from canine hepatocytes, and mitochondrial DAMPs (containing approx 75% mitochondrial proteins) were prepared. Mitochondrial DAMPs and the nuclear cytokine high-mobility group box protein 1 were applied to splenocytes, bone marrow-differentiated dendritic cells, and a canine myelomonocytic cell (DH82) line for 6 or 24 hours. Cell culture supernatants from splenocytes, dendritic cells, and DH82 cells were assayed for tumor necrosis factor α with an ELISA. Expression of tumor necrosis factor α mRNA in splenocytes was evaluated with a quantitative real-time PCR assay. RESULTS In all cell populations evaluated, production of tumor necrosis factor α was consistently increased by mitochondrial DAMPs at 6 hours (as measured by an ELISA). In contrast, high-mobility group box protein 1 did not have any independent proinflammatory effects in this experimental system. CONCLUSIONS AND CLINICAL RELEVANCE The study revealed an in vitro inflammatory effect of mitochondrial DAMPs (containing approx 75% mitochondrial proteins) in canine cells and validated the use of an in vitro splenocyte model to assess DAMP-induced inflammation in dogs. This experimental system may aid in understanding the contribution of DAMPs to sepsis and the systemic inflammatory response syndrome in humans. Further studies in dogs are needed to validate the biological importance of these findings and to evaluate the in vivo role of mitochondrial DAMPs in triggering and perpetuating systemic inflammatory states.


Dogs , Mitochondria/metabolism , Spleen/cytology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cadaver , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation , Humans , Inflammation/metabolism , Mitochondria/genetics , Mitochondria/pathology , Tumor Necrosis Factor-alpha/genetics
9.
J Immunotoxicol ; 13(5): 652-65, 2016 09.
Article En | MEDLINE | ID: mdl-27075513

The potential to engineer extracellular vesicles (EV) that target specific cells and deliver a therapeutic payload has propelled a growing interest in their development as promising therapeutics. These EV are often produced from cultured cells. Very little is known about the interaction of cell culture-derived EV with cells of the immune system and their potential immunomodulatory effects. The present study evaluated potential immunotoxic effects of HEK293T-derived EV on the human monocytic cell lines THP-1 and U937. Incubation of cells with different doses of EV for 16-24 h was followed by assessment of cytotoxicity and cell function by flow cytometry. Changes in cell functionality were evaluated by the capacity of cells to phagocytize fluorescent microspheres. In addition, the internalization of labeled EV in THP-1 and U937 cells was evaluated. Exposure to EV did not affect the viability of THP-1 or U937 cells. Although lower doses of the EV increased phagocytic capacity in both cell lines, phagocytic efficiency of individual cells was not affected by EV exposure at any of the doses evaluated. This study also demonstrated that THP-1 and U937 monocytic cells are highly permissive to EV entry in a dose-response manner. These results suggest that, although HEK293T-derived EV are efficiently internalized by human monocytic cells, they do not exert a cytotoxic effect or alter phagocytic efficiency on the cell lines evaluated.


Cell Death , Extracellular Vesicles/immunology , Monocytes/immunology , Cell Culture Techniques , Cell Survival , Drug Delivery Systems/methods , HEK293 Cells , Humans , Phagocytosis , U937 Cells
10.
PLoS One ; 10(7): e0132921, 2015.
Article En | MEDLINE | ID: mdl-26218271

OBJECTIVE: Immune suppression during critical illness predisposes to serious infections. We sought to determine the mechanisms regulating tolerance and cross-tolerance to common pro-inflammatory danger signals in a model that recapitulates the intact in vivo immune response. MATERIALS AND METHODS: Flt3-expanded splenocytes obtained from wild-type or matching IRAK-M knockout (IRAK-M-/-), C57BL/6, male mice (8-10 weeks old) were treated repeatedly or alternately with either LPS or CpGA DNA, agonists of Toll-like receptor (TLR)-4 and -9, respectively, over successive 24-hour periods. Supernatants were collected following each 24-hour period with cytokine release (ELISA) and splenocyte IRAK-M expression (Western blot) determined. Tolerance and cross-tolerance were assessed in the absence or presence of programmed death receptor (PD)-1 blocking antibody or IL-7 pre-treatment. MAIN RESULTS: Splenocytes notably exhibited both tolerance and cross-tolerance to subsequent treatments with either LPS or CpGA DNA. The character of tolerance and cross-tolerance in this model was distinct following initial LPS or CpGA treatment in that TNFα and IFNγ release (not IL-10) were suppressed following LPS; whereas, initial CpGA treatment suppressed TNFα, IFNγ and IL-10 release in response to subsequent stimulation (LPS or CpGA). Tolerance and cross-tolerance were unrelated to IL-10 release or PD-1 but were attenuated in IRAK-M-/- splenocytes. IL-7 significantly suppressed IRAK-M expression and restored TNFα and IFNγ production without influencing IL-10 release. CONCLUSIONS: In summary, acute immune tolerance and cross-tolerance in response to LPS or CpGA were distinct in that LPS selectively suppressed pro-inflammatory cytokine responses; whereas, CpGA suppressed both pro- and anti-inflammatory responses. The induction of tolerance and cross-tolerance in response to common danger signals was mechanistically unrelated to IL-10 or PD-1 but was directly influenced by IRAK-M expression. IL-7 reduced IRAK-M expression and attenuated immune tolerance induced by either LPS or CpGA, and thus may be useful for reversal of immune tolerance in the setting of critical illness.


Immune Tolerance , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-7/immunology , Spleen/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/immunology , Animals , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-7/genetics , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Spleen/cytology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
12.
BMC Immunol ; 15: 8, 2014 Feb 18.
Article En | MEDLINE | ID: mdl-24548459

BACKGROUND: Myeloid cells (MC) have potent immunoregulatory abilities that can be therapeutically useful to treat inflammatory disease. However, the factors which promote regulatory myeloid cell differentiation remain poorly understood. We have previously shown that estriol (E3) induces mature regulatory dendritic cells in vivo. To determine whether additional steroid hormones could induce mature regulatory myeloid cells, we investigated the effects of retinoic acid (RA) on MCs. Retinoic acid is a steroid hormone important in regulating mucosal immunity in the gut and promoting myeloid differentiation. We hypothesized that the presence of RA during differentiation would promote the formation of mature regulatory myeloid cells (MCregs). METHODS: To determine RA's ability to induce regulatory myeloid cells, we differentiated bone marrow progenitor cells with granulocytic-macrophage colony-stimulating factor (GM-CSF) under the influence of RA. We found that day 7 MCs differentiated in the presence of RA had an increase in the percent positive and relative expression levels of both maturation (CD80, CD86, and MHCII) and inhibitory (PD-L1 and PD-L2) markers compared to control cells. Functionally, these day 7 RA MCs expressed increased intracellular IL-10, induced regulatory T cells in vitro compared to controls and suppressed the proliferation of responder immune cells even after inflammatory challenge with LPS. CONCLUSION: RA induced mature regulatory myeloid cells that were suppressive and had a CD11b+ CD11c-Ly6C low/intermediate monocyte phenotype. Surprisingly, RA CD11c+ dendritic cells were not suppressive and could contribute to enhanced proliferation. These results suggest that continuous RA has unique effects on different myeloid populations during monopoeisis and dendropoiesis and promotes a population of regulatory monocytes.


Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Monocytes/cytology , Monocytes/drug effects , Tretinoin/pharmacology , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Differentiation/immunology , Cell Line , Dendritic Cells/immunology , Immunophenotyping , Mice , Mice, Transgenic , Monocytes/immunology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Mol Pharm ; 11(3): 828-35, 2014 Mar 03.
Article En | MEDLINE | ID: mdl-24433027

Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system that can cause loss of motor function and is thought to result, in part, from chronic inflammation due to an antigen-specific T cell immune response. Current treatments suppress the immune system without antigen specificity, increasing the risks of cancer, chronic infection, and other long-term side effects. In this study, we show treatment of experimental autoimmune encephalomyelitis (EAE), a model of MS, by coencapsulating the immunodominant peptide of myelin oligodendrocyte glycoprotein (MOG) with dexamethasone (DXM) into acetalated dextran (Ac-DEX) microparticles (DXM/MOG/MPs) and administering the microparticles subcutaneously. The clinical score of the mice was reduced from 3.4 to 1.6 after 3 injections 3 days apart with the coencapsulated microparticulate formulation (MOG 17.6 µg and DXM 8 µg). This change in clinical score was significantly greater than observed with phosphate-buffered saline (PBS), empty MPs, free DXM and MOG, DXM/MPs, and MOG/MPs. Additionally, treatment with DXM/MOG/MPs significantly inhibited disease-associated cytokine (e.g., IL-17, GM-CSF) expression in splenocytes isolated in treated mice. Here we show a promising approach for the therapeutic treatment of MS using a polymer-based microparticle delivery platform.


Dexamethasone/administration & dosage , Dextrans/chemistry , Drug Delivery Systems , Encephalomyelitis, Autoimmune, Experimental/therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/administration & dosage , Polymers/chemistry , Animals , Cell Proliferation/drug effects , Combined Modality Therapy , Cytokines/metabolism , Dexamethasone/pharmacokinetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/metabolism , Nitric Oxide/metabolism , Peptide Fragments/immunology , Tissue Distribution
14.
J Antimicrob Chemother ; 69(1): 168-75, 2014 Jan.
Article En | MEDLINE | ID: mdl-23956375

OBJECTIVES: The imidazoquinoline family of drugs are Toll-like receptor 7/8 agonists that have previously been used in the treatment of cutaneous leishmaniasis. Because of the hydrophobic nature of imidazoquinolines, they are traditionally not administered systemically for the treatment of visceral leishmaniasis. We formulated liposomal resiquimod, an imidazoquinoline, for the systemic treatment of visceral leishmaniasis. METHODS: By using lipid film hydration with extrusion, we encapsulated resiquimod in liposomes. These liposomes were then injected intravenously to treat BALB/c mice infected with Leishmania donovani. RESULTS: Treatment with liposomal resiquimod significantly decreased the parasite load in the liver, spleen and bone marrow. In addition, resiquimod treatment increased interferon-γ and interleukin-10 production in an antigen recall assay. Resiquimod was shown to be non-toxic in histology and in vitro culture experiments. CONCLUSIONS: FDA-approved resiquimod, in a liposomal formulation, displays promising results in treating visceral leishmaniasis.


Antiprotozoal Agents/administration & dosage , Imidazoles/administration & dosage , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Liposomes/administration & dosage , Administration, Intravenous , Animals , Bone Marrow/parasitology , Disease Models, Animal , Interferon-gamma/metabolism , Interleukin-10/metabolism , Leishmania donovani/isolation & purification , Leishmaniasis, Visceral/parasitology , Leukocytes, Mononuclear/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Parasite Load , Spleen/parasitology , Treatment Outcome
15.
Eur J Immunol ; 44(2): 450-9, 2014 Feb.
Article En | MEDLINE | ID: mdl-24242758

We and others have previously shown that IL-12 is indispensable for immunity and is required for the optimal antiparasitic activity of antimonials in experimental visceral leishmaniasis caused by Leishmania donovani. Here we investigated the role of STAT4 in immunity against L. donovani using STAT4 knockout mice and also determined the effect of STAT4 deficiency in response to antimonial therapy. Upon infection with L. donovani, stat4⁻/⁻ BALB/c and C57BL/6 mice showed enhanced susceptibility to Leishmania during late time points of infection which was associated with a marked reduction in Th1 responses and hepatic immunopathology. Interestingly, these defects in Th1 responses in stat4⁻/⁻ did not impair the antimonial chemotherapy as both stat4⁻/⁻ and WT mice showed comparable levels of parasite clearance from the liver and spleen. These findings highlight the role of STAT4 in immunity to L. donovani infection and also provide evidence that STAT4 is dispensable for antimonial-based chemotherapy.


Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , Animals , Interleukin-10/immunology , Interleukin-4/immunology , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/parasitology , Liver/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , STAT4 Transcription Factor/deficiency , Spleen/immunology , Spleen/parasitology , Th1 Cells/immunology
16.
PLoS One ; 8(8): e72354, 2013.
Article En | MEDLINE | ID: mdl-23951313

OBJECTIVE: Mitochondrial transcription factor A (TFAM) is normally bound to and remains associated with mitochondrial DNA (mtDNA) when released from damaged cells. We hypothesized that TFAM, bound to mtDNA (or equivalent CpG-enriched DNA), amplifies TNFα release from TLR9-expressing plasmacytoid dendritic cells (pDCs) by engaging RAGE. MATERIALS AND METHODS: Murine Flt3 ligand-expanded splenocytes obtained from C57BL/6 mice were treated with recombinant human TFAM, alone or in combination with CpG-enriched DNA with subsequent TNFα release measured by ELISA. The role of RAGE was determined by pre-treatment with soluble RAGE or heparin or by employing matching RAGE (-/-) splenocytes. TLR9 signaling was evaluated using a specific TLR9-blocking oligonucleotide and by inhibiting endosomal processing, PI3K and NF-κB. Additional studies examined whether heparin sulfate moieties or endothelin converting enzyme-1 (ECE-1)-dependent recycling of endosomal receptors were required for TFAM and CpG DNA recognition. MAIN RESULTS: TFAM augmented splenocyte TNFα release in response to CpGA DNA, which was strongly dependent upon pDCs and regulated by RAGE and TLR9 receptors. Putative TLR9 signaling pathways, including endosomal acidification and signaling through PI3K and NF-κB, were essential for splenocyte TNFα release in response to TFAM+CpGA DNA. Interestingly, TNFα release depended upon endothelin converting enzyme (ECE)-1, which cleaves and presumably activates TLR9 within endosomes. Recognition of the TFAM-CpGA DNA complex was dependent upon heparin sulfate moieties, and recombinant TFAM Box 1 and Box 2 proteins were equivalent in terms of augmenting TNFα release. CONCLUSIONS: TFAM promoted TNFα release in a splenocyte culture model representing complex cell-cell interactions in vivo with pDCs playing a critical role. To our knowledge, this study is the first to incriminate ECE-1-dependent endosomal cleavage of TLR9 as a critical step in the signaling pathway leading to TNFα release. These findings, and others reported herein, significantly advance our understanding of sterile immune responses triggered by mitochondrial danger signals.


DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Toll-Like Receptor 9/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , CpG Islands , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Dendritic Cells/immunology , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Humans , Male , Mice , Models, Biological , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Signal Transduction , Spleen/cytology , Spleen/metabolism
17.
BMC Vet Res ; 8: 209, 2012 Oct 31.
Article En | MEDLINE | ID: mdl-23110794

BACKGROUND: Myeloid derived suppressor cells (MDSCs) are a recently described population of immune cells that significantly contribute to the immunosuppression seen in cancer patients. MDSCs are one of the most important factors that limit the efficacy of cancer immunotherapy (e.g. cancer vaccines) and MDSC levels are increased in cancer in multiple species. Identifying and targeting MDSCs is actively being investigated in the field of human oncology and is increasingly being investigated in veterinary oncology. The treatment of canine cancer not only benefits dogs, but is being used for translational studies evaluating and modifying candidate therapies for use in humans. Thus, it is necessary to understand the immune alterations seen in canine cancer patients which, to date, have been relatively limited. This study investigates the use of commercially available canine antibodies to detect an immunosuppressive (CD11b low/CADO48 low) cell population that is increased in the peripheral blood of tumor-bearing dogs. RESULTS: Commercially available canine antibodies CD11b and CADO48A were used to evaluate white blood cells from the peripheral blood cells of forty healthy control dogs and forty untreated, tumor-bearing dogs. Tumor-bearing dogs had a statistically significant increase in CD11b low/CADO48A low cells (7.9%) as compared to the control dogs (3.6%). Additionally, sorted CD11b low/CADO48A low generated in vitro suppressed the proliferation of canine lymphocytes. CONCLUSIONS: The purpose of this study was aimed at identifying potential canine specific markers for identifying MDSCs in the peripheral blood circulation of dogs. This study demonstrates an increase in a unique CD11b low/CADO48A low cell population in tumor-bearing dogs. This immunophenotype is consistent with described phenotypes of MDSCs in other species (i.e. mice) and utilizes commercially available canine-specific antibodies. Importantly, CD11b low/CADO48A low from a tumor environment suppress the proliferation of lymphocytes. These results provide a useful phenotype of cells increased in canine cancer patients that may serve as a useful prognostic marker for assessing immune status and functional response to cancer immunotherapies in dogs. Understanding MDSCs in dogs will allow for increased effectiveness of cancer immunotherapy in both dogs and humans.


Biomarkers, Tumor/blood , CD11b Antigen/blood , Dog Diseases/blood , Myeloid Cells/pathology , Neoplasms/veterinary , Animals , Dog Diseases/immunology , Dog Diseases/pathology , Dogs , Flow Cytometry/veterinary , Immunophenotyping/veterinary , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Myeloid Cells/immunology , Neoplasms/blood , Neoplasms/immunology , Neoplasms/pathology , Prospective Studies , Statistics, Nonparametric
18.
J Immunol ; 189(1): 433-43, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22675199

Plasmacytoid dendritic cells (pDC) are potent APCs known to regulate immune responses to self-Ags, particularly DNA. The mitochondrial fraction of necrotic cells was found to most potently promote human pDC activation, as reflected by type I IFN release, which was dependent upon the presence of mitochondrial DNA and involved TLR9 and receptors for advanced glycation end products. Mitochondrial transcription factor A (TFAM), a highly abundant mitochondrial protein that is functionally and structurally homologous to high mobility group box protein 1, was observed to synergize with CpG-containing oligonucleotide, type A, DNA to promote human pDC activation. pDC type I IFN responses to TFAM and CpG-containing oligonucleotide, type A, DNA indicated their engagement with receptors for advanced glycation end products and TLR9, respectively, and were dependent upon endosomal processing and PI3K, ERK, and NF-κB signaling. Taken together, these results indicate that pDC contribute to sterile immune responses by recognizing the mitochondrial component of necrotic cells and further incriminate TFAM and mitochondrial DNA as likely mediators of pDC activation under these circumstances.


Adjuvants, Immunologic/physiology , CpG Islands/immunology , DNA, Mitochondrial/genetics , DNA-Binding Proteins/physiology , Dendritic Cells/immunology , Dendritic Cells/pathology , Mitochondrial Proteins/physiology , Signal Transduction/immunology , Transcription Factors/physiology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Animals , CpG Islands/genetics , DNA-Binding Proteins/genetics , Dendritic Cells/metabolism , Gene Amplification/immunology , Hep G2 Cells , Humans , Interferon-alpha/metabolism , Mice , Mitochondrial Proteins/genetics , Necrosis , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/immunology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Toll-Like Receptor 9/physiology , Transcription Factors/genetics
19.
J Biol Chem ; 287(33): 28017-26, 2012 Aug 10.
Article En | MEDLINE | ID: mdl-22733812

Dimethyl fumarate (DMF) is an effective novel treatment for multiple sclerosis in clinical trials. A reduction of IFN-γ-producing CD4(+) T cells is observed in DMF-treated patients and may contribute to its clinical efficacy. However, the cellular and molecular mechanisms behind this clinical observation are unclear. In this study, we investigated the effects of DMF on dendritic cell (DC) maturation and subsequent DC-mediated T cell responses. We show that DMF inhibits DC maturation by reducing inflammatory cytokine production (IL-12 and IL-6) and the expression of MHC class II, CD80, and CD86. Importantly, this immature DC phenotype generated fewer activated T cells that were characterized by decreased IFN-γ and IL-17 production. Further molecular studies demonstrated that DMF impaired nuclear factor κB (NF-κB) signaling via reduced p65 nuclear translocalization and phosphorylation. NF-κB signaling was further decreased by DMF-mediated suppression of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and its downstream kinase mitogen stress-activated kinase 1 (MSK1). MSK1 suppression resulted in decreased p65 phosphorylation at serine 276 and reduced histone phosphorylation at serine 10. As a consequence, DMF appears to reduce p65 transcriptional activity both directly and indirectly by promoting a silent chromatin environment. Finally, treatment of DCs with the MSK1 inhibitor H89 partially mimicked the effects of DMF on the DC signaling pathway and impaired DC maturation. Taken together, these studies indicate that by suppression of both NF-κB and ERK1/2-MSK1 signaling, DMF inhibits maturation of DCs and subsequently Th1 and Th17 cell differentiation.


Dendritic Cells/immunology , Fumarates/pharmacology , Immunosuppressive Agents/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/immunology , Ribosomal Protein S6 Kinases, 90-kDa/immunology , Transcription Factor RelA/immunology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , B7-2 Antigen/biosynthesis , B7-2 Antigen/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dimethyl Fumarate , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , MAP Kinase Signaling System/immunology , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factor RelA/metabolism
20.
Cancer Res ; 71(24): 7608-16, 2011 Dec 15.
Article En | MEDLINE | ID: mdl-22009537

Patients with advanced hepatocellular carcinoma (HCC) face a dismal prognosis because of a lack of any effective therapies. To address this situation, we conducted a preclinical investigation of the therapeutic efficacy of oligonucleotides directed against the oncogenic microRNA miR-221, which has been implicated in HCC. Of 9 chemistries evaluated, we determined that a 2'-O-methyl phosphorothioate-modified anti-miR-221 oligonucleotide was most effective at reducing proliferation in vitro. A cholesterol-modified isoform of anti-miR-221 (chol-anti-miR-221) exhibited improved pharmacokinetics and liver tissue distribution compared with unmodified oligonucleotide. Chol-anti-miR-221 significantly reduced miR-221 levels in liver within a week of intravenous administration and in situ hybridization studies confirmed accumulation of the oligonucleotide in tumor cells in vivo. Within the same period, chol-anti-miR-221 reduced tumor cell proliferation and increased markers of apoptosis and cell-cycle arrest, elevating the tumor doubling time and increasing mouse survival. Taken together, our findings offer a preclinical proof of efficacy for chol-anti-miR-221 in a valid orthotopic mouse model of HCC, suggesting that this targeted agent could benefit treatment for patients with advanced HCC.


Carcinoma, Hepatocellular/genetics , Gene Silencing , Liver Neoplasms, Experimental/genetics , MicroRNAs/genetics , Xenograft Model Antitumor Assays/methods , Animals , Apoptosis/drug effects , Blotting, Northern , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cholesterol/chemistry , Female , Humans , In Situ Hybridization , Kaplan-Meier Estimate , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/therapy , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods
...