253 results on '"Lenardo MJ"'
Search Results
52. Metabolically inactive insulin analogue does not prevent autoimmune diabetes in NOD mice.
- Author
-
Grönholm J, Pagni PP, Pham MN, Gibson CB, Macomber PF, Vela JL, von Herrath M, and Lenardo MJ
- Subjects
- Animals, Autoantibodies immunology, CD4-Positive T-Lymphocytes metabolism, Female, Flow Cytometry, Hybridomas metabolism, Insulin analogs & derivatives, Mice, Mice, Inbred NOD, Mice, Transgenic, Diabetes Mellitus, Type 1 immunology, Diabetes Mellitus, Type 1 prevention & control, Insulin therapeutic use
- Abstract
Aims/hypothesis: Insulin is widely considered to be a driver antigen in type 1 diabetes in humans and in mouse models of the disease. Therefore, insulin or insulin analogues are candidates for tolerogenic drugs to prevent disease onset in individuals with risk of diabetes. Previous experiments have shown that autoimmune diabetes can be prevented in NOD mice by repeated doses of insulin administered via an oral, nasal or parenteral route, but clinical trials in humans have not succeeded. The hypoglycaemic activity of insulin is dose-limiting in clinical studies attempting tolerance and disease prevention. Here, we aimed to investigate the therapeutic potential of metabolically inactive insulin analogue (MII) in NOD mice., Methods: The tolerogenic potential of MII to prevent autoimmune diabetes was studied by administering multiple i.v. or s.c. injections of MII to non-diabetic 7-12-week-old female NOD mice in three geographical colony locations. The incidence of diabetes was assessed from daily or weekly blood glucose measurements. The effect of MII on insulin autoantibody levels was studied using an electrochemiluminescence-based insulin autoantibody assay. The effect on the number of insulin-reactive CD8
+ and CD4+ T lymphocytes in peripheral lymphoid tissue was studied with MHC class I and MHC class II tetramers, respectively., Results: We found that twice-weekly s.c. administration of MII accelerates rather than prevents diabetes. High-dose i.v. treatment did not prevent disease or affect insulin autoantibody levels, but it increased the amount of insulin-reactive CD4+ T lymphocytes in peripheral lymphoid tissue., Conclusions/interpretation: Our data suggest that parenteral MII, even when used in high doses, has little or no therapeutic potential in NOD mice and may exacerbate disease.- Published
- 2017
- Full Text
- View/download PDF
53. CD55 Deficiency, Early-Onset Protein-Losing Enteropathy, and Thrombosis.
- Author
-
Ozen A, Comrie WA, Ardy RC, Domínguez Conde C, Dalgic B, Beser ÖF, Morawski AR, Karakoc-Aydiner E, Tutar E, Baris S, Ozcay F, Serwas NK, Zhang Y, Matthews HF, Pittaluga S, Folio LR, Unlusoy Aksu A, McElwee JJ, Krolo A, Kiykim A, Baris Z, Gulsan M, Ogulur I, Snapper SB, Houwen RHJ, Leavis HL, Ertem D, Kain R, Sari S, Erkan T, Su HC, Boztug K, and Lenardo MJ
- Subjects
- CD55 Antigens blood, Child, Child, Preschool, Complement Activation drug effects, Complement Inactivating Agents pharmacology, Female, Homozygote, Humans, Immunoglobulin A blood, Infant, Intestine, Small pathology, Male, Pedigree, Protein-Losing Enteropathies complications, Statistics, Nonparametric, Syndrome, T-Lymphocytes metabolism, CD55 Antigens genetics, Complement Activation genetics, Complement System Proteins metabolism, Mutation, Protein-Losing Enteropathies genetics, Thrombosis genetics
- Abstract
Background: Studies of monogenic gastrointestinal diseases have revealed molecular pathways critical to gut homeostasis and enabled the development of targeted therapies., Methods: We studied 11 patients with abdominal pain and diarrhea caused by early-onset protein-losing enteropathy with primary intestinal lymphangiectasia, edema due to hypoproteinemia, malabsorption, and less frequently, bowel inflammation, recurrent infections, and angiopathic thromboembolic disease; the disorder followed an autosomal recessive pattern of inheritance. Whole-exome sequencing was performed to identify gene variants. We evaluated the function of CD55 in patients' cells, which we confirmed by means of exogenous induction of expression of CD55., Results: We identified homozygous loss-of-function mutations in the gene encoding CD55 (decay-accelerating factor), which lead to loss of protein expression. Patients' T lymphocytes showed increased complement activation causing surface deposition of complement and the generation of soluble C5a. Costimulatory function and cytokine modulation by CD55 were defective. Genetic reconstitution of CD55 or treatment with a complement-inhibitory therapeutic antibody reversed abnormal complement activation., Conclusions: CD55 deficiency with hyperactivation of complement, angiopathic thrombosis, and protein-losing enteropathy (the CHAPLE syndrome) is caused by abnormal complement activation due to biallelic loss-of-function mutations in CD55. (Funded by the National Institute of Allergy and Infectious Diseases and others.).
- Published
- 2017
- Full Text
- View/download PDF
54. BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency.
- Author
-
Afzali B, Grönholm J, Vandrovcova J, O'Brien C, Sun HW, Vanderleyden I, Davis FP, Khoder A, Zhang Y, Hegazy AN, Villarino AV, Palmer IW, Kaufman J, Watts NR, Kazemian M, Kamenyeva O, Keith J, Sayed A, Kasperaviciute D, Mueller M, Hughes JD, Fuss IJ, Sadiyah MF, Montgomery-Recht K, McElwee J, Restifo NP, Strober W, Linterman MA, Wingfield PT, Uhlig HH, Roychoudhuri R, Aitman TJ, Kelleher P, Lenardo MJ, O'Shea JJ, Cooper N, and Laurence ADJ
- Subjects
- Adrenal Cortex Hormones therapeutic use, Adult, Autoimmune Diseases complications, Colitis complications, Colitis genetics, Colitis pathology, Female, Fever complications, Fever drug therapy, Fever genetics, Haploinsufficiency, Heterozygote, Humans, Immunologic Deficiency Syndromes complications, Lymphopenia complications, Lymphopenia genetics, Male, Middle Aged, Mutation, Pancytopenia complications, Pancytopenia drug therapy, Pancytopenia genetics, Pedigree, Polymorphism, Single Nucleotide, Recurrence, Respiratory Tract Infections complications, Respiratory Tract Infections diagnostic imaging, Respiratory Tract Infections genetics, Splenomegaly complications, Splenomegaly genetics, Syndrome, Tomography, X-Ray Computed, Young Adult, Autoimmune Diseases genetics, Basic-Leucine Zipper Transcription Factors genetics, Immunologic Deficiency Syndromes genetics
- Abstract
The transcriptional programs that guide lymphocyte differentiation depend on the precise expression and timing of transcription factors (TFs). The TF BACH2 is essential for T and B lymphocytes and is associated with an archetypal super-enhancer (SE). Single-nucleotide variants in the BACH2 locus are associated with several autoimmune diseases, but BACH2 mutations that cause Mendelian monogenic primary immunodeficiency have not previously been identified. Here we describe a syndrome of BACH2-related immunodeficiency and autoimmunity (BRIDA) that results from BACH2 haploinsufficiency. Affected subjects had lymphocyte-maturation defects that caused immunoglobulin deficiency and intestinal inflammation. The mutations disrupted protein stability by interfering with homodimerization or by causing aggregation. We observed analogous lymphocyte defects in Bach2-heterozygous mice. More generally, we observed that genes that cause monogenic haploinsufficient diseases were substantially enriched for TFs and SE architecture. These findings reveal a previously unrecognized feature of SE architecture in Mendelian diseases of immunity: heterozygous mutations in SE-regulated genes identified by whole-exome/genome sequencing may have greater significance than previously recognized.
- Published
- 2017
- Full Text
- View/download PDF
55. 30 Years of NF-κB: A Blossoming of Relevance to Human Pathobiology.
- Author
-
Zhang Q, Lenardo MJ, and Baltimore D
- Subjects
- Animals, History, 20th Century, History, 21st Century, Humans, Inflammation immunology, Mice, Mutation, NF-kappa B chemistry, NF-kappa B genetics, Signal Transduction, Inflammation metabolism, NF-kappa B history, NF-kappa B metabolism
- Abstract
NF-κB was discovered 30 years ago as a rapidly inducible transcription factor. Since that time, it has been found to have a broad role in gene induction in diverse cellular responses, particularly throughout the immune system. Here, we summarize elaborate regulatory pathways involving this transcription factor and use recent discoveries in human genetic diseases to place specific proteins within their relevant medical and biological contexts., (Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
56. Combined immunodeficiency and Epstein-Barr virus-induced B cell malignancy in humans with inherited CD70 deficiency.
- Author
-
Abolhassani H, Edwards ES, Ikinciogullari A, Jing H, Borte S, Buggert M, Du L, Matsuda-Lennikov M, Romano R, Caridha R, Bade S, Zhang Y, Frederiksen J, Fang M, Bal SK, Haskologlu S, Dogu F, Tacyildiz N, Matthews HF, McElwee JJ, Gostick E, Price DA, Palendira U, Aghamohammadi A, Boisson B, Rezaei N, Karlsson AC, Lenardo MJ, Casanova JL, Hammarström L, Tangye SG, Su HC, and Pan-Hammarström Q
- Subjects
- Adolescent, Adult, CD27 Ligand genetics, CD8-Positive T-Lymphocytes immunology, Child, Cytotoxicity, Immunologic, Female, Herpesvirus 4, Human immunology, Humans, Immunologic Memory, Male, Mutation, Tumor Necrosis Factor Receptor Superfamily, Member 7 physiology, B-Lymphocytes immunology, CD27 Ligand deficiency, Epstein-Barr Virus Infections complications, Hodgkin Disease etiology, Immunologic Deficiency Syndromes complications
- Abstract
In this study, we describe four patients from two unrelated families of different ethnicities with a primary immunodeficiency, predominantly manifesting as susceptibility to Epstein-Barr virus (EBV)-related diseases. Three patients presented with EBV-associated Hodgkin's lymphoma and hypogammaglobulinemia; one also had severe varicella infection. The fourth had viral encephalitis during infancy. Homozygous frameshift or in-frame deletions in CD70 in these patients abolished either CD70 surface expression or binding to its cognate receptor CD27. Blood lymphocyte numbers were normal, but the proportions of memory B cells and EBV-specific effector memory CD8
+ T cells were reduced. Furthermore, although T cell proliferation was normal, in vitro-generated EBV-specific cytotoxic T cell activity was reduced because of CD70 deficiency. This reflected impaired activation by, rather than effects during killing of, EBV-transformed B cells. Notably, expression of 2B4 and NKG2D, receptors implicated in controlling EBV infection, on memory CD8+ T cells from CD70-deficient individuals was reduced, consistent with their impaired killing of EBV-infected cells. Thus, autosomal recessive CD70 deficiency is a novel cause of combined immunodeficiency and EBV-associated diseases, reminiscent of inherited CD27 deficiency. Overall, human CD70-CD27 interactions therefore play a nonredundant role in T and B cell-mediated immunity, especially for protection against EBV and humoral immunity., (© 2017 Abolhassani et al.)- Published
- 2017
- Full Text
- View/download PDF
57. Clinical Genomics - Molecular Pathogenesis Revealed.
- Author
-
Lenardo MJ
- Subjects
- Animals, Gene Expression Regulation, Humans, Infant, Newborn, Male, Receptors, Antigen, T-Cell physiology, Repressor Proteins deficiency, Tumor Suppressor Proteins deficiency, Zebrafish growth & development, Abnormalities, Multiple genetics, Disease Models, Animal, Hematopoietic Stem Cells physiology, Mutation, Missense, Neonatal Screening methods, Repressor Proteins genetics, Severe Combined Immunodeficiency genetics, Tumor Suppressor Proteins genetics
- Published
- 2016
- Full Text
- View/download PDF
58. CHAI and LATAIE: new genetic diseases of CTLA-4 checkpoint insufficiency.
- Author
-
Lo B, Fritz JM, Su HC, Uzel G, Jordan MB, and Lenardo MJ
- Subjects
- Humans, Immune System Diseases therapy, Immunotherapy, Mutation genetics, Phenotype, CTLA-4 Antigen genetics, Cell Cycle Checkpoints genetics, Immune System Diseases genetics
- Abstract
CTLA-4 is a critical inhibitory "checkpoint" molecule of immune activation. Several recent reports have described patients with immune dysregulation and lymphoproliferative disease resulting from 2 different genetic diseases that directly or indirectly cause CTLA-4 deficiency. Numerous articles have also been published describing CTLA-4 blockade in cancer immunotherapy and its side effects, which are ultimately the consequence of treatment-induced CTLA-4 deficiency. Here, we review these 2 diseases and CTLA-4 blockade therapy, emphasizing the crucial role of CTLA-4 in immune checkpoint regulation.
- Published
- 2016
- Full Text
- View/download PDF
59. Life with a Primary Immune Deficiency: a Systematic Synthesis of the Literature and Proposed Research Agenda.
- Author
-
Similuk MN, Wang A, Lenardo MJ, and Erby LH
- Subjects
- Humans, Immunologic Deficiency Syndromes epidemiology, Immunologic Deficiency Syndromes psychology, Quality of Life
- Abstract
Purpose: The clinical immunology literature is punctuated with research on psychosocial dimensions of illness. Studies investigating the lived experiences and stated needs of patients with primary immune deficiencies and their families are essential to improving clinical management and determining the research questions that matter to patients and other stakeholders. Yet, to move the field forward, a systematic review of literature and proposed agenda is needed., Methods: A systematic review was conducted via PubMed and Scopus to include original research on psychological, social, or behavioral aspects of primary immune deficiencies published between 1999 and 2015. A Title/Abstract keyword search was conducted, 317 candidate article abstracts were manually reviewed, and forward/backward reference searches were completed., Results: Twenty-nine studies met inclusion criteria. These illuminate the complex psychological, social, and emotional experiences of primary immune deficiency. Themes included the potential for negative psychosocial impact from disease; adaptation over time; the multi-dimensional assessments of quality of life; familial impact; the important roles of hope, developing a sense of control, social support; and addressing anxiety/depression in our patients and their families. Methodological considerations and areas for improvement are discussed., Conclusion: We propose the research agenda focus on study creativity and rigor, with improved engagement with existing literature and critical study design (e.g., methodology with adequate statistical power, careful variable selection, etc.). This review highlights opportunities to advance psychosocial research and bring a brighter future to clinicians, researchers, and families affected by primary immune deficiency.
- Published
- 2016
- Full Text
- View/download PDF
60. Characterization of a genetically engineered mouse model of hemophilia A with complete deletion of the F8 gene.
- Author
-
Chao BN, Baldwin WH, Healey JF, Parker ET, Shafer-Weaver K, Cox C, Jiang P, Kanellopoulou C, Lollar P, Meeks SL, and Lenardo MJ
- Subjects
- Animals, Antibodies blood, Blood Coagulation Tests, Chromogenic Compounds, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Factor VIII immunology, Factor VIII metabolism, Factor VIII pharmacology, Genetic Predisposition to Disease, Hemophilia A blood, Hemophilia A drug therapy, Hemophilia A immunology, Hemostatics immunology, Hemostatics pharmacology, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Severity of Illness Index, Factor VIII genetics, Gene Deletion, Hemophilia A genetics, Hemostasis drug effects, Hemostasis genetics
- Abstract
Unlabelled: ESSENTIALS: Anti-factor VIII (FVIII) inhibitory antibody formation is a severe complication in hemophilia A therapy. We genetically engineered and characterized a mouse model with complete deletion of the F8 coding region. F8(TKO) mice exhibit severe hemophilia, express no detectable F8 mRNA, and produce FVIII inhibitors. The defined background and lack of FVIII in F8(TKO) mice will aid in studying FVIII inhibitor formation., Background: The most important complication in hemophilia A treatment is the development of inhibitory anti-Factor VIII (FVIII) antibodies in patients after FVIII therapy. Patients with severe hemophilia who express no endogenous FVIII (i.e. cross-reacting material, CRM) have the greatest incidence of inhibitor formation. However, current mouse models of severe hemophilia A produce low levels of truncated FVIII. The lack of a corresponding mouse model hampers the study of inhibitor formation in the complete absence of FVIII protein., Objectives: We aimed to generate and characterize a novel mouse model of severe hemophilia A (designated the F8(TKO) strain) lacking the complete coding sequence of F8 and any FVIII CRM., Methods: Mice were created on a C57BL/6 background using Cre-Lox recombination and characterized using in vivo bleeding assays, measurement of FVIII activity by coagulation and chromogenic assays, and anti-FVIII antibody production using ELISA., Results: All F8 exonic coding regions were deleted from the genome and no F8 mRNA was detected in F8(TKO) mice. The bleeding phenotype of F8(TKO) mice was comparable to E16 mice by measurements of factor activity and tail snip assay. Similar levels of anti-FVIII antibody titers after recombinant FVIII injections were observed between F8(TKO) and E16 mice., Conclusions: We describe a new C57BL/6 mouse model for severe hemophilia A patients lacking CRM. These mice can be directly bred to the many C57BL/6 strains of genetically engineered mice, which is valuable for studying the impact of a wide variety of genes on FVIII inhibitor formation on a defined genetic background., (© 2015 International Society on Thrombosis and Haemostasis.)
- Published
- 2016
- Full Text
- View/download PDF
61. Mitochondrial Protein PGAM5 Regulates Mitophagic Protection against Cell Necroptosis.
- Author
-
Lu W, Sun J, Yoon JS, Zhang Y, Zheng L, Murphy E, Mattson MP, and Lenardo MJ
- Subjects
- Animals, Brain metabolism, Mice, Mice, Knockout, Myocardial Ischemia genetics, Myocardial Ischemia metabolism, Necrosis genetics, Phosphoprotein Phosphatases, Phosphoric Monoester Hydrolases genetics, Reactive Oxygen Species metabolism, Reperfusion Injury genetics, Reperfusion Injury metabolism, Mitophagy physiology, Necrosis metabolism, Phosphoric Monoester Hydrolases metabolism
- Abstract
Necroptosis as a molecular program, rather than simply incidental cell death, was established by elucidating the roles of receptor interacting protein (RIP) kinases 1 and 3, along with their downstream partner, mixed lineage kinase-like domain protein (MLKL). Previous studies suggested that phosphoglycerate mutase family member 5 (PGAM5), a mitochondrial protein that associates with RIP1/RIP3/MLKL complex, promotes necroptosis. We have generated mice deficient in the pgam5 gene and surprisingly found PGAM5-deficiency exacerbated rather than reduced necroptosis in response to multiple in vitro and in vivo necroptotic stimuli, including ischemic reperfusion injury (I/R) in the heart and brain. Electron microscopy, biochemical, and confocal analysis revealed that PGAM5 is indispensable for the process of PINK1 dependent mitophagy which antagonizes necroptosis. The loss of PGAM5/PINK1 mediated mitophagy causes the accumulation of abnormal mitochondria, leading to the overproduction of reactive oxygen species (ROS) that worsen necroptosis. Our results revise the former proposal that PGAM5 acts downstream of RIP1/RIP3 to mediate necroptosis. Instead, PGAM5 protects cells from necroptosis by independently promoting mitophagy. PGAM5 promotion of mitophagy may represent a therapeutic target for stroke, myocardial infarction and other diseases caused by oxidative damage and necroptosis.
- Published
- 2016
- Full Text
- View/download PDF
62. Reprogramming of Polycomb-Mediated Gene Silencing in Embryonic Stem Cells by the miR-290 Family and the Methyltransferase Ash1l.
- Author
-
Kanellopoulou C, Gilpatrick T, Kilaru G, Burr P, Nguyen CK, Morawski A, Lenardo MJ, and Muljo SA
- Subjects
- Animals, Cell Line, Cells, Cultured, DNA-Binding Proteins, Embryonic Stem Cells cytology, Gene Silencing, Genes, Homeobox, Mice, Cellular Reprogramming, Embryonic Stem Cells metabolism, Histone-Lysine N-Methyltransferase genetics, MicroRNAs genetics, Polycomb-Group Proteins metabolism
- Abstract
Members of the miR-290 family are the most abundantly expressed microRNAs (miRNAs) in mouse embryonic stem cells (ESCs). They regulate aspects of differentiation, pluripotency, and proliferation of ESCs, but the molecular program that they control has not been fully delineated. In the absence of Dicer, ESCs fail to express mature miR-290 miRNAs and have selective aberrant overexpression of Hoxa, Hoxb, Hoxc, and Hoxd genes essential for body plan patterning during embryogenesis, but they do not undergo a full differentiation program. Introduction of mature miR-291 into DCR(-/-) ESCs restores Hox gene silencing. This was attributed to the unexpected regulation of Polycomb-mediated gene targeting by miR-291. We identified the methyltransferase Ash1l as a pivotal target of miR-291 mediating this effect. Collectively, our data shed light on the role of Dicer in ESC homeostasis by revealing a facet of molecular regulation by the miR-290 family., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
63. Identifying genetic determinants of autoimmunity and immune dysregulation.
- Author
-
Lucas CL and Lenardo MJ
- Subjects
- Animals, Disease Susceptibility, Gene-Environment Interaction, Genetic Predisposition to Disease, Humans, Immunomodulation genetics, Mendelian Randomization Analysis, Mutation genetics, Penetrance, Polymorphism, Genetic, Autoimmune Diseases genetics, Autoimmunity genetics
- Abstract
Common autoimmune diseases are relatively heterogeneous with both genetic and environmental factors influencing disease susceptibility and progression. As the populations in developed countries age, these chronic diseases will become an increasing burden in human suffering and health care costs. By contrast, rare immune diseases that are severe and develop early in childhood are frequently monogenic and fully penetrant, often with a Mendelian inheritance pattern. Although these may be incompatible with survival or cured by hematopoietic stem cell transplantation, we will argue that they constitute a rich source of genetic insights into immunological diseases. Here, we discuss five examples of well-studied Mendelian disease-causing genes and their known or predicted roles in conferring susceptibility to common, polygenic diseases of autoimmunity. Mendelian disease mutations, as experiments of nature, reveal human loci that are indispensable for immune regulation and, therefore, most promising as therapeutic targets., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
64. Bill Paul: The heart of immunology.
- Author
-
Lenardo MJ and Staudt LM
- Subjects
- Animals, History, 20th Century, History, 21st Century, Humans, Portraits as Topic, Allergy and Immunology history
- Published
- 2015
- Full Text
- View/download PDF
65. Genomics is rapidly advancing precision medicine for immunological disorders.
- Author
-
Zhang Y, Su HC, and Lenardo MJ
- Subjects
- Humans, Immune System Diseases diagnosis, Immune System Diseases therapy, Genomics, Immune System Diseases genetics, Precision Medicine
- Published
- 2015
- Full Text
- View/download PDF
66. AUTOIMMUNE DISEASE. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy.
- Author
-
Lo B, Zhang K, Lu W, Zheng L, Zhang Q, Kanellopoulou C, Zhang Y, Liu Z, Fritz JM, Marsh R, Husami A, Kissell D, Nortman S, Chaturvedi V, Haines H, Young LR, Mo J, Filipovich AH, Bleesing JJ, Mustillo P, Stephens M, Rueda CM, Chougnet CA, Hoebe K, McElwee J, Hughes JD, Karakoc-Aydiner E, Matthews HF, Price S, Su HC, Rao VK, Lenardo MJ, and Jordan MB
- Subjects
- Abatacept, Adaptor Proteins, Signal Transducing genetics, Adolescent, Autoimmune Diseases metabolism, CTLA-4 Antigen genetics, Child, Chloroquine pharmacology, Common Variable Immunodeficiency metabolism, Endosomes metabolism, Female, Forkhead Transcription Factors analysis, Gene Knockdown Techniques, HEK293 Cells, Humans, Lung Diseases, Interstitial drug therapy, Lung Diseases, Interstitial metabolism, Lymphocyte Activation, Lysosomes metabolism, Male, Proteolysis, T-Lymphocytes drug effects, T-Lymphocytes immunology, Young Adult, Adaptor Proteins, Signal Transducing metabolism, Autoimmune Diseases drug therapy, CTLA-4 Antigen deficiency, Common Variable Immunodeficiency drug therapy, Immunoconjugates therapeutic use
- Abstract
Mutations in the LRBA gene (encoding the lipopolysaccharide-responsive and beige-like anchor protein) cause a syndrome of autoimmunity, lymphoproliferation, and humoral immune deficiency. The biological role of LRBA in immunologic disease is unknown. We found that patients with LRBA deficiency manifested a dramatic and sustained improvement in response to abatacept, a CTLA4 (cytotoxic T lymphocyte antigen-4)-immunoglobulin fusion drug. Clinical responses and homology of LRBA to proteins controlling intracellular trafficking led us to hypothesize that it regulates CTLA4, a potent inhibitory immune receptor. We found that LRBA colocalized with CTLA4 in endosomal vesicles and that LRBA deficiency or knockdown increased CTLA4 turnover, which resulted in reduced levels of CTLA4 protein in FoxP3(+) regulatory and activated conventional T cells. In LRBA-deficient cells, inhibition of lysosome degradation with chloroquine prevented CTLA4 loss. These findings elucidate a mechanism for CTLA4 trafficking and control of immune responses and suggest therapies for diseases involving the CTLA4 pathway., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
67. Clinical utility gene card for: X-linked immunodeficiency with magnesium defect, Epstein-Barr virus infection, and neoplasia (XMEN).
- Author
-
Li FY, Chaigne-Delalande B, Rao VK, Zhang Y, Matthews H, Kuijpers TT, Su H, Uzel G, and Lenardo MJ
- Subjects
- Epstein-Barr Virus Infections diagnosis, Female, Humans, Magnesium Deficiency diagnosis, Male, Neoplastic Syndromes, Hereditary diagnosis, X-Linked Combined Immunodeficiency Diseases diagnosis, Cation Transport Proteins genetics, Epstein-Barr Virus Infections genetics, Magnesium Deficiency genetics, Neoplastic Syndromes, Hereditary genetics, X-Linked Combined Immunodeficiency Diseases genetics
- Published
- 2015
- Full Text
- View/download PDF
68. Novel diagnostic and therapeutic approaches for autoimmune diabetes--a prime time to treat insulitis as a disease.
- Author
-
Grönholm J and Lenardo MJ
- Subjects
- Humans, Immunosuppression Therapy, Immunosuppressive Agents therapeutic use, Insulin metabolism, Autoantibodies immunology, Diabetes Mellitus, Type 1 diagnosis, Diabetes Mellitus, Type 1 prevention & control, Immunomodulation drug effects, Insulin-Secreting Cells drug effects, Insulin-Secreting Cells immunology, Lymphocytes immunology
- Abstract
Type 1 diabetes is a progressive autoimmune disease with no curative treatment, making prevention critical. At the time of diagnosis, a majority of the insulin secreting β-cells have already been destroyed. Insulitis, lymphocytic infiltration to the pancreatic islets, is believed to begin months to years before the clinical symptoms of insulin deficiency appear. Insulitis should be treated as its own disease, for it is a known precursor to autoimmune diabetes. Because it is difficult to detect insulitic cellular infiltrates noninvasively, considerable interest has been focused on the levels of islet autoantibodies in blood as measurable diagnostic markers for islet autoimmunity. The traditional islet autoantibody detection assays have many limitations. New electrochemiluminescence-based autoantibody detection assays have the potential to overcome these challenges and they offer promising, cost-effective screening tools in identifying high-risk individuals for trials of preventive interventions. Here, we outline diagnostic and therapeutic strategies to overcome pancreatic β-cell destroying insulitis., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
69. Dual proteolytic pathways govern glycolysis and immune competence.
- Author
-
Lu W, Zhang Y, McDonald DO, Jing H, Carroll B, Robertson N, Zhang Q, Griffin H, Sanderson S, Lakey JH, Morgan NV, Reynard LN, Zheng L, Murdock HM, Turvey SE, Hackett SJ, Prestidge T, Hall JM, Cant AJ, Matthews HF, Koref MF, Simon AK, Korolchuk VI, Lenardo MJ, Hambleton S, and Su HC
- Subjects
- Amino Acid Sequence, Aminopeptidases chemistry, Animals, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases chemistry, Female, Humans, Immunologic Deficiency Syndromes immunology, Lysosomes metabolism, Male, Models, Molecular, Molecular Sequence Data, Pedigree, Sequence Alignment, Serine Endopeptidases chemistry, Adaptive Immunity, Aminopeptidases metabolism, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases metabolism, Glycolysis, Immunity, Innate, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes metabolism, Proteolysis, Serine Endopeptidases metabolism
- Abstract
Proteasomes and lysosomes constitute the major cellular systems that catabolize proteins to recycle free amino acids for energy and new protein synthesis. Tripeptidyl peptidase II (TPPII) is a large cytosolic proteolytic complex that functions in tandem with the proteasome-ubiquitin protein degradation pathway. We found that autosomal recessive TPP2 mutations cause recurrent infections, autoimmunity, and neurodevelopmental delay in humans. We show that a major function of TPPII in mammalian cells is to maintain amino acid levels and that TPPII-deficient cells compensate by increasing lysosome number and proteolytic activity. However, the overabundant lysosomes derange cellular metabolism by consuming the key glycolytic enzyme hexokinase-2 through chaperone-mediated autophagy. This reduces glycolysis and impairs the production of effector cytokines, including IFN-γ and IL-1β. Thus, TPPII controls the balance between intracellular amino acid availability, lysosome number, and glycolysis, which is vital for adaptive and innate immunity and neurodevelopmental health., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
70. Heterozygous splice mutation in PIK3R1 causes human immunodeficiency with lymphoproliferation due to dominant activation of PI3K.
- Author
-
Lucas CL, Zhang Y, Venida A, Wang Y, Hughes J, McElwee J, Butrick M, Matthews H, Price S, Biancalana M, Wang X, Richards M, Pozos T, Barlan I, Ozen A, Rao VK, Su HC, and Lenardo MJ
- Subjects
- Adolescent, Adult, Antibody Formation, Base Sequence, CD8-Positive T-Lymphocytes immunology, Catalytic Domain, Cell Differentiation, Child, Preschool, Class Ia Phosphatidylinositol 3-Kinase, Enzyme Activation, Exons genetics, Female, Heterozygote, Humans, Immunologic Deficiency Syndromes immunology, Lymphoproliferative Disorders enzymology, Lymphoproliferative Disorders immunology, Male, Molecular Sequence Data, Pedigree, Phosphatidylinositol 3-Kinases chemistry, Protein Structure, Tertiary, Sequence Deletion, Signal Transduction, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases metabolism, Telomere metabolism, Alternative Splicing genetics, Genes, Dominant, Immunologic Deficiency Syndromes enzymology, Immunologic Deficiency Syndromes genetics, Lymphoproliferative Disorders genetics, Mutation genetics, Phosphatidylinositol 3-Kinases genetics
- Abstract
Class IA phosphatidylinositol 3-kinases (PI3K), which generate PIP3 as a signal for cell growth and proliferation, exist as an intracellular complex of a catalytic subunit bound to a regulatory subunit. We and others have previously reported that heterozygous mutations in PIK3CD encoding the p110δ catalytic PI3K subunit cause a unique disorder termed p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency (PASLI) disease. We report four patients from three families with a similar disease who harbor a recently reported heterozygous splice site mutation in PIK3R1, which encodes the p85α, p55α, and p50α regulatory PI3K subunits. These patients suffer from recurrent sinopulmonary infections and lymphoproliferation, exhibit hyperactive PI3K signaling, and have prominent expansion and skewing of peripheral blood CD8(+) T cells toward terminally differentiated senescent effector cells with short telomeres. The PIK3R1 splice site mutation causes skipping of an exon, corresponding to loss of amino acid residues 434-475 in the inter-SH2 domain. The mutant p85α protein is expressed at low levels in patient cells and activates PI3K signaling when overexpressed in T cells from healthy subjects due to qualitative and quantitative binding changes in the p85α-p110δ complex and failure of the C-terminal region to properly inhibit p110δ catalytic activity.
- Published
- 2014
- Full Text
- View/download PDF
71. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4.
- Author
-
Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, Schickel JN, Tran DQ, Stoddard J, Zhang Y, Frucht DM, Dumitriu B, Scheinberg P, Folio LR, Frein CA, Price S, Koh C, Heller T, Seroogy CM, Huttenlocher A, Rao VK, Su HC, Kleiner D, Notarangelo LD, Rampertaap Y, Olivier KN, McElwee J, Hughes J, Pittaluga S, Oliveira JB, Meffre E, Fleisher TA, Holland SM, Lenardo MJ, Tangye SG, and Uzel G
- Subjects
- Adult, Animals, B-Lymphocytes immunology, Female, Forkhead Transcription Factors immunology, Humans, Male, Mice, Mice, Mutant Strains, Pedigree, T-Lymphocytes, Regulatory immunology, Young Adult, CTLA-4 Antigen genetics, Germ-Line Mutation, Haploinsufficiency, Immune System Diseases genetics, Immunity genetics
- Abstract
Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an inhibitory receptor found on immune cells. The consequences of mutations in CTLA4 in humans are unknown. We identified germline heterozygous mutations in CTLA4 in subjects with severe immune dysregulation from four unrelated families. Whereas Ctla4 heterozygous mice have no obvious phenotype, human CTLA4 haploinsufficiency caused dysregulation of FoxP3(+) regulatory T (Treg) cells, hyperactivation of effector T cells, and lymphocytic infiltration of target organs. Patients also exhibited progressive loss of circulating B cells, associated with an increase of predominantly autoreactive CD21(lo) B cells and accumulation of B cells in nonlymphoid organs. Inherited human CTLA4 haploinsufficiency demonstrates a critical quantitative role for CTLA-4 in governing T and B lymphocyte homeostasis., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
72. Divalent cation signaling in immune cells.
- Author
-
Chaigne-Delalande B and Lenardo MJ
- Subjects
- Animals, Humans, Immune System Diseases therapy, Molecular Targeted Therapy, Second Messenger Systems immunology, Signal Transduction, Cations, Divalent metabolism, Immune System, Immune System Diseases metabolism
- Abstract
Divalent cations of two alkaline earth metals Ca(2+) and Mg(2+) and the transition metal Zn(2+) play vital roles in the immune system, and several immune disorders are associated with disturbances of their function. Until recently only Ca(2+) was considered to serve as a second messenger. However, signaling roles for Mg(2+) and Zn(2+) have been recently described, leading to a reevaluation of their role as potential second messengers. We review here the roles of these cations as second messengers in light of recent advances in Ca(2+), Mg(2+), and Zn(2+) signaling in the immune system. Developing a better understanding of these signaling cations may lead to new therapeutic strategies for immune disorders., (Copyright © 2014. Published by Elsevier Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
73. XMEN disease: a new primary immunodeficiency affecting Mg2+ regulation of immunity against Epstein-Barr virus.
- Author
-
Li FY, Chaigne-Delalande B, Su H, Uzel G, Matthews H, and Lenardo MJ
- Subjects
- Adult, Epstein-Barr Virus Infections diagnosis, Epstein-Barr Virus Infections genetics, Humans, Magnesium Deficiency diagnosis, Magnesium Deficiency genetics, Neoplasms diagnosis, Neoplasms genetics, Syndrome, X-Linked Combined Immunodeficiency Diseases diagnosis, X-Linked Combined Immunodeficiency Diseases genetics, Epstein-Barr Virus Infections complications, Herpesvirus 4, Human immunology, Immunity, Innate drug effects, Magnesium pharmacology, Magnesium Deficiency complications, Neoplasms complications, X-Linked Combined Immunodeficiency Diseases complications
- Abstract
Epstein-Barr virus (EBV) is an oncogenic gammaherpesvirus that infects and persists in 95% of adults worldwide and has the potential to cause fatal disease, especially lymphoma, in immunocompromised hosts. Primary immunodeficiencies (PIDs) that predispose to EBV-associated malignancies have provided novel insights into the molecular mechanisms of immune defense against EBV. We have recently characterized a novel PID now named "X-linked immunodeficiency with magnesium defect, EBV infection, and neoplasia" (XMEN) disease characterized by loss-of-function mutations in the gene encoding magnesium transporter 1 (MAGT1), chronic high-level EBV with increased EBV-infected B cells, and heightened susceptibility to EBV-associated lymphomas. The genetic etiology of XMEN disease has revealed an unexpected quantitative role for intracellular free magnesium in immune functions and has led to novel diagnostic and therapeutic strategies. Here, we review the clinical presentation, genetic mutation spectrum, molecular mechanisms of pathogenesis, and diagnostic and therapeutic considerations for this previously unrecognized disease.
- Published
- 2014
- Full Text
- View/download PDF
74. Natural history of autoimmune lymphoproliferative syndrome associated with FAS gene mutations.
- Author
-
Price S, Shaw PA, Seitz A, Joshi G, Davis J, Niemela JE, Perkins K, Hornung RL, Folio L, Rosenberg PS, Puck JM, Hsu AP, Lo B, Pittaluga S, Jaffe ES, Fleisher TA, Rao VK, and Lenardo MJ
- Subjects
- Adolescent, Adult, Autoimmune Lymphoproliferative Syndrome pathology, Cell Proliferation, Child, Disease Progression, Female, Follow-Up Studies, Humans, Lymphocytes pathology, Lymphocytes physiology, Male, Middle Aged, Penetrance, Young Adult, Autoimmune Lymphoproliferative Syndrome genetics, Autoimmune Lymphoproliferative Syndrome therapy, Mutation, fas Receptor genetics
- Abstract
Autoimmune lymphoproliferative syndrome (ALPS) presents in childhood with nonmalignant lymphadenopathy and splenomegaly associated with a characteristic expansion of mature CD4 and CD8 negative or double negative T-cell receptor αβ(+) T lymphocytes. Patients often present with chronic multilineage cytopenias due to autoimmune peripheral destruction and/or splenic sequestration of blood cells and have an increased risk of B-cell lymphoma. Deleterious heterozygous mutations in the FAS gene are the most common cause of this condition, which is termed ALPS-FAS. We report the natural history and pathophysiology of 150 ALPS-FAS patients and 63 healthy mutation-positive relatives evaluated in our institution over the last 2 decades. Our principal findings are that FAS mutations have a clinical penetrance of <60%, elevated serum vitamin B12 is a reliable and accurate biomarker of ALPS-FAS, and the major causes of morbidity and mortality in these patients are the overwhelming postsplenectomy sepsis and development of lymphoma. With longer follow-up, we observed a significantly greater relative risk of lymphoma than previously reported. Avoiding splenectomy while controlling hypersplenism by using corticosteroid-sparing treatments improves the outcome in ALPS-FAS patients. This trial was registered at www.clinicaltrials.gov as #NCT00001350.
- Published
- 2014
- Full Text
- View/download PDF
75. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110δ result in T cell senescence and human immunodeficiency.
- Author
-
Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, Avery DT, Moens L, Cannons JL, Biancalana M, Stoddard J, Ouyang W, Frucht DM, Rao VK, Atkinson TP, Agharahimi A, Hussey AA, Folio LR, Olivier KN, Fleisher TA, Pittaluga S, Holland SM, Cohen JI, Oliveira JB, Tangye SG, Schwartzberg PL, Lenardo MJ, and Uzel G
- Subjects
- Antibiotics, Antineoplastic therapeutic use, Cell Differentiation genetics, Cells, Cultured, Class I Phosphatidylinositol 3-Kinases, Cytomegalovirus Infections blood, Cytomegalovirus Infections genetics, Cytomegalovirus Infections virology, Epstein-Barr Virus Infections blood, Epstein-Barr Virus Infections genetics, Epstein-Barr Virus Infections virology, Female, Genes, Dominant, Humans, Immunoblotting, Immunologic Deficiency Syndromes drug therapy, Male, Pedigree, Phosphatidylinositol 3-Kinases chemistry, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, Sirolimus therapeutic use, TOR Serine-Threonine Kinases metabolism, Viremia drug therapy, Viremia genetics, Viremia virology, Cellular Senescence genetics, Germ-Line Mutation, Immunologic Deficiency Syndromes genetics, Phosphatidylinositol 3-Kinases genetics, T-Lymphocytes metabolism
- Abstract
The p110δ subunit of phosphatidylinositol-3-OH kinase (PI(3)K) is selectively expressed in leukocytes and is critical for lymphocyte biology. Here we report fourteen patients from seven families who were heterozygous for three different germline, gain-of-function mutations in PIK3CD (which encodes p110δ). These patients presented with sinopulmonary infections, lymphadenopathy, nodular lymphoid hyperplasia and viremia due to cytomegalovirus (CMV) and/or Epstein-Barr virus (EBV). Strikingly, they had a substantial deficiency in naive T cells but an over-representation of senescent effector T cells. In vitro, T cells from patients exhibited increased phosphorylation of the kinase Akt and hyperactivation of the metabolic checkpoint kinase mTOR, enhanced glucose uptake and terminal effector differentiation. Notably, treatment with rapamycin to inhibit mTOR activity in vivo partially restored the abundance of naive T cells, largely 'rescued' the in vitro T cell defects and improved the clinical course.
- Published
- 2014
- Full Text
- View/download PDF
76. Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D.
- Author
-
Chaigne-Delalande B, Li FY, O'Connor GM, Lukacs MJ, Jiang P, Zheng L, Shatzer A, Biancalana M, Pittaluga S, Matthews HF, Jancel TJ, Bleesing JJ, Marsh RA, Kuijpers TW, Nichols KE, Lucas CL, Nagpal S, Mehmet H, Su HC, Cohen JI, Uzel G, and Lenardo MJ
- Subjects
- Cation Transport Proteins genetics, Cation Transport Proteins metabolism, Humans, NK Cell Lectin-Like Receptor Subfamily K genetics, X-Linked Combined Immunodeficiency Diseases immunology, CD8-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic, Epstein-Barr Virus Infections immunology, Killer Cells, Natural immunology, Magnesium immunology, Magnesium Deficiency immunology, NK Cell Lectin-Like Receptor Subfamily K metabolism
- Abstract
The magnesium transporter 1 (MAGT1) is a critical regulator of basal intracellular free magnesium (Mg(2+)) concentrations. Individuals with genetic deficiencies in MAGT1 have high levels of Epstein-Barr virus (EBV) and a predisposition to lymphoma. We show that decreased intracellular free Mg(2+) causes defective expression of the natural killer activating receptor NKG2D in natural killer (NK) and CD8(+) T cells and impairs cytolytic responses against EBV. Notably, magnesium supplementation in MAGT1-deficient patients restores intracellular free Mg(2+) and NKG2D while concurrently reducing EBV-infected cells in vivo, demonstrating a link between NKG2D cytolytic activity and EBV antiviral immunity in humans. Moreover, these findings reveal a specific molecular function of free basal intracellular Mg(2+) in eukaryotic cells.
- Published
- 2013
- Full Text
- View/download PDF
77. A rapid ex vivo clinical diagnostic assay for fas receptor-induced T lymphocyte apoptosis.
- Author
-
Lo B, Ramaswamy M, Davis J, Price S, Rao VK, Siegel RM, and Lenardo MJ
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Autoimmune Lymphoproliferative Syndrome genetics, Autoimmune Lymphoproliferative Syndrome immunology, Child, Child, Preschool, Flow Cytometry, Humans, Middle Aged, Mutation, Reproducibility of Results, Sensitivity and Specificity, T-Lymphocytes immunology, Young Adult, fas Receptor genetics, Apoptosis immunology, Autoimmune Lymphoproliferative Syndrome diagnosis, Cytotoxicity Tests, Immunologic methods, T-Lymphocytes metabolism, fas Receptor metabolism
- Abstract
Deleterious mutations in genes involved in the Fas apoptosis pathway lead to Autoimmune Lymphoproliferative Syndrome (ALPS). Demonstration of an apoptosis defect is critical for the diagnosis and study of ALPS. The traditional in vitro apoptosis assay, however, requires a week of experimental procedures. Here, we show that defects in Fas-induced apoptosis in PBMCs can be evaluated directly ex vivo using multicolor flow cytometry to analyze the apoptosis of effector memory T cells, a Fas-sensitive subset of PBMCs. This method allowed us to sensitively quantify defective apoptosis in ALPS patients within a few hours. Some ALPS patients (ALPS-sFAS) without germline mutations have somatic mutations in Fas specifically in double-negative αβ T cells (DNTs), an unusual lymphocyte population that is characteristically expanded in ALPS. Since DNTs have been notoriously difficult to culture, defective apoptosis has not been previously demonstrated for ALPS-sFAS patients. Using our novel ex vivo apoptosis assay, we measured Fas-induced apoptosis of DNTs for the first time and found that ALPS-sFAS patients had significant apoptosis defects in these cells compared to healthy controls. Hence, this rapid apoptosis assay can expedite the diagnosis of new ALPS patients, including those with somatic mutations, and facilitate clinical and molecular investigation of these diseases.
- Published
- 2013
- Full Text
- View/download PDF
78. Congenital B cell lymphocytosis explained by novel germline CARD11 mutations.
- Author
-
Snow AL, Xiao W, Stinson JR, Lu W, Chaigne-Delalande B, Zheng L, Pittaluga S, Matthews HF, Schmitz R, Jhavar S, Kuchen S, Kardava L, Wang W, Lamborn IT, Jing H, Raffeld M, Moir S, Fleisher TA, Staudt LM, Su HC, and Lenardo MJ
- Subjects
- Base Sequence, Cluster Analysis, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Gene Expression Profiling, Germ-Line Mutation genetics, High-Throughput Nucleotide Sequencing, Humans, Immunoblotting, Lymphocytosis complications, Microscopy, Confocal, Molecular Sequence Data, Mutation, Missense genetics, NF-kappa B metabolism, Pedigree, Splenomegaly complications, B-Lymphocytes metabolism, CARD Signaling Adaptor Proteins genetics, Genetic Predisposition to Disease genetics, Guanylate Cyclase genetics, Lymphocytosis genetics
- Abstract
Nuclear factor-κB (NF-κB) controls genes involved in normal lymphocyte functions, but constitutive NF-κB activation is often associated with B cell malignancy. Using high-throughput whole transcriptome sequencing, we investigated a unique family with hereditary polyclonal B cell lymphocytosis. We found a novel germline heterozygous missense mutation (E127G) in affected patients in the gene encoding CARD11, a scaffolding protein required for antigen receptor (AgR)-induced NF-κB activation in both B and T lymphocytes. We subsequently identified a second germline mutation (G116S) in an unrelated, phenotypically similar patient, confirming mutations in CARD11 drive disease. Like somatic, gain-of-function CARD11 mutations described in B cell lymphoma, these germline CARD11 mutants spontaneously aggregate and drive constitutive NF-κB activation. However, these CARD11 mutants rendered patient T cells less responsive to AgR-induced activation. By reexamining this rare genetic disorder first reported four decades ago, our findings provide new insight into why activating CARD11 mutations may induce B cell expansion and preferentially predispose to B cell malignancy without dramatically perturbing T cell homeostasis.
- Published
- 2012
- Full Text
- View/download PDF
79. The role of IL-15 in activating STAT5 and fine-tuning IL-17A production in CD4 T lymphocytes.
- Author
-
Pandiyan P, Yang XP, Saravanamuthu SS, Zheng L, Ishihara S, O'Shea JJ, and Lenardo MJ
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Cell Differentiation genetics, Cell Differentiation immunology, Cells, Cultured, Coculture Techniques, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental pathology, Interleukin-15 deficiency, Interleukin-17 antagonists & inhibitors, Interleukin-17 genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Phosphorylation genetics, Phosphorylation immunology, Promoter Regions, Genetic immunology, Signal Transduction genetics, Signal Transduction immunology, Th17 Cells cytology, Up-Regulation genetics, Up-Regulation immunology, Interleukin-15 physiology, Interleukin-17 biosynthesis, STAT5 Transcription Factor metabolism, Th17 Cells immunology, Th17 Cells metabolism
- Abstract
IL-15 is an important IL-2-related cytokine whose role in Th17 cell biology has not been fully elucidated. In this study, we show that exogenous IL-15 decreased IL-17A production in Th17 cultures. Neutralization of IL-15 using an Ab led to increases in IL-17A production in Th17 cultures. Both Il15(-/-) and Il15r(-/-) T cell cultures displayed higher frequency of IL-17A producers and higher amounts of IL-17A in the supernatants compared with those of wild-type (WT) cells in vitro. IL-15 down-modulated IL-17A production independently of retinoic acid-related orphan receptor-γt, Foxp3, and IFN-γ expression. Both Th17 cells and APCs produced IL-15, which induced binding of STAT5, an apparent repressor to the Il17 locus in CD4 T cells. Also, in a model of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE), Il15(-/-) mice displayed exacerbated inflammation-correlating with increased IL-17A production by their CD4(+) T cells-compared with WT controls. Exogenous IL-15 administration and IL-17A neutralization reduced the severity of EAE in Il15(-/-) mice. Taken together, these data indicate that IL-15 has a negative regulatory role in fine-tuning of IL-17A production and Th17-mediated inflammation.
- Published
- 2012
- Full Text
- View/download PDF
80. The role of LRRK2 in inflammatory bowel disease.
- Author
-
Liu Z and Lenardo MJ
- Subjects
- Animals, Colitis genetics, Colitis metabolism, Humans, Inflammatory Bowel Diseases genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Protein Serine-Threonine Kinases genetics, Inflammatory Bowel Diseases metabolism, Protein Serine-Threonine Kinases metabolism
- Published
- 2012
- Full Text
- View/download PDF
81. Antibodies against insulin measured by electrochemiluminescence predicts insulitis severity and disease onset in non-obese diabetic mice and can distinguish human type 1 diabetes status.
- Author
-
Lo B, Swafford AD, Shafer-Weaver KA, Jerome LF, Rakhlin L, Mathern DR, Callahan CA, Jiang P, Davison LJ, Stevens HE, Lucas CL, White J, von Borstel R, Todd JA, and Lenardo MJ
- Subjects
- Animals, Autoantibodies blood, Diabetes Mellitus, Type 1 pathology, Disease Progression, Female, Humans, Insulin-Secreting Cells metabolism, Mice, Mice, Inbred NOD, ROC Curve, Radioligand Assay, Reproducibility of Results, Sensitivity and Specificity, Diabetes Mellitus, Type 1 blood, Diabetes Mellitus, Type 1 diagnosis, Electrochemistry methods, Insulin Antibodies blood, Insulin-Secreting Cells pathology, Luminescent Measurements methods
- Abstract
Background: The detection of insulin autoantibodies (IAA) aids in the prediction of autoimmune diabetes development. However, the long-standing, gold standard 125I-insulin radiobinding assay (RBA) has low reproducibility between laboratories, long sample processing times and requires the use of newly synthesized radiolabeled insulin for each set of assays. Therefore, a rapid, non-radioactive, and reproducible assay is highly desirable., Methods: We have developed electrochemiluminescence (ECL)-based assays that fulfill these criteria in the measurement of IAA and anti-insulin antibodies (IA) in non-obese diabetic (NOD) mice and in type 1 diabetic individuals, respectively. Using the murine IAA ECL assay, we examined the correlation between IAA, histopathological insulitis, and blood glucose in a cohort of female NOD mice from 4 up to 36 weeks of age. We developed a human IA ECL assay that we compared to conventional RBA and validated using samples from 34 diabetic and 59 non-diabetic individuals in three independent laboratories., Results: Our ECL assays were rapid and sensitive with a broad dynamic range and low background. In the NOD mouse model, IAA levels measured by ECL were positively correlated with insulitis severity, and the values measured at 8-10 weeks of age were predictive of diabetes onset. Using human serum and plasma samples, our IA ECL assay yielded reproducible and accurate results with an average sensitivity of 84% at 95% specificity with no statistically significant difference between laboratories., Conclusions: These novel, non-radioactive ECL-based assays should facilitate reliable and fast detection of antibodies to insulin and its precursors sera and plasma in a standardized manner between laboratories in both research and clinical settings. Our next step is to evaluate the human IA assay in the detection of IAA in prediabetic subjects or those at risk of type 1 diabetes and to develop similar assays for other autoantibodies that together are predictive for the diagnosis of this common disorder, in order to improve prediction and facilitate future therapeutic trials.
- Published
- 2011
- Full Text
- View/download PDF
82. The molecular mechanisms of regulatory T cell immunosuppression.
- Author
-
Pandiyan P, Zheng L, and Lenardo MJ
- Abstract
CD4⁺CD25⁺Foxp3⁺ T lymphocytes, known as regulatory T cells or T(regs), have been proposed to be a lineage of professional immune suppressive cells that exclusively counteract the effects of the immunoprotective "helper" and "cytotoxic" lineages of T lymphocytes. Here we discuss new concepts on the mechanisms and functions of T(regs). There are several key points we emphasize: 1. Tregs exert suppressive effects both directly on effector T cells and indirectly through antigen-presenting cells; 2. Regulation can occur through a novel mechanism of cytokine consumption to regulate as opposed to the usual mechanism of cytokine/chemokine production; 3. In cases where CD4⁺ effector T cells are directly inhibited by T(regs), it is chiefly through a mechanism of lymphokine withdrawal apoptosis leading to polyclonal deletion; and 4. Contrary to the current view, we discuss new evidence that T(regs), similar to other T-cells lineages, can promote protective immune responses in certain infectious contexts (Chen et al., 2011; Pandiyan et al., 2011). Although these points are at variance to varying degrees with the standard model of T(reg) behavior, we will recount developing findings that support these new concepts.
- Published
- 2011
- Full Text
- View/download PDF
83. The kinase LRRK2 is a regulator of the transcription factor NFAT that modulates the severity of inflammatory bowel disease.
- Author
-
Liu Z, Lee J, Krummey S, Lu W, Cai H, and Lenardo MJ
- Subjects
- Active Transport, Cell Nucleus, Animals, Cell Line, Colitis chemically induced, Colitis genetics, Colitis immunology, Crohn Disease genetics, Disease Models, Animal, Genetic Predisposition to Disease, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2, Macrophage Activation genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Processing, Post-Translational immunology, Protein Serine-Threonine Kinases genetics, RNA, Long Noncoding, Transgenes genetics, Cell Nucleus metabolism, Colitis metabolism, NFATC Transcription Factors metabolism, Protein Serine-Threonine Kinases metabolism, RNA, Untranslated metabolism
- Abstract
Leucine-rich repeat kinase 2 (LRRK2) has been identified by genome-wide association studies as being encoded by a major susceptibility gene for Crohn's disease. Here we found that LRRK2 deficiency conferred enhanced susceptibility to experimental colitis in mice. Mechanistic studies showed that LRRK2 was a potent negative regulator of the transcription factor NFAT and was a component of a complex that included the large noncoding RNA NRON (an NFAT repressor). Furthermore, the risk-associated allele encoding LRRK2 Met2397 identified by a genome-wide association study for Crohn's disease resulted in less LRRK2 protein post-translationally. Severe colitis in LRRK2-deficient mice was associated with enhanced nuclear localization of NFAT1. Thus, our study defines a new step in the control of NFAT activation that involves an immunoregulatory function of LRRK2 and has important implications for inflammatory bowel disease.
- Published
- 2011
- Full Text
- View/download PDF
84. Loss of MAGT1 abrogates the Mg2+ flux required for T cell signaling and leads to a novel human primary immunodeficiency.
- Author
-
Li FY, Lenardo MJ, and Chaigne-Delalande B
- Subjects
- Calcium metabolism, Cation Transport Proteins metabolism, Humans, Primary Immunodeficiency Diseases, Cation Transport Proteins deficiency, Immunologic Deficiency Syndromes immunology, Magnesium metabolism, Signal Transduction immunology, T-Lymphocytes immunology
- Abstract
Although Mg(2+) has a well-recognized role as an essential cofactor for all ATP-binding enzymes, its role as a signaling ion, like Ca(2+), has been controversial. A requirement for Mg(2+)for optimal T lymphocyte stimulation was demonstrated more than 30 years ago, but the mechanism of its synergistic effect with Ca(2+)in T cell activation remains elusive. Here, we summarize our recent discovery of a signaling role for Mg(2+)in the T cell antigen receptor (TCR) signaling pathway from the study of a novel primary immunodeficiency, now named X-linked immunodeficiency with Mg(2+)defect, EBV infection and neoplasia (XMEN). XMEN patients were found to have a deficiency in magnesium transporter 1 (MAGT1), an Mg(2+)-specific transporter, which leads to the absence of a TCR-stimulated Mg(2+)flux and an attenuation of T cell activation. We further showed that this Mg(2+)flux is required proximally for the temporal orchestration of phospholipase C-γ1 (PLCγ1) activation. Thus, our study not only provides a second messenger role for Mg(2+)to explain its synergism with calcium in T cell signaling, it also identifies a potential extracellular therapeutic target for T cell-specific immunomodulation.
- Published
- 2011
- Full Text
- View/download PDF
85. Second messenger role for Mg2+ revealed by human T-cell immunodeficiency.
- Author
-
Li FY, Chaigne-Delalande B, Kanellopoulou C, Davis JC, Matthews HF, Douek DC, Cohen JI, Uzel G, Su HC, and Lenardo MJ
- Subjects
- Calcium immunology, Cation Transport Proteins genetics, Female, Gene Knockdown Techniques, HEK293 Cells, Humans, Male, Phospholipase C gamma genetics, Phospholipase C gamma metabolism, T-Lymphocytopenia, Idiopathic CD4-Positive genetics, Magnesium immunology, Second Messenger Systems immunology, T-Lymphocytes immunology, T-Lymphocytopenia, Idiopathic CD4-Positive immunology
- Abstract
The magnesium ion, Mg(2+), is essential for all life as a cofactor for ATP, polyphosphates such as DNA and RNA, and metabolic enzymes, but whether it plays a part in intracellular signalling (as Ca(2+) does) is unknown. Here we identify mutations in the magnesium transporter gene, MAGT1, in a novel X-linked human immunodeficiency characterized by CD4 lymphopenia, severe chronic viral infections, and defective T-lymphocyte activation. We demonstrate that a rapid transient Mg(2+) influx is induced by antigen receptor stimulation in normal T cells and by growth factor stimulation in non-lymphoid cells. MAGT1 deficiency abrogates the Mg(2+) influx, leading to impaired responses to antigen receptor engagement, including defective activation of phospholipase Cγ1 and a markedly impaired Ca(2+) influx in T cells but not B cells. These observations reveal a role for Mg(2+) as an intracellular second messenger coupling cell-surface receptor activation to intracellular effectors and identify MAGT1 as a possible target for novel therapeutics.
- Published
- 2011
- Full Text
- View/download PDF
86. Human immunodeficiency virus type 1 Vif causes dysfunction of Cdk1 and CyclinB1: implications for cell cycle arrest.
- Author
-
Sakai K, Barnitz RA, Chaigne-Delalande B, Bidère N, and Lenardo MJ
- Subjects
- CDC2 Protein Kinase antagonists & inhibitors, Cyclin B1 antagonists & inhibitors, HIV-1, Humans, Jurkat Cells, T-Lymphocytes physiology, T-Lymphocytes virology, CDC2 Protein Kinase metabolism, Cell Cycle, Cyclin B1 metabolism, Virulence Factors metabolism, vif Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
The two major cytopathic factors in human immunodeficiency virus type 1 (HIV-1), the accessory proteins viral infectivity factor (Vif) and viral protein R (Vpr), inhibit cell-cycle progression at the G2 phase of the cell cycle. Although Vpr-induced blockade and the associated T-cell death have been well studied, the molecular mechanism of G2 arrest by Vif remains undefined. To elucidate how Vif induces arrest, we infected synchronized Jurkat T-cells and examined the effect of Vif on the activation of Cdk1 and CyclinB1, the chief cell-cycle factors for the G2 to M phase transition. We found that the characteristic dephosphorylation of an inhibitory phosphate on Cdk1 did not occur in infected cells expressing Vif. In addition, the nuclear translocation of Cdk1 and CyclinB1 was disregulated. Finally, Vif-induced cell cycle arrest was correlated with proviral expression of Vif. Taken together, our results suggest that Vif impairs mitotic entry by interfering with Cdk1-CyclinB1 activation.
- Published
- 2011
- Full Text
- View/download PDF
87. Spinster is required for autophagic lysosome reformation and mTOR reactivation following starvation.
- Author
-
Rong Y, McPhee CK, Deng S, Huang L, Chen L, Liu M, Tracy K, Baehrecke EH, Yu L, and Lenardo MJ
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Carbohydrate Metabolism, Drosophila genetics, Drosophila metabolism, Drosophila Proteins antagonists & inhibitors, Drosophila Proteins genetics, Gene Knockdown Techniques, Genes, Insect, Humans, Lysosomal-Associated Membrane Protein 1 metabolism, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Mice, Molecular Sequence Data, Mutation, RNA Interference, Rats, Sequence Homology, Amino Acid, Autophagy physiology, Drosophila Proteins metabolism, Lysosomes metabolism, Membrane Proteins metabolism, TOR Serine-Threonine Kinases metabolism
- Abstract
Autophagy is a conserved cellular process to degrade and recycle cytoplasmic components. During autophagy, lysosomes fuse with an autophagosome to form an autolysosome. Sequestered components are degraded by lysosomal hydrolases and presumably released into the cytosol by lysosomal efflux permeases. Following starvation-induced autophagy, lysosome homeostasis is restored by autophagic lysosome reformation (ALR) requiring activation of the "target of rapamycin" (TOR) kinase. Spinster (Spin) encodes a putative lysosomal efflux permease with the hallmarks of a sugar transporter. Drosophila spin mutants accumulate lysosomal carbohydrates and enlarged lysosomes. Here we show that defects in spin lead to the accumulation of enlarged autolysosomes. We find that spin is essential for mTOR reactivation and lysosome reformation following prolonged starvation. Further, we demonstrate that the sugar transporter activity of Spin is essential for ALR.
- Published
- 2011
- Full Text
- View/download PDF
88. IKKβ phosphorylation regulates RPS3 nuclear translocation and NF-κB function during infection with Escherichia coli strain O157:H7.
- Author
-
Wan F, Weaver A, Gao X, Bern M, Hardwidge PR, and Lenardo MJ
- Subjects
- Active Transport, Cell Nucleus, Amino Acid Sequence, Animals, Cell Nucleus metabolism, Escherichia coli Infections genetics, Escherichia coli Infections metabolism, Escherichia coli Infections virology, Escherichia coli O157 genetics, Escherichia coli O157 metabolism, Escherichia coli O157 physiology, Escherichia coli Proteins genetics, HEK293 Cells, HeLa Cells, Host-Pathogen Interactions, Humans, I-kappa B Kinase genetics, Immunoblotting, Jurkat Cells, Molecular Sequence Data, Mutation, Phosphorylation, Protein Binding, RNA Interference, Ribosomal Proteins genetics, Sequence Homology, Amino Acid, Serine genetics, Serine metabolism, Swine, Escherichia coli Proteins metabolism, I-kappa B Kinase metabolism, NF-kappa B metabolism, Ribosomal Proteins metabolism
- Abstract
NF-κB is a major gene regulator in immune responses, and ribosomal protein S3 (RPS3) is an NF-κB subunit that directs specific gene transcription. However, it is unknown how nuclear translocation of RPS3 is regulated. Here we report that phosphorylation of RPS3 Ser209 by the kinase IKKβ was crucial for nuclear localization of RPS3 in response to activating stimuli. Moreover, virulence protein NleH1 of the foodborne pathogen Escherichia coli strain O157:H7 specifically inhibited phosphorylation of RPS3 Ser209 and blocked RPS3 function, thereby promoting bacterial colonization and diarrhea but resulting in less mortality in a gnotobiotic piglet-infection model. Thus, the IKKβ-dependent modification of a specific amino acid in RPS3 promoted specific NF-κB functions that underlie the molecular pathogenetic mechanisms of E. coli O157:H7.
- Published
- 2011
- Full Text
- View/download PDF
89. CD4(+)CD25(+)Foxp3(+) regulatory T cells promote Th17 cells in vitro and enhance host resistance in mouse Candida albicans Th17 cell infection model.
- Author
-
Pandiyan P, Conti HR, Zheng L, Peterson AC, Mathern DR, Hernández-Santos N, Edgerton M, Gaffen SL, and Lenardo MJ
- Subjects
- Animals, Candida albicans immunology, Cell Differentiation, Disease Models, Animal, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Th17 Cells cytology, CD4 Antigens immunology, Candidiasis immunology, Forkhead Transcription Factors immunology, Immunity, Innate, Interleukin-2 Receptor alpha Subunit immunology, T-Lymphocytes, Regulatory immunology, Th17 Cells immunology
- Abstract
Th17 cells and CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells are thought to promote and suppress inflammatory responses, respectively. Here we explore why under Th17 cell polarizing conditions, Treg cells did not suppress, but rather upregulated, the expression of interleukin-17A (IL-17A), IL-17F, and IL-22 from responding CD4(+) T cells (Tresp cells). Upregulation of IL-17 cytokines in Tresp cells was dependent on consumption of IL-2 by Treg cells, especially at early time points both in vitro and in vivo. During an oral Candida albicans infection in mice, Treg cells induced IL-17 cytokines in Tresp cells, which markedly enhanced fungal clearance and recovery from infection. These findings show how Treg cells can promote acute Th17 cell responses to suppress mucosal fungus infections and reveal that Treg cells have a powerful capability to fight infections besides their role in maintaining tolerance or immune homeostasis., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
90. An allele of IKZF1 (Ikaros) conferring susceptibility to childhood acute lymphoblastic leukemia protects against type 1 diabetes.
- Author
-
Swafford AD, Howson JM, Davison LJ, Wallace C, Smyth DJ, Schuilenburg H, Maisuria-Armer M, Mistry T, Lenardo MJ, and Todd JA
- Subjects
- Adult, Alleles, Case-Control Studies, Female, Genotype, Humans, Logistic Models, Male, Odds Ratio, Polymorphism, Single Nucleotide, White People genetics, Diabetes Mellitus, Type 1 genetics, Genetic Predisposition to Disease, Ikaros Transcription Factor genetics
- Abstract
Objective: IKZF1 encoding Ikaros, an essential regulator of lymphopoiesis and immune homeostasis, has been implicated in the development of childhood acute lymphoblastic leukemia (C-ALL). Because recent genome-wide association (GWA) studies have linked a region of the 3'-UTR of IKZF1 with C-ALL susceptibility, we tested whether IKZF1 is associated with the autoimmune disease type 1 diabetes., Research Design and Methods: rs10272724 (T>C) near IKZF1 at 7p12 was genotyped in 8,333 individuals with type 1 diabetes, 9,947 control subjects, and 3,997 families of European ancestry. Association was tested using logistic regression in the case-control data and by the transmission disequilibrium test in the families. Expression data for IKZF1 by rs10272724 genotype were obtained using quantitative PCR of mRNA/cDNA generated from peripheral blood mononuclear cells from 88 individuals, whereas expression data for five other neighboring genes were obtained from the online Genevar dataset., Results: The minor allele of rs10272724 (C) was found to be protective from type 1 diabetes (odds ratio 0.87 [95% CI 0.83-0.91]; P = 1.1 × 10(-11)). rs10272724 was not correlated with levels of two transcripts of IKZF1 in peripheral blood mononuclear cells., Conclusions: The major susceptibility genotype for C-ALL confers protection from type 1 diabetes. Our finding strengthens the link between autoimmunity and lymphoid cancers. Further investigation is warranted for the genetic effect marked by rs10272724, its impact on IKZF1, and the role of Ikaros and other family members, Ailios (IKZF3) and Eos (IKZF4), in autoimmunity.
- Published
- 2011
- Full Text
- View/download PDF
91. Exposed hydrophobic residues in human immunodeficiency virus type 1 Vpr helix-1 are important for cell cycle arrest and cell death.
- Author
-
Barnitz RA, Chaigne-Delalande B, Bolton DL, and Lenardo MJ
- Subjects
- Cell Nucleus genetics, Cell Nucleus metabolism, Cell Nucleus virology, Cells, Cultured, HIV Infections genetics, HIV Infections metabolism, HIV Infections virology, HIV-1, Humans, Immunoblotting, Mutation genetics, Protein Structure, Secondary, vpr Gene Products, Human Immunodeficiency Virus genetics, Apoptosis, Cell Division physiology, G2 Phase physiology, Hydrophobic and Hydrophilic Interactions, Virion physiology, vpr Gene Products, Human Immunodeficiency Virus chemistry, vpr Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
The human immunodeficiency virus type 1 (HIV-1) accessory protein viral protein R (Vpr) is a major determinant for virus-induced G2/M cell cycle arrest and cytopathicity. Vpr is thought to perform these functions through the interaction with partner proteins. The NMR structure of Vpr revealed solvent exposed hydrophobic amino acids along helices 1 and 3 of Vpr, which could be putative protein binding domains. We previously showed that the hydrophobic patch along helix-3 was important for G2/M blockade and cytopathicity. Mutations of the exposed hydrophobic residues along helix-1 were found to reduce Vpr-induced cell cycle arrest and cell death as well. The levels of toxicity during virion delivery of Vpr correlated with G2/M arrest. Thus, the exposed hydrophobic amino acids in the amino-terminal helix-1 are important for the cell cycle arrest and cytopathicity functions of Vpr.
- Published
- 2011
- Full Text
- View/download PDF
92. Gene defects in the soma: some get it and some don't!
- Author
-
Lo B and Lenardo MJ
- Subjects
- Autoimmune Lymphoproliferative Syndrome immunology, Genetic Variation, Humans, Lymphocytes, Null immunology, Models, Genetic, Penetrance, Protein Structure, Tertiary, Signal Transduction genetics, fas Receptor chemistry, fas Receptor genetics, Autoimmune Lymphoproliferative Syndrome genetics, Mutation
- Abstract
Advances in DNA sequencing technologies have increased attention on genetic variation in somatic tissues. Although long known to cause neoplastic diseases, somatic variation is now being investigated as a pathogenetic mechanism for other diseases. Somatic changes are genomic DNA variations that were not inherited but arise in tissues throughout life. In this issue of the JCI, Magerus-Chatinet et al. explore somatic changes in patients with autoimmune lymphoproliferative syndrome (ALPS), a congenital disease of defective apoptosis and autoimmunity that is usually associated with germline heterozygous mutations in the gene encoding the Fas death receptor. They explain why certain individuals have severe disease manifestations by documenting somatic alterations in the germline normal FAS allele in an unusual population of "double-negative" T cells found in ALPS. Thus, the oncological concept of somatic loss of heterozygosity leading to selected cell expansion also applies to autoimmune diseases.
- Published
- 2011
- Full Text
- View/download PDF
93. Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop.
- Author
-
Oliveira JB, Bleesing JJ, Dianzani U, Fleisher TA, Jaffe ES, Lenardo MJ, Rieux-Laucat F, Siegel RM, Su HC, Teachey DT, and Rao VK
- Subjects
- Autoimmune Lymphoproliferative Syndrome classification, Child, Humans, United States, Autoimmune Lymphoproliferative Syndrome diagnosis
- Abstract
Lymphadenopathy in children for which no infectious or malignant cause can be ascertained constitutes a challenging diagnostic dilemma. Autoimmune lymphoproliferative syndrome (ALPS) is a human genetic disorder of lymphocyte apoptosis resulting in an accumulation of lymphocytes and childhood onset chronic lymphadenopathy, splenomegaly, multilineage cytopenias, and an increased risk of B-cell lymphoma. In 1999, investigators at the National Institutes of Health (NIH) suggested criteria to establish the diagnosis of ALPS. Since then, with approximately 500 patients with ALPS studied worldwide, significant advances in our understanding of the disease have prompted the need for revisions to the existing diagnostic criteria and classification scheme. The rationale and recommendations outlined here stem from an international workshop held at NIH on September 21 and 22, 2009, attended by investigators from the United States, Europe, and Australia engaged in clinical and basic science research on ALPS and related disorders. It is hoped that harmonizing the diagnosis and classification of ALPS will foster collaborative research and better understanding of the pathogenesis of autoimmune cytopenias and B-cell lymphomas.
- Published
- 2010
- Full Text
- View/download PDF
94. The power and the promise of restimulation-induced cell death in human immune diseases.
- Author
-
Snow AL, Pandiyan P, Zheng L, Krummey SM, and Lenardo MJ
- Subjects
- Animals, Apoptosis Regulatory Proteins metabolism, Bcl-2-Like Protein 11, Genetic Diseases, X-Linked, Humans, Lymphoproliferative Disorders genetics, Membrane Proteins metabolism, Models, Immunological, Proto-Oncogene Proteins metabolism, T-Lymphocytes metabolism, fas Receptor metabolism, Apoptosis immunology, Lymphoproliferative Disorders immunology, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
Controlled expansion and contraction of lymphocytes both during and after an adaptive immune response are imperative to sustain a healthy immune system. Both extrinsic and intrinsic pathways of lymphocyte apoptosis are programmed to eliminate cells at the proper time to ensure immune homeostasis. Genetic disorders of apoptosis described in mice and humans have established Fas and Bim as critical pro-apoptotic molecules responsible for T-cell death in response to T-cell receptor restimulation and cytokine withdrawal, respectively. Emerging evidence prompts revision of this classic paradigm, especially for our understanding of restimulation-induced cell death (RICD) and its physiological purpose. Recent work indicates that RICD employs both Fas and Bim for T-cell deletion, dispelling the notion that these molecules are assigned to mutually exclusive apoptotic pathways. Furthermore, new mouse model data combined with our discovery of defective RICD in X-linked lymphoproliferative disease (XLP) patient T cells suggest that RICD is essential for precluding excess T-cell accumulation and associated immunopathology during the course of certain infections. Here, we review how these advances offer a refreshing new perspective on the phenomenon of T-cell apoptosis induced through antigen restimulation, including its relevance to immune homeostasis and potential for therapeutic interventions.
- Published
- 2010
- Full Text
- View/download PDF
95. Protein kinase A phosphorylation activates Vpr-induced cell cycle arrest during human immunodeficiency virus type 1 infection.
- Author
-
Barnitz RA, Wan F, Tripuraneni V, Bolton DL, and Lenardo MJ
- Subjects
- Cell Line, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Humans, Lymphocytes virology, Phosphorylation, Serine metabolism, Cell Cycle, Cyclic AMP-Dependent Protein Kinases metabolism, HIV-1 pathogenicity, vpr Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
Infection with human immunodeficiency virus type 1 (HIV-1) causes an inexorable depletion of CD4(+) T cells. The loss of these cells is particularly pronounced in the mucosal immune system during acute infection, and the data suggest that direct viral cytopathicity is a major factor. Cell cycle arrest caused by the HIV-1 accessory protein Vpr is strongly correlated with virus-induced cell death, and phosphorylation of Vpr serine 79 (S79) is required to activate G(2)/M cell cycle blockade. However, the kinase responsible for phosphorylating Vpr remains unknown. Our bioinformatic analyses revealed that S79 is part of a putative phosphorylation site recognized by protein kinase A (PKA). We show here that PKA interacts with Vpr and directly phosphorylates S79. Inhibition of PKA activity during HIV-1 infection abrogates Vpr cell cycle arrest. These findings provide new insight into the signaling event that activates Vpr cell cycle arrest, ultimately leading to the death of infected T cells.
- Published
- 2010
- Full Text
- View/download PDF
96. Termination of autophagy and reformation of lysosomes regulated by mTOR.
- Author
-
Yu L, McPhee CK, Zheng L, Mardones GA, Rong Y, Peng J, Mi N, Zhao Y, Liu Z, Wan F, Hailey DW, Oorschot V, Klumperman J, Baehrecke EH, and Lenardo MJ
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, HeLa Cells, Homeostasis physiology, Humans, Lysosomes ultrastructure, Rats, Signal Transduction, TOR Serine-Threonine Kinases, Vero Cells, Autophagy physiology, Intracellular Signaling Peptides and Proteins metabolism, Lysosomes metabolism, Nutritional Physiological Phenomena, Protein Serine-Threonine Kinases metabolism
- Abstract
Autophagy is an evolutionarily conserved process by which cytoplasmic proteins and organelles are catabolized. During starvation, the protein TOR (target of rapamycin), a nutrient-responsive kinase, is inhibited, and this induces autophagy. In autophagy, double-membrane autophagosomes envelop and sequester intracellular components and then fuse with lysosomes to form autolysosomes, which degrade their contents to regenerate nutrients. Current models of autophagy terminate with the degradation of the autophagosome cargo in autolysosomes, but the regulation of autophagy in response to nutrients and the subsequent fate of the autolysosome are poorly understood. Here we show that mTOR signalling in rat kidney cells is inhibited during initiation of autophagy, but reactivated by prolonged starvation. Reactivation of mTOR is autophagy-dependent and requires the degradation of autolysosomal products. Increased mTOR activity attenuates autophagy and generates proto-lysosomal tubules and vesicles that extrude from autolysosomes and ultimately mature into functional lysosomes, thereby restoring the full complement of lysosomes in the cell-a process we identify in multiple animal species. Thus, an evolutionarily conserved cycle in autophagy governs nutrient sensing and lysosome homeostasis during starvation.
- Published
- 2010
- Full Text
- View/download PDF
97. ALPS-ten lessons from an international workshop on a genetic disease of apoptosis.
- Author
-
Lenardo MJ, Oliveira JB, Zheng L, and Rao VK
- Subjects
- Animals, Autoimmune Lymphoproliferative Syndrome immunology, Genetic Predisposition to Disease, Humans, Mutation, Apoptosis, Autoimmune Lymphoproliferative Syndrome genetics
- Abstract
An international group of researchers investigating the molecular, cellular, immunological, and clinical aspects of the autoimmune lymphoproliferative syndrome (ALPS) met in Bethesda, Maryland on September 21-22, 2009 to discuss advances made over the past 15 years. Their discussions yielded ten broad messages applicable to genetic and immunological investigations of human disease.
- Published
- 2010
- Full Text
- View/download PDF
98. The open reading frame 3a protein of severe acute respiratory syndrome-associated coronavirus promotes membrane rearrangement and cell death.
- Author
-
Freundt EC, Yu L, Goldsmith CS, Welsh S, Cheng A, Yount B, Liu W, Frieman MB, Buchholz UJ, Screaton GR, Lippincott-Schwartz J, Zaki SR, Xu XN, Baric RS, Subbarao K, and Lenardo MJ
- Subjects
- ADP-Ribosylation Factor 1 genetics, ADP-Ribosylation Factor 1 metabolism, Animals, Cell Death, Chlorocebus aethiops, Cytoplasmic Vesicles metabolism, Cytoplasmic Vesicles virology, Gene Deletion, Golgi Apparatus metabolism, Golgi Apparatus pathology, Humans, Severe acute respiratory syndrome-related coronavirus genetics, Severe acute respiratory syndrome-related coronavirus metabolism, Transfection, Vero Cells pathology, Cell Membrane pathology, Cell Membrane virology, Host-Pathogen Interactions, Severe acute respiratory syndrome-related coronavirus pathogenicity, Viral Structural Proteins metabolism
- Abstract
The genome of the severe acute respiratory syndrome-associated coronavirus (SARS-CoV) contains eight open reading frames (ORFs) that encode novel proteins. These accessory proteins are dispensable for in vitro and in vivo replication and thus may be important for other aspects of virus-host interactions. We investigated the functions of the largest of the accessory proteins, the ORF 3a protein, using a 3a-deficient strain of SARS-CoV. Cell death of Vero cells after infection with SARS-CoV was reduced upon deletion of ORF 3a. Electron microscopy of infected cells revealed a role for ORF 3a in SARS-CoV induced vesicle formation, a prominent feature of cells from SARS patients. In addition, we report that ORF 3a is both necessary and sufficient for SARS-CoV-induced Golgi fragmentation and that the 3a protein accumulates and localizes to vesicles containing markers for late endosomes. Finally, overexpression of ADP-ribosylation factor 1 (Arf1), a small GTPase essential for the maintenance of the Golgi apparatus, restored Golgi morphology during infection. These results establish an important role for ORF 3a in SARS-CoV-induced cell death, Golgi fragmentation, and the accumulation of intracellular vesicles.
- Published
- 2010
- Full Text
- View/download PDF
99. The nuclear signaling of NF-kappaB: current knowledge, new insights, and future perspectives.
- Author
-
Wan F and Lenardo MJ
- Subjects
- Animals, Humans, Nuclear Proteins genetics, Protein Subunits genetics, Ribosomal Proteins genetics, Signal Transduction immunology, Transcription Factors genetics, Transcriptional Activation genetics, Cell Nucleus genetics, Immunity genetics, Inflammation genetics, NF-kappa B metabolism, Signal Transduction genetics
- Abstract
The nuclear factor-kappa B (NF-kappaB) transcription factor plays a critical role in diverse cellular processes associated with proliferation, cell death, development, as well as innate and adaptive immune responses. NF-kappaB is normally sequestered in the cytoplasm by a family of inhibitory proteins known as inhibitors of NF-kappaB (IkappaBs). The signal pathways leading to the liberation and nuclear accumulation of NF-kappaB, which can be activated by a wide variety of stimuli, have been extensively studied in the past two decades. After gaining access to the nucleus, NF-kappaB must be actively regulated to execute its fundamental function as a transcription factor. Recent studies have highlighted the importance of nuclear signaling in the regulation of NF-kappaB transcriptional activity. A non-Rel subunit of NF-kappaB, ribosomal protein S3 (RPS3), and numerous other nuclear regulators of NF-kappaB, including Akirin, Nurr1, SIRT6, and others, have recently been identified, unveiling novel and exciting layers of regulatory specificity for NF-kappaB in the nucleus. Further insights into the nuclear events that govern NF-kappaB function will deepen our understanding of the elegant control of its transcriptional activity and better inform the potential rational design of therapeutics for NF-kappaB-associated diseases.
- Published
- 2010
- Full Text
- View/download PDF
100. Dock8 mutations cripple B cell immunological synapses, germinal centers and long-lived antibody production.
- Author
-
Randall KL, Lambe T, Johnson AL, Treanor B, Kucharska E, Domaschenz H, Whittle B, Tze LE, Enders A, Crockford TL, Bouriez-Jones T, Alston D, Cyster JG, Lenardo MJ, Mackay F, Deenick EK, Tangye SG, Chan TD, Camidge T, Brink R, Vinuesa CG, Batista FD, Cornall RJ, and Goodnow CC
- Subjects
- Amino Acid Sequence, Animals, B-Lymphocytes metabolism, Base Sequence, Germinal Center metabolism, Guanine Nucleotide Exchange Factors chemistry, Guanine Nucleotide Exchange Factors metabolism, Humans, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Models, Molecular, Molecular Sequence Data, Protein Structure, Quaternary, Sequence Alignment, Antibody Formation, B-Lymphocytes immunology, Germinal Center immunology, Guanine Nucleotide Exchange Factors genetics, Guanine Nucleotide Exchange Factors immunology, Mutation, Synapses immunology
- Abstract
To identify genes and mechanisms involved in humoral immunity, we did a mouse genetic screen for mutations that do not affect the first wave of antibody to immunization but disrupt response maturation and persistence. The first two mutants identified had loss-of-function mutations in the gene encoding a previously obscure member of a family of Rho-Rac GTP-exchange factors, DOCK8. DOCK8-mutant B cells were unable to form marginal zone B cells or to persist in germinal centers and undergo affinity maturation. Dock8 mutations disrupted accumulation of the integrin ligand ICAM-1 in the B cell immunological synapse but did not alter other aspects of B cell antigen receptor signaling. Humoral immunodeficiency due to Dock8 mutation provides evidence that organization of the immunological synapse is critical for signaling the survival of B cell subsets required for long-lasting immunity.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.