Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 46(5): 1291-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26865269

ABSTRACT

Humanized mice harboring human immune systems (HIS) represent a platform to study immune responses against pathogens and to screen vaccine candidates and novel immunotherapeutics. Innate and adaptive immune responses are suboptimal in HIS mice, possibly due to poor reconstitution of human antigen-presenting cells, including dendritic cells (DCs). DC homeostasis is regulated by cytokine availability, and Flt3-ligand (Flt3L) is one factor that conditions this process. Mouse myelopoiesis is essentially normal in most current HIS models. As such, developing mouse myeloid cells may limit human DC reconstitution by reducing available Flt3L and by cellular competition for specific "niches." To address these issues, we created a novel HIS model that compromises host myeloid cell development via deficiency in the receptor tyrosine kinase Flk2/Flt3. In Balb/c Rag2(-/-) Il2rg(-/-) Flt3(-/-) (BRGF) recipients, human conventional DCs and plasmacytoid DCs develop from hCD34(+) precursors and can be specifically boosted with exogenous Flt3L. Human DCs that develop in this context normally respond to TLR stimulation, and improved human DC homeostasis is associated with increased numbers of human NK and T cells. This new HIS-DC model should provide a means to dissect human DC differentiation and represents a novel platform to screen immune adjuvants and DC targeting therapies.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/physiology , fms-Like Tyrosine Kinase 3/deficiency , Adjuvants, Immunologic , Animals , Homeostasis , Humans , Immunity, Innate , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/physiology , Natural Killer T-Cells/physiology , T-Lymphocytes/physiology , fms-Like Tyrosine Kinase 3/genetics
2.
Proc Natl Acad Sci U S A ; 108(15): 6217-22, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21444793

ABSTRACT

Cytokine immunotherapies targeting T lymphocytes are attractive clinical interventions against viruses and tumors. In the mouse, the homeostasis of memory α/ß CD8(+) T cells and natural killer (NK) cells is significantly improved with increased IL-15 bioavailability. In contrast, the role of "transpresented" IL-15 on human T-cell development and homeostasis in vivo is unknown. We found that both CD8 and CD4 T cells in human immune system (HIS) mice are highly sensitive to transpresented IL-15 in vivo, with both naïve (CD62L(+)CD45RA(+)) and memory phenotype (CD62L(-)CD45RO(+)) subsets being significantly increased following IL-15 "boosting." The unexpected global improvement in human T-cell homeostasis involved enhanced proliferation and survival of both naïve and memory phenotype peripheral T cells, which potentiated B-cell responses by increasing the frequency of antigen-specific responses following immunization. Transpresented IL-15 did not modify T-cell activation patterns or alter the global T-cell receptor (TCR) repertoire diversity. Our results indicate an unexpected effect of IL-15 on human T cells in vivo, in particular on CD4(+) T cells. As IL-15 promotes human peripheral T-cell homeostasis and increases the frequency of neutralizing antibody responses in HIS mice, IL-15 immunotherapy could be envisaged as a unique approach to improve vaccine responses in the clinical setting.


Subject(s)
Antibody Formation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-15/immunology , Animals , Cell Proliferation , Humans , Mice , Mice, Mutant Strains , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Interleukin-15/antagonists & inhibitors
3.
Proc Natl Acad Sci U S A ; 108(8): 3330-5, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21300860

ABSTRACT

αß T-cell repertoire selection is mediated by peptide-MHC complexes presented by thymic epithelial or myeloid cells, and by lipid-CD1 complexes expressed by thymocytes. γδ T-cell repertoire selection, by contrast, is largely unresolved. Mice mutant for Skint-1, a unique Ig superfamily gene, do not develop canonical Vγ5Vδ1(+) dendritic epidermal T cells. This study shows that transgenic Skint-1, across a broad range of expression levels, precisely and selectively determines the Vγ5Vδ1(+) dendritic epidermal T-cell compartment. Skint-1 is expressed by medullary thymic epithelial cells, and unlike lipid-CD1 complexes, must be expressed by stromal cells to function efficiently. Its unusual transmembrane-cytoplasmic regions severely limit cell surface expression, yet increasing this or, conversely, retaining Skint1 intracellularly markedly compromises function. Each Skint1 domain appears nonredundant, including a unique decamer specifying IgV-domain processing. This investigation of Skint-1 biology points to complex events underpinning the positive selection of an intraepithelial γδ repertoire.


Subject(s)
Epidermis/immunology , Immunoglobulins/immunology , T-Lymphocytes/immunology , Animals , Dendritic Cells , Epidermal Cells , Epithelial Cells , Gene Expression/immunology , Immunoglobulins/genetics , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets
4.
J Immunol ; 184(11): 5949-53, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20439914

ABSTRACT

Thymic epithelial cells (TECs) are the predominant intrathymic source of the essential thymopoietin IL-7. Whether thymocyte-TEC interactions have a role in the regulation of IL-7 expression is not known. By exploiting IL-7 reporter mice in which yellow fluorescent protein expression identifies TECs expressing high levels of IL-7 (Il7(+) TECs), we show that Il7(+) TECs segregate from emerging medullary TECs during thymic organogenesis. Although Il7(+) TECs normally diminish with age, we found that Il7(+) TECs are markedly retained in alymphoid Rag2(-/-)Il2rg(-/-) IL-7 reporter mice that manifest a profound thymopoietic arrest. Transfer of Tcra(-/-) or wild-type (but not Rag2(-/-)) hematopoietic progenitors to alymphoid IL-7 reporter recipients normalizes the frequency of Il7(+) TECs and re-establishes cortical TEC/medullary TEC segregation. Although thymocyte-derived signals are often considered stimulatory for TEC maturation, our findings identify a negative feedback mechanism in which signals derived from TCRbeta-selected thymocytes modulate TEC-dependent IL-7 expression.


Subject(s)
Epithelial Cells/metabolism , Gene Expression Regulation/immunology , Interleukin-7/biosynthesis , Receptor Cross-Talk/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes/cytology , Animals , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Gene Expression , Immunohistochemistry , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
5.
J Clin Invest ; 120(6): 2131-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20440074

ABSTRACT

Enteropathy-associated T cell lymphoma is a severe complication of celiac disease (CD). One mechanism suggested to underlie its development is chronic exposure of intraepithelial lymphocytes (IELs) to potent antiapoptotic signals initiated by IL-15, a cytokine overexpressed in the enterocytes of individuals with CD. However, the signaling pathway by which IL-15 transmits these antiapoptotic signals has not been firmly established. Here we show that the survival signals delivered by IL-15 to freshly isolated human IELs and to human IEL cell lines derived from CD patients with type II refractory CD (RCDII) - a clinicopathological entity considered an intermediary step between CD and enteropathy-associated T cell lymphoma - depend on the antiapoptotic factors Bcl-2 and/or Bcl-xL. The signals also required IL-15Rbeta, Jak3, and STAT5, but were independent of PI3K, ERK, and STAT3. Consistent with these data, IELs from patients with active CD and RCDII contained increased amounts of Bcl-xL, phospho-Jak3, and phospho-STAT5. Furthermore, incubation of patient duodenal biopsies with a fully humanized human IL-15-specific Ab effectively blocked Jak3 and STAT5 phosphorylation. In addition, treatment with this Ab induced IEL apoptosis and wiped out the massive IEL accumulation in mice overexpressing human IL-15 in their gut epithelium. Together, our results delineate the IL-15-driven survival pathway in human IELs and demonstrate that IL-15 and its downstream effectors are meaningful therapeutic targets in RCDII.


Subject(s)
Apoptosis/immunology , Celiac Disease/immunology , Inflammation/immunology , Interleukin-15/immunology , Lymphocytes/immunology , Adult , Apoptosis/drug effects , Celiac Disease/complications , Celiac Disease/metabolism , Cytokines/immunology , Cytokines/metabolism , Cytokines/pharmacology , Enterocytes/immunology , Enterocytes/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Interleukin-15/metabolism , Interleukin-15/pharmacology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestines/immunology , Janus Kinase 3/immunology , Janus Kinase 3/metabolism , Leukemia/complications , Leukemia/immunology , Leukemia/metabolism , Lymphocytes/metabolism , Phosphorylation , Protein Binding/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology
6.
Cell Host Microbe ; 6(1): 5-9, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19616761

ABSTRACT

Over 800 million people worldwide are infected with hepatitis viruses, human immunodeficiency virus (HIV), and malaria, resulting in more than 5 million deaths annually. Here we discuss the potential and challenges of humanized mouse models for developing effective and affordable therapies and vaccines, which are desperately needed to combat these diseases.


Subject(s)
Biomedical Research/trends , Communicable Diseases , Disease Models, Animal , Animals , Humans , Mice
7.
Immunity ; 29(6): 958-70, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19084435

ABSTRACT

Natural killer (NK) cells are innate lymphocytes with spontaneous antitumor activity, and they produce interferon-gamma (IFN-gamma) that primes immune responses. Whereas T helper cell subsets differentiate from naive T cells via specific transcription factors, evidence for NK cell diversification is limited. In this report, we characterized intestinal lymphocytes expressing the NK cell natural cytotoxicity receptor NKp46. Gut NKp46+ cells were distinguished from classical NK cells by limited IFN-gamma production and absence of perforin, whereas several subsets expressed the nuclear hormone receptor retinoic acid receptor-related orphan receptor t (RORgammat) and interleukin-22 (IL-22). Intestinal NKp46+IL-22+ cells were generated via a local process that was conditioned by commensal bacteria and required RORgammat. Mice lacking IL-22-producing NKp46+ cells showed heightened susceptibility to the pathogen Citrobacter rodentium, consistent with a role for intestinal NKp46+ cells in immune protection. RORgammat-driven diversification of intestinal NKp46+ cells thereby specifies an innate cellular defense mechanism that operates at mucosal surfaces.


Subject(s)
Antigens, Ly/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Interleukins/immunology , Intestines/immunology , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Animals , Antigens, Ly/metabolism , Enterobacteriaceae Infections/microbiology , Immunity, Innate , Immunity, Mucosal/immunology , Interleukins/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Perforin/immunology , Perforin/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , Signal Transduction/immunology , Interleukin-22
8.
Gastroenterology ; 132(3): 994-1008, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17324400

ABSTRACT

BACKGROUND AND AIMS: Interleukin (IL)-15 delivers signals that drive chronic inflammation in several diseases, including celiac disease. Smad3-transforming growth factor-beta (TGF-beta) signaling is instrumental to counteract proinflammatory signals and maintain immune homeostasis. Our goal has been to investigate why the proinflammatory effects of IL-15 cannot be efficiently controlled by TGF-beta in celiac disease. METHODS: The impact of IL-15 on TGF-beta signaling in T cells and in the intestinal mucosa of celiac disease patients was analyzed by combining cell and organ cultures, immunohistochemistry, flow cytometry, real-time polymerase chain reaction, electromobility gel shift, and Western blot. RESULTS: IL-15 impaired Smad3-dependent TGF-beta signaling in human T lymphocytes downstream from Smad3 nuclear translocation. IL-15-mediated inhibition was associated with a long-lasting activation of c-jun-N-terminal kinase and reversed by c-jun antisense oligonucleotides, consistent with the demonstrated inhibitory effect of phospho-c-jun on the formation of Smad3-DNA complexes. In active celiac disease, intestinal lymphocytes showed impaired TGF-beta-Smad3-dependent transcriptional responses and up-regulation of phospho-c-jun. Anti-IL-15 antibody and c-jun antisense both downmodulated phospho-c-jun expression and restored TGF-beta-Smad-dependent transcription in biopsies of active celiac disease. c-jun antisense decreased interferon gamma transcription. CONCLUSIONS: Impairment of TGF-beta-mediated signaling by IL-15 might promote and sustain intestinal inflammation in celiac disease. More generally, our data provide a new rationale for the potent proinflammatory effects of IL-15, and further support the concept that IL-15 is a meaningful therapeutic target in inflammatory diseases associated with irreducible elevation of IL-15.


Subject(s)
Celiac Disease/metabolism , Interleukin-15/biosynthesis , Intestinal Mucosa/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/metabolism , Active Transport, Cell Nucleus , Adult , Aged , Celiac Disease/genetics , Celiac Disease/immunology , Celiac Disease/pathology , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Activation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeostasis/immunology , Humans , Immunity, Mucosal , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-15/pharmacology , Interleukin-2/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Middle Aged , Organ Culture Techniques , Phosphorylation , RNA, Messenger/biosynthesis , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction/drug effects , Smad3 Protein/metabolism , Smad7 Protein/biosynthesis , Smad7 Protein/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Transcription, Genetic , Transforming Growth Factor beta1/pharmacology , Tristetraprolin/biosynthesis , Tristetraprolin/genetics , Up-Regulation
9.
J Immunol ; 177(12): 8835-43, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17142786

ABSTRACT

We previously reported the clinical phenotype of two siblings with a novel inherited developmental and immunodeficiency syndrome consisting of severe intrauterine growth retardation and the impaired development of specific lymphoid lineages, including transient CD8 alphabeta T lymphopenia and a persistent lack of blood NK cells. We describe here the elucidation of a plausible underlying pathogenic mechanism, with a cellular phenotype of impaired survival of both fresh and herpesvirus saimiri-transformed T cells, in the surviving child. Clearly, NK cells could not be studied. However, peripheral blood T lymphocytes displayed excessive apoptosis ex vivo. Moreover, the survival rates of CD4 and CD8 alphabeta T cell blasts generated in vitro, and herpesvirus saimiri-transformed T cells cultured in vitro, were low, but not nil, following treatment with IL-2 and IL-15. In contrast, Fas-mediated activation-induced cell death was not enhanced, indicating a selective excess of cytokine deprivation-mediated apoptosis. In keeping with the known roles of IL-2 and IL-15 in the development of NK and CD8 T cells in the mouse model, these data suggest that an impaired, but not abolished, survival response to IL-2 and IL-15 accounts for the persistent lack of NK cells and the transient CD8 alphabeta T lymphopenia documented in vivo. Impaired cytokine-mediated lymphocyte survival is likely to be the pathogenic mechanism underlying this novel form of inherited and selective NK deficiency in humans.


Subject(s)
Cell Survival/drug effects , Immunologic Deficiency Syndromes/etiology , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Killer Cells, Natural/pathology , Lymphocytes/pathology , Adolescent , Apoptosis/drug effects , Case-Control Studies , Child , Child, Preschool , Family Health , Female , Humans , Infant , Lymphopenia/etiology
10.
Immunity ; 21(3): 367-77, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15357948

ABSTRACT

MICA molecules interact with the NKG2D-activating receptor on human NK and CD8 T cells. We investigated the participation of the MICA/NKG2D pathway in the destruction of intestinal epithelium by intraepithelial T lymphocytes (IEL) in Celiac disease and its premalignant complication, refractory sprue. We show that MICA is strongly expressed at epithelial cell surface in patients with active disease and is induced by gliadin or its p31-49 derived peptide upon in vitro challenge, an effect relayed by IL-15. This triggers direct activation and costimulation of IEL through engagement of NKG2D, leading to an innate-like cytotoxicity toward epithelial targets and enhanced TCR-dependent CD8 T cell-mediated adaptive response. Villous atrophy in Celiac disease might thus be ascribed to an IEL-mediated damage to enterocytes involving NKG2D/MICA interaction after gliadin-induced expression of MICA on gut epithelium. This supports a key role for MIC/NKG2D in the activation of intraepithelial immunity in response to danger.


Subject(s)
Celiac Disease/immunology , Intestinal Mucosa/immunology , Proteins/immunology , Receptors, Immunologic/immunology , Atrophy/immunology , Atrophy/pathology , Celiac Disease/blood , Celiac Disease/pathology , Cytotoxicity Tests, Immunologic , Enterocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gliadin/pharmacology , HeLa Cells , Humans , Immunohistochemistry , Interleukin-15/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/pathology , NK Cell Lectin-Like Receptor Subfamily K , Protein Biosynthesis , Proteins/drug effects , RNA, Long Noncoding , RNA, Untranslated , Receptors, Natural Killer Cell , T-Lymphocytes/immunology
12.
Gastroenterology ; 125(3): 730-45, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949719

ABSTRACT

BACKGROUND & AIMS: The mechanism of intraepithelial lymphocyte hyperplasia, a hallmark of celiac disease, is unknown. We have investigated the role of epithelium-derived interleukin (IL)-15 in the alterations of epithelial homeostasis in refractory celiac sprue, a privileged situation to study the first step of lymphoid transformation and the contribution of intraepithelial lymphocytes to villous atrophy in celiac disease. METHODS: IL-15 expression was assessed in biopsy specimens and isolated enterocytes by combining immunohistochemistry, flow cytometry, and real-time quantitative polymerase chain reaction. The ability of IL-15 to induce growth and survival of clonal intraepithelial lymphocytes lacking surface CD3 and to induce their cytotoxicity and secretion of interferon gamma was tested using soluble IL-15 and coculture in the presence of epithelial cell lines expressing membrane IL-15. RESULTS: IL-15 was massively overexpressed not only in lamina propria but also in the intestinal epithelium of patients with active celiac disease and refractory celiac sprue. IL-15 was not secreted but delivered at the surface of enterocytes. IL-15 specifically induced the expansion and survival of the clonal abnormal intraepithelial lymphocytes that characterize refractory celiac sprue and triggered their secretion of interferon gamma and their cytotoxicity against intestinal epithelial cells. Comparable activating signals could be delivered by IL-15 expressed at the membrane of the T84 enterocyte cell line. CONCLUSIONS: These data provide strong evidence that uncontrolled overexpression of IL-15 in refractory celiac sprue perpetuates epithelial damage and promotes the emergence of T-cell clonal proliferations. Blocking IL-15 might prove useful to treat this severe complication of celiac disease.


Subject(s)
Celiac Disease/immunology , Interleukin-15/physiology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Lymphoma/etiology , Adult , Aged , Aged, 80 and over , Antigens, CD/analysis , Granzymes , Homeostasis , Humans , Integrin alpha Chains/analysis , Interferon-gamma/genetics , Middle Aged , Serine Endopeptidases/genetics
13.
Gastroenterology ; 125(1): 40-6, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12851869

ABSTRACT

BACKGROUND & AIMS: Refractory celiac sprue, a low-grade intraepithelial lymphoma characterized by expansion of clonal intraepithelial lymphocytes with intracellular CD3 epsilon but no surface CD3-T-cell receptor complexes, can be an intermediary step between celiac disease and overt T-cell lymphoma. To gain insight into the mechanisms of lymphomagenesis in celiac disease, we have performed the first cytogenetic study in refractory celiac sprue. METHODS: Karyotypes were performed on: (1) 7 cell lines derived from clonal intraepithelial lymphocytes of patients with refractory celiac sprue; (2) 14 control T-cell lines, either from 4 of 7 patients with refractory celiac sprue or from 10 patients with uncomplicated celiac disease; and (3) bone marrow and peripheral blood lymphocytes in 1 of 7 patients with refractory celiac sprue. Rearrangements were confirmed by in situ hybridization using whole-chromosome painting probes and by comparative genomic hybridization in one patient. RESULTS: A recurrent structural chromosomal aberration leading to partial trisomy of the long arm of chromosome 1 was found in 6 of 7 cell lines from patients with refractory celiac sprue but in none of the control T-cell lines. In one patient with circulating abnormal intraepithelial lymphocytes, the partial trisomy 1q was confirmed on cells freshly isolated from bone marrow and blood. CONCLUSIONS: Refractory celiac sprue is strongly associated with partial trisomy of the 1q region. Gain of chromosome 1q, recently found in 16% of enteropathy-type T-cell lymphoma, may be an early event in lymphomagenesis related to celiac disease and provides a key to investigating molecular mechanisms of lymphoid transformation in this disease.


Subject(s)
Celiac Disease/genetics , Chromosomes, Human, Pair 1 , Lymphocytes/cytology , Trisomy , Adult , Aged , Celiac Disease/immunology , Cell Line , Clone Cells , Female , Gene Rearrangement , Genes, T-Cell Receptor gamma , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Male , Middle Aged , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...