Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Surg Res ; 208: 33-39, 2017 02.
Article in English | MEDLINE | ID: mdl-27993215

ABSTRACT

BACKGROUND: Sepsis remains a leading cause of death in most intensive care units. Many deaths in sepsis are due to nosocomial infections in patients who have entered the immunosuppressive phase of the disorder. One cause of immunosuppression in sepsis is T-cell exhaustion mediated by programmed cell death-1 (PD-1) interaction with its ligand (PD-L1). Studies demonstrated that blocking the interaction of PD-1 with PD-L1 with knockout mice or inhibitory antibodies reversed T-cell dysfunction and improved sepsis survival. This study assessed the efficacy of a novel short-acting peptide (compound 8) that inhibits PD-1:PD-L1 signaling in a clinically relevant second-hit fungal sepsis model. METHODS: Mice underwent cecal ligation and puncture to induce peritonitis. Three days later, mice received intravenous injection of Candida albicans. Forty-eight hours after Candida infection, mice were treated with compound 8 or inactive peptide. The effect of Candida infection on expression of coinhibitory molecules, PD-1, and PD-L1 were quantitated by flow cytometry on CD4+ cells, CD8+ cells, natural killer (NK) cells, and natural killer T-cells (NKT). The effect of compound 8 on survival was also examined. RESULTS: Four days after fungal infection, PD-1 and PD-L1 expressions were markedly increased on CD4+, NK, and NKT cells in septic versus sham-operated mice (%PD-1 on CD4+, 11.9% versus 2.8%; and %PD-L1 on NKT, 14.8% versus 0.5%). Compared with control, compound 8 caused a 2-fold increase in survival from 30% to 60%, P < 0.05. CONCLUSIONS: Compound 8 significantly improved survival in a clinically relevant immunosuppressive model of sepsis. These results support immunoadjuvant therapy targeting T-cell exhaustion in this lethal disease.


Subject(s)
B7-H1 Antigen/metabolism , Candidemia/drug therapy , Peptides/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Animals , Candidemia/metabolism , Drug Evaluation, Preclinical , Male , Mice , Peptides/pharmacology , Spleen/metabolism
2.
PLoS One ; 8(10): e77203, 2013.
Article in English | MEDLINE | ID: mdl-24204769

ABSTRACT

World conditions place large populations at risk from ionizing radiation (IR) from detonation of dirty bombs or nuclear devices. In a subgroup of patients, ionizing radiation exposure would be followed by a secondary infection. The effects of radiation combined injury are potentially more lethal than either insult in isolation. The purpose of this study was to determine mechanisms of mortality and possible therapeutic targets in radiation combined injury. Mice were exposed to IR with 2.5 Gray (Gy) followed four days later by intratracheal methicillin-resistant Staphylococcus aureus (MRSA). While either IR or MRSA alone yielded 100% survival, animals with radiation combined injury had 53% survival (p = 0.01). Compared to IR or MRSA alone, mice with radiation combined injury had increased gut apoptosis, local and systemic bacterial burden, decreased splenic CD4 T cells, CD8 T cells, B cells, NK cells, and dendritic cells, and increased BAL and systemic IL-6 and G-CSF. In contrast, radiation combined injury did not alter lymphocyte apoptosis, pulmonary injury, or intestinal proliferation compared to IR or MRSA alone. In light of the synergistic increase in gut apoptosis following radiation combined injury, transgenic mice that overexpress Bcl-2 in their intestine and wild type mice were subjected to IR followed by MRSA. Bcl-2 mice had decreased gut apoptosis and improved survival compared to WT mice (92% vs. 42%; p<0.01). These data demonstrate that radiation combined injury results in significantly higher mortality than could be predicted based upon either IR or MRSA infection alone, and that preventing gut apoptosis may be a potential therapeutic target.


Subject(s)
Apoptosis/immunology , Intestinal Mucosa/immunology , Radiation Injuries, Experimental/immunology , Staphylococcal Infections/immunology , Animals , Apoptosis/genetics , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , B-Lymphocytes/radiation effects , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/radiation effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Dendritic Cells/radiation effects , Gamma Rays , Gene Expression , Intestinal Mucosa/microbiology , Intestinal Mucosa/radiation effects , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/radiation effects , Lung/immunology , Lung/microbiology , Lung/radiation effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Radiation Injuries, Experimental/complications , Radiation Injuries, Experimental/mortality , Radiation Injuries, Experimental/pathology , Staphylococcal Infections/complications , Staphylococcal Infections/mortality , Staphylococcal Infections/pathology , Survival Analysis , Whole-Body Counting
3.
Crit Care ; 17(3): R85, 2013 May 11.
Article in English | MEDLINE | ID: mdl-23663657

ABSTRACT

INTRODUCTION: Fungal sepsis is an increasingly common problem in intensive care unit patients.Mortality from fungal sepsis remains high despite antimicrobial therapy that is highly active against most fungal pathogens, a finding consistent with defective host immunity that is present in many patients with disseminated fungemia.One recently recognized immunologic defect that occurs in patients with sepsis is T cell "exhaustion" due to increased expression of programmed cell death -1 (PD-1).This study tested the ability of anti-PD-1 and anti-programmed cell death ligand -1 (anti-PD-L1) antagonistic antibodies to improve survival and reverse sepsis-induced immunosuppression in two mouse models of fungal sepsis. METHODS: Fungal sepsis was induced in mice using two different models of infection, that is, primary fungal sepsis and secondary fungal sepsis occurring after sub-lethal cecal ligation and puncture (CLP).Anti-PD-1 and anti-PD-L1 were administered 24 to 48 h after fungal infection and effects on survival, interferon gamma production, and MHC II expression were examined. RESULTS: Anti-PD-1 and anti-PD-L1 antibodies were highly effective at improving survival in primary and secondary fungal sepsis.Both antibodies reversed sepsis-induced suppression of interferon gamma and increased expression of MHC II on antigen presenting cells.Blockade of cytotoxic T-lymphocyte antigen-4 (CTLA-4), a second negative co-stimulatory molecule that is up-regulated in sepsis and acts like PD-1 to suppress T cell function, also improved survival in fungal sepsis. CONCLUSIONS: Immuno-adjuvant therapy with anti-PD-1, anti-PD-L1 and anti-CTLA-4 antibodies reverse sepsis-induced immunosuppression and improve survival in fungal sepsis.The present results are consistent with previous studies showing that blockade of PD-1 and CTLA-4 improves survival in bacterial sepsis.Thus, immuno-adjuvant therapy represents a novel approach to sepsis and may have broad applicability in the disorder.Given the relative safety of anti-PD-1 antibody in cancer clinical trials to date, therapy with anti-PD-1 in patients with life-threatening sepsis who have demonstrable immunosuppression should be strongly considered.


Subject(s)
Antibodies/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Fungemia/immunology , Fungemia/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Candidiasis/immunology , Candidiasis/therapy , Disease Models, Animal , Genes, MHC Class I , HLA-DR Antigens/biosynthesis , Immunocompromised Host , Interferon-gamma/biosynthesis , Male , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Survival Analysis
4.
Am J Pathol ; 176(5): 2367-77, 2010 May.
Article in English | MEDLINE | ID: mdl-20304952

ABSTRACT

In the adult intestine, luminal microbiota induce cryptopatches to transform into isolated lymphoid follicles (ILFs), which subsequently act as sites for the generation of IgA responses. The events leading to this conversion are incompletely understood. Dendritic cells (DCs) are components of cryptopatches (CPs) and ILFs and were therefore evaluated in this process. We observed that the adult murine intestine contains clusters of DCs restricted to the CP/ILF continuum. A numerical and cell associative hierarchy in the adult intestine and a chronologic hierarchy in the neonatal intestine demonstrated that these clusters form after the coalescence of CD90+ cells to form CPs and before the influx of B220+ B lymphocytes to form ILFs. Cluster formation was dependent on lymphotoxin and the lymphotoxin beta receptor and independent of lymphocytes. The ILF DC population was distinguished from that of the lamina propria by the absence of CD4+CD11c+ cells and an increased proportion of CD11c+B220+ cells. The formation of clusters was not limited by DC numbers but was induced by luminal microbiota. Moreover, in the absence of the chemokine CXCL13, CP transformation into ILF was arrested. Furthermore, ILF DCs express CXCL13, and depletion of DCs resulted in regression of ILFs and disorganization of CPs. These results reveal DC participation in ILF transformation and maintenance and suggest that in part this may be due to CXCL13 production by these cells.


Subject(s)
Chemokine CXCL13/metabolism , Dendritic Cells/cytology , Gene Expression Regulation , Intestine, Small/metabolism , Lymphoid Tissue/metabolism , Animals , B-Lymphocytes/cytology , Flow Cytometry/methods , Immune System , Leukocyte Common Antigens/metabolism , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mucous Membrane/pathology , Thy-1 Antigens/biosynthesis
5.
J Immunol ; 184(7): 3768-79, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20200277

ABSTRACT

Sepsis is a highly lethal disorder characterized by widespread apoptosis-induced depletion of immune cells and the development of a profound immunosuppressive state. IL-7 is a potent antiapoptotic cytokine that enhances immune effector cell function and is essential for lymphocyte survival. In this study, recombinant human IL-7 (rhIL-7) efficacy and potential mechanisms of action were tested in a murine peritonitis model. Studies at two independent laboratories showed that rhIL-7 markedly improved host survival, blocked apoptosis of CD4 and CD8 T cells, restored IFN-gamma production, and improved immune effector cell recruitment to the infected site. Importantly, rhIL-7 also prevented a hallmark of sepsis (i.e., the loss of delayed-type hypersensitivity), which is an IFN-gamma- and T cell-dependent response. Mechanistically, rhIL-7 significantly increased the expression of the leukocyte adhesion markers LFA-1 and VLA-4, consistent with its ability to improve leukocyte function and trafficking to the infectious focus. rhIL-7 also increased the expression of CD8. The potent antiapoptotic effect of rhIL-7 was due to increased Bcl-2, as well as to a dramatic decrease in sepsis-induced PUMA, a heretofore unreported effect of IL-7. If additional animal studies support its efficacy in sepsis and if current clinical trials continue to confirm its safety in diverse settings, rhIL-7 should be strongly considered for clinical trials in sepsis.


Subject(s)
Apoptosis/drug effects , Chemotaxis, Leukocyte/immunology , Interleukin-17/immunology , Sepsis/immunology , T-Lymphocytes/immunology , Animals , Cell Survival , Chemotaxis, Leukocyte/drug effects , Humans , Hypersensitivity, Delayed/immunology , Interleukin-17/pharmacology , Mice , Mice, Inbred C57BL , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/drug therapy , T-Lymphocytes/drug effects
6.
J Leukoc Biol ; 86(2): 219-27, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19369639

ABSTRACT

Sepsis is the leading cause of death in critically ill patients in the United States with over 210,000 deaths annually. One stumbling block to an effective therapy of sepsis has been the lack of a clinically relevant animal model. There are important distinctions in the mouse versus human immune system regarding the host response to invading pathogens. These differences may explain the disappointing results in many sepsis clinical trials despite the clear efficacy of these agents in mouse models of sepsis. The purpose of the present study was to develop a "humanized" mouse model of sepsis and to determine if the model recapitulated the major findings of lymphocyte apoptosis and cytokine response that exist in patients with sepsis. Two-day-old NOD-scid IL2rgamma(null) mice received an adoptive transfer of hCD34(+) hematopoietic cord blood stem cells. These mice acquired a functional human innate and adaptive immune system, as evidenced by the development of all lineages of human immune cells as well as by mounting a DTH response. Eight weeks post-transfer, mice were made septic using the highly clinical relevant CLP model of sepsis, and sepsis induced marked elevations in human pro- and anti-inflammatory cytokines as well as a dramatic increase in human T and B cell apoptosis. Collectively, these results show that the humanized mouse model recapitulates many of the classic findings in patients with sepsis. Therefore, it represents an advanced, clinically relevant model for mechanistic studies of sepsis and testing of novel therapies.


Subject(s)
Apoptosis/immunology , Cytokines/metabolism , Hematopoietic Stem Cell Transplantation/methods , Inflammation/immunology , Lymphocytes/immunology , Systemic Inflammatory Response Syndrome/immunology , Adoptive Transfer/methods , Animals , Animals, Newborn , Cell Lineage/immunology , Cell Proliferation , Cells, Cultured , Cord Blood Stem Cell Transplantation/methods , Disease Models, Animal , Graft Survival/immunology , Hematopoietic Stem Cells/immunology , Humans , Immunity, Innate/immunology , Inflammation/physiopathology , Lymphocyte Activation/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Mice, SCID , Systemic Inflammatory Response Syndrome/physiopathology
7.
J Leukoc Biol ; 85(3): 382-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19088177

ABSTRACT

Sepsis is a severe, life-threatening infection and a leading cause of death in hospitals. A hallmark of sepsis is the profound apoptosis-induced depletion of lymphocytes generating a lymphopenic environment. As lymphopenia can induce nonantigen-driven homeostatic proliferation (HP), we examined this process during sepsis. CD4(+) and CD8(+) T cells, which were depleted within 24 h of sepsis induction, remained at significantly reduced levels until Day 21 when normal numbers were detected. When HP was examined, naïve CD8(+) T cells proliferated between Day 7 and Day 21 post-cecal ligation and puncture, developing into memory cells with relatively few cells expressing an activation phenotype. Conversely, naïve CD4(+) T cells did not undergo HP, but proportionally higher numbers expressed activation markers. Adoptive transfer studies revealed that T cells from mice that had recovered from sepsis were not protective when transferred to naïve mice undergoing sepsis. In addition, the TCR repertoire was not skewed toward any specific Vbeta type but resembled the repertoire found in normal mice, suggesting that T cells were not primed to antigens resulting from the infection. Interestingly, depletion of endogenous CD8(+) but not CD4(+) T cells restored the ability of naive CD4(+) T cells to undergo HP, increasing the number of CD4(+) T cells with memory but not activation markers. We conclude that homeostatic control in the postseptic environment permits recovery of the T cell repertoire to normal levels without generating antigen-specific memory or aberrant T cell specificities. Restoration of homeostatic control mechanisms might be a rational therapy for this disorder.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Homeostasis/immunology , Lymphopenia/immunology , Sepsis/immunology , Adoptive Transfer , Animals , Immunologic Memory , Lymphocyte Activation/immunology , Mice , Time Factors
8.
Shock ; 31(4): 359-66, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18838943

ABSTRACT

This study examined the fate of dendritic cells (DCs) and macrophages (M Phi) in vivo in a murine model of sepsis. Wild-type, knockout, and transgenic mice were used to examine the role of Bcl-2 family members on the regulation of splenic DCs and M Phi survival. Bim knockout (Bim) mice and mice overexpressing Bcl-2 in selected hematopoietic cells were used: (a) overexpression of Bcl-2 in all hematopoietic cells using a vav promoter (Vav-Bcl-2) and (b) overexpression of Bcl-2 in all Major histocompatibility complex (MHC) class I cells (H-2K-Bcl-2). Mice underwent sham surgery or cecal ligation and puncture, and absolute numbers of splenic DCs and M Phi were determined. Importantly, two distinct M Phi populations, that is, well-differentiated "mature" M Phi population and a less differentiated "immature," "monocyte-like" (IM Phi) population were identified that demonstrated differential susceptibility to apoptosis. In wild-type mice, sepsis induced a 64% +/- 7% and a 77% +/- 3% decrease in absolute cell numbers of splenic DCs and IM Phi, respectively (n = 7, P < 0.05). Mature M Phi were not depleted in sepsis. No significant cell depletion was evident in Vav-Bcl-2, H-2K-Bcl-2, or Bim mice. We conclude that sepsis induces a major depletion of developing M Phi as well as DCs, and this depletion may be an important mechanism of immune suppression in sepsis.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , Proto-Oncogene Proteins c-bcl-2/physiology , Sepsis/immunology , Animals , Dendritic Cells/pathology , Flow Cytometry , Gene Expression Regulation , Lymphocyte Depletion , Macrophages/pathology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/genetics , Sepsis/pathology , Spleen/immunology , Spleen/pathology
9.
Shock ; 32(2): 131-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19033888

ABSTRACT

Immune suppression is a major cause of morbidity and mortality in the patients with sepsis. Apoptotic loss of immune effector cells such as CD4 T and B cells is a key component in the loss of immune competence in sepsis. Inhibition of lymphocyte apoptosis has led to improved survival in animal models of sepsis. Using quantitative real-time polymerase chain reaction of isolated splenic CD4 T and B cells, we determined that Bim and PUMA, two key cell death proteins, are markedly upregulated during sepsis. Lymphocytes have been notoriously difficult to transfect with small interfering RNA (siRNA). Consequently a novel, cyclodextrin polymer-based, transferrin receptor-targeted, delivery vehicle was used to coadminister siRNA to Bim and PUMA to mice immediately after cecal ligation and puncture. Antiapoptotic siRNA-based therapy markedly decreased lymphocyte apoptosis and prevented the loss of splenic CD4 T and B cells. Flow cytometry confirmed in vivo delivery of siRNA to CD4 T and B cells and also demonstrated decreases in intracellular Bim and PUMA protein. In conclusion, Bim and PUMA are two critical mediators of immune cell death in sepsis. Use of a novel cyclodextrin polymer-based, transferrin receptor-targeted siRNA delivery vehicle enables effective administration of antiapoptotic siRNAs to lymphocytes and reverses the immune cell depletion that is a hallmark of this highly lethal disorder.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , RNA, Small Interfering/immunology , Sepsis/immunology , Transfection/methods , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , B-Lymphocytes/immunology , Bcl-2-Like Protein 11 , Cellulose/pharmacology , Cyclodextrins/pharmacology , Humans , Lymphocyte Depletion , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Transferrin/agonists , Receptors, Transferrin/genetics , Receptors, Transferrin/immunology , Sepsis/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
10.
J Immunol ; 181(6): 4052-61, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768861

ABSTRACT

The alpha(4) integrins alpha(4)beta(7) and alpha(4)beta(1), and their ligands mucosal vascular addressin cell adhesion molecule 1 (MAdCAM-1) and VCAM-1, have diverse functions, including roles in the formation of secondary lymphoid tissues at early time points during the colonization and clustering of the fetal lymphoid tissue inducer (LTi) cells and at later time points during the recruitment of lymphocytes. In this study, we evaluated the role of alpha(4) integrins in the development of a recently appreciated class of intestinal lymphoid tissues, isolated lymphoid follicles (ILFs). We observed that diverse ILF cellular populations express alpha(4)beta(7) and alpha(4)beta(1), including the LTi-like cells and lymphocytes, while ILF stromal cells and vessels within ILFs express VCAM-1 and MAdCAM-1, respectively. Evaluation of adult and neonatal beta(7)(-/-) mice and adult and neonatal mice given blocking Abs to alpha(4)beta(7), MAdCAM-1, or VCAM-1 did not identify a role for alpha(4) integrins in cryptopatch (CP) development; however, these studies demonstrated that alpha(4)beta(7) and MAdCAM-1 are required for the transitioning of CP into lymphoid tissues containing lymphocytes or ILFs. Competitive bone marrow transfers demonstrated that beta(7)(-/-) LTi-like cells had a reduced but not significantly impaired ability to localize to CP. Bone marrow transfers and adoptive transfers of B lymphocytes revealed that beta(7) expression by B lymphocytes was essential for their entry into the developing ILFs. These findings demonstrate an essential role for alpha(4)beta(7)/MAdCAM-1 in ILF development corresponding to the influx of beta(7)-expressing lymphocytes and a nonessential role for beta(7)-localizing LTi-like cells to the small intestine.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Communication/immunology , Cell Movement/immunology , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Integrins/metabolism , Peyer's Patches/immunology , Animals , Animals, Newborn , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/physiology , Cell Aggregation/immunology , Integrin alpha Chains/physiology , Integrin beta Chains/biosynthesis , Integrin beta Chains/genetics , Integrin beta Chains/physiology , Integrins/biosynthesis , Integrins/physiology , Intestinal Mucosa/blood supply , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mucoproteins , Peyer's Patches/blood supply , Peyer's Patches/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
11.
Shock ; 30(2): 127-34, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18197142

ABSTRACT

To assess the degree of lymphocyte apoptosis and survival in mice treated with small interfering RNA (siRNA) targeted to Bim, a proapoptotic molecule from the Bcl-2 family, within a clinically relevant model of sepsis. C57BL/6 mice were treated with a single dose of Bim siRNA complexed in cationic liposomes via tail vein injection. Approximately 24 h later, mice were subjected to either cecal ligation and puncture (CLP) or sham surgery. Animals were killed at 20 h postsurgery, and spleens were harvested for fluorescence-activated cell sorting analysis using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling as a marker for apoptosis. A second cohort of mice was followed for survival for 7 days. The degree of lymphocyte apoptosis in Bim siRNA-treated mice was markedly decreased compared with controls. Fluorescent activated cell sorter analysis demonstrated 13.1% +/- 1.2% B-cell apoptosis and 11.5% +/- 1.5% T-cell apoptosis in control mice compared with 2.7% +/- 0.4% B-cell apoptosis and 3.9% +/- 0.3% T-cell apoptosis in Bim siRNA-treated mice after CLP (P < 0.001 and P < 0.01, respectively). This striking difference in lymphocyte apoptosis correlated with a significant survival advantage in Bim siRNA-treated mice. At 7 days, there was 90% overall survival in Bim siRNA-treated septic mice compared with 50% overall survival in control septic mice (P < 0.05). Treatment with Bim siRNA in vivo has the potential to be an effective therapy in the treatment of sepsis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Lymphocytes/pathology , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/physiology , Sepsis/mortality , Sepsis/therapy , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/deficiency , Bcl-2-Like Protein 11 , Cells, Cultured , Disease Models, Animal , Gamma Rays , Humans , Lymphocytes/metabolism , Lymphocytes/radiation effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peritonitis/metabolism , Peritonitis/mortality , Peritonitis/therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/deficiency , Sepsis/metabolism , Survival Analysis
12.
Am J Pathol ; 170(4): 1229-40, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392163

ABSTRACT

Isolated lymphoid follicles (ILFs) are organized lymphoid structures that facilitate the efficient interaction of antigen, antigen-presenting cells, and lymphocytes to generate controlled adaptive immune responses within the intestine. Because CC chemokine receptor 6 (CCR6) deficiency affects the generation of mucosal immune responses, we evaluated a potential role for CCR6 in the development of ILFs. We observed that CCR6 and its ligand CCL20 are highly expressed within ILFs and that B lymphocytes are the largest CCR6-expressing population within ILFs. ILF development was profoundly arrested in the absence of CCR6. Concordant with a block in ILF development at a stage corresponding to the influx of B lymphocytes, we observed that CCR6-deficient mice had a diminished population of intestinal B lymphocytes. Bone marrow reconstitution studies demonstrated that ILF development is dependent on CCR6-sufficient B lymphocytes, and adoptive transfers demonstrated that CCR6(-/-) B lymphocytes were inefficient at localizing to intestinal lymphoid structures. Paralleling these findings, we observed that CCR6-deficient mice had a reduced proportion of Peyer's patch B lymphocytes and an associated re-duction in the number and size of Peyer's patch follicular domes. These observations define an essential role for CCR6 expression by B lymphocytes in localizing to intestinal lymphoid structures and in ILF development.


Subject(s)
B-Lymphocytes/metabolism , Peyer's Patches/metabolism , Receptors, Chemokine/metabolism , Adoptive Transfer/methods , Animals , B-Lymphocytes/cytology , B-Lymphocytes/transplantation , Chemokine CCL20 , Chemokines, CC/metabolism , Female , Flow Cytometry , Gastrointestinal Tract/metabolism , Immunohistochemistry , Lymphocyte Count , Lymphoid Tissue/growth & development , Lymphoid Tissue/metabolism , Macrophage Inflammatory Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Peyer's Patches/growth & development , Receptors, CCR6 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Thy-1 Antigens/metabolism
13.
Am J Physiol Gastrointest Liver Physiol ; 291(4): G595-604, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16782693

ABSTRACT

Isolated lymphoid follicles (ILFs) are organized intestinal lymphoid structures whose formation can be induced by luminal stimuli. ILFs have been demonstrated to act as inductive sites for the generation of immune responses directed toward luminal stimuli; however, the phenotype of the immune response initiated within ILFs has largely been uninvestigated. To gain a better understanding of the immune responses initiated within ILFs, we examined phenotypic and functional aspects of the largest cellular component of the murine ILF lymphocyte population, B lymphocytes. We observed that murine ILF B lymphocytes are composed of a relatively homogenous population of follicular B-2 B lymphocytes. Consistent with their proximity to multiple stimuli, ILF B lymphocytes displayed a more activated phenotype compared with their counterparts in the spleen and Peyer's patch (PP). ILF B lymphocytes also expressed higher levels of immunomodulatory B7 and CD28 family members B7X and programmed death-1 compared with their counterparts in the spleen and PP. ILF B lymphocytes preferentially differentiate into IgA-producing plasma cells and produce more IL-4 and IL-10 and less interferon-gamma compared with their counterparts in the spleen. Immunoglobulin repertoire analysis from individual ILFs demonstrated that ILFs contain a polyclonal population of B lymphocytes. These findings indicate that murine ILFs contain a polyclonal population of follicular B-2 B lymphocytes with a phenotype similar to PP B lymphocytes and that, in unchallenged animals, ILFs promote immune responses with a homeostatic phenotype.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Immunity, Mucosal/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Animals , Antigens, CD/metabolism , Cell Differentiation , Cytokines/metabolism , Female , Gene Expression Regulation , Immunoglobulin A/metabolism , Mice , Mice, Inbred BALB C , Plasma Cells/metabolism
14.
J Immunol ; 174(9): 5720-8, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843574

ABSTRACT

Isolated lymphoid follicles (ILFs) are recently appreciated members of the mucosal immune system. The architecture, composition, and inducible nature of these structures indicates that these structures are tertiary lymphoid structures. The process leading to the formation of tertiary lymphoid structures, lymphoid neogenesis, has been observed in a number of inflammatory and autoimmune conditions. Given this association, there is considerable interest in identifying the factors promoting lymphoid neogenesis, and understanding the steps in this process. Using murine ILF formation as a model, we have examined the roles of different cellular sources of lymphotoxin (LT) and the adaptive immune response in lymphoid neogenesis. In this study, we report that, although other cellular sources of LT may supplant B lymphocytes in the formation of immature ILFs (loosely organized clusters of B lymphocytes), LT-sufficient B lymphocytes are required for the progression of immature ILFs to mature ILFs (organized lymphoid aggregates with a follicle-associated epithelium). ILF formation occurs in the absence of T lymphocytes and Ag-specific B lymphocyte responses, and ILF B lymphocytes express elevated levels of LT in the absence of antigenic stimulation. Consistent with a role for chemokines inducing LT expression in Ag-naive B lymphocytes, and a chemokine-driven positive-feedback loop driving mature ILF formation, mature ILFs express elevated levels of B lymphocyte chemoattractant in the absence of Ag-specific B lymphocyte stimulation. These observations indicate that ILFs contain Ag-naive lymphocytes, and suggest that events occurring within ILFs shape subsequent immune responses mediated by these lymphocytes.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Epitopes, B-Lymphocyte/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphotoxin-alpha/physiology , Animals , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Chemokines/biosynthesis , Epitopes, B-Lymphocyte/genetics , Female , Homeostasis/genetics , Homeostasis/immunology , Immunity, Innate/genetics , Immunophenotyping , Intestine, Small/cytology , Intestine, Small/immunology , Intestine, Small/metabolism , Lymphocyte Activation/genetics , Lymphoid Tissue/metabolism , Lymphotoxin-alpha/deficiency , Lymphotoxin-alpha/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology
15.
J Immunol ; 170(11): 5475-82, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12759424

ABSTRACT

The gastrointestinal mucosa contains a complex network of lymphoid compartments that have evolved to efficiently protect the host from invading pathogens. Recently, an additional lymphoid structure resembling Peyer's patches (PP) in composition and architecture has been identified in the murine small intestine, the isolated lymphoid follicle (ILF). In this study we examine the nature and factors required for ILF formation. We observed a spectrum of structures fitting the previous descriptions of ILFs, ranging from clusters of B220(+) cells (which we have termed immature ILFs) to well-organized lymphoid nodules (which we have termed mature ILFs). Here we demonstrate that that similar to PP formation, ILF formation requires lymphotoxin (LT)- and LT beta receptor-dependent events. However unlike PP formation, the LT- and LT beta receptor-dependent events required for ILF formation can occur in adulthood and require LT-sufficient B lymphocytes. We demonstrate that mature ILF formation occurs in response to lumenal stimuli, including normal bacterial flora, and requires TNF receptor I function. These findings suggest that ILFs are organized intestinal lymphoid structures whose formation can be induced and whose mass can be expanded in response to mucosal challenges.


Subject(s)
Antigens, CD/physiology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Lymphoid Tissue/growth & development , Lymphoid Tissue/immunology , Lymphotoxin-alpha/physiology , Receptors, Tumor Necrosis Factor/physiology , Aging/genetics , Aging/immunology , Animals , Antigens, CD/genetics , B-Lymphocyte Subsets/cytology , Cell Aggregation/genetics , Cell Aggregation/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/growth & development , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Leukocyte Common Antigens/biosynthesis , Lymphoid Tissue/metabolism , Lymphoid Tissue/microbiology , Lymphotoxin beta Receptor , Lymphotoxin-alpha/biosynthesis , Lymphotoxin-alpha/deficiency , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Stromal Cells/immunology , Stromal Cells/metabolism
16.
J Immunol ; 168(10): 4988-97, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11994450

ABSTRACT

Lymphotoxin (LT), a cytokine belonging to the TNF family, has established roles in the formation of secondary lymphoid structures and in the compartmentalization of T and B lymphocyte areas of the spleen. In this study, we examine the role of LT in directing the composition of intestinal lymphocytes. We report that mice deficient in LT have a normal composition of intestinal lamina propria (LP) T lymphocytes, and an absence of intestinal LP B lymphocytes. We further refine this observation to demonstrate that the interaction of LT with the LTbetaR is essential for the presence LP B lymphocytes. The LT/LTbetaR-dependent events relevant for the presence of LP B lymphocytes occur after birth, do not require the presence of Peyer's patches, lymph nodes, or the spleen; and therefore, are distinct and independent from the previously identified roles of LT/LTbetaR. The LT-dependent signal relevant for the presence of LP B lymphocytes is optimally supplied by a LT-sufficient B lymphocyte, and requires a LTbetaR-sufficient radio-resistant, non-bone marrow-derived cell. Based upon the severity of the deficit of LP B lymphocytes we observed, these novel LT/LTbetaR-dependent events are of primary importance in directing the entry and residence of LP B lymphocytes.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphotoxin-alpha/metabolism , Lymphotoxin-alpha/physiology , Membrane Proteins/metabolism , Postpartum Period/immunology , Receptors, Tumor Necrosis Factor/physiology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/radiation effects , Bone Marrow Cells/immunology , Female , Gamma Rays , Intestinal Mucosa/cytology , Intestinal Mucosa/radiation effects , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/radiation effects , Lymphotoxin beta Receptor , Lymphotoxin-beta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Radiation Chimera/immunology , Radiation Tolerance/immunology , Receptors, Tumor Necrosis Factor/radiation effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL