Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Crit Rev Immunol ; 40(6): 485-496, 2020.
Article in English | MEDLINE | ID: mdl-33900693

ABSTRACT

In December 2019, outbreak of a novel coronavirus flared in Wuhan, the capital city of Hubei province, China. The identified pathogen was an enveloped RNA betacoronavirus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The outbreak was declared a pandemic by the World Health Organization (WHO), because the continual spread of this deadly and highly infectious virus is a health emergency for all world nations. SARS-CoV-2 is associated with severe atypical pneumonia coronavirus disease-19. Typical symptoms of this disease include fever, malaise, cough, shortness of breath, and in severe cases, death. As the virus continues to invade host cells deep into alveoli, infection severity mostly depends on the undeterred immune response that is triggered by elevated levels of inflammation-inducing cytokines, called a cytokine storm. In this article, we provide a comprehensive review of the viral life cycle and immunological responses associated with the SARS-CoV-2 infection.


Subject(s)
COVID-19/etiology , Disease Susceptibility , Host-Pathogen Interactions/immunology , SARS-CoV-2/physiology , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/therapy , Cytokine Release Syndrome/diagnosis , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/therapy , Disease Susceptibility/immunology , Humans , Immune Evasion , Immunity, Cellular , Immunity, Innate , Severity of Illness Index
2.
Gut ; 65(3): 456-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25645662

ABSTRACT

OBJECTIVE: ATG16L1 is an autophagy gene known to control host immune responses to viruses and bacteria. Recently, a non-synonymous single-nucleotide polymorphism in ATG16L1 (Thr300Ala), previously identified as a risk factor in Crohn's disease (CD), was associated with more favourable clinical outcomes in thyroid cancer. Mechanisms underlying this observation have not been proposed, nor is it clear whether an association between Thr300Ala and clinical outcomes will be observed in other cancers. We hypothesised that Thr300Ala influences clinical outcome in human colorectal cancer (CRC) and controls innate antiviral pathways in colon cancer cells. DESIGN: We genotyped 460 patients with CRC and assessed for an association between ATG16L1 Thr300Ala and overall survival and clinical stage. Human CRC cell lines were targeted by homologous recombination to examine the functional consequence of loss of ATG16L1, or introduction of the Thr300Ala variant. RESULTS: We found an association between longer overall survival, reduced metastasis and the ATG16L1 Ala/Ala genotype. Tumour sections from ATG16L1 Ala/Ala patients expressed elevated type I interferons (IFN-I)-inducible, MxA, suggesting that differences in cytokine production may influence disease progression. When introduced into human CRC cells by homologous recombination, the Thr300Ala variant did not affect bulk autophagy, but increased basal production of type I IFN. Introduction of Thr300Ala resulted in increased sensitivity to the dsRNA mimic poly(I:C) through a mitochondrial antiviral signalling (MAVS)-dependent pathway. CONCLUSIONS: The CD-risk allele, Thr300Ala, in ATG16L1 is associated with improved overall survival in human CRC, generating a rationale to genotype ATG16L1 Thr300Ala in patients with CRC. We found that Thr300A alters production of MAVS-dependent type I IFN in CRC cells, providing a mechanism that may influence clinical outcomes.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carrier Proteins/genetics , Colorectal Neoplasms/genetics , Interferon Type I/metabolism , Polymorphism, Single Nucleotide , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Autophagy-Related Proteins , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Female , Follow-Up Studies , Humans , Male , Middle Aged , Survival Analysis
3.
Inflamm Bowel Dis ; 21(10): 2393-2402, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26197453

ABSTRACT

BACKGROUND: A peptide derived from Antrum Mucosal Protein (AMP)-18 (gastrokine-1) reduces the extent of mucosal erosions and clinical severity in mice with dextran sulfate sodium-induced colonic injury. This study set out to determine if AMP peptide was also therapeutic for immune- and cytokine-mediated mouse models of intestinal injury and inflammatory bowel diseases by enhancing and stabilizing tight junctions. METHODS: Therapeutic effects of AMP peptide were examined in interleukin-10-deficient and a T-cell adoptive transfer models of colitis in immunodeficient recombinase activating gene-1 knock-out (RAG-1-/-) mice. Mechanisms by which AMP peptide enhances barrier function and structure were studied ex vivo using intestine and colon from mice given lipopolysaccharide and in AMP-18-deficient mice given dextran sulfate sodium. RESULTS: In interleukin-10-deficient mice given piroxicam, AMP peptide enhanced recovery after weight loss, protected against colon shortening and segmental dilation, and reduced the colitis activity score. In the T-cell transfer model, treatment with the peptide protected against colon shortening. In mice given lipopolysaccharide in vivo to induce gut injury, AMP peptide prevented the onset of, and reversed established intestinal hyperpermeability by targeting TJ proteins and perijunctional actin. AMP-18-deficient mice challenged with dextran sulfate sodium exhibited increased mortality, developed erosions in the colon, and had lower levels of ZO-1 in TJs than heterozygous littermates or wild-type mice. CONCLUSIONS: The results indicate that AMP-18/peptide may serve a protective role against injury along the gastrointestinal mucosal barrier, and recommend further development of AMP peptide as a novel agent to treat patients with inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Peptide Hormones/pharmacology , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Actins/metabolism , Adoptive Transfer , Animals , Anti-Inflammatory Agents, Non-Steroidal , Colitis/chemically induced , Colitis/metabolism , Colon/injuries , Cytokines/metabolism , Dextran Sulfate/toxicity , Interleukin-10/deficiency , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Peptide Hormones/genetics , Peptide Hormones/metabolism , Permeability/drug effects , Piroxicam , Protective Agents/pharmacology , Severity of Illness Index
4.
Am J Physiol Gastrointest Liver Physiol ; 307(9): G871-82, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25234043

ABSTRACT

Tumor necrosis factor-induced protein 3 (TNFAIP3; also known as A20) negatively regulates NF-κB and MAPK signals to control inflammatory responses. TNFAIP3 also protects against TNF-induced cell death. Intestinal epithelial cell (IEC) expression of TNFAIP3 improves barrier function and tight junction integrity and prevents dextran sulfate sodium (DSS)-induced IEC death and colitis. We therefore investigated the effects of TNFAIP3 expression in IEC on immune homeostasis in the intestines of immune-compromised mice. Villin-TNFAIP3 (v-TNFAIP3) transgenic mice were interbred with IL-10(-/-) mice (v-TNFAIP3 × IL-10(-/-)) and incidence, onset, and severity of colitis was assessed. v-TNFAIP3 × IL-10(-/-) mice displayed severe, early onset, and highly penetrant colitis that was not observed in IL-10(-/-) or v-TNFAIP3 mice. V-TNFAIP3 mice displayed altered expression of mucosal cytokines, increased numbers of mucosal regulatory T cells, and altered expression of mucosal antimicrobial peptides (AMPs). Microbial colonization of the inner mucus layer of v-TNFAIP3 mice was observed, along with alterations in the microbiome, but this was not sufficient to induce colitis in v-TNFAIP3 mice. The relative sterility of the inner mucus layer observed in wild-type and IL-10(-/-) mice was lost in v-TNFAIP3 × IL-10(-/-) mice. Thus IEC-derived factors, induced by signals that are inhibited by TNFAIP3, suppress the onset of inflammatory bowel disease in IL-10(-/-) mice. Our results indicate that IEC expression of TNFAIP3 alters AMP expression and allows microbial colonization of the inner mucus layer, which activates an IL-10-dependent anti-inflammatory process that is necessary to prevent colitis.


Subject(s)
Colitis, Ulcerative/metabolism , Cysteine Endopeptidases/metabolism , Interleukin-10/genetics , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microbiota , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Cysteine Endopeptidases/genetics , Gene Deletion , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/genetics , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Pancreatitis-Associated Proteins , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
5.
J Biochem Pharmacol Res ; 1(3): 161-167, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24286022

ABSTRACT

Linear polyubiquitin is processed at LRLRGG sequences by deubiquitinating enzymes to make free monomeric ubiquitin. This LRLRGG ubiquitin-like motif is found in a limited number of mammalian non-ubiquitin proteins, including the MAP3K Apoptosis Signal-Regulating Kinase-1 (ASK1), which activates MAPK signaling pathways. The c-terminus of ASK1 binds to the 19S cap of the proteasome allowing ASK1 to phosphorylate and inhibit proteasomal activity. We investigated whether the ubiquitin-like sequence in the c-terminus of ASK1 mediates its association with and inhibition of the proteasome. To test this we generated ASK1 with substitutions or deletions in this ubiquitin-like domain and examined the activation of cellular signaling and the association of ASK1 with the 19S cap of the proteasome. We show that ASK1 mutants have reduced association with the 19S cap of the proteasome, reduced capacity to inhibit the proteasome, and diminished ability to inhibit TNF-induced NF-κB activation. Mutant forms of ASK1 also had reduced capacity to activate JNK signaling, suggesting that the ubiquitin-like motif in ASK1 is also important for coordinating the balance between JNK and NF-κB signaling. Together these results demonstrate that the ubiquitin-like sequence of ASK1 is important for binding to and inhibition of the proteasome, and for the coordinated activation of cellular NF-κB and JNK signaling.

6.
BMJ Open ; 3(6)2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23794574

ABSTRACT

OBJECTIVE: A common genetic coding variant in the core autophagy gene ATG16L1 is associated with increased susceptibility to Crohn's disease (CD). The variant encodes an amino acid change in ATG16L1 such that the threonine at position 300 is substituted with an alanine (ATG16L1 T300A). How this variant contributes to increased risk of CD is not known, but studies with transfected cell lines and gene-targeted mice have demonstrated that ATG16L1 is required for autophagy, control of interleukin-1-ß and autophagic clearance of intracellular microbes. In addition, studies with human cells expressing ATG16L1 T300A indicate that this variant reduces the autophagic clearance of intracellular microbes. DESIGN/RESULTS: We demonstrate, using somatically gene-targeted human cells that the ATG16L1 T300A variant confers protection from cellular invasion by Salmonella. In addition, we show that ATG16L1-deficient cells are resistant to bacterial invasion. CONCLUSIONS: These results suggest that cellular expression of ATG16L1 facilitates bacterial invasion and that the CD-associated ATG16L1 T300A variant may confer protection from bacterial infection.

7.
Am J Physiol Gastrointest Liver Physiol ; 303(2): G220-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22595989

ABSTRACT

Intestinal epithelial cells (IEC) maintain gastrointestinal homeostasis by providing a physical and functional barrier between the intestinal lumen and underlying mucosal immune system. The activation of NF-κB and prevention of apoptosis in IEC are required to maintain the intestinal barrier and prevent colitis. How NF-κB activation in IEC prevents colitis is not fully understood. TNFα-induced protein 3 (TNFAIP3) is a NF-κB-induced gene that acts in a negative-feedback loop to inhibit NF-κB activation and also to inhibit apoptosis; therefore, we investigated whether TNFAIP3 expression in the intestinal epithelium impacts susceptibility of mice to colitis. Transgenic mice expressing TNFAIP3 in IEC (villin-TNFAIP3 Tg mice) were exposed to dextran sodium sulfate (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS), and the severity and characteristics of mucosal inflammation and barrier function were compared with wild-type mice. Villin-TNFAIP3 Tg mice were protected from DSS-induced colitis and displayed reduced production of NF-κB-dependent inflammatory cytokines. Villin-TNFAIP3 Tg mice were also protected from DSS-induced increases in intestinal permeability and induction of IEC death. Villin-TNFAIP3 Tg mice were not protected from colitis induced by TNBS. These results indicate that TNFAIP3 expression in IEC prevents colitis involving DSS-induced IEC death, but not colitis driven by T cell-mediated inflammation. As TNFAIP3 inhibits NF-κB activation and IEC death, expression of TNFAIP3 in IEC may provide an avenue to inhibit IEC NF-κB activation without inducing IEC death and inflammation.


Subject(s)
Colitis/metabolism , Cysteine Endopeptidases/metabolism , Dextran Sulfate/adverse effects , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Trinitrobenzenesulfonic Acid/toxicity , Animals , Apoptosis/drug effects , Colitis/chemically induced , Cytokines/biosynthesis , Intestinal Mucosa/drug effects , Mice , Mice, Transgenic , NF-kappa B/metabolism , Severity of Illness Index , Tumor Necrosis Factor alpha-Induced Protein 3
8.
PLoS One ; 6(10): e26352, 2011.
Article in English | MEDLINE | ID: mdl-22031828

ABSTRACT

Tight junctions between intestinal epithelial cells mediate the permeability of the intestinal barrier, and loss of intestinal barrier function mediated by TNF signaling is associated with the inflammatory pathophysiology observed in Crohn's disease and celiac disease. Thus, factors that modulate intestinal epithelial cell response to TNF may be critical for the maintenance of barrier function. TNF alpha-induced protein 3 (TNFAIP3) is a cytosolic protein that acts in a negative feedback loop to regulate cell signaling induced by Toll-like receptor ligands and TNF, suggesting that TNFAIP3 may play a role in regulating the intestinal barrier. To investigate the specific role of TNFAIP3 in intestinal barrier function we assessed barrier permeability in TNFAIP3(-/-) mice and LPS-treated villin-TNFAIP3 transgenic mice. TNFAIP3(-/-) mice had greater intestinal permeability compared to wild-type littermates, while villin-TNFAIP3 transgenic mice were protected from increases in permeability seen within LPS-treated wild-type littermates, indicating that barrier permeability is controlled by TNFAIP3. In cultured human intestinal epithelial cell lines, TNFAIP3 expression regulated both TNF-induced and myosin light chain kinase-regulated tight junction dynamics but did not affect myosin light chain kinase activity. Immunohistochemistry of mouse intestine revealed that TNFAIP3 expression inhibits LPS-induced loss of the tight junction protein occludin from the apical border of the intestinal epithelium. We also found that TNFAIP3 deubiquitinates polyubiquitinated occludin. These in vivo and in vitro studies support the role of TNFAIP3 in promoting intestinal epithelial barrier integrity and demonstrate its novel ability to maintain intestinal homeostasis through tight junction protein regulation.


Subject(s)
Cysteine Endopeptidases/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Tight Junctions/metabolism , Animals , Cell Line , Cysteine Endopeptidases/genetics , DNA-Binding Proteins , HCT116 Cells , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Tight Junctions/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/metabolism
9.
J Immunol ; 184(12): 7001-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483768

ABSTRACT

The TNF alpha-induced protein 3 (TNFAIP3) is an ubiquitin-modifying enzyme and an essential negative regulator of inflammation. Genome-wide association studies have implicated the TNFAIP3 locus in susceptibility to autoimmune disorders in European cohorts, including rheumatoid arthritis, coronary artery disease, psoriasis, celiac disease, type 1 diabetes, inflammatory bowel disease, and systemic lupus erythematosus (SLE). There are two nonsynonymous coding polymorphisms in the deubiquitinating (DUB) domain of TNFAIP3: F127C, which is in high-linkage disequilibrium with reported SLE-risk variants, and A125V, which has not been previously studied. We conducted a case-control study in African-American SLE patients using these coding variants, along with tagging polymorphisms in TNFAIP3, and identified a novel African-derived risk haplotype that is distinct from previously reported risk variants (odds ratio=1.6, p=0.006). In addition, a rare protective haplotype was defined by A125V (odds ratio=0.31, p=0.027). Although A125V was associated with protection from SLE, surprisingly the same allele was associated with increased risk of inflammatory bowel disease. We tested the functional activity of nonsynonymous coding polymorphisms within TNFAIP3, and found that the A125V coding-change variant alters the DUB activity of the protein. Finally, we used computer modeling to depict how the A125V amino acid change in TNFAIP3 may affect the three-dimensional structure of the DUB domain to a greater extent than F127C. This is the first report of an association between TNFAIP3 polymorphisms and autoimmunity in African-Americans.


Subject(s)
Autoimmunity/genetics , Genetic Predisposition to Disease , Intracellular Signaling Peptides and Proteins/genetics , Lupus Erythematosus, Systemic/genetics , Nuclear Proteins/genetics , Black or African American/genetics , Case-Control Studies , DNA-Binding Proteins , Genome-Wide Association Study , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/chemistry , Nuclear Proteins/chemistry , Polymerase Chain Reaction , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Protein Structure, Quaternary , Transfection , Tumor Necrosis Factor alpha-Induced Protein 3
10.
Biochem Biophys Res Commun ; 394(2): 254-9, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20188700

ABSTRACT

Gastrointestinal (GI) anthrax, caused by the bacterial infection of Bacillus anthracis, posts a significant bioterrorism threat by its relatively high mortality rate in humans. Different from inhalational anthrax by the route of infection, accumulating evidence indicates the bypass of vegetative bacteria across GI epithelium is required to initiate GI anthrax. Previously, we reported that purified anthrolysin O (ALO), instead of tripartite anthrax edema and lethal toxins, is capable of disrupting gut epithelial tight junctions and barrier function in cultured cells. Here, we show that ALO can disrupt intestinal tissue barrier function in an ex vivo mouse model. To explore the effects of ALO in a cell culture model of B. anthracis infection, we showed that anthrax bacteria can effectively reduce the monolayer integrity of human Caco-2 brush-border expressor (C2BBE) cells based on the reduced transepithelial resistance and the increased leakage of fluorescent dye. This disruption is likely caused by tight junction dysfunction observed by the reorganization of the tight junction protein occludin. Consequently, we observe significant passage of vegetative anthrax bacteria across C2BBE cells. This barrier disruption and bacterial crossover requires ALO since ALO-deficient B. anthracis strains fail to induce monolayer dysfunction and allow the passage of anthrax bacteria. Together these findings point to a pivotal role for ALO within the establishment of GI anthrax infection and the initial bypass of the epithelial barrier.


Subject(s)
Anthrax/pathology , Bacillus anthracis/pathogenicity , Bacterial Proteins/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/pathology , Membrane Glycoproteins/metabolism , Animals , Anthrax/metabolism , Anthrax/microbiology , Bacillus anthracis/metabolism , Cell Line , Disease Models, Animal , Female , Humans , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , Intestines/pathology , Mice , Mice, Inbred C57BL , Protein Transport , Tight Junctions/metabolism , Tight Junctions/microbiology , Tight Junctions/pathology
11.
J Exp Med ; 197(8): 977-84, 2003 Apr 21.
Article in English | MEDLINE | ID: mdl-12695489

ABSTRACT

Natural killer (NK) cells protect hosts against viral pathogens and transformed cells. IL-15 is thought to play a critical role in NK cell development, but its role in the regulation of peripheral NK cells is less well defined. We now find that adoptive transfer of normal NK cells into mice lacking the high affinity interleukin (IL)-15 receptor, IL-15Ralpha, surprisingly results in the abrupt loss of these cells. Moreover, IL-15Ralpha-deficient NK cells can differentiate successfully in radiation bone marrow chimera bearing normal cells. Finally, adoptively transferred IL-15Ralpha-deficient NK cells survive in normal but not IL-15Ralpha-deficient mice. These findings demonstrate that NK cell-independent IL-15Ralpha expression is critical for maintaining peripheral NK cells, while IL-15Ralpha expression on NK cells is not required for this function.


Subject(s)
Cell Survival/physiology , Killer Cells, Natural/metabolism , Receptors, Interleukin-2/metabolism , Adoptive Transfer , Animals , Homeostasis , Interleukin-15/genetics , Interleukin-15/metabolism , Killer Cells, Natural/cytology , Mice , Mice, Inbred Strains , Mice, Knockout , Radiation Chimera/physiology , Receptors, Interleukin-15 , Receptors, Interleukin-2/genetics , Signal Transduction/physiology , Spleen/cytology
12.
Am J Physiol Gastrointest Liver Physiol ; 285(2): G382-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12702495

ABSTRACT

IL-2 receptor alpha-deficient (IL2Ralpha-/-) mice spontaneously accumulate vast numbers of intestinal lamina propria (LP) T cells and develop bowel inflammation. The accumulation of T cells in IL2Ralpha-/- mice is thought to result, in part, from defective Fas-induced cell death. To understand the role of cell proliferation and death in regulating LP T cells in IL2Ralpha-/- mice, we have directly examined the proliferation and Fas sensitivity of wild-type, lpr/lpr, and IL2Ralpha-/- LP T cells. In wild-type mice, 5'-bromodeoxyuridine (BrdU) labeling and Fas susceptibility are greatest in CD44Hi LP T cells. Fas-deficient lpr/lpr mice have normal total numbers of LP T cells, despite an increased proportion of BrdU+ T cells. By contrast, IL2Ralpha-/- mice possess increased total numbers of LP T cells, despite normal proportions of BrdU+ LP T cells. Finally, wild-type and IL2Ralpha-/- LP T cells are equivalently Fas sensitive. These results demonstrate that LP T cells proliferate and are Fas-sensitive cells. IL2Ralpha-/- mice accumulate a large number of these Fas-sensitive LP T cells and clearly differ from Fas-deficient lpr/lpr mice in this regard. Thus our studies reveal that Fas is dispensable for LP T cell homeostasis and suggest that the intestinal inflammation observed in IL2Ralpha-/- mice is independent of defective Fas-induced cell death.


Subject(s)
Intestines/pathology , T-Lymphocytes/physiology , fas Receptor/physiology , Animals , Apoptosis , Bromodeoxyuridine/metabolism , Cell Division , Homeostasis , Hyaluronan Receptors/analysis , Hypertrophy , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/physiology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymectomy , fas Receptor/genetics
13.
Mol Immunol ; 39(9): 537-44, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12431387

ABSTRACT

Interleukin-15 (IL-15) is a cytokine that plays unique roles in both innate and adaptive immune cell homeostasis. While early studies suggested that IL-15 resembled IL-2, more recent work suggests that IL-15 may play multiple unique roles in immune homeostasis befitting its pleiotropic expression pattern. This review will focus on recent studies that highlight some of these functions.


Subject(s)
Interleukin-15/physiology , Lymphocytes/immunology , Animals , Homeostasis , Immunologic Memory , Interleukin-2/physiology , Lymphocyte Activation , Receptors, Interleukin-15 , Receptors, Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes/immunology
14.
Cytokine Growth Factor Rev ; 13(6): 429-39, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12401478

ABSTRACT

Interleukin (IL)-15 is a member of the common gamma chain family of cytokines, and is closely related to IL-2. While these two cytokines share several important biological functions in vitro, recent mouse models have demonstrated unique roles for these two cytokines in supporting lymphoid homeostasis in vivo. IL-15 has been shown to regulate the homeostasis of both innate and adaptive immune cells, and this review will discuss several ways in which this pleiotropic cytokine may support lymphoid homeostasis.


Subject(s)
Interleukin-15/metabolism , Interleukin-15/physiology , Lymphocytes/metabolism , Animals , Cell Division , Humans , Immunologic Memory , Models, Biological , Receptors, Interleukin-15 , Receptors, Interleukin-2/metabolism , Signal Transduction
15.
Inflamm Bowel Dis ; 8(1): 35-42, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11837936

ABSTRACT

The intestinal lamina propria contains lymphocytes that are chronically activated by exposure to luminal antigens. Dysregulation of these cells is thought to be central to the pathogenesis of bowel inflammation in experimental models of inflammatory bowel disease. CD28 signals on peripheral T cells provide important costimulatory signals that enhance T-cell proliferation and activation responses to antigens. However, the role of CD28 signals in lamina propria T cells or models of inflammatory bowel disease have not been determined. Accordingly, we examined T lymphocyte activation and proliferation in CD28-deficient (CD28-/-) mice to examine the in vivo roles of CD28 signals in lamina propria T-cell homeostasis. We further generated CD28-/- interleukin (IL)-2-/- double mutant mice to assess the role of CD28 signals in supporting the spontaneously activated and pathogenic T cells that accumulate in IL-2-/- mice. CD28-/- lamina propria T cells displayed reduced activation markers, but were present in normal numbers and proliferated normally. IL-2-/- lymphocytes expressed high levels of bcl-xL, whereas CD28-/- IL-2-/- cells had substantially less bcl-xL. However, lymphadenopathy and ulcerative colitis-like disease occurred in both IL-2-/- and CD28-/- IL-2-/- mice. Thus, CD28 provides a functional costimulatory signal to lamina propria T cells but is not required for homeostasis of these cells. In addition, neither CD28 nor bcl-xL appears to be required for the spontaneous accumulation of T cells in IL-2-/- mice. This suggests that other costimulatory molecules or T-cell receptor ligation alone drive lymphocyte expansion in IL-2-deficient mice.


Subject(s)
CD28 Antigens/genetics , CD28 Antigens/physiology , Colitis/genetics , Colitis/physiopathology , Interleukin-2/deficiency , Interleukin-2/physiology , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Animals , Disease Models, Animal , Interleukin-2/genetics , Intestinal Mucosa/physiopathology , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Signal Transduction/physiology , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...