Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(11): 113323, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37889750

ABSTRACT

Intestinal colonization by antigenically foreign microbes necessitates expanded peripheral immune tolerance. Here we show commensal microbiota prime expansion of CD4 T cells unified by the Kruppel-like factor 2 (KLF2) transcriptional regulator and an essential role for KLF2+ CD4 cells in averting microbiota-driven intestinal inflammation. CD4 cells with commensal specificity in secondary lymphoid organs and intestinal tissues are enriched for KLF2 expression, and distinct from FOXP3+ regulatory T cells or other differentiation lineages. Mice with conditional KLF2 deficiency in T cells develop spontaneous rectal prolapse and intestinal inflammation, phenotypes overturned by eliminating microbiota or reconstituting with donor KLF2+ cells. Activated KLF2+ cells selectively produce IL-10, and eliminating IL-10 overrides their suppressive function in vitro and protection against intestinal inflammation in vivo. Together with reduced KLF2+ CD4 cell accumulation in Crohn's disease, a necessity for the KLF2+ subpopulation of T regulatory type 1 (Tr1) cells in sustaining commensal tolerance is demonstrated.


Subject(s)
CD4-Positive T-Lymphocytes , Microbiota , Mice , Animals , Interleukin-10/metabolism , T-Lymphocytes, Regulatory , Transcription Factors/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism
2.
Science ; 381(6664): 1324-1330, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37733857

ABSTRACT

Pregnancy confers partner-specific protection against complications in future pregnancy that parallel persistence of fetal microchimeric cells (FMcs) in mothers after parturition. We show that preexisting FMcs become displaced by new FMcs during pregnancy and that FMc tonic stimulation is essential for expansion of protective fetal-specific forkhead box P3 (FOXP3)-positive regulatory T cells (Treg cells). Maternal microchimeric cells and accumulation of Treg cells with noninherited maternal antigen (NIMA) specificity are similarly overturned in daughters after pregnancy, highlighting a fixed microchimeric cell niche. Whereas NIMA-specific tolerance is functionally erased by pregnancy, partner-specific resiliency against pregnancy complications persists in mothers despite paternity changes in intervening pregnancy. Persistent fetal tolerance reflects FOXP3 expression plasticity, which allows mothers to more durably remember their babies, whereas daughters forget their mothers with new pregnancy-imprinted immunological memories.


Subject(s)
Chimerism , Fetus , Immune Tolerance , Immunologic Memory , Maternal-Fetal Exchange , Pregnancy , Animals , Female , Mice , Pregnancy/immunology , Antigens/immunology , Cell Plasticity , Fetus/cytology , Fetus/immunology , Forkhead Transcription Factors/immunology , Maternal-Fetal Exchange/immunology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology
3.
Hepatology ; 78(4): 1035-1049, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37078450

ABSTRACT

BACKGROUND AND AIMS: Although a dysregulated type 1 immune response is integral to the pathogenesis of biliary atresia, studies in both humans and mice have uncovered a type 2 response, primarily driven by type 2 innate lymphoid cells. In nonhepatic tissues, natural type 2 innate lymphoid cell (nILC2s) regulate epithelial proliferation and tissue repair, whereas inflammatory ILC2s (iIlC2s) drive tissue inflammation and injury. The aim of this study is to determine the mechanisms used by type 2 innate lymphoid cell (ILC2) subpopulations to regulate biliary epithelial response to an injury. APPROACH AND RESULTS: Using Spearman correlation analysis, nILC2 transcripts, but not those of iILC2s, are positively associated with cholangiocyte abundance in biliary atresia patients at the time of diagnosis. nILC2s are identified in the mouse liver through flow cytometry. They undergo expansion and increase amphiregulin production after IL-33 administration. This drives epithelial proliferation dependent on the IL-13/IL-4Rα/STAT6 pathway as determined by decreased nILC2s and reduced epithelial proliferation in knockout strains. The addition of IL-2 promotes inter-lineage plasticity towards a nILC2 phenotype. In experimental biliary atresia induced by rotavirus, this pathway promotes epithelial repair and tissue regeneration. The genetic loss or molecular inhibition of any part of this circuit switches nILC2s to inflammatory type 2 innate lymphoid cell-like, resulting in decreased amphiregulin production, decreased epithelial proliferation, and the full phenotype of experimental biliary atresia. CONCLUSIONS: These findings identify a key function of the IL-13/IL-4Rα/STAT6 pathway in ILC2 plasticity and an alternate circuit driven by IL-2 to promote nILC2 stability and amphiregulin expression. This pathway induces epithelial homeostasis and repair in experimental biliary atresia.


Subject(s)
Biliary Atresia , Humans , Animals , Mice , Biliary Atresia/pathology , Immunity, Innate , Interleukin-13/metabolism , Interleukin-2/metabolism , Lymphocytes , Amphiregulin/genetics , Amphiregulin/metabolism
4.
Clin Liver Dis (Hoboken) ; 19(3): 111-113, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35355844

ABSTRACT

Content available: Author Audio Recording.

6.
Med Educ Online ; 23(1): 1505401, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30343644

ABSTRACT

BACKGROUND: The continuity provided by longitudinal clerkships has documented benefits to medical student education. Yet, little quantitative data exist on the association between longitudinal clerkships and patient outcomes. OBJECTIVE: This study compares screening metrics of a longitudinal clerkship called the education-centered medical home (ECMH) with the standard clinical model at a student-volunteer free clinic (SVFC). In the ECMH model, the same attending physician staffs one half-day of clinic with same group of students weekly for 4 years. Standard clinical models are staffed with students and physicians who come to the SVFC based on availability. DESIGN: ECMH students aimed to increase human immunodeficiency virus (HIV) screening rates in their patient panel as part of a quality improvement project. Students prepared individualized care plans prior to patient visits that included whether screening had been performed. They were also reminded to confirm completion of testing. Percentages of patients screened for HIV before and after establishment of the ECMH were compared with four standard clinical models. Screening rates for breast, colon, and cervical cancer, as well as hepatitis C, served as secondary endpoints. RESULTS: While screening rates were initially similar between models (43.2% and 34.8% for the ECMH and standard clinical panels, respectively, p = 0.32), HIV screening rates increased from 43.2% to 95.0% in the ECMH compared with a significantly smaller increase from 35.0% to 50.0% in the standard clinical panel (p < 0.0001). Additionally, the ECMH resulted in statistically significantly increased screening rates for cervical cancer (p < 0.001) and hepatitis C (p < 0.0001). CONCLUSIONS: This study demonstrates an association between a longitudinal ECMH clerkship and improved quality metrics at an SVFC. Even measures not targeted for intervention, such as colorectal cancer and hepatitis C, showed significant improvement in screening rates when compared with the standard clinical model.


Subject(s)
Clinical Clerkship/organization & administration , Models, Educational , Patient-Centered Care/organization & administration , Quality Improvement/organization & administration , Student Run Clinic/organization & administration , Adult , Breast Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , Female , HIV Infections/diagnosis , Hepatitis C/diagnosis , Humans , Male , Mass Screening/organization & administration , Middle Aged , Primary Health Care/organization & administration , Uterine Cervical Neoplasms/diagnosis
7.
Proc Natl Acad Sci U S A ; 115(7): E1520-E1529, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29378942

ABSTRACT

The cellular and molecular basis of sex-dimorphic autoimmune diseases, such as the CNS demyelinating disease multiple sclerosis (MS), remains unclear. Our studies in the SJL mouse model of MS, experimental autoimmune encephalomyelitis (EAE), reveal that sex-determined differences in Il33 expression by innate immune cells in response to myelin peptide immunization regulate EAE susceptibility. IL-33 is selectively induced in PLP139-151-immunized males and activates type 2 innate lymphoid cells (ILC2s), cells that promote and sustain a nonpathogenic Th2 myelin-specific response. Without this attenuating IL-33 response, females generate an encephalitogenic Th17-dominant response, which can be reversed by IL-33 treatment. Mast cells are one source of IL-33 and we provide evidence that testosterone directly induces Il33 gene expression and also exerts effects on the potential for Il33 gene expression during mast cell development. Thus, in contrast to their pathogenic role in allergy, we propose a sex-specific role for both mast cells and ILC2s as attenuators of the pathogenic Th response in CNS inflammatory disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-33/metabolism , Mast Cells/immunology , Sex Characteristics , Th17 Cells/immunology , Animals , Cytokines/metabolism , Female , Interleukin-33/genetics , Interleukin-33/immunology , Male , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Testosterone/blood
8.
Clin Immunol ; 189: 14-22, 2018 04.
Article in English | MEDLINE | ID: mdl-27108197

ABSTRACT

Inflammasomes are multiprotein complexes that assemble in response to microbial and other danger signals and regulate the secretion of biologically active IL-1ß and IL-18. Although they are important in protective immunity against bacterial, viral and parasitic infections, aberrant inflammasome activity promotes chronic inflammation associated with autoimmune disease. Inflammasomes have been described in many immune cells, but the majority of studies have focused on their activity in macrophages. Here we discuss an important role for mast cell-inflammasome activity in EAE, the rodent model of multiple sclerosis, a CNS demyelinating disease. We review our evidence that mast cells in the meninges, tissues that surround the brain and spinal cord, interact with infiltrating myelin-specific T cells in early disease. This interaction elicits IL-1ß expression by mast cells, which in turn, promotes GM-CSF expression by T cells. In view of the essential role that GM-CSF plays in T cell encephalitogenicity, we propose this mast cell-T cell crosstalk in the meninges is critical for EAE disease development.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammasomes/immunology , Mast Cells/immunology , Meninges/immunology , Multiple Sclerosis/immunology , Animals , Cell Communication/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Inflammation/immunology , Multiple Sclerosis/metabolism , T-Lymphocytes/immunology
9.
Nat Immunol ; 18(10): 1063-1064, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28926546

Subject(s)
Lymphocytes
10.
J Autoimmun ; 73: 100-10, 2016 09.
Article in English | MEDLINE | ID: mdl-27396526

ABSTRACT

GM-CSF is a cytokine produced by T helper (Th) cells that plays an essential role in orchestrating neuroinflammation in experimental autoimmune encephalomyelitis, a rodent model of multiple sclerosis. Yet where and how Th cells acquire GM-CSF expression is unknown. In this study we identify mast cells in the meninges, tripartite tissues surrounding the brain and spinal cord, as important contributors to antigen-specific Th cell accumulation and GM-CSF expression. In the absence of mast cells, Th cells do not accumulate in the meninges nor produce GM-CSF. Mast cell-T cell co-culture experiments and selective mast cell reconstitution of the meninges of mast cell-deficient mice reveal that resident meningeal mast cells are an early source of caspase-1-dependent IL-1ß that licenses Th cells to produce GM-CSF and become encephalitogenic. We also provide evidence of mast cell-T cell co-localization in the meninges and CNS of recently diagnosed acute MS patients indicating similar interactions may occur in human demyelinating disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1beta/metabolism , Mast Cells/immunology , Meninges/cytology , Multiple Sclerosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Adult , Aged , Animals , Caspase 1/metabolism , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Humans , Male , Meninges/immunology , Meninges/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Young Adult
11.
J Immunol ; 194(12): 5609-13, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972476

ABSTRACT

Multiple sclerosis preferentially affects women, and this sexual dimorphism is recapitulated in the SJL mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). In this study, we demonstrate that signaling through c-Kit exerts distinct effects on EAE susceptibility in male and female SJL mice. Previous studies in females show that Kit mutant (W/W(v)) mice are less susceptible to EAE than are wild-type mice. However, male W/W(v) mice exhibit exacerbated disease, a phenotype independent of mast cells and corresponding to a shift from a Th2- to a Th17-dominated T cell response. We demonstrate a previously undescribed deficit in c-Kit(+) type 2 innate lymphoid cells (ILC2s) in W/W(v) mice. ILC2s are also significantly reduced in EAE-susceptible wild-type females, indicating that both c-Kit signals and undefined male-specific factors are required for ILC2 function. We propose that deficiencies in Th2-promoting ILC2s remove an attenuating influence on the encephalitogenic T cell response and therefore increases disease susceptibility.


Subject(s)
Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Disease Susceptibility , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Immunophenotyping , Inflammation Mediators/metabolism , Lymphocyte Count , Male , Mast Cells/immunology , Mice , Mice, Transgenic , Phenotype , Proto-Oncogene Proteins c-kit/genetics , Sex Factors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
12.
Transl Res ; 165(2): 255-69, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25241937

ABSTRACT

The central nervous system (CNS) largely comprises nonregenerating cells, including neurons and myelin-producing oligodendrocytes, which are particularly vulnerable to immune cell-mediated damage. To protect the CNS, mechanisms exist that normally restrict the transit of peripheral immune cells into the brain and spinal cord, conferring an "immune-specialized" status. Thus, there has been a long-standing debate as to how these restrictions are overcome in several inflammatory diseases of the CNS, including multiple sclerosis (MS). In this review, we highlight the role of the meninges, tissues that surround and protect the CNS and enclose the cerebral spinal fluid, in promoting chronic inflammation that leads to neuronal damage. Although the meninges have traditionally been considered structures that provide physical protection for the brain and spinal cord, new data have established these tissues as sites of active immunity. It has been hypothesized that the meninges are important players in normal immunosurveillance of the CNS but also serve as initial sites of anti-myelin immune responses. The resulting robust meningeal inflammation elicits loss of localized blood-brain barrier (BBB) integrity and facilitates a large-scale influx of immune cells into the CNS parenchyma. We propose that targeting the cells and molecules mediating these inflammatory responses within the meninges offers promising therapies for MS that are free from the constraints imposed by the BBB. Importantly, such therapies may avoid the systemic immunosuppression often associated with the existing treatments.


Subject(s)
Meninges/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Blood-Brain Barrier/immunology , Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Inflammation/immunology , Mast Cells/immunology , Mice , Models, Immunological , Neutrophil Infiltration/immunology , T-Lymphocytes/immunology , Translational Research, Biomedical
SELECTION OF CITATIONS
SEARCH DETAIL
...