Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Haematologica ; 108(8): 2155-2166, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36861411

ABSTRACT

Multiple Myeloma (MM) is an incurable plasma cell malignancy often treated by autologous stem cell transplant (ASCT). Clinical response to ASCT has been associated with DNA repair efficiency. Here we interrogated the role of the base excision DNA repair (BER) pathway in MM response to ASCT. Across 450 clinical samples and six disease stages, expression levels of genes in the BER pathway were found to be highly upregulated during the development of MM. In a separate cohort of 559 patients with MM treated with ASCT, expression of BER pathway members MPG and PARP3 was positively associated with overall survival (OS) while expression of PARP1, POLD1, and POLD2 was negatively associated with OS. In a validation cohort of 356 patients with MM treated with ASCT, PARP1 and POLD2 findings were replicated. In patients with MM who never received ASCT (n=319), PARP1 and POLD2 were not associated with OS, suggesting that the prognostic effect of these genes may be treatment-dependent. In preclinical models of MM, synergy was observed in anti-tumor activity when poly (ADPribose) polymerase (PARP) inhibitors (olaparib, talazoparib) were used in combination with melphalan. The negative prognosis associated with PARP1 and POLD2 expression along with the apparent melphalan-sensitizing effect of PARP inhibition may suggest this pathway as a potential biomarker in patients with MM in the setting of ASCT. Further understanding of the role of the BER pathway in MM is vital to improve therapeutic strategies related to ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Melphalan/therapeutic use , Prognosis , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Transplantation, Autologous , Stem Cell Transplantation , Retrospective Studies , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/therapeutic use , DNA Polymerase III
2.
Front Pharmacol ; 14: 1334440, 2023.
Article in English | MEDLINE | ID: mdl-38259277

ABSTRACT

Introduction: Polymorphisms in genes responsible for the metabolism and transport of tacrolimus have been demonstrated to influence clinical outcomes for patients following allogeneic hematologic stem cell transplant (allo-HSCT). However, the clinical impact of germline polymorphisms specifically for oral formulations of tacrolimus is not fully described. Methods: To investigate the clinical impact of genetic polymorphisms in CYP3A4, CYP3A5, and ABCB1 on oral tacrolimus pharmacokinetics and clinical outcomes, we prospectively enrolled 103 adult patients receiving oral tacrolimus for the prevention of graft-versus-host disease (GVHD) following allo-HSCT. Patients were followed in the inpatient and outpatient phase of care for the first 100 days of tacrolimus therapy. Patients were genotyped for CYP3A5 *3 (rs776746), CYP3A4 *1B (rs2740574), ABCB1 exon 12 (rs1128503), ABCB1 exon 21 (rs2032582), ABCB1 exon 26 (rs1045642). Results: Expression of CYP3A5 *1 was highly correlated with tacrolimus pharmacokinetics in the inpatient phase of care (p < 0.001) and throughout the entirety of the study period (p < 0.001). Additionally, Expression of CYP3A5 *1 was associated with decreased risk of developing AKI as an inpatient (p = 0.06). Variants in ABCB1 were not associated with tacrolimus pharmacokinetics in this study. We were unable to discern an independent effect of CYP3A4 *1B or *22 in this population. Conclusion: Expression of CYP3A5 *1 is highly influential on the pharmacokinetics and clinical outcomes for patients receiving oral tacrolimus as GVHD prophylaxis following allo-HSCT.

3.
Anticancer Res ; 40(10): 5707-5713, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988896

ABSTRACT

BACKGROUND/AIM: Genetic variations of the non-coding RNA gene, ANRIL, have been associated with human diseases including cancer, type-2 diabetes, and atherosclerosis. In the present study, we investigated the potential associations of select ANRIL single nucleotide polymorphisms (SNPs) with overall survival and other clinical outcomes in adult patients with hematologic malignancies after allogeneic hematopoietic stem cell transplantation (allo-HSCT). PATIENTS AND METHODS: Genomic DNA was extracted from whole blood samples from 103 adult patients with hematologic malignancies who had received allo-HSCT followed by oral tacrolimus therapy. The genotypes of four select ANRIL SNPs, rs564398, rs1063192, rs2151280, and rs2157719 were determined using qRT-PCR-based genotyping assays. RESULTS: rs2151280 (C->T) in ANRIL was associated with worse overall survival in these patients (CT/CC vs. TT). Contrarily, rs2151280 and the other select ANRIL SNPs were not associated with death at Day-100 after transplantation, the incidence of graft-versus-host disease (GVHD), acute kidney injury (AKI), and neurotoxicity in the study cohort. CONCLUSION: rs2151280 represents a potential prognostic biomarker for overall survival in adult patients with hematologic malignancies after allo-HSCT.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Hematologic Neoplasms/genetics , RNA, Long Noncoding/genetics , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Aged , Female , Genotype , Graft vs Host Disease/etiology , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Hematologic Neoplasms/complications , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Progression-Free Survival , Tacrolimus/pharmacokinetics , Transplantation, Homologous/adverse effects
4.
Anticancer Res ; 40(1): 67-73, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31892553

ABSTRACT

BACKGROUND/AIM: Aberrant expression of the SEI1 oncogene has been prevalently found in a variety of human cancers, including oral squamous cell carcinoma (OSCC). Recent studies have shown that cisplatin up-regulates the expression of SEI1 in breast and bladder cancer cells, thus inhibiting apoptosis and cell death in these cells. In the present study, we investigated the impact of cisplatin on the expression of SEI1 in OSCC cells. MATERIALS AND METHODS: Four OSCC cell lines, CAL27, SCC4, SCC15, and SCC22A were treated with cisplatin and 5-fluorouracil, and changes in SEI1 expression in these cells were evaluated using quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR) analyses. RESULTS: Cisplatin significantly induced SEI1 expression in the tested OSCC cells. Contrarily, cisplatin treatment did not affect the expression of gankyrin and BMI1, two oncogenes frequently overexpressed in a coordinate manner with SEI1 in OSCC. Additionally, 5-fluorouracil did not bring about any detectable changes in SEI1 expression in these cells. CONCLUSION: Cisplatin-induced up-regulation of SEI1 expression in OSCC is specific, and such induction could underlie the development of resistance to cisplatin in OSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Mouth Neoplasms/genetics , Oncogenes , Transcription Factors/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Humans
5.
Anticancer Res ; 40(1): 133-141, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31892561

ABSTRACT

BACKGROUND/AIM: Aberrant expression of the BMI1 oncogene has been prevalently found in a variety of human cancers, including cervical cancer. Recent studies have shown that PTC209, a specific BMI1 inhibitor, exhibits high potency in inhibiting the growth of colon, breast, oral cancer cells and cancer-initiating cells, indicative of its chemotherapeutic potential. In the current study, we evaluated the inhibitory abilities of PTC209 in cervical cancer cells. MATERIALS AND METHODS: Three cervical cell lines, C33A, HeLa, and SiHa were treated with PTC209. The impacts of PTC209 on BMI1 were investigated using quantitative reverse-transcription PCR assay (qRT-PCR) and western blotting; changes in cell viability, cell cycle distribution, and apoptosis were assessed using cell viability testing, colony formation assay and flow cytometry analyses, respectively. RESULTS: PTC209 exhibited considerably high short-term and long-term cytotoxicities in all tested cervical cancer cell lines regardless of their HPV infection status, TP53 and pRb statuses. PTC209 significantly downregulated the expression of BMI1 in cervical cancer cell lines, and such downregulation led to G0/G1 arrest (p<0.05). Moreover, PTC209 drove more cells into apoptosis (p<0.05). CONCLUSION: PTC209 (BMI1-targeting agents, in general) represents a novel chemotherapeutic agent with potential in cervical cancer therapy.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Enzyme Inhibitors/pharmacology , Polycomb Repressive Complex 1/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
6.
Bioanalysis ; 11(6): 471-483, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30892061

ABSTRACT

Aim: To develop and validate a reliable, robust and efficient assay to detect and quantify biologic compounds in vitro and in vivo during early stage of a biotherapeutic agent discovery. Methodology & results: An enrichment-free immunoassay method was developed to quantify a polyhistidine N- and FLAG C-terminally-tagged recombinant protein of ∼55 kDa. The target proteins were purified by a nickel-based matrix via tag affinity, followed by probing with biotinylated antitag antibody and subsequently detected by streptavidin-horseradish peroxidase conjugate using an automated capillary-based western system. Conclusion: A simple, highly sensitive and efficient immunoassay protocol was established to assess the in vitro stability and pharmacokinetic properties of propitious recombinant proteins in vivo in mouse to support early stage development of a biotherapeutic lead.


Subject(s)
Epitopes/chemistry , Immunoassay/methods , Recombinant Proteins/blood , Animals , Biotinylation , Blotting, Western/methods , Histidine/chemistry , Indicators and Reagents/chemistry , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Nickel/chemistry , Oligopeptides/blood , Oligopeptides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Streptavidin/chemistry
7.
Mol Carcinog ; 56(7): 1722-1732, 2017 07.
Article in English | MEDLINE | ID: mdl-28150872

ABSTRACT

Multiple myeloma (MM) is a hematologic malignancy characterized by clonal proliferation of plasma cells and overproduction of monoclonal immunoglobins. Treatment with melphalan is currently standard of care for younger and fit patients when followed by hematopoietic stem cell transplantation (HSCT), and in transplant ineligible patients when used in combination regimens. It has been previously shown that changes in the p53 pathway are associated with melphalan efficacy, but the regulatory role of the p14ARF-MDM2-p53 axis has yet to be fully explored. Recently, a non-coding RNA, ANRIL (antisense non-coding RNA in the INK4-ARF locus) has been shown to negatively regulate the transcription of the entire INK4-ARF locus and simultaneously modulate the p53 and pRb pathways. Moreover, some single nucleotide polymorphisms (SNPs) in ANRIL have previously been associated with susceptibility to several malignancies. Here we investigated select ANRIL SNPs in DNA from patient-derived peripheral blood mononuclear cells obtained from 108 MM patients treated with high-dose melphalan followed by HSCT. Our results show that the rs2151280 (CàT) SNP in ANRIL was associated with worse progression-free survival (TC/CC vs TT: HR = 0.53, 95%CI, [0.26, 1.07], P = 0.07; adjusted HR = 0.39, 95%CI, [0.18, 0.84], P = 0.016), and the TT variant had higher ANRIL expression and lower p15, p14ARF, and p16 expression compared to the TC/CC variants. Our results indicate that ANRIL may be involved in melphalan-mediated apoptosis via down-regulating p14ARF and subsequent p53, and that the rs2151280 polymorphism may be a potential prognostic biomarker for relapse in melphalan-treated MM patients.


Subject(s)
Leukocytes, Mononuclear/pathology , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Polymorphism, Single Nucleotide/genetics , RNA, Long Noncoding/genetics , Stem Cell Transplantation/adverse effects , Adult , Aged , Antineoplastic Agents, Alkylating/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Follow-Up Studies , Humans , Leukocytes, Mononuclear/metabolism , Male , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Prognosis , Prospective Studies , Survival Rate , Transplantation, Autologous , Tumor Suppressor Protein p14ARF/metabolism
8.
Cell Transplant ; 26(1): 11-21, 2017 01 24.
Article in English | MEDLINE | ID: mdl-27393133

ABSTRACT

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.


Subject(s)
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
9.
Am J Vet Res ; 77(6): 604-12, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27227498

ABSTRACT

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.


Subject(s)
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
10.
BMC Immunol ; 15: 8, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24548459

ABSTRACT

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.


Subject(s)
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
11.
Mol Pharm ; 11(3): 828-35, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24433027

ABSTRACT

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.


Subject(s)
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
12.
PLoS One ; 8(8): e72354, 2013.
Article in English | MEDLINE | ID: mdl-23951313

ABSTRACT

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.


Subject(s)
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
13.
Mol Pharm ; 10(8): 2849-57, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23768126

ABSTRACT

To enhance the immune activity of vaccine adjuvants polyinosinic:polycytidylic acid (poly I:C) and CpG acetalated dextran (Ac-DEX) microparticles can be used. Ac-DEX is a biodegradable and water-insoluble polymer that degrades significantly faster at pH 5.0 (phagosomal pH) than at pH 7.4 and has tunable degradation rates that can range from hours to months. This is an ideal characteristic for delivery of an antigen and adjuvant within the lysosomal compartment of a phagocytic cell. We evaluated poly I:C and CpG encapsulated in Ac-DEX microparticles using RAW macrophages as a model antigen-presenting cell. These cells were cultured with poly I:C or CpG in their free form, encapsulated in a fast degrading Ac-DEX, in slow degrading Ac-DEX, or in the Food and Drug Administration-approved polymer poly(lactic-co-glycolic acid) (PLGA). Ac-DEX had higher encapsulation efficiencies for both poly I:C and CpG than PLGA. Furthermore, poly I:C or CpG encapsulated in Ac-DEX also showed, in general, a significantly stronger immunostimulatory response than PLGA and unencapsulated CpG or poly I:C, which was indicated by a higher rate of nitric oxide release and increased levels of cytokines such as TNF-α, IL-6, IL-10, and IFN-γ. Overall, we have illustrated a method for enhancing the delivery of these vaccine adjuvants to further enhance the development of Ac-DEX vaccine formulations.


Subject(s)
Dinucleoside Phosphates/metabolism , Poly I-C/metabolism , Toll-Like Receptors/agonists , Animals , Cell Line , Dextrans/chemistry , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Atomic Force
14.
Biochem Genet ; 51(9-10): 686-97, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23695555

ABSTRACT

The northeastern bulrush, Scirpus ancistrochaetus, is a federally endangered wetland plant species found primarily in Pennsylvania, USA. Data on the population genetic structure of this species are needed by conservation managers to prioritize conservation efforts. In this study, we used two genetic marker systems to examine diversity and structure of this species in populations throughout Pennsylvania. The first simple and inexpensive approach utilized RAPD primers; our second, more detailed approach relied on DNA sequencing of single nucleotide polymorphisms. We found genetic variation using both RAPDs and sequencing and found some overlap in information between the two methods, including clusters of related populations and the identification of a genetically unique population. Future studies will seek to examine variation across the full geographic range of the species.


Subject(s)
Cyperaceae/genetics , DNA, Plant/genetics , Endangered Species , Genetic Variation , Conservation of Natural Resources , Cyperaceae/classification , Genetic Markers , Pennsylvania , Polymorphism, Single Nucleotide , Random Amplified Polymorphic DNA Technique , Sequence Analysis, DNA
15.
J Immunol ; 189(1): 433-43, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22675199

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...