Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Hum Reprod ; 39(5): 1023-1041, 2024 May 02.
Article En | MEDLINE | ID: mdl-38511208

STUDY QUESTION: How does ovarian stimulation (OS), which is used to mature multiple oocytes for ART procedures, impact the principal cellular compartments and transcriptome of the human endometrium in the periovulatory and mid-secretory phases? SUMMARY ANSWER: During the mid-secretory window of implantation, OS alters the abundance of endometrial immune cells, whereas during the periovulatory period, OS substantially changes the endometrial transcriptome and impacts both endometrial glandular and immune cells. WHAT IS KNOWN ALREADY: Pregnancies conceived in an OS cycle are at risk of complications reflective of abnormal placentation and placental function. OS can alter endometrial gene expression and immune cell populations. How OS impacts the glandular, stromal, immune, and vascular compartments of the endometrium, in the periovulatory period as compared to the window of implantation, is unknown. STUDY DESIGN, SIZE, DURATION: This prospective cohort study carried out between 2020 and 2022 included 25 subjects undergoing OS and 25 subjects in natural menstrual cycles. Endometrial biopsies were performed in the proliferative, periovulatory, and mid-secretory phases. PARTICIPANTS/MATERIALS, SETTING, METHODS: Blood samples were processed to determine serum estradiol and progesterone levels. Both the endometrial transcriptome and the principal cellular compartments of the endometrium, including glands, stroma, immune, and vasculature, were evaluated by examining endometrial dating, differential gene expression, protein expression, cell populations, and the three-dimensional structure in endometrial tissue. Mann-Whitney U tests, unpaired t-tests or one-way ANOVA and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE: In the periovulatory period, OS induced high levels of differential gene expression, glandular-stromal dyssynchrony, and an increase in both glandular epithelial volume and the frequency of endometrial monocytes/macrophages. In the window of implantation during the mid-secretory phase, OS induced changes in endometrial immune cells, with a greater frequency of B cells and a lower frequency of CD4 effector T cells. LARGE SCALE DATA: The data underlying this article have been uploaded to the Genome Expression Omnibus/National Center for Biotechnology Information with accession number GSE220044. LIMITATIONS, REASONS FOR CAUTION: A limited number of subjects were included in this study, although the subjects within each group, natural cycle or OS, were homogenous in their clinical characteristics. The number of subjects utilized was sufficient to identify significant differences; however, with a larger number of subjects and additional power, we may detect additional differences. Another limitation of the study is that proliferative phase biopsies were collected in natural cycles, but not in OS cycles. Given that the OS cycle subjects did not have known endometrial factor infertility, and the comparisons involved subjects who had a similar and robust response to stimulation, the findings are generalizable to women with a normal response to OS. WIDER IMPLICATIONS OF THE FINDINGS: OS substantially altered the periovulatory phase endometrium, with fewer transcriptomic and cell type-specific changes in the mid-secretory phase. Our findings show that after OS, the endometrial microenvironment in the window of implantation possesses many more similarities to that of a natural cycle than does the periovulatory endometrium. Further investigation of the immune compartment and the functional significance of this cellular compartment under OS conditions is warranted. STUDY FUNDING/COMPETING INTERESTS: Research reported in this publication was supported by the National Institute of Allergy and Infectious Diseases (R01AI148695 to A.M.B. and N.C.D.), Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01HD109152 to R.A.), and the March of Dimes (5-FY20-209 to R.A.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or March of Dimes. All authors declare no conflict of interest.


Endometrium , Ovulation Induction , Transcriptome , Humans , Female , Endometrium/metabolism , Adult , Cellular Microenvironment , Prospective Studies , Estradiol/blood , Embryo Implantation/physiology , Progesterone/blood , Progesterone/metabolism , Pregnancy , Menstrual Cycle
2.
Proc Natl Acad Sci U S A ; 119(35): e2123267119, 2022 08 30.
Article En | MEDLINE | ID: mdl-35994660

The pregnant uterus is an immunologically rich organ, with dynamic changes in the inflammatory milieu and immune cell function underlying key stages of pregnancy. Recent studies have implicated dysregulated expression of the interleukin-1 (IL-1) family cytokine, IL-33, and its receptor, ST2, in poor pregnancy outcomes in women, including recurrent pregnancy loss, preeclampsia, and preterm labor. How IL-33 supports pregnancy progression in vivo is not well understood. Here, we demonstrate that maternal IL-33 signaling critically regulates uterine tissue remodeling and immune cell function during early pregnancy in mice. IL-33-deficient dams exhibit defects in implantation chamber formation and decidualization, and abnormal vascular remodeling during early pregnancy. These defects coincide with delays in early embryogenesis, increased resorptions, and impaired fetal and placental growth by late pregnancy. At a cellular level, myometrial fibroblasts, and decidual endothelial and stromal cells, are the main IL-33+ cell types in the uterus during decidualization and early placentation, whereas ST2 is expressed by uterine immune populations associated with type 2 immune responses, including ILC2s, Tregs, CD4+ T cells, M2- and cDC2-like myeloid cells, and mast cells. Early pregnancy defects in IL-33-deficient dams are associated with impaired type 2 cytokine responses by uterine lymphocytes and fewer Arginase-1+ macrophages in the uterine microenvironment. Collectively, our data highlight a regulatory network, involving crosstalk between IL-33-producing nonimmune cells and ST2+ immune cells at the maternal-fetal interface, that critically supports pregnancy progression in mice. This work has the potential to advance our understanding of how IL-33 signaling may support optimal pregnancy outcomes in women.


Interleukin-33 , Placenta , Placentation , Uterus , Animals , Decidua/blood supply , Decidua/cytology , Decidua/growth & development , Decidua/immunology , Female , Fetus/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/deficiency , Interleukin-33/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Placenta/immunology , Placenta/metabolism , Pregnancy , Uterus/blood supply , Uterus/growth & development , Uterus/immunology , Uterus/metabolism
3.
Immunol Rev ; 300(1): 125-133, 2021 03.
Article En | MEDLINE | ID: mdl-33491231

Natural killer (NK) cells are cytotoxic innate lymphocytes with key roles in host protection against viruses and malignancy. Notwithstanding their historical classification as innate immune cells, NK cells are now understood to have some capacity to mount memory or memory-like immune responses in which effector cells undergo antigen-driven expansion and give rise to long-lived memory cells with enhanced functionality. Understanding how antigen-specific effector and memory NK responses are regulated is an important and active area of research in the field. Here, we discuss key transcription factors and epigenetic processes involved in antigen-specific effector and memory NK cell differentiation.


Epigenesis, Genetic , Immunologic Memory , Antigens , Killer Cells, Natural , Lymphocyte Activation
4.
Int J Mol Sci ; 21(18)2020 Sep 05.
Article En | MEDLINE | ID: mdl-32899448

Maternal spiral arteries and newly formed decidual capillaries support embryonic development prior to placentation. Previous studies demonstrated that Notch signaling is active in endothelial cells of both decidual capillaries and spiral arteries, however the role of Notch signaling in physiologic decidual angiogenesis and maintenance of the decidual vasculature in early mouse pregnancy has not yet been fully elucidated. We used the Cdh5-CreERT2;Jagged1(Jag1)flox/flox (Jag1∆EC) mouse model to delete Notch ligand, Jag1, in maternal endothelial cells during post-implantation, pre-placentation mouse pregnancy. Loss of endothelial Jag1 leads to increased expression of Notch effectors, Hey2 and Nrarp, and increased endothelial Notch signaling activity in areas of the decidua with remodeling angiogenesis. This correlated with an increase in Dll4 expression in capillary endothelial cells, but not spiral artery endothelial cells. Consistent with increased Dll4/Notch signaling, we observed decreased VEGFR2 expression and endothelial cell proliferation in angiogenic decidual capillaries. Despite aberrant Dll4 expression and Notch activation in Jag1∆EC mutants, pregnancies were maintained and the decidual vasculature was not altered up to embryonic day 7.5. Thus, Jag1 functions in the newly formed decidual capillaries as an antagonist of endothelial Dll4/Notch signaling during angiogenesis, but Jag1 signaling is not necessary for early uterine angiogenesis.


Jagged-1 Protein/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Animals , Calcium-Binding Proteins/metabolism , Cell Proliferation , Decidua/metabolism , Embryo Implantation/physiology , Embryonic Development , Endometrium/metabolism , Endothelial Cells/metabolism , Female , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Placentation , Pregnancy , Receptors, Notch/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Nat Immunol ; 21(10): 1181-1193, 2020 10.
Article En | MEDLINE | ID: mdl-32807943

Type 2 cytokine responses promote parasitic immunity and initiate tissue repair; however, they can also result in immunopathologies when not properly restricted. Although basophilia is recognized as a common feature of type 2 inflammation, the roles basophils play in regulating these responses are unknown. Here, we demonstrate that helminth-induced group 2 innate lymphoid cell (ILC2) responses are exaggerated in the absence of basophils, resulting in increased inflammation and diminished lung function. Additionally, we show that ILC2s from basophil-depleted mice express reduced amounts of the receptor for the neuropeptide neuromedin B (NMB). Critically, NMB stimulation inhibited ILC2 responses from control but not basophil-depleted mice, and basophils were sufficient to directly enhance NMB receptor expression on ILC2s. These studies suggest that basophils prime ILC2s to respond to neuron-derived signals necessary to maintain tissue integrity. Further, these data provide mechanistic insight into the functions of basophils and identify NMB as a potent inhibitor of type 2 inflammation.


Basophils/immunology , Lung/metabolism , Lymphocytes/immunology , Nippostrongylus/physiology , Strongylida Infections/immunology , Animals , Cell Communication , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Th2 Cells/immunology , Tryptases/genetics
6.
Front Immunol ; 11: 1355, 2020.
Article En | MEDLINE | ID: mdl-32733452

[This corrects the article DOI: 10.3389/fimmu.2020.00309.].

7.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Article En | MEDLINE | ID: mdl-32421753

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Erythroid Precursor Cells/immunology , Erythropoiesis/immunology , Mast Cells/immunology , Mastocytosis/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Carbonic Anhydrase I/genetics , Carbonic Anhydrase I/immunology , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/pathology , Female , Mast Cells/parasitology , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mice , Mice, Transgenic , Trichinellosis/genetics , Trichinellosis/pathology
8.
Article En | MEDLINE | ID: mdl-32269968

NK cells are cytotoxic lymphocytes that provide systemic defense against pathogens and malignancy. Although historically considered cells of the innate immune system, NK cells are now known to be capable of memory or memory-like immune responses in certain settings. Memory NK responses were initially reported over a decade ago in studies involving mouse models of cytomegalovirus infection and delayed-type hypersensitivity reactions to chemical haptens and viral antigens. Since then, a growing body of literature suggests that memory or memory-like NK cell responses may occur in a broader range of immunological settings, including in response to various viral and bacterial infections, and some immunization protocols. Memory-like NK cell responses have also now been reported in humans and non-human primates. Here, we summarize recent studies demonstrating memory or memory-like responses by NK cells in settings of infection and immunization against infectious agents.


Immunologic Memory , Killer Cells, Natural , Animals , Antigens, Viral , Disease Models, Animal , Primates
9.
Front Immunol ; 11: 309, 2020.
Article En | MEDLINE | ID: mdl-32161593

Natural killer (NK) cells are cytotoxic innate lymphocytes that are well-known for their ability to kill infected or malignant cells. Beyond their roles in tumor surveillance and anti-pathogen defense, more recent studies have highlighted key roles for NK cells in a broad range of biological processes, including metabolic homeostasis, immunomodulation of T cells, contact hypersensitivity, and pregnancy. Consistent with the breadth and diversity of these functions, it is now appreciated that NK cells are a heterogeneous population, comprised of specialized and sometimes tissue-specific subsets with distinct phenotypes and effector functions. Indeed, in addition to the conventional NK cells (cNKs) that are abundant and have been well-studied in the blood and spleen, distinct subsets of tissue-resident NK cells (trNKs) and "helper" Group 1 innate lymphoid cells (ILC1s) have now been described in multiple organs and tissues, including the liver, uterus, thymus, adipose tissue, and skin, among others. The cNK, trNK, and/or helper ILC1 populations that co-exist in these various tissues exhibit both common and distinct developmental requirements, suggesting that a combination of lineage-, subset-, and tissue-specific differentiation processes may contribute to the unique functional properties of these various populations. Here, we provide an overview of the transcriptional regulatory pathways known to instruct the development and differentiation of cNK, trNK, and helper ILC1 populations in specific tissues in mice.


Killer Cells, Natural/metabolism , Transcription Factors/metabolism , Animals , Bone Marrow/metabolism , Cell Differentiation , Gene Expression Regulation , Humans , Immunity, Innate , Lymphocytes/metabolism , Mice , T-Lymphocytes, Helper-Inducer/metabolism
10.
J Histochem Cytochem ; 68(4): 253-267, 2020 04.
Article En | MEDLINE | ID: mdl-32108542

Interleukin-33 (IL-33) is an IL-1 family cytokine with pleiotropic effects on diverse cell types. Dysregulated IL-33 signaling has been implicated in pregnancy-related disorders, including preeclampsia and recurrent pregnancy loss, and in ovarian function in women undergoing controlled ovarian stimulation for in vitro fertilization. To date, expression of IL-33 and its receptor subunit, ST2, in the female reproductive tract remains poorly characterized. We identify IL-33-expressing oocytes surrounded by ST2-expressing granulosa cells at all stages of follicular development, in addition to IL-33+ and ST2+ non-endothelial cells in the ovarian stroma and theca layer in ovaries from adult mice. These expression patterns are similar in estrus- and diestrus-stage adults and in pubescent mice, suggesting a role for IL-33 signaling in ovarian function throughout development and in the estrous cycle. In the uterus, we find expression of IL-33 and ST2 in glandular and luminal epithelia during estrus and at the initiation of pregnancy. Uterine IL-33 expression was modulated by the estrous cycle and was reduced in pubescent females. Last, superovulation increases transcripts for IL-33 and the soluble form of ST2 (sST2) in ovaries, and for IL-33 in uteri. Collectively, our findings lay the foundation for studies identifying cell type-specific requirements for IL-33/ST2 signaling in the establishment and maintenance of mouse pregnancy.


Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Ovary/metabolism , Superovulation , Uterus/metabolism , Animals , Female , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Male , Mice , Mice, Inbred C57BL , Ovary/cytology , Pregnancy , Uterus/cytology
11.
Immunology ; 156(2): 111-119, 2019 02.
Article En | MEDLINE | ID: mdl-30450565

Natural killer (NK) cells are highly specialized cytotoxic lymphocytes that provide protection against pathogens and malignant cells. They develop from common lymphoid progenitors via a multi-stage lineage commitment and differentiation process that gives rise to mature NK cells with potent cytotoxic functionality. Although generally considered cells of the innate immune system, recent studies have demonstrated that NK cells have the capacity to mount immune responses with features of adaptive immunity, including robust antigen-specific clonal-like expansion and the generation of long-lived memory cells that mediate enhanced recall responses. Here, we discuss specific transcription factors that have been shown to commonly and uniquely regulate NK cell development and effector and memory responses in experimental mouse models.


Immunity, Cellular , Immunity, Innate , Immunologic Memory , Killer Cells, Natural/immunology , Models, Immunological , Transcription, Genetic/immunology , Animals , Humans , Mice
12.
J Leukoc Biol ; 104(6): 1087-1096, 2018 12.
Article En | MEDLINE | ID: mdl-30156708

NK cells are important mediators of immunological defense against pathogens and cancer, owing in part to their ability to directly kill infected and malignant host cells. Although historically considered cells of the innate immune system, a growing body of literature indicates that NK cells have the capacity to mount immune responses with features of immunological memory, including enhanced recall responses that are long-lived and Ag-specific. Anamnestic NK cell responses in mice have now been described in a broad range of immunological settings, including viral and bacterial infections, hapten-induced contact hypersensitivity (CHS) reactions, and alloantigen responses. Memory-like NK cell populations have also been identified in humans, most notably in the context of human cytomegalovirus (HCMV) infection. Here, an overview of these studies is provided with discussion of the molecular, transcriptional, and epigenetic pathways that regulate adaptive NK cell responses.


Immunologic Memory , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Adaptive Immunity , Animals , Gene Expression Profiling , Humans , Hypersensitivity/immunology , Infections/immunology , Isoantigens/immunology , Killer Cells, Natural/transplantation , Lymphocyte Activation , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred Strains , Mice, Knockout , Neoplasms/immunology , Primates/immunology
13.
Methods Mol Biol ; 1441: 1-12, 2016.
Article En | MEDLINE | ID: mdl-27177652

In C57BL/6 mice, NK cells expressing the activating receptor Ly49H proliferate robustly in response to mouse cytomegalovirus (MCMV) infection. The expansion of Ly49H(+) NK cells peaks at approximately 1 week post-infection, and is then followed by a distinct contraction phase that ultimately leaves a small but long-lived pool of MCMV-experienced Ly49H(+) NK cells that are capable of mediating enhanced memory-like responses during subsequent encounters with MCMV. Here we describe an adoptive transfer model in which the expansion, contraction, and memory cell persistence of transferred Ly49H(+) NK cells are tracked in congenic C57BL/6 hosts following MCMV infection.


Cell Tracking/methods , Cytomegalovirus Infections/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/transplantation , Adoptive Transfer , Animals , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Cytomegalovirus Infections/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism
14.
Adv Exp Med Biol ; 850: 81-91, 2015.
Article En | MEDLINE | ID: mdl-26324348

Immunological memory is a hallmark of the adaptive immune system. Although natural killer (NK) cells have traditionally been classified as a component of the innate immune system, they have recently been shown in mice and humans to exhibit certain features of immunological memory, including an ability to undergo a clonal-like expansion during virus infection, generate long-lived progeny (i.e. memory cells), and mediate recall responses against previously encountered pathogens--all characteristics previously ascribed only to adaptive immune responses by B and T cells in mammals. To date, the molecular events that govern the generation of NK cell memory are not completely understood. Using a mouse model of cytomegalovirus infection, we demonstrate that individual pro-inflammatory IL-12, IL-18, and type I-IFN signaling pathways are indispensible and play non-redundant roles in the generation of virus-specific NK cell memory. Furthermore, we discovered that antigen-specific proliferation and protection by NK cells is mediated by the transcription factor Zbtb32, which is induced by pro-inflammatory cytokines and promotes a cell cycle program in activated NK cells. A greater understanding of the molecular mechanisms controlling NK cell responses will provide novel strategies for tailoring vaccines to target infectious disease.


Adaptive Immunity , Herpesviridae Infections/immunology , Immunologic Memory/genetics , Killer Cells, Natural/immunology , Signal Transduction/immunology , Animals , Cell Proliferation , Gene Expression Regulation , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Mice , Muromegalovirus/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology
15.
Nat Immunol ; 15(6): 546-53, 2014 Jun.
Article En | MEDLINE | ID: mdl-24747678

Natural killer (NK) cells are innate lymphocytes that exhibit many features of adaptive immunity, including clonal proliferation and long-lived memory. Here we demonstrate that the BTB-ZF transcription factor Zbtb32 (also known as ROG, FAZF, TZFP and PLZP) was essential for the proliferative burst and protective capacity of virus-specific NK cells. Signals from proinflammatory cytokines were both necessary and sufficient to induce high expression of Zbtb32 in NK cells. Zbtb32 facilitated NK cell proliferation during infection by antagonizing the anti-proliferative factor Blimp-1 (Prdm1). Our data support a model in which Zbtb32 acts as a cellular 'hub' through which proinflammatory signals instruct a 'proliferation-permissive' state in NK cells, thereby allowing their prolific expansion in response to viral infection.


Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Repressor Proteins/immunology , Adaptive Immunity , Animals , Cell Proliferation , Cell Survival/immunology , Cytokines/immunology , Immunologic Memory , Inflammation/immunology , Inflammation/virology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/immunology , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/genetics , Transcription Factors/antagonists & inhibitors
16.
J Exp Med ; 210(13): 2981-90, 2013 Dec 16.
Article En | MEDLINE | ID: mdl-24277151

Development of the natural killer (NK) cell lineage is dependent on the transcription factor Nfil3 (or E4BP4), which is thought to act downstream of IL-15 signaling. Nfil3-deficient mice lack NK cells, whereas other lymphocyte lineages (B, T, and NKT cells) remain largely intact. We report the appearance of Ly49H-expressing NK cells in Nfil3(-/-) mice infected with mouse cytomegalovirus (MCMV) or recombinant viruses expressing the viral m157 glycoprotein. Nfil3(-/-) NK cells at the peak of antigen-driven expansion were functionally similar to NK cells from infected wild-type mice with respect to IFN-γ production and cytotoxicity, and could comparably produce long-lived memory NK cells that persisted in lymphoid and nonlymphoid tissues for >60 d. We demonstrate that generation and maintenance of NK cell memory is an Nfil3-independent but IL-15-dependent process. Furthermore, specific ablation of Nfil3 in either immature NK cells in the bone marrow or mature peripheral NK cells had no observable effect on NK cell lineage maintenance or homeostasis. Thus, expression of Nfil3 is crucial only early in the development of NK cells, and signals through activating receptors and proinflammatory cytokines during viral infection can bypass the requirement for Nfil3, promoting the proliferation and long-term survival of virus-specific NK cells.


Basic-Leucine Zipper Transcription Factors/genetics , Killer Cells, Natural/cytology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Lineage , Cell Separation , Cell Survival , Cytokines/metabolism , Flow Cytometry , Gene Expression Regulation , Homeostasis , Inflammation , Interferon-gamma/metabolism , Interleukin-15/metabolism , Killer Cells, Natural/virology , Ligands , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Spleen/cytology , Tamoxifen/chemistry
17.
Immunol Rev ; 253(1): 40-52, 2013 May.
Article En | MEDLINE | ID: mdl-23550637

The important role of microRNAs in directing immune responses has become increasingly clear. Here, we highlight discoveries uncovering the role of specific microRNAs in regulating the development and function of natural killer (NK) cells. Furthermore, we discuss the impact of NK cells on the entire immune system during global and specific microRNA ablation in the settings of inflammation, infection, and immune dysregulation.


Immune System Diseases/immunology , Killer Cells, Natural/immunology , MicroRNAs/immunology , Neoplasms/immunology , Animals , Cell Differentiation/genetics , Cytotoxicity, Immunologic/genetics , Homeostasis , Humans , Immune System Diseases/genetics , Immunomodulation , MicroRNAs/genetics , Neoplasms/genetics , RNA Interference/immunology
18.
Proc Natl Acad Sci U S A ; 110(17): 6967-72, 2013 Apr 23.
Article En | MEDLINE | ID: mdl-23572582

Natural killer (NK) cells function in the recognition and destruction of host cells infected with pathogens. Many regulatory mechanisms govern the potent responses of NK cells, both at the cellular and molecular level. Ablation of microRNA (miRNA) processing enzymes demonstrated that miRNAs play critical roles in NK cell differentiation and function; however, the role of individual miRNAs requires further investigation. Using mice containing a targeted deletion of microRNA-155 (miR-155), we observed defects in NK cell maintenance and maturation at steady state, as well as in homeostatic proliferation in lymphopenic mice. In addition, we discovered that miR-155 is up-regulated in activated NK cells during mouse cytomegalovirus (MCMV) infection in response to signals from the proinflammatory cytokines IL-12 and IL-18 and through signal transducer and activator of transcription 4 (STAT4) signaling. Although miR-155 was found to be dispensable for cytotoxicity and cytokine production when triggered through activating receptors, NK cells lacking miR-155 exhibited severely impaired effector and memory cell numbers in both lymphoid and nonlymphoid tissues after MCMV infection. We demonstrate that miR-155 differentially targets Noxa and suppressor of cytokine signaling 1 (SOCS1) in NK cells at distinct stages of homeostasis and activation. NK cells constitutively expressing Noxa and SOCS1 exhibit profound defects in expansion during the response to MCMV infection, suggesting that their regulation by miR-155 promotes antiviral immunity.


Gene Expression Regulation/immunology , Herpesviridae Infections/immunology , Homeostasis/immunology , Killer Cells, Natural/immunology , MicroRNAs/metabolism , Muromegalovirus/immunology , Adoptive Transfer , Animals , Chromatin Immunoprecipitation , Gene Deletion , Interleukin-12/metabolism , Interleukin-18/metabolism , Luciferases , Mice , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Real-Time Polymerase Chain Reaction , STAT4 Transcription Factor/metabolism , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
19.
PLoS One ; 6(9): e24441, 2011.
Article En | MEDLINE | ID: mdl-21915328

In mice, the transcription factor, PLZF, controls the development of effector functions in invariant NKT cells and a subset of NKT cell-like, γδ T cells. Here, we show that in human lymphocytes, in addition to invariant NKT cells, PLZF was also expressed in a large percentage of CD8+ and CD4+ T cells. Furthermore, PLZF was also found to be expressed in all γδ T cells and in all NK cells. Importantly, we show that in a donor lacking functional PLZF, all of these various lymphocyte populations were altered. Therefore, in contrast to mice, PLZF appears to control the development and/or function of a wide variety of human lymphocytes that represent more than 10% of the total PBMCs. Interestingly, the PLZF-expressing CD8+ T cell population was found to be expanded in the peripheral blood of patients with metastatic melanoma but was greatly diminished in patients with autoimmune disease.


CD8-Positive T-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/metabolism , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Cells, Cultured , Flow Cytometry , Humans , Kruppel-Like Transcription Factors/genetics , Melanoma/immunology , Melanoma/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Real-Time Polymerase Chain Reaction
20.
J Immunol ; 187(6): 2841-7, 2011 Sep 15.
Article En | MEDLINE | ID: mdl-21900183

Successful immunity depends upon the activity of multiple cell types. Commitment of pluripotent precursor cells to specific lineages, such as T or B cells, is obviously fundamental to this process. However, it is also becoming clear that continued differentiation and specialization of lymphoid cells is equally important for immune system integrity. Several members of the BTB-ZF family have emerged as critical factors that control development of specific lineages and also of specific effector subsets within these lineages. For example, BTB-ZF genes have been shown to control T cell versus B cell commitment and CD4 versus CD8 lineage commitment. Others, such as PLZF for NKT cells and Bcl-6 for T follicular helper cells, are necessary for the acquisition of effector functions. In this review, we summarize current findings concerning the BTB-ZF family members with a reported role in the immune system.


Cell Differentiation/physiology , Cell Lineage/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Transcription Factors/immunology , Zinc Fingers/immunology , Animals , Humans
...