Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Dev Med Child Neurol ; 64(12): 1470-1476, 2022 12.
Article in English | MEDLINE | ID: mdl-35441707

ABSTRACT

AIM: To define clinical common data elements (CDEs) and a mandatory minimum data set (MDS) for genomic studies of cerebral palsy (CP). METHOD: Candidate data elements were collated following a review of the literature and existing CDEs. An online, three-round Delphi survey was used to rate each data element as either 'core', 'recommended', 'exploratory', or 'not required'. Members of the International Cerebral Palsy Genomics Consortium (ICPGC) rated the core CDEs as either mandatory or not, to form the MDS. For both the CDEs and the MDS, a data element was considered to have reached consensus if more than 75% of respondents agreed. RESULTS: Forty-six individuals from around the world formed the Delphi panel: consumers (n=2), scientists/researchers (n=17), medical (n=19), and allied health professionals (n=8). The CDEs include 107 data elements across six categories: demographics, diagnostics, family history, antenatal and neonatal details, clinical traits, and CP-specific assessments. Of these, 10 are mandatory, 42 core, 41 recommended, and 14 are exploratory. INTERPRETATION: The ICPGC CDEs provide a foundation for the standardization of phenotype data captured in CP genomic studies and will benefit international collaborations and pooling of data, particularly in rare conditions. WHAT THIS PAPER ADDS: A set of 107 common data elements (CDEs) for genomics studies in cerebral palsy is provided. The CDEs include standard definitions and data values domains. The CDEs will facilitate international data sharing, collaboration, and improved clinical interpretation of findings.


OBJETIVO: Definir elementos de dados clínicos comuns (DCC) e um conjunto mínimo de dados obrigatórios (CMDO) para estudos genômicos de paralisia cerebral (PC). MÉTODO: Os elementos de dados do candidato foram coletados seguindo uma revisão da literatura e através dos DCC existentes. Uma pesquisa on-line de três rodadas Delphi foi usada para classificar cada elemento de dados como 'essencial', 'recomendado', 'exploratório' ou 'não obrigatório'. Os Membros do Consorcio Internacional de Genoma na Paralisia Cerebral (MCIGPC) classificaram os DCC do núcleo como obrigatórios ou não, para formar o CMDO. Tanto para os DCC quanto para o CMDO, um elemento de dados foi considerado como tendo chegado a um consenso se mais de 75% dos respondentes concordassem. RESULTADOS: Quarenta e seis indivíduos de todo o mundo formaram o painel Delphi: consumidores (n=2), cientistas/pesquisadores (n=17), médicos (n=19) e profissionais de saúde aliados (n=8). Os DCC incluem 107 elementos de dados em seis categorias: demografia, diagnóstico, história familiar, detalhes pré-natais e neonatais, características clínicas e avaliações específicas de PC. Destes, 10 são obrigatórios, 42 essenciais, 41 recomendados e 14 são exploratórios INTERPRETAÇÃO: Os DCC do MCIGPC fornecem uma base para a padronização de dados de fenótipo capturados em estudos genômicos de PC e beneficiarão colaborações internacionais e agrupamento de dados, particularmente em condições raras.


Subject(s)
Biomedical Research , Cerebral Palsy , Female , Pregnancy , United States , Humans , Common Data Elements , National Institute of Neurological Disorders and Stroke (U.S.) , Cerebral Palsy/diagnosis , Cerebral Palsy/genetics , Genomics
2.
Blood ; 119(24): 5898-908, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22538855

ABSTRACT

FoxP3(+) confers suppressive properties and is confined to regulatory T cells (T(reg)) that potently inhibit autoreactive immune responses. In the transplant setting, natural CD4(+) T(reg) are critical in controlling alloreactivity and the establishment of tolerance. We now identify an important CD8(+) population of FoxP3(+) T(reg) that convert from CD8(+) conventional donor T cells after allogeneic but not syngeneic bone marrow transplantation. These CD8(+) T(reg) undergo conversion in the mesenteric lymph nodes under the influence of recipient dendritic cells and TGF-ß. Importantly, this population is as important for protection from GVHD as the well-studied natural CD4(+)FoxP3(+) population and is more potent in exerting class I-restricted and antigen-specific suppression in vitro and in vivo. Critically, CD8(+)FoxP3(+) T(reg) are exquisitely sensitive to inhibition by cyclosporine but can be massively and specifically expanded in vivo to prevent GVHD by coadministering rapamycin and IL-2 antibody complexes. CD8(+)FoxP3(+) T(reg) thus represent a new regulatory population with considerable potential to preferentially subvert MHC class I-restricted T-cell responses after bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/cytology , Forkhead Transcription Factors/metabolism , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes/immunology , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immune Tolerance/drug effects , Interleukin-2/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Phenotype , Sirolimus/administration & dosage , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/pharmacology , Transplantation, Homologous
3.
Blood ; 119(24): 5918-30, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22415754

ABSTRACT

Alloreactivity after transplantation is associated with profound immune suppression, and consequent opportunistic infection results in high morbidity and mortality. This immune suppression is most profound during GVHD after bone marrow transplantation where an inflammatory cytokine storm dominates. Contrary to current dogma, which avers that this is a T-cell defect, we demonstrate that the impairment lies within conventional dendritic cells (cDCs). Significantly, exogenous antigens can only be presented by the CD8(-) cDC subset after bone marrow transplantation, and inflammation during GVHD specifically renders the MHC class II presentation pathway in this population incompetent. In contrast, both classic and cross-presentation within MHC class I remain largely intact. Importantly, this defect in antigen processing can be partially reversed by TNF inhibition or the adoptive transfer of donor cDCs generated in the absence of inflammation.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Immunosuppression Therapy , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cross-Priming/immunology , Graft vs Host Disease/pathology , Histocompatibility Antigens Class II/immunology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/metabolism , Isoantigens/immunology , Mice , Mice, Transgenic , Peptides/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
4.
Blood ; 118(12): 3399-409, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21719602

ABSTRACT

Although the effects of type II-IFN (IFN-γ) on GVHD and leukemia relapse are well studied, the effects of type I-interferon (type I-IFN, IFN-α/ß) remain unclear. We investigated this using type I-IFN receptor-deficient mice and exogenous IFN-α administration in established models of GVHD and GVL. Type I-IFN signaling in host tissue prevented severe colon-targeted GVHD in CD4-dependent models of GVHD directed toward either major histocompatibility antigens or multiple minor histocompatibility antigens. This protection was the result of suppression of donor CD4(+) T-cell proliferation and differentiation. Studies in chimeric recipients demonstrated this was due to type I-IFN signaling in hematopoietic tissue. Consistent with this finding, administration of IFN-α during conditioning inhibited donor CD4(+) proliferation and differentiation. In contrast, CD8-dependent GVHD and GVL effects were enhanced when type I-IFN signaling was intact in the host or donor, respectively. This finding reflected the ability of type I-IFN to both sensitize host target tissue/leukemia to cell-mediated cytotoxicity and augment donor CTL function. These data confirm that type I-IFN plays an important role in defining the balance of GVHD and GVL responses and suggests that administration of the cytokine after BM transplantation could be studied prospectively in patients at high risk of relapse.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Interferon-alpha , Leukemia/immunology , Receptor, Interferon alpha-beta/deficiency , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Female , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/drug effects , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Interferon-beta/immunology , Leukemia/mortality , Leukemia/pathology , Leukemia/therapy , Lymphocyte Activation/immunology , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptor, Interferon alpha-beta/immunology , Signal Transduction , Survival Rate , Transplantation, Homologous , Whole-Body Irradiation
6.
Blood ; 116(5): 819-28, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20435882

ABSTRACT

The recent shift to the use of stem cells mobilized by granulocyte colony-stimulating factor (G-CSF) for hematopoietic transplantation has increased chronic graftversus-host disease (GVHD), although the mechanisms of this are unclear. We have found that G-CSF invokes potent type 17 rather than type 1 or type 2 differentiation. The amplification of interleukin-17 (IL-17) production by G-CSF occurs in both CD4 and CD8 conventional T cells and is dependent on, and downstream of, G-CSF-induced IL-21 signaling. Importantly, donor IL-17A controls the infiltration of macrophages into skin and cutaneous fibrosis, manifesting late after transplantation as scleroderma. Interestingly, donor CD8 T cells were the predominant source of IL-17A after transplantation and could mediate scleroderma independently of CD4 T cells. This study provides a logical explanation for the propensity of allogeneic stem cell transplantation to invoke sclerodermatous GVHD and suggests a therapeutic strategy for intervention.


Subject(s)
Bone Marrow Transplantation/adverse effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Mobilization/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Interleukin-17/physiology , Interleukins/physiology , Scleroderma, Localized/etiology , T-Lymphocyte Subsets/transplantation , Animals , Cell Differentiation/drug effects , Crosses, Genetic , Cytokines/biosynthesis , Female , Fibrosis , Humans , Interleukin-17/metabolism , Macrophages/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Radiation Chimera , Signal Transduction , Skin/immunology , Skin/pathology , Transplantation, Homologous/adverse effects
7.
Blood ; 116(2): 287-96, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20435883

ABSTRACT

Suppressor of cytokine signaling-3 (SOCS3) is the main intracellular regulator of signaling by granulocyte colony-stimulating factor, an immune-modulatory cytokine used to mobilize stem cells for transplantation. We have therefore studied the contribution of SOCS3 to the spectrum of graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (SCT). Grafts from SOCS3(-/Deltavav) donor mice in which SOCS3 deficiency is restricted to the hematopoietic compartment had an augmented capacity to induce acute GVHD. With the use of SOCS3(-/DeltaLysM) and SOCS3(-/Deltalck) donors in which SOCS3 deficiency was restricted to the myeloid or T-cell lineage, respectively, we confirmed SOCS3 deficiency promoted acute GVHD mortality and histopathology within the gastrointestinal tract by effects solely within the donor T cell. SOCS3(-/Deltalck) donor T cells underwent enhanced alloantigen-dependent proliferation and generation of interleukin-10 (IL-10), IL-17, and interferon-gamma (IFNgamma) after SCT. The enhanced capacity of the SOCS3(-/Deltalck) donor T cell to induce acute GVHD was dependent on IFNgamma but independent of IL-10 or IL-17. Surprisingly, SOCS3(-/Deltalck) donor T cells also induced severe, transforming growth factor beta- and IFNgamma-dependent, sclerodermatous GVHD. Thus, the delivery of small molecule SOCS3 mimetics may prove to be useful for the inhibition of both acute and chronic GVHD.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/immunology , Animals , Cell Separation , Cytokines/biosynthesis , Female , Flow Cytometry , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Mice, Inbred C57BL , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/metabolism , Transplantation, Homologous
8.
Blood ; 115(1): 122-32, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19789388

ABSTRACT

Tumor necrosis factor (TNF) is a key cytokine in the effector phase of graft-versus-host disease (GVHD) after bone marrow transplantation, and TNF inhibitors have shown efficacy in clinical and experimental GVHD. TNF signals through the TNF receptors (TNFR), which also bind soluble lymphotoxin (LTalpha3), a TNF family member with a previously unexamined role in GVHD pathogenesis. We have used preclinical models to investigate the role of LT in GVHD. We confirm that grafts deficient in LTalpha have an attenuated capacity to induce GVHD equal to that seen when grafts lack TNF. This is not associated with other defects in cytokine production or T-cell function, suggesting that LTalpha3 exerts its pathogenic activity directly via TNFR signaling. We confirm that donor-derived LTalpha is required for graft-versus-leukemia (GVL) effects, with equal impairment in leukemic clearance seen in recipients of LTalpha- and TNF-deficient grafts. Further impairment in tumor clearance was seen using Tnf/Lta(-/-) donors, suggesting that these molecules play nonredundant roles in GVL. Importantly, donor TNF/LTalpha were only required for GVL where the recipient leukemia was susceptible to apoptosis via p55 TNFR signaling. These data suggest that antagonists neutralizing both TNF and LTalpha3 may be effective for treatment of GVHD, particularly if residual leukemia lacks the p55 TNFR.


Subject(s)
Graft vs Host Disease/immunology , Lymphotoxin-alpha/immunology , Animals , Apoptosis , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Graft vs Host Disease/pathology , Inflammation Mediators/metabolism , Mice , Protein Multimerization , Receptors, Antigen, T-Cell/immunology , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/immunology , Solubility , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/immunology
9.
J Immunol ; 185(6): 3632-42, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20720206

ABSTRACT

Cerebral malaria is a severe complication of malaria. Sequestration of parasitized RBCs in brain microvasculature is associated with disease pathogenesis, but our understanding of this process is incomplete. In this study, we examined parasite tissue sequestration in an experimental model of cerebral malaria (ECM). We show that a rapid increase in parasite biomass is strongly associated with the induction of ECM, mediated by IFN-gamma and lymphotoxin alpha, whereas TNF and IL-10 limit this process. Crucially, we discovered that host CD4(+) and CD8(+) T cells promote parasite accumulation in vital organs, including the brain. Modulation of CD4(+) T cell responses by helminth coinfection amplified CD4(+) T cell-mediated parasite sequestration, whereas vaccination could generate CD4(+) T cells that reduced parasite biomass and prevented ECM. These findings provide novel insights into immune-mediated mechanisms of ECM pathogenesis and highlight the potential of T cells to both prevent and promote infectious diseases.


Subject(s)
Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Plasmodium berghei/immunology , Animals , Brain/blood supply , Brain/immunology , Brain/parasitology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Erythrocytes/immunology , Erythrocytes/parasitology , Erythrocytes/pathology , Female , Gastrointestinal Tract/blood supply , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology , Kidney/blood supply , Kidney/immunology , Kidney/parasitology , Liver/blood supply , Liver/immunology , Liver/parasitology , Lung/blood supply , Lung/immunology , Lung/parasitology , Malaria, Cerebral/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Organ Specificity/immunology , Plasmodium berghei/growth & development , Severity of Illness Index , Spleen/blood supply , Spleen/immunology , Spleen/parasitology
10.
J Infect Dis ; 204(12): 1893-901, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22043019

ABSTRACT

BACKGROUND: Live attenuated varicella vaccine is considered a safe vaccine with serious adverse effects reported only in immunocompromised children. We describe a severe life-threatening infection with varicella vaccine virus causing rash and pneumonitis in a 6-year-old boy with no apparent immunodeficiency. METHODS AND RESULTS: Polymerase chain reaction (PCR) analysis of vesicle swab samples demonstrated varicella zoster virus (VZV). Sequencing of the PCR product demonstrated 100% homology with human herpesvirus 3 strain VZV-Oka ORF62 gene. Routine immunologic investigations failed to demonstrate any abnormality. Total leukocyte, lymphocyte, and neutrophil counts and lymphocyte subsets were normal. Immunoglobulins, C3, C4, and CH50 were intact. Specific IgG to protein and polysaccharide antigens and to Epstein-Barr virus and cytomegalovirus were present. Normal lymphocyte proliferation to phytohemagglutinin and VZV antigens was detected. Neutrophil function and natural killer (NK) cell activity were normal. The analysis of invariant NK T (iNKT) cell numbers and function revealed diminished iNKT cells, reported once previously and unique to our patient, deficient expression of the cognate receptor, CD1d. CONCLUSIONS: This report provides a further link between deficiency of the iNKT/CD1d pathway and increased susceptibility to varicella vaccine virus, suggesting an important role of this innate pathway in host defense against yet another member of the herpesvirus family.


Subject(s)
Antigens, CD1d/metabolism , Chickenpox Vaccine/adverse effects , Chickenpox/immunology , Herpesvirus 3, Human/immunology , Natural Killer T-Cells/immunology , Vaccination/adverse effects , Cell Proliferation , Chickenpox/drug therapy , Chickenpox/virology , Child , Humans , Interferon-gamma/metabolism , Lymphocyte Count , Male , Natural Killer T-Cells/metabolism , Vaccines, Attenuated/adverse effects
11.
Blood ; 113(22): 5644-9, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19336758

ABSTRACT

We have quantified the relative contribution of donor antigen-presenting cell populations to alloantigen presentation after bone marrow transplantation (BMT) by using transgenic T cells that can respond to host-derived alloantigen presented within the donor major histocompatibility complex. We also used additional transgenic/knockout donor mice and/or monoclonal antibodies that allowed conditional depletion of conventional dendritic cells (cDCs), plasmacytoid DC (pDCs), macrophages, or B cells. Using these systems, we demonstrate that donor cDCs are the critical population presenting alloantigen after BMT, whereas pDCs and macrophages do not make a significant contribution in isolation. In addition, alloantigen presentation was significantly enhanced in the absence of donor B cells, confirming a regulatory role for these cells early after transplantation. These data have major implications for the design of therapeutic strategies post-BMT, and suggest that cDC depletion and the promotion of B-cell reconstitution may be beneficial tools for the control of alloreactivity.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , Dendritic Cells/immunology , Isoantigens/immunology , Animal Experimentation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD11 Antigens/genetics , Dendritic Cells/physiology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
12.
Sci Rep ; 7(1): 16786, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196744

ABSTRACT

Water intake is essential for survival and thus under strong regulation. Here, we describe a simple high throughput system to monitor water intake over time in Drosophila. The design of the assay involves dehydrating fly food and then adding water back separately so flies either eat or drink. Water consumption is then evaluated by weighing the water vessel and comparing this back to an evaporation control. Our system is high throughput, does not require animals to be artificially dehydrated, and is simple both in design and implementation. Initial characterisation of homeostatic water consumption shows high reproducibility between biological replicates in a variety of experimental conditions. Water consumption was dependent on ambient temperature and humidity and was equal between sexes when corrected for mass. By combining this system with the Drosophila genetics tools, we could confirm a role for ppk28 and DopR1 in promoting water consumption, and through functional investigation of RNAseq data from dehydrated animals, we found DopR1 expression in the mushroom body was sufficient to drive consumption and enhance water taste sensitivity. Together, we provide a simple high throughput water consumption assay that can be used to dissect the cellular and molecular machinery regulating water homeostasis in Drosophila.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Epithelial Sodium Channels/genetics , Receptors, Dopamine/genetics , Water/metabolism , Animals , Drinking , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eating , Epithelial Sodium Channels/metabolism , High-Throughput Screening Assays , Receptors, Dopamine/metabolism , Sequence Analysis, RNA
13.
Cell Metab ; 24(1): 75-90, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27411010

ABSTRACT

Non-nutritive sweeteners like sucralose are consumed by billions of people. While animal and human studies have demonstrated a link between synthetic sweetener consumption and metabolic dysregulation, the mechanisms responsible remain unknown. Here we use a diet supplemented with sucralose to investigate the long-term effects of sweet/energy imbalance. In flies, chronic sweet/energy imbalance promoted hyperactivity, insomnia, glucose intolerance, enhanced sweet taste perception, and a sustained increase in food and calories consumed, effects that are reversed upon sucralose removal. Mechanistically, this response was mapped to the ancient insulin, catecholamine, and NPF/NPY systems and the energy sensor AMPK, which together comprise a novel neuronal starvation response pathway. Interestingly, chronic sweet/energy imbalance promoted increased food intake in mammals as well, and this also occurs through an NPY-dependent mechanism. Together, our data show that chronic consumption of a sweet/energy imbalanced diet triggers a conserved neuronal fasting response and increases the motivation to eat.


Subject(s)
Eating/drug effects , Fasting , Neurons/metabolism , Neuropeptide Y/metabolism , Sucrose/analogs & derivatives , Adenylate Kinase/metabolism , Animals , Appetite/drug effects , Dopamine/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/drug effects , Drosophila melanogaster/physiology , Energy Intake/drug effects , Enzyme Activation/drug effects , Homeostasis/drug effects , Hunger/drug effects , Insulin/metabolism , Male , Neurons/drug effects , Octopamine/metabolism , Receptors, Cell Surface/metabolism , Sucrose/pharmacology , Sweetening Agents/pharmacology , Taste/drug effects
14.
Nat Commun ; 6: 8570, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26490707

ABSTRACT

Disease incidences increase with age, but the molecular characteristics of ageing that lead to increased disease susceptibility remain inadequately understood. Here we perform a whole-blood gene expression meta-analysis in 14,983 individuals of European ancestry (including replication) and identify 1,497 genes that are differentially expressed with chronological age. The age-associated genes do not harbor more age-associated CpG-methylation sites than other genes, but are instead enriched for the presence of potentially functional CpG-methylation sites in enhancer and insulator regions that associate with both chronological age and gene expression levels. We further used the gene expression profiles to calculate the 'transcriptomic age' of an individual, and show that differences between transcriptomic age and chronological age are associated with biological features linked to ageing, such as blood pressure, cholesterol levels, fasting glucose, and body mass index. The transcriptomic prediction model adds biological relevance and complements existing epigenetic prediction models, and can be used by others to calculate transcriptomic age in external cohorts.


Subject(s)
Aging/blood , Transcriptome , Biomarkers/blood , DNA Methylation , Gene Expression Profiling , Humans , White People
15.
Nat Med ; 18(1): 135-42, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22127134

ABSTRACT

The presentation pathways by which allogeneic peptides induce graft-versus-host disease (GVHD) are unclear. We developed a bone marrow transplant (BMT) system in mice whereby presentation of a processed recipient peptide within major histocompatibility complex (MHC) class II molecules could be spatially and temporally quantified. Whereas donor antigen presenting cells (APCs) could induce lethal acute GVHD via MHC class II, recipient APCs were 100-1,000 times more potent in this regard. After myeloablative irradiation, T cell activation and memory differentiation occurred in lymphoid organs independently of alloantigen. Unexpectedly, professional hematopoietic-derived recipient APCs within lymphoid organs had only a limited capacity to induce GVHD, and dendritic cells were not required. In contrast, nonhematopoietic recipient APCs within target organs induced universal GVHD mortality and promoted marked alloreactive donor T cell expansion within the gastrointestinal tract and inflammatory cytokine generation. These data challenge current paradigms, suggesting that experimental lethal acute GVHD can be induced by nonhematopoietic recipient APCs.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/cytology , Cytokines/immunology , Dendritic Cells/immunology , Hematopoietic System/immunology , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Transplant Donor Site , Transplantation, Homologous
16.
Nat Med ; 15(4): 436-41, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19330008

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is often used to hasten neutrophil recovery after allogeneic bone marrow transplantation (BMT), but the clinical and immunological consequences evoked remain unclear. We examined the effect of G-CSF administration after transplantation in mouse models and found that exposure to either standard G-CSF or pegylated-G-CSF soon after BMT substantially increased graft-versus-host disease (GVHD). This effect was dependent on total body irradiation (TBI) rendering host dendritic cells (DCs) responsive to G-CSF by upregulating their expression of the G-CSF receptor. Stimulation of host DCs by G-CSF subsequently unleashed a cascade of events characterized by donor natural killer T cell (NKT cell) activation, interferon-gamma secretion and CD40-dependent amplification of donor cytotoxic T lymphocyte function during the effector phase of GVHD. Crucially, the detrimental effects of G-CSF were only present when it was administered after TBI conditioning and at a time when residual host antigen presenting cells were still present, perhaps explaining the conflicting and somewhat controversial clinical studies from the large European and North American BMT registries. These data have major implications for the use of G-CSF in disease states where NKT cell activation may have effects on outcome.


Subject(s)
Bone Marrow Transplantation/immunology , Granulocyte Colony-Stimulating Factor/therapeutic use , Killer Cells, Natural/immunology , Neutrophils/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Humans , Interferon-gamma/physiology , Lymphocyte Activation/drug effects , Lymphocyte Transfusion , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Neutrophils/drug effects , T-Lymphocytes/transplantation , Transplantation, Homologous/immunology , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL