Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Prev Alzheimers Dis ; 10(3): 581-594, 2023.
Article in English | MEDLINE | ID: mdl-37357300

ABSTRACT

BACKGROUND: The results of clinical trials for Alzheimer's disease (AD) patients treated with Intravenous immunoglobulin (IVIG) revealed inconsistency in efficacy. OBJECTIVE: To explore the neuroprotective effects and possible mechanisms of different IVIG in 3xTg-AD mice. METHODS: 3-month-old 3xTg-AD mice were administered intraperitoneally with different IVIG (A/B/C) for 3 months and then the therapeutic effects were observed and tested at 9 months of age. The bioavailability of IVIG and Aß40/42 concentrations in parietotemporal cortex was measured by ELISA. Behavioral tests were performed to examine cognitive functions. Immunohistochemistry was utilized to examine the deposition of Aß, the phosphorylation of tau, the levels of GFAP and Iba-1 in the hippocampus. Proteomics, Luminex assay and parallel reaction monitoring were performed to identify and verify the proteins that showed a marked change in the hippocampus. RESULTS: IVIG-C was more effective than IVIG-A and IVIG-B in counteracting cognitive deficits, ameliorating Aß deposits and tau phosphorylation, attenuating the activation of microglia and astrocytes in the hippocampus and inhibiting the secretion of pro-inflammatory factors. IVIG-C affected innate immunity and suppressed the activation of antigen processing and presentation by MHC class I molecule (APP-MHC-I). CONCLUSION: The efficacy of different IVIG on AD was significantly different, and only IVIG-C has been confirmed to possess significant neuroprotective effects, which are related to the inhibition of APP-MHC-I. IVIG may be a potential therapeutic for AD but further research is needed to evaluate the functional of IVIG before clinical trials of AD treatment.


Subject(s)
Alzheimer Disease , Neuroprotective Agents , Mice , Humans , Animals , Immunoglobulins, Intravenous/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Histocompatibility Antigens Class I/therapeutic use , Antigen Presentation , tau Proteins/metabolism , Mice, Transgenic
2.
Sci Rep ; 11(1): 21358, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725399

ABSTRACT

Respiratory syncytial virus (RSV) is one of the main pathogens associated with lower respiratory tract infections in infants and young children worldwide. Exosomes secreted by antigen presenting cells (APCs) can elicit immune responses by carrying major histocompatibility complex (MHC) class I molecules complexed with antigenic peptides and other co-stimulating factors. Therefore, we developed novel immunomagnetic nanographene particles to sequentially isolate, surface engineer, and release intact dendritic cell (DC) exosomes for use as a potential vaccine platform against RSV. The H-2Db-restricted, immunodominant peptides from RSV (M187-195 and NS161-75) were introduced to MHC-I on DC-derived exosomes to express peptide/MHC-I (pMHC-I) complexes. A mouse model of RSV infection was used to define the immunogenicity of surface engineered exosomes for activating virus-specific immune responses. Ex vivo assays demonstrated that engineered exosomes carrying RSV-specific peptides can elicit interferon-gamma (IFN-γ) production by virus-specific CD8+ T cells isolated from RSV-infected C57BL/6 mice. In vivo assays demonstrated that subcutaneous administration of both M187-195 and NS161-75 engineered exosomes to mice, with or without additional adjuvant, appeared safe and well tolerated, however, did not prime antigen-specific CD8+ T cell responses. Surface engineered exosomes are immunogenic and promising for further development as a vaccine platform.


Subject(s)
Exosomes/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Exosomes/transplantation , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/therapeutic use , Humans , Mice, Inbred C57BL , Peptides/immunology , Peptides/therapeutic use , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/therapeutic use , Viral Proteins/immunology , Viral Proteins/therapeutic use
3.
Clin Transl Sci ; 14(1): 362-372, 2021 01.
Article in English | MEDLINE | ID: mdl-33064927

ABSTRACT

Enzyme replacement with ectonucleotide pyrophosphatase phospodiesterase-1 (ENPP1) eliminates mortality in a murine model of the lethal calcification disorder generalized arterial calcification of infancy. We used protein engineering, glycan optimization, and a novel biomanufacturing platform to enhance potency by using a three-prong strategy. First, we added new N-glycans to ENPP1; second, we optimized pH-dependent cellular recycling by protein engineering of the Fc neonatal receptor; finally, we used a two-step process to improve sialylation by first producing ENPP1-Fc in cells stably transfected with human α-2,6-sialyltransferase (ST6) and further enhanced terminal sialylation by supplementing production with 1,3,4-O-Bu3 ManNAc. These steps sequentially increased the half-life of the parent compound in rodents from 37 hours to ~ 67 hours with an added N-glycan, to ~ 96 hours with optimized pH-dependent Fc recycling, to ~ 204 hours when the therapeutic was produced in ST6-overexpressing cells with 1,3,4-O-Bu3 ManNAc supplementation. The alterations were demonstrated to increase drug potency by maintaining efficacious levels of plasma phosphoanhydride pyrophosphate in ENPP1-deficient mice when the optimized biologic was administered at a 10-fold lower mass dose less frequently than the parent compound-once every 10 days vs. 3 times a week. We believe these improvements represent a general strategy to rationally optimize protein therapeutics.


Subject(s)
Histocompatibility Antigens Class I/therapeutic use , Phosphoric Diester Hydrolases/pharmacology , Protein Engineering , Pyrophosphatases/pharmacology , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/pharmacology , Vascular Calcification/drug therapy , Animals , Area Under Curve , Disease Models, Animal , Enzyme Replacement Therapy/methods , Glycosylation , Half-Life , Histocompatibility Antigens Class I/genetics , Humans , Male , Mice, Transgenic , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/isolation & purification , Phosphoric Diester Hydrolases/therapeutic use , Protein Structure, Tertiary/genetics , Pyrophosphatases/genetics , Pyrophosphatases/isolation & purification , Pyrophosphatases/therapeutic use , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/therapeutic use , Vascular Calcification/genetics
4.
Curr Opin Allergy Clin Immunol ; 20(6): 557-564, 2020 12.
Article in English | MEDLINE | ID: mdl-33044340

ABSTRACT

PURPOSE OF REVIEW: This review describes how plasma is sourced for fractionation into plasma-derived medicinal products (PDMPs), such as immunoglobulin (Ig) together with differences between plasma from whole blood (recovered plasma) and from plasmapheresis (source plasma) in terms of global plasma supply. Specific areas of growth in immunoglobulin use are identified alongside novel therapies, which may reduce demand for some immunoglobulin indications. RECENT FINDINGS: There has been a 6--8% annual growth in immunoglobulin use. Secondary immunodeficiency alongside improved recognition and diagnosis primary immunodeficiency disorders are drivers whereas the novel neonatal Fc receptor inhibitors (FcRni) may reduce demand for some immunomodulatory indications. SUMMARY: There is a significant geographical imbalance in global supply of plasma with 65% collected in the United States. This results in a dependency of other countries on United States supply and argues for both more plasma supply and greater regionally balanced plasma collection. In addition, progress towards a transparent, regulated and well tolerated framework for the coexistence of unpaid and compensated plasma donations is needed as unpaid donation will not be sufficient. These discussions should be informed by the needs of patients for this life-saving therapy, the care of donors and the safety of plasma and PDMPs.


Subject(s)
Histocompatibility Antigens Class I/therapeutic use , Immunoglobulins/therapeutic use , Immunologic Deficiency Syndromes/therapy , Plasma/physiology , Receptors, Fc/therapeutic use , Blood Donors , Humans , Immunomodulation
5.
Acta Trop ; 178: 55-60, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29111138

ABSTRACT

Killer immunoglobulin-like receptors (KIRs) genomic regions have been suggested to influence malaria pathogenesis and infection susceptibility. KIRs are known as activating natural killer (NK) cell receptors, which upon binding to their corresponding human leukocyte antigen (HLA) ligands cause lysis of any infected cell. We have examined the potential association of KIR genes with complicated malaria (CM) among north Indians in this study and further evaluated the KIR receptor-HLA ligand association on the severity of the disease considering the uncomplicated malaria (UCM) subjects as control. Molecular profiling of KIR and HLA was carried out using the PCR-SSP method. Susceptible association was found for individuals possessing KIR2DS2 (OR=1.76, p-value=0.0390), KIR2DL1 (OR=2.87, p-value=0.0005) and KIR2DL3 (OR=2.74, p-value=0.0011) genes with CM. This was supported by the strong linkage disequilibrium observed for 2DS2-2DL2 (D́=0.87, r2=0.54) with CM. Whereas the receptor-ligand association has revealed risk association against KIR2DS2-HLAC1 (OR=2.08, p-value=0.0229), KIR2DL3-HLAC1 (OR=1.79, p-value=0.0301), and KIR2DL1-HLAC2 (OR=2.10, p-value=0.0175) combinations for complicated malaria. The frequency of different KIR genes are more or less similar to that observed in African population showing not much genetic diversity at KIR level in context to malarial infection. In conclusion, our data indicates KIR gene loci differentially influenced the malarial outcome in north Indians and in particular the KIR2DS2 gene appeared to be associated with disease severity.


Subject(s)
Apoptosis/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/therapeutic use , Malaria/therapy , Receptors, KIR/genetics , Receptors, KIR/immunology , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Adult , Female , Gene Frequency , Genetic Variation , Genotype , Humans , India , Male , Middle Aged
6.
Nat Commun ; 8: 15314, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561044
7.
Cancer Immunol Res ; 5(8): 654-665, 2017 08.
Article in English | MEDLINE | ID: mdl-28637877

ABSTRACT

Manipulation of human natural killer (NK) cell repertoires promises more effective strategies for NK cell-based cancer immunotherapy. A subset of highly differentiated NK cells, termed adaptive NK cells, expands naturally in vivo in response to human cytomegalovirus (HCMV) infection, carries unique repertoires of inhibitory killer cell immunoglobulin-like receptors (KIR), and displays strong cytotoxicity against tumor cells. Here, we established a robust and scalable protocol for ex vivo generation and expansion of adaptive NK cells for cell therapy against pediatric acute lymphoblastic leukemia (ALL). Culture of polyclonal NK cells together with feeder cells expressing HLA-E, the ligand for the activating NKG2C receptor, led to selective expansion of adaptive NK cells with enhanced alloreactivity against HLA-mismatched targets. The ex vivo expanded adaptive NK cells gradually obtained a more differentiated phenotype and were specific and highly efficient killers of allogeneic pediatric T- and precursor B-cell acute lymphoblastic leukemia (ALL) blasts, previously shown to be refractory to killing by autologous NK cells and the NK-cell line NK92 currently in clinical testing. Selective expansion of NK cells that express one single inhibitory KIR for self-HLA class I would allow exploitation of the full potential of NK-cell alloreactivity in cancer immunotherapy. In summary, our data suggest that adaptive NK cells may hold utility for therapy of refractory ALL, either as a bridge to transplant or for patients that lack stem cell donors. Cancer Immunol Res; 5(8); 654-65. ©2017 AACR.


Subject(s)
Immunotherapy , Killer Cells, Natural/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, KIR/immunology , Adaptive Immunity , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Child , Cytomegalovirus/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/therapeutic use , Humans , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, KIR/therapeutic use , HLA-E Antigens
8.
J Biol Chem ; 292(20): 8498-8506, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28258216

ABSTRACT

Antibody therapeutics offer effective treatment options for a broad range of diseases. One of the greatest benefits of antibody therapeutics is their extraordinarily long serum half-life, allowing infrequent dosing with long-lasting effects. A characteristic of antibodies that drives long half-life is the ability to interact with the recycling receptor, FcRn, in a pH-dependent manner. The benefit of long half-life, however, carries with it liabilities. Although the positive effects of antibody therapeutics are long-lasting, any acute adverse events or chronic negative impacts, such as immunosuppression in the face of an infection, are also long-lasting. Therefore, we sought to develop antibodies with a chemical handle that alone would enjoy the long half-life of normal antibodies but, upon addition of a small-molecule antidote, would interact with the chemical handle and inhibit the antibody recycling mechanism, thus leading to rapid degradation and shortened half-life in vivo Here we present a proof of concept study where we identify sites to incorporate a non-natural amino acid that can be chemically modified to modulate FcRn interaction in vitro and antibody half-life in vivo This is an important first step in developing safer therapeutics, and the next step will be development of technology that can perform the modifying chemistry in vivo.


Subject(s)
Antibodies/chemistry , Antidotes/chemistry , Histocompatibility Antigens Class I/chemistry , Receptors, Fc/chemistry , Antibodies/therapeutic use , Antidotes/therapeutic use , Histocompatibility Antigens Class I/therapeutic use , Humans , Receptors, Fc/therapeutic use
9.
Pharmacol Ther ; 161: 22-39, 2016 05.
Article in English | MEDLINE | ID: mdl-27016466

ABSTRACT

Advances in the understanding of neonatal Fc receptor (FcRn) biology and function have demonstrated that this receptor, primarily identified for the transfer of passive immunity from mother infant, is involved in several biological and immunological processes. In fact, FcRn is responsible for the long half-life of IgG and albumin in the serum, by creating an intracellular protein reservoir, which is protected from lysosomal degradation and, importantly, trafficked across the cell. Such discovery has led researchers to hypothesize the role for this unique receptor in the controlled delivery of therapeutic agents. A great amount of FcRn-based strategies are already under extensive investigation, in which FcRn reveals to have profound impact on the biodistribution and half-life extension of therapeutic agents. This review summarizes the main findings on FcRn biology, function and distribution throughout different tissues, together with the main advances on the FcRn-based therapeutic opportunities and model systems, which indicate that this receptor is a potential target for therapeutic regimen modification.


Subject(s)
Drug Delivery Systems/methods , Histocompatibility Antigens Class I/physiology , Receptors, Fc/physiology , Albumins/metabolism , Animals , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/therapeutic use , Humans , Immunoglobulin G/metabolism , Molecular Targeted Therapy/methods , Receptors, Fc/biosynthesis , Receptors, Fc/metabolism , Receptors, Fc/therapeutic use , Tissue Distribution
11.
Immunol Cell Biol ; 91(5): 350-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23528729

ABSTRACT

Nanoparticles (NPs) coated with ß-cell-specific peptide major histocompatibility complex (pMHC) class I molecules can effectively restore normoglycemia in spontaneously diabetic nonobese diabetic mice. They do so by expanding pools of cognate memory autoreactive regulatory CD8+ T cells that arise from naive low-avidity T-cell precursors to therapeutic levels. Here we develop our previously constructed mathematical model to explore the effects of compound design parameters (NP dose and pMHC valency) on therapeutic efficacy with the underlying hypothesis that the functional correlates of the therapeutic response (expansion of autoregulatory T cells and deletion of autoantigen-loaded antigen-presenting cells by these T cells) are biphasic. We show, using bifurcation analysis, that the model exhibits a 'resonance'-like behavior for a given range of NP dose in which bistability between the healthy state (possessing zero level of effector T-cell population) and autoimmune state (possessing elevated level of the same population) disappears. A heterogeneous population of model mice subjected to several treatment protocols under these new conditions is conducted to quantify both the average percentage of autoregulatory T cells in responsive and nonresponsive model mice, and the average valency-dependent minimal optimal dose needed for effective therapy. Our results reveal that a moderate increase (≥1.6-fold) in the NP-dependent expansion rate of autoregulatory T-cell population leads to a significant increase in the efficacy and the area corresponding to the effective treatment regimen, provided that NP dose ≥8 µg. We expect the model developed here to generalize to other autoimmune diseases and serve as a computational tool to understand and optimize pMHC-NP-based therapies.


Subject(s)
Autoantigens/administration & dosage , CD8-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/therapy , Drug Design , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/therapeutic use , Models, Theoretical , Nanoparticles/chemistry , Animals , Autoimmune Diseases/therapy , CD8-Positive T-Lymphocytes/immunology , Clinical Protocols , Computational Biology , Diabetes Mellitus, Type 1/immunology , Humans , Hyperglycemia/drug therapy , Hyperglycemia/immunology , Insulin-Secreting Cells/immunology , Mice , Mice, Inbred NOD , Nanoparticles/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
12.
Blood ; 119(13): 3024-30, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22246033

ABSTRACT

Despite proven benefits, prophylactic treatment for hemophilia A is hampered by the short half-life of factor VIII. A recombinant factor VIII-Fc fusion protein (rFVIIIFc) was constructed to determine the potential for reduced frequency of dosing. rFVIIIFc has an ∼ 2-fold longer half-life than rFVIII in hemophilia A (HemA) mice and dogs. The extension of rFVIIIFc half-life requires interaction of Fc with the neonatal Fc receptor (FcRn). In FcRn knockout mice, the extension of rFVIIIFc half-life is abrogated, and is restored in human FcRn transgenic mice. The Fc fusion has no impact on FVIII-specific activity. rFVIIIFc has comparable acute efficacy as rFVIII in treating tail clip injury in HemA mice, and fully corrects whole blood clotting time (WBCT) in HemA dogs immediately after dosing. Furthermore, consistent with prolonged half-life, rFVIIIFc shows 2-fold longer prophylactic efficacy in protecting HemA mice from tail vein transection bleeding induced 24-48 hours after dosing. In HemA dogs, rFVIIIFc also sustains partial correction of WBCT 1.5- to 2-fold longer than rFVIII. rFVIIIFc was well tolerated in both species. Thus, the rescue of FVIII by Fc fusion to provide prolonged protection presents a novel pathway for FVIII catabolism, and warrants further investigation.


Subject(s)
Factor VIII/pharmacokinetics , Hemophilia A/metabolism , Histocompatibility Antigens Class I/pharmacology , Recombinant Fusion Proteins/pharmacokinetics , Animals , Coagulants/pharmacokinetics , Coagulants/therapeutic use , Disease Models, Animal , Dog Diseases/drug therapy , Dog Diseases/metabolism , Dogs , Factor VIII/chemistry , Factor VIII/genetics , Factor VIII/therapeutic use , HEK293 Cells , Half-Life , Hemophilia A/drug therapy , Hemophilia A/pathology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Fc/chemistry , Receptors, Fc/metabolism , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Whole Blood Coagulation Time
14.
BioDrugs ; 25(1): 1-11, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21033767

ABSTRACT

Since the establishment of monoclonal antibody production using hybridoma technology in the mid-1970s, there has been expanding progress and continuous technological improvement in the development of therapeutic antibodies. The initial technological breakthroughs involved reduction of immunogenicity and thus enabled repeated administration. The establishment of chimeric, humanized, and fully human antibodies has led to the great success of several 'second-generation' therapeutic antibodies, such as rituximab, trastuzumab, cetuximab, and bevacizumab. However, there still exists an urgent demand for improvement in the efficacy of the current antibody therapeutics, which is not yet fully satisfactory for patients. Based on the current understanding of the clinical mechanisms of several therapeutic antibodies, many now believe that Fc-mediated functions (e.g. antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and neonatal Fc receptor [FcRn]-mediated storage) will improve the clinical outcomes of therapeutic antibodies. The present review focuses on the recent progress in the development of 'Fc engineering,' which dramatically improves (and sometimes silences) Fc-mediated functions. These achievements can be classified into two technological approaches: (i) introducing amino acid mutations and (ii) modifying Fc-linked oligosaccharide structures. The effectiveness of multiple third-generation therapeutic antibodies armed with various engineered Fcs is now ready to be tested in clinical trials.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Histocompatibility Antigens Class I/genetics , Immunoglobulin Fc Fragments/genetics , Protein Engineering/trends , Receptors, Fc/genetics , Animals , Antibodies, Monoclonal/physiology , Histocompatibility Antigens Class I/physiology , Histocompatibility Antigens Class I/therapeutic use , Humans , Immunoglobulin Fc Fragments/therapeutic use , Mutation/genetics , Protein Engineering/standards , Protein Structure, Secondary/genetics , Receptors, Fc/physiology , Receptors, Fc/therapeutic use
15.
Cell Mol Life Sci ; 68(3): 397-404, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21103908

ABSTRACT

HLA-G plays a particular role during pregnancy in which its expression at the feto-maternal barrier participates into the tolerance of the allogenic foetus. HLA-G has also been demonstrated to be expressed in some transplanted patients, suggesting that it regulates the allogenic response. In vitro data indicate that HLA-G modulates NK cells, T cells, and DC maturation through its interactions with various inhibitory receptors. In this paper, we will review the data reporting the HLA-G involvement of HLA-G in human organ transplantation, then factors that can modulate HLA-G, and finally the use of HLA-G as a therapeutic tool in organ transplantation.


Subject(s)
HLA Antigens/immunology , HLA Antigens/therapeutic use , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/therapeutic use , Gene Expression Regulation , HLA Antigens/genetics , HLA-G Antigens , Histocompatibility Antigens Class I/genetics , Humans , Organ Transplantation
16.
Ann N Y Acad Sci ; 1202: 221-5, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20712796

ABSTRACT

Hepcidin (HAMP) negatively regulates iron absorption, degrading the iron exporter ferroportin at the level of enterocytes and macrophages. We showed that mice with beta-thalassemia intermedia (th3/+) have increased anemia and iron overload. However, their hepcidin expression is relatively low compared to their iron burden. We also showed that the iron metabolism gene Hfe is down-regulated in concert with hepcidin in th3/+ mice. These observations suggest that low hepcidin levels are responsible for abnormal iron absorption in thalassemic mice and that down-regulation of Hfe might be involved in the pathway that controls hepcidin synthesis in beta-thalassemia. Therefore, these studies suggest that increasing hepcidin and/or Hfe expression could be a strategy to reduces iron overload in these animals. The goal of this paper is to review recent findings that correlate hepcidin, Hfe, and iron metabolism in beta-thalassemia and to discuss potential novel therapeutic approaches based on these recent discoveries.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Histocompatibility Antigens Class I/metabolism , Iron Overload/metabolism , Membrane Proteins/metabolism , beta-Thalassemia/metabolism , Anemia/etiology , Anemia/metabolism , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/therapeutic use , Genetic Therapy , Genetic Vectors/genetics , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/therapeutic use , Humans , Iron Overload/etiology , Iron Overload/therapy , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , beta-Thalassemia/complications , beta-Thalassemia/genetics , beta-Thalassemia/therapy
17.
Parkinsonism Relat Disord ; 15 Suppl 3: S189-94, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20082988

ABSTRACT

A large body of evidence from postmortem brain tissue and genetic analysis in humans, as well as biochemical and pathological studies in animal models of neurodegeneration suggest that mitochondrial dysfunction is a key pathological mechanism in Parkinson's Disease (PD). Mitochondrial dysfunction leads to oxidative stress, damage to mitochondrial DNA, mitochondrial DNA deletions, altered mitochondrial morphology, alterations in mitochondrial fission and fusion and ultimately neuronal demise. Therapeutic approaches targeting mitochondrial dysfunction and oxidative damage, therefore, hold great promise in PD. A number of agents, which target energy metabolism, are presently in therapeutic trials in PD. Both creatine and Coenzyme Q10 (CoQ10) are being tested in phase III clinical trials. In addition, preclinical studies in animal models have shown efficacy of mitochondrial-targeted antioxidants and the SS peptides. A promising approach for increasing antioxidant defenses is to transcriptionally increase the activity of the Nrf2/ARE pathway, which activates transcription of anti-inflammatory and antioxidant genes. A number of agents including sulforaphane, curcumin and triterpenoids have been shown to activate this pathway and to produce neuroprotective effects. Lastly, newly identified therapeutic targets include peroxisomal proliferator activated receptor gamma-coactivator (PGC-1alpha) and sirtuins. These pathways provide promise for future therapeutic developments in the treatment of PD.


Subject(s)
Mitochondrial Diseases/etiology , Mitochondrial Diseases/therapy , Parkinson Disease/complications , Animals , Clinical Trials, Phase III as Topic , Creatine/pharmacology , Creatine/therapeutic use , Heat-Shock Proteins/pharmacology , Heat-Shock Proteins/therapeutic use , Histocompatibility Antigens Class I/pharmacology , Histocompatibility Antigens Class I/therapeutic use , Humans , Mitochondrial Diseases/genetics , NF-E2-Related Factor 2/metabolism , Parkinson Disease/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction/drug effects , Transcription Factors/pharmacology , Transcription Factors/therapeutic use
18.
Transplant Proc ; 40(5): 1618-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18589161

ABSTRACT

BACKGROUND: [corrected] The present study was undertaken to determine the role of costimulatory blockade in a murine cardiac transplant model. MATERIALS AND METHODS: We blocked the CD28/B7 and CD154/CD40 costimulatory pathways by transient administration of CTLA4-Ig and MR1 antibody to study the effects on allograft survival time, deviation of Th1 and Th2 cytokine secretion, and other mechanisms related to prolonged survival. RESULTS: Costimulatory blockade prolonged the mean survival time (MST) of cardiac allografts to 43 days for the treated group vs 8 days for the untreated group (P < .01). The costimulatory blockade down-regulated the expression of 2 Th1 cytokines (interferon-gamma [IFN-gamma] and interleukin-2 [IL-2]) and 2 Th2 cytokines (IL-4 and IL-10), reduced the numbers of graft-infiltrating CD4+ and CD8+ lymphocytes, and inhibited the expression of both perforin/GrB and FasL in allografts. CONCLUSIONS: Combined administration of CTLA4-Ig/MR1 inhibited acute rejection reactions in murine cardiac allografts, prolonging the survival of cardiac grafts through several mechanisms, including inhibition of Th1 and Th2 cytokine expression, graft infiltration by CD4+ and CD8+ T lymphocytes, and reduced both perforin/GrB and Fas-FasL.


Subject(s)
Graft Survival/immunology , Heart Transplantation/immunology , Histocompatibility Antigens Class I/therapeutic use , Immunoconjugates/therapeutic use , Immunosuppressive Agents/therapeutic use , Abatacept , Abdomen , Animals , Graft Survival/drug effects , Mice , Minor Histocompatibility Antigens , Recombinant Proteins/therapeutic use , Transplantation, Heterotopic , Transplantation, Homologous
19.
Blood ; 111(10): 4862-70, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18334671

ABSTRACT

Human leukocyte antigen G (HLA-G) is a nonclassic major histocompatibility complex (MHC) class I molecule that functions as an immunomodulatory molecule capable of protecting fetal tissues from the maternal immune system. The relevance of HLA-G in other contexts was investigated soon afterward. Numerous studies have sought (and some have shown) the relevance of HLA-G in pathologic conditions, such as transplantation, autoimmunity, and cancer and hematologic malignancies. One of the main goals of the current research on HLA-G is now to use it in the clinic, either for diagnosis or as a therapeutic tool/target. For this, precise knowledge on the nature and functions of HLA-G is critical. We highlight here what we consider are recent key basic findings on the immunomodulatory function of HLA-G. These strengthen the case for considering HLA-G as clinically relevant.


Subject(s)
HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Immunologic Factors , Female , HLA Antigens/chemistry , HLA Antigens/genetics , HLA Antigens/therapeutic use , HLA-G Antigens , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/therapeutic use , Humans , Immune System/cytology , Killer Cells, Natural/immunology , Male , Pregnancy , Receptors, Immunologic
20.
Trends Immunol ; 29(3): 125-32, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18249584

ABSTRACT

The relevance of the nonclassical human leukocyte antigen (HLA) class I molecule HLA-G in human physiological and pathological contexts has been the center of intense investigation. In light of the recent advances, we report here the clinical implications of HLA-G as a tolerogenic molecule promoting uterine implantation of the embryo or acceptance of solid allografts while allowing the evasion of tumors or viruses from the immune response. These recent findings are important in terms of clinical benefits at both diagnostic and therapeutic levels.


Subject(s)
HLA Antigens/physiology , HLA Antigens/therapeutic use , Histocompatibility Antigens Class I/physiology , Histocompatibility Antigens Class I/therapeutic use , Animals , Embryo Implantation/immunology , HLA-G Antigens , Humans , Transplantation Tolerance/immunology , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...