Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
1.
J Reprod Immunol ; 163: 104223, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38489930

ABSTRACT

Autophagy is a process that occurs in almost all eukaryotic cells and this process is controlled by several molecular processes. Its biological roles include the provision of energy, the maintenance of cell homeostasis, and the promotion of aberrant cell death. The importance of autophagy in pregnancy is gradually becoming recognized. In literature, it has been indicated that autophagy has three different effects on the onset and maintenance of pregnancy: embryo (embryonic development), feto-maternal immune crosstalk, and maternal (decidualization). In humans, proper decidualization is a major predictor of pregnancy accomplishment and it can be influenced by different factors. This review highlights the genes, pathways, regulation, and function of autophagy in endometrial decidualization and other involved factors in this process.


Subject(s)
Autophagy , Decidua , Endometrium , Pregnancy Complications , Signal Transduction , Humans , Female , Pregnancy , Autophagy/immunology , Signal Transduction/immunology , Pregnancy Complications/immunology , Decidua/immunology , Decidua/metabolism , Endometrium/immunology , Endometrium/metabolism , Animals , Embryonic Development/immunology , Embryonic Development/genetics , Embryo Implantation/immunology
2.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36902134

ABSTRACT

Autoimmune thyroid disease (AITD) is one of the most common endocrinopathies and is more prevalent in women. It becomes evident that the circulating antithyroid antibodies that often follow AITD have effects on many tissues, including ovaries, and therefore that this common morbidity might have an impact on female fertility, the investigation of which is the aim of the present research. Ovarian reserve, ovarian response to stimulation and early embryo development in infertile patients with thyroid autoimmunity were assessed in 45 women with thyroid autoimmunity and 45 age-matched control patients undergoing infertility treatment. It was demonstrated that the presence of anti-thyroid peroxidase antibodies is associated with lower serum anti-Müllerian hormone levels and antral follicle count. Further investigation revealed the higher prevalence of sub-optimal response to ovarian stimulation in TAI-positive women, lower fertilization rate and lower number of high-quality embryos in this group of patients. The cut-off value for follicular fluid anti-thyroid peroxidase antibody affecting the above-mentioned parameters was determined to be 105.0 IU/mL, highlighting the necessity of closer monitoring in couples seeking infertility treatment with ART.


Subject(s)
Autoantibodies , Embryonic Development , Infertility, Female , Iodide Peroxidase , Ovarian Reserve , Sperm Injections, Intracytoplasmic , Thyroiditis, Autoimmune , Female , Humans , Infertility, Female/etiology , Infertility, Female/therapy , Ovarian Reserve/immunology , Embryonic Development/immunology , Autoantibodies/immunology , Iodide Peroxidase/immunology , Thyroiditis, Autoimmune/complications
3.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638989

ABSTRACT

The latest vaccination campaign has actualized the potential impact of antigenic stimuli on reproductive functions. To address this, we mimicked vaccination's effects by administering keyhole limpet hemocyanin (KLH ) to CD1 male mice and used their sperm for in vitro fertilization (IVF). Two-cell embryos after IVF with spermatozoa from control (C) or KLH-treated (Im) male mice were transferred to surrogate mothers mated with vasectomized control (C) or KLH-treated (Im) male mice, resulting in four experimental groups: C-C, Im-C, C-Im, and Im-Im. The pre-implantation losses were significantly lower in the Im-C group than in the C-Im group. At the same time, the resorption rates reduced markedly in the C-Im compared to the Im-C group. Embryo and placenta weights were significantly higher in the Im-Im group. Although the GM-CSF levels were lower in the amniotic fluid of the gestating surrogate mothers in the Im-Im group, they were strongly correlated with embryo mass. The number-size trade-off was only significant in the Im-Im group. This suggests a positive, cooperative effect of spermatozoa and seminal fluid from immune-primed males on embryo growth and the optimal distribution of surrogate mother maternal resources despite the negative impact of males' antigenic challenge on the IVF success rate.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Embryo Transfer/methods , Embryonic Development/immunology , Fertilization in Vitro/methods , Hemocyanins/administration & dosage , Semen/immunology , Spermatozoa/immunology , Vaccination/methods , Animals , Antibodies/blood , Blastocyst/immunology , Blastocyst/metabolism , Cell Division/immunology , Embryo Implantation/immunology , Female , Hemocyanins/immunology , Immunoglobulin G/blood , Male , Mice , Pregnancy , Vasectomy/methods
4.
FASEB J ; 35(10): e21938, 2021 10.
Article in English | MEDLINE | ID: mdl-34547143

ABSTRACT

Uterine glands are essential for the establishment of pregnancy and have critical roles in endometrial receptivity to blastocyst implantation, stromal cell decidualization, and placentation. Uterine gland dysfunction is considered a major contributing factor to pregnancy loss, however our understanding of how glands impact embryo survival and stromal cell decidualization is incomplete. Forkhead box A2 (FOXA2) is expressed only in the glandular epithelium and regulates its development and function. Mice with a conditional deletion of FOXA2 in the uterus are infertile due to defective embryo implantation arising from a lack of leukemia inhibitory factor (LIF), a critical factor of uterine gland origin. Here, a glandless FOXA2-deficient mouse model, coupled with LIF repletion to rescue the implantation defect, was used to investigate the roles of uterine glands in embryo survival and decidualization. Studies found that embryo survival and decidualization were compromised in glandless FOXA2-deficient mice on gestational day 6.5, resulting in abrupt pregnancy loss by day 7.5. These findings strongly support the hypothesis that uterine glands secrete factors other than LIF that impact embryo survival and stromal cell decidualization for pregnancy success.


Subject(s)
Decidua/metabolism , Embryo Loss , Embryo, Mammalian/embryology , Embryonic Development , Stromal Cells/metabolism , Uterus/metabolism , Animals , Decidua/immunology , Embryo Loss/immunology , Embryo, Mammalian/immunology , Embryonic Development/immunology , Female , Hepatocyte Nuclear Factor 3-beta/deficiency , Leukemia Inhibitory Factor , Mice , Pregnancy , Pregnancy Outcome , Stromal Cells/immunology , Transcriptome , Uterus/immunology
5.
Front Immunol ; 12: 714090, 2021.
Article in English | MEDLINE | ID: mdl-34497610

ABSTRACT

Although most causes of death and morbidity in premature infants are related to immune maladaptation, the premature immune system remains poorly understood. We provide a comprehensive single-cell depiction of the neonatal immune system at birth across the spectrum of viable gestational age (GA), ranging from 25 weeks to term. A mass cytometry immunoassay interrogated all major immune cell subsets, including signaling activity and responsiveness to stimulation. An elastic net model described the relationship between GA and immunome (R=0.85, p=8.75e-14), and unsupervised clustering highlighted previously unrecognized GA-dependent immune dynamics, including decreasing basal MAP-kinase/NFκB signaling in antigen presenting cells; increasing responsiveness of cytotoxic lymphocytes to interferon-α; and decreasing frequency of regulatory and invariant T cells, including NKT-like cells and CD8+CD161+ T cells. Knowledge gained from the analysis of the neonatal immune landscape across GA provides a mechanistic framework to understand the unique susceptibility of preterm infants to both hyper-inflammatory diseases and infections.


Subject(s)
Biomarkers , Embryonic Development/immunology , Immune System Phenomena , Single-Cell Analysis , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Communication , Disease Susceptibility/immunology , Gene Expression Regulation , Gestational Age , Humans , Immunomodulation , Infant, Newborn , Premature Birth , Signal Transduction , Single-Cell Analysis/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34370006

ABSTRACT

B1 lymphocytes are a small but unique component of the innate immune-like cells. However, their ontogenic origin is still a matter of debate. Although it is widely accepted that B1 cells originate early in fetal life, whether or not they arise from hematopoietic stem cells (HSCs) is still unclear. In order to shed light on the B1 cell origin, we set out to determine whether their lineage specification is dependent on Notch signaling, which is essential for the HSC generation and, therefore, all derivatives lineages. Using mouse embryonic stem cells (mESCs) to recapitulate murine embryonic development, we have studied the requirement for Notch signaling during the earliest B-cell lymphopoiesis and found that Rbpj-deficient mESCs are able to generate B1 cells. Their Notch independence was confirmed in ex vivo experiments using Rbpj-deficient embryos. In addition, we found that upregulation of Notch signaling induced the emergence of B2 lymphoid cells. Taken together, these findings indicate that control of Notch signaling dose is crucial for different B-cell lineage specification from endothelial cells and provides pivotal information for their in vitro generation from PSCs for therapeutic applications. This article has an associated 'The people behind the papers' interview.


Subject(s)
B-Lymphocyte Subsets/immunology , Embryonic Development/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/immunology , Endothelial Cells/immunology , Hematopoietic Stem Cells/immunology , Mice , Mice, Inbred C57BL
7.
Semin Cell Dev Biol ; 120: 133-146, 2021 12.
Article in English | MEDLINE | ID: mdl-34147339

ABSTRACT

Research using avian embryos has led to major conceptual advances in developmental biology, virology, immunology, genetics and cell biology. The avian embryo has several significant advantages, including ready availability and ease of accessibility, rapid development with marked similarities to mammals and a high amenability to manipulation. As mechanical forces are increasingly recognised as key drivers of morphogenesis, this powerful model system is shedding new light on the mechanobiology of embryonic development. Here, we highlight progress in understanding how mechanical forces direct key morphogenetic processes in the early avian embryo. Recent advances in quantitative live imaging and modelling are elaborating upon traditional work using physical models and embryo manipulations to reveal cell dynamics and tissue forces in ever greater detail. The recent application of transgenic technologies further increases the strength of the avian model and is providing important insights about previously intractable developmental processes.


Subject(s)
Bird Diseases/embryology , Embryonic Development/immunology , Animals , Gastrulation
8.
Eur J Immunol ; 51(4): 764-772, 2021 04.
Article in English | MEDLINE | ID: mdl-33569778

ABSTRACT

The development of the human immune system during embryonic and fetal life has historically been difficult to research due to limited access to human tissue. Experimental animal models have been widely used to study development but cellular and molecular programmes may not be conserved across species. The advent of multiomic single-cell technologies and an increase in human developmental tissue biobank resources have facilitated single-cell multiomic studies focused on human immune development. A critical question in the near future is "How do we best reconcile scientific findings across multiple omic modalities, developmental time, and organismic space?" In this review, we discuss the application of single-cell multiomic technologies to unravel the major cellular lineages in the prenatal human immune system. We also identify key areas where the combined power of multiomics technologies can be leveraged to address specific immunological gaps in our current knowledge and explore new research horizons in human development.


Subject(s)
Embryonic Development/immunology , Epigenomics/methods , Gene Expression Profiling/methods , Genomics/methods , Proteomics/methods , Single-Cell Analysis/methods , Cell Lineage/genetics , Cell Lineage/immunology , Embryo, Mammalian/embryology , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , High-Throughput Nucleotide Sequencing/methods , Humans
9.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33452132

ABSTRACT

OCT4 is a fundamental component of the molecular circuitry governing pluripotency in vivo and in vitro. To determine how OCT4 establishes and protects the pluripotent lineage in the embryo, we used comparative single-cell transcriptomics and quantitative immunofluorescence on control and OCT4 null blastocyst inner cell masses at two developmental stages. Surprisingly, activation of most pluripotency-associated transcription factors in the early mouse embryo occurs independently of OCT4, with the exception of the JAK/STAT signaling machinery. Concurrently, OCT4 null inner cell masses ectopically activate a subset of trophectoderm-associated genes. Inspection of metabolic pathways implicates the regulation of rate-limiting glycolytic enzymes by OCT4, consistent with a role in sustaining glycolysis. Furthermore, up-regulation of the lysosomal pathway was specifically detected in OCT4 null embryos. This finding implicates a requirement for OCT4 in the production of normal trophectoderm. Collectively, our findings uncover regulation of cellular metabolism and biophysical properties as mechanisms by which OCT4 instructs pluripotency.


Subject(s)
Cell Lineage/genetics , Embryonic Development/immunology , Octamer Transcription Factor-3/genetics , STAT3 Transcription Factor/genetics , Animals , Blastocyst Inner Cell Mass/metabolism , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Glycolysis/genetics , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics , Single-Cell Analysis
10.
Immunol Med ; 44(1): 23-29, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32649853

ABSTRACT

Recently, great advancements have been made towards understanding the mechanisms underlying dermatomyositis (DM). Many novel autoantibodies, such as anti-MDA5, anti-TIF1γ, anti-NXP2, and anti-SAE, have been reported to be involved in DM. DM is now classified based on these myositis-specific autoantibodies. Anti-TIF1γ antibodies are closely associated with juvenile DM and adult cancer-associated DM. Anti-TIF1γ antibody-positive DM tends to present severe cutaneous manifestations, mild myositis, and dysphagia. TIF1γ (also known as TRIM33) plays a role in transcriptional elongation, DNA repair, differentiation of cells, embryonic development, and mitosis. Moreover, TIF1γ has been shown to suppress various tumors via the TGF-ß/Smad and the Wnt/ß-Catenin signaling pathways. In this review, we explore the relationship between TIF1γ, cancer, and DM. We also discuss the pathogenesis of anti-TIF1γ antibody-positive DM.


Subject(s)
Autoantibodies , Dermatomyositis/immunology , Nuclear Proteins/immunology , Transcription Factors/immunology , DNA Repair/immunology , Embryonic Development/immunology , Humans , Mitosis/immunology , Transcription Elongation, Genetic/immunology , Transcription Factors/physiology , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/immunology
11.
J Reprod Immunol ; 143: 103240, 2021 02.
Article in English | MEDLINE | ID: mdl-33166807

ABSTRACT

Insufficient experimental studies have reported the effect of ovalbumin (OVA) as an allergen towards embryonic growth in asthma mouse model. The impact of 10 µg/200 µL OVA on maternal inflammatory and oxidative stress (OS) responses, and preimplantation embryonic development was investigated in this study. We first established OVA-induced asthma mouse model, and following superovulation, mated the females and challenged them with Methacholine (Mch) test. Upon embryo retrieval, only those with the highest implantation potential were cultured in vitro. Significant reduction in the number of embryos at each preimplantation stage was noted in the treated group. Uneven sized blastomeres at 2-, 4- and 8-cell stages were also evident in this group. Embryo fragmentation was significant at only 2-, 4- and 8-cell stages. We also found that OVA tended to raise maternal inflammatory and OS biomarker levels as well as to cause inappropriate levels of pregnancy hormones progesterone (P4) and estrogen (E2) although insignificant. The combined results indicate that 10 µg/200 µL OVA had altered both quality and quantity of the embryos in asthma mouse model although its effect on pregnancy hormones, inflammatory and OS responses were non-pathological.


Subject(s)
Asthma/immunology , Embryonic Development/immunology , Pregnancy Complications/immunology , Animals , Asthma/blood , Disease Models, Animal , Estrogens/blood , Female , Humans , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , Oxidative Stress/immunology , Pregnancy , Pregnancy Complications/blood , Progesterone/blood
12.
J Genet Genomics ; 47(9): 547-561, 2020 09 20.
Article in English | MEDLINE | ID: mdl-33309050

ABSTRACT

Suppressive regulatory T cells (Treg cells) play a vital role in preventing autoimmunity and restraining excessive immune response to both self- and non-self-antigens. Studies on humans and mice show that the Forkhead box p3 (Foxp3) is a key regulatory gene for the development and function of Treg cells. In zebrafish, Treg cells have been identified by using foxp3a as a reliable marker. However, little is known about the function of foxp3a and Treg cells in gonadal development and sex differentiation. Here, we show that foxp3a is essential for maintaining immune homeostasis in zebrafish testis development. We found that foxp3a was specifically expressed in a subset of T cells in zebrafish testis, while knockout of foxp3a led to deficiency of foxp3a-positive Treg cells in the testis. More than 80% of foxp3a-/- mutants developed as subfertile males, and the rest of the mutants developed as fertile females with decreased ovulation. Further study revealed that foxp3a-/- mutants had a delayed juvenile ovary-to-testis transition in definite males and sex reversal in about half of the definite females, which led to a dominance of later male development. Owing to the absence of foxp3a-positive Treg cells in the differentiating testis of foxp3a-/- mutants, abundant T cells and macrophages expand to disrupt an immunosuppressive milieu, resulting in defective development of germ cells and gonadal somatic cells and leading to development of infertile males. Therefore, our study reveals that foxp3a-positive Treg cells play an essential role in the orchestration of gonadal development and sex differentiation in zebrafish.


Subject(s)
Embryonic Development/genetics , Forkhead Transcription Factors/genetics , Testis/growth & development , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Embryonic Development/immunology , Female , Homeostasis/genetics , Homeostasis/immunology , Humans , Immune System/growth & development , Immune System/metabolism , Male , Ovary/growth & development , Sex Differentiation/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology , Testis/immunology , Testis/metabolism , Zebrafish/growth & development
13.
Front Immunol ; 11: 555414, 2020.
Article in English | MEDLINE | ID: mdl-33329514

ABSTRACT

It is generally understood that the entry of semen into the female reproductive tract provokes molecular and cellular changes facilitating conception and pregnancy. We show a broader picture of the participation of prostaglandins in the fertilization, implantation and maintenance of the embryo. A large number of cells and molecules are related to signaling networks, which regulate tolerance to implantation and maintenance of the embryo and fetus. In this work, many of those cells and molecules are analyzed. We focus on platelets, polymorphonuclear leukocytes, and group 2 innate lymphoid cells involved in embryo tolerance in order to have a wider view of how prostaglandins participate. The combination of platelets and neutrophil extracellular traps (Nets), uterine innate lymphoid cells (uILC), Treg cells, NK cells, and sex hormones have an important function in immunological tolerance. In both animals and humans, the functions of these cells can be regulated by prostaglandins and soluble factors in seminal plasma to achieve an immunological balance, which maintains fetal-maternal tolerance. Prostaglandins, such as PGI2 and PGE2, play an important role in the suppression of the previously mentioned cells. PGI2 inhibits platelet aggregation, in addition to IL-5 and IL-13 expression in ILC2, and PGE2 inhibits some neutrophil functions, such as chemotaxis and migration processes, leukotriene B4 (LTB4) biosynthesis, ROS production, and the formation of extracellular traps, which could help prevent trophoblast injury and fetal loss. The implications are related to fertility in female when seminal fluid is deposited in the vagina or uterus.


Subject(s)
Embryonic Development/genetics , Embryonic Development/immunology , Immune Tolerance , Prostaglandins/metabolism , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Embryo, Mammalian , Female , Fertilization , Genitalia, Female , Humans , Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Maternal-Fetal Exchange/immunology , Pregnancy , Semen , Signal Transduction
14.
Mol Biol Rep ; 47(12): 9959-9965, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33226564

ABSTRACT

In-vitro fertilization is a routine livestock-breeding technique widely used around the world. Several studies have reported the interaction of bovine viral-diarrhea virus (BVDV) with gametes and in-vitro-produced (IVP) bovine embryos. Since, gene expression in BVDV-infected IVP bovine embryos is scarcely addressed. The aim of this work was to evaluate the differential expression of genes involved in immune and inflammatory response. Groups of 20-25 embryos on Day 6 (morula stage) were exposed (infected) or not (control) to an NCP-BVDV strain in SOF medium. After 24 h, embryos that reached expanded blastocyst stage were washed. Total RNA of each embryo group was extracted to determine the transcription levels of 9 specific transcripts related with antiviral and inflammatory response by SYBR Green real time quantitative (RT-qPCR). Culture media and an aliquot of the last embryos wash on Day 7 were analyzed by titration and virus isolation, respectively. A conventional PCR confirmed BVDV presence in IVP embryos. A significantly higher expression of interferon-α was observed in blastocysts exposed to NCP-BVDV compared to the controls (p < 0.05). In this study, the upregulation of INFα and TLR7 genes involved in inflammatory and immune response in BVDV-infected IVP bovine embryos is a new finding in this field. This differential expression suggest that embryonic cells could function in a manner like immune cells by recognizing and responding early to interaction with viral pathogens. These results provide new insights into the action of BVDV on the complex molecular pathways controlling bovine early embryonic development.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease , Cattle , Diarrhea Viruses, Bovine Viral/immunology , Embryonic Development/immunology , Gene Expression/immunology , Interferon-alpha , Animals , Bovine Virus Diarrhea-Mucosal Disease/embryology , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle/embryology , Cattle/immunology , Diarrhea Viruses, Bovine Viral/isolation & purification , Embryo, Mammalian/immunology , Embryo, Mammalian/virology , Female , Fertilization in Vitro , Interferon-alpha/immunology , Toll-Like Receptor 7/immunology
15.
Front Immunol ; 11: 592010, 2020.
Article in English | MEDLINE | ID: mdl-33193435

ABSTRACT

During pregnancy, the maternal uterus and fetus form a special microenvironment at the maternal-fetal interface to support fetal development. Extravillous trophoblasts (EVTs), differentiated from the fetus, invade into the decidua and interact with maternal cells. Human leukocyte antigen (HLA)-G is a non-classical MHC-I molecule that is expressed abundantly and specifically on EVTs in physiological conditions. Soluble HLA-G (sHLA-G) is also found in maternal blood, amniotic fluid, and cord blood. The abnormal expression and polymorphisms of HLA-G are related to adverse pregnancy outcomes such as preeclampsia (PE) and recurrent spontaneous abortion (RSA). Here we summarize current findings about three main roles of HLA-G during pregnancy, namely its promotion of spiral artery remodeling, immune tolerance, and fetal growth, all resulting from its interaction with immune cells. These findings are not only of great significance for the treatment of pregnancy-related diseases but also provide clues to tumor immunology research since HLA-G functions as a checkpoint in tumors.


Subject(s)
Cellular Microenvironment/immunology , HLA-G Antigens/immunology , Histocompatibility, Maternal-Fetal , Maternal-Fetal Exchange/immunology , Placenta/immunology , Placenta/metabolism , Animals , Biomarkers , Embryonic Development/immunology , Female , Gene Expression Regulation, Developmental , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Pregnancy , Signal Transduction , Trophoblasts/immunology , Trophoblasts/metabolism
16.
Mol Immunol ; 126: 129-135, 2020 10.
Article in English | MEDLINE | ID: mdl-32823237

ABSTRACT

Kalliklectin is a unique fish-specific lectin, whose sequence is similar to the heavy chain of mammalian plasma kallikrein and coagulation factor XI. In this study, we aimed to evaluate dynamic expression profiles of the lectin gene, during early developmental stages, in fugu, Takifugu rubripes. Reverse transcription-polymerase chain reaction (RT-PCR) showed that the kalliklectin gene was not expressed until 14 h post-fertilization (hpf), while the mRNA was detected after 30 hpf. In real-time quantitative PCR (qPCR), the gene was first expressed at 10.5 hpf; then, the expression level increased with a peak at 30 hpf and then gradually decreased. On the other hand, western blotting with specific antibody detected the lectin protein at all tested stages, including the unfertilized egg, which suggests that the lectin detected in the early stages was a maternal factor. Immunohistochemistry demonstrated that kalliklectin was localized at the basement membranes of the newly hatched larvae, while the lectin was widely detected in epidermal cells in larva at 5 dph. A 40-kDa lectin was partially purified from unfertilized eggs using mannose-affinity chromatography, and the lectin was determined as kalliklectin by liquid chromatography with quadrupole time-of-flight tandem mass spectrometry (LC/Q-TOF-MS) analysis, which indicated that the lectin is functional in the eggs. The egg lectin can bind to Gram-positive bacterial pathogens of fish, such as Lactococcus garvieae and Streptococcus iniae. We conclude that fugu kalliklectin might be an important immunocomponent, transferred from mother to offspring.


Subject(s)
Embryonic Development/immunology , Fish Proteins/metabolism , Immunity, Maternally-Acquired , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Takifugu/growth & development , Animals , Embryo, Nonmammalian , Female , Fish Proteins/immunology , Gene Expression Profiling , Lactococcus/immunology , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/immunology , Ovum/immunology , Ovum/metabolism , Receptors, Cell Surface/immunology , Streptococcus iniae/immunology , Takifugu/immunology , Takifugu/microbiology
17.
Science ; 369(6503): 530-537, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32732419

ABSTRACT

Microglia, immune cells of the central nervous system (CNS), are important for tissue development and maintenance and are implicated in CNS disease, but we lack understanding of human fetal microglia development. Single-cell gene expression and bulk chromatin profiles of microglia at 9 to 18 gestational weeks (GWs) of human fetal development were generated. Microglia were heterogeneous at all studied GWs. Microglia start to mature during this developmental period and increasingly resemble adult microglia with CNS-surveilling properties. Chromatin accessibility increases during development with associated transcriptional networks reflective of adult microglia. Thus, during early fetal development, microglia progress toward a more mature, immune-sensing competent phenotype, and this might render the developing human CNS vulnerable to environmental perturbations during early pregnancy.


Subject(s)
Brain/embryology , Embryonic Development/immunology , Fetus/immunology , Microglia/immunology , Phagocytosis/immunology , Brain/cytology , Cell Separation , Cells, Cultured , Embryonic Development/genetics , Gene Regulatory Networks , Humans , Phagocytosis/genetics , Transcriptome
18.
Int J Mol Sci ; 21(14)2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32650396

ABSTRACT

The large family of C-type lectin (CLEC) receptors comprises carbohydrate-binding proteins that require Ca2+ to bind a ligand. The prototypic receptor is the asialoglycoprotein receptor-1 (ASGR1, CLEC4H1) that is expressed primarily by hepatocytes. The early work on ASGR1, which is highly specific for N-acetylgalactosamine (GalNAc), established the foundation for understanding the overall function of CLEC receptors. Cells of the immune system generally express more than one CLEC receptor that serve diverse functions such as pathogen-recognition, initiation of cellular signaling, cellular adhesion, glycoprotein turnover, inflammation and immune responses. The receptor CLEC10A (C-type lectin domain family 10 member A, CD301; also called the macrophage galactose-type lectin, MGL) contains a carbohydrate-recognition domain (CRD) that is homologous to the CRD of ASGR1, and thus, is also specific for GalNAc. CLEC10A is most highly expressed on immature DCs, monocyte-derived DCs, and alternatively activated macrophages (subtype M2a) as well as oocytes and progenitor cells at several stages of embryonic development. This receptor is involved in initiation of TH1, TH2, and TH17 immune responses and induction of tolerance in naïve T cells. Ligand-mediated endocytosis of CLEC receptors initiates a Ca2+ signal that interestingly has different outcomes depending on ligand properties, concentration, and frequency of administration. This review summarizes studies that have been carried out on these receptors.


Subject(s)
Asialoglycoprotein Receptor/metabolism , Lectins, C-Type/metabolism , Animals , Asialoglycoprotein Receptor/immunology , Calcium Signaling/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Embryonic Development/immunology , Humans , Immunity/immunology , Lectins, C-Type/immunology , Ligands , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Front Immunol ; 11: 727, 2020.
Article in English | MEDLINE | ID: mdl-32411141

ABSTRACT

In this study we have assessed the effects of increased cortisol levels during early embryonic development on immune function in zebrafish (Danio rerio) larvae. Fertilized eggs were exposed to either a cortisol-containing, a dexamethasone-containing (to stimulate the glucocorticoid receptor selectively) or a control medium for 6 h post-fertilization (0-6 hpf). First, we measured baseline expression of a number of immune-related genes (socs3a, mpeg1.1, mpeg1.2, and irg1l) 5 days post-fertilization (dpf) in larvae of the AB and TL strain to assess the effectiveness of our exposure procedure and potential strain differences. Cortisol and dexamethasone strongly up-regulated baseline expression of these genes independent of strain. The next series of experiments were therefore carried out in larvae of the AB strain only. We measured neutrophil/macrophage recruitment following tail fin amputation (performed at 3 dpf) and phenotypical changes as well as survival following LPS-induced sepsis (150 µg/ml; 4-5 dpf). Dexamethasone, but not cortisol, exposure at 0-6 hpf enhanced neutrophil recruitment 4 h post tail fin amputation. Cortisol and dexamethasone exposure at 0-6 hpf led to a milder phenotype (e.g., less tail fin damage) and enhanced survival following LPS challenge compared to control exposure. Gene-expression analysis showed accompanying differences in transcript abundance of tlr4bb, cxcr4a, myd88, il1ß, and il10. These data show that early-life exposure to cortisol, which may be considered to be a model or proxy of maternal stress, induces an adaptive response to immune challenges, which seems mediated via the glucocorticoid receptor.


Subject(s)
Embryonic Development/drug effects , Embryonic Development/immunology , Hydrocortisone/pharmacology , Immune System/drug effects , Larva/drug effects , Animals , Dexamethasone/pharmacology , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Neutrophils/drug effects , Receptors, Glucocorticoid/drug effects , Survival , Zebrafish
20.
Dev Comp Immunol ; 111: 103748, 2020 10.
Article in English | MEDLINE | ID: mdl-32442442

ABSTRACT

Two galactose-binding proteins were purified from the eggs of Takifugu rubripes by affinity chromatography. These proteins were detected at 26 and 23 kDa under reducing and at 40 and 45 kDa under non-reducing conditions at SDS-PAGE. The peptide sequences from both proteins matched to short-type pentraxin. The 26-kDa lectin was glycosylated, while the other one was not, indicating that these could be glycoforms of pentraxin. Messenger RNA of pentraxin was detected in eggs and embryos at 1-cell stage, was undetectable till blastula, and finally detected again after gastrula, suggesting that the mRNAs in eggs and 1-cell embryos were maternal in origin, and autologous transcription of the gene occurred after blastula. Since they bind to pathogenic bacteria, egg pentraxins may have immunological functions during embryogenesis. This is the first study to show the presence of short-type pentraxin in fish eggs and the diversity of fish egg lectins.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , C-Reactive Protein/metabolism , Fish Proteins/metabolism , Serum Amyloid P-Component/metabolism , Takifugu/immunology , Animals , C-Reactive Protein/genetics , Carbohydrates/immunology , Cells, Cultured , Chromatography, Affinity , Embryonic Development/immunology , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Life Cycle Stages , Ovum , Protein Binding , Serum Amyloid P-Component/genetics
SELECTION OF CITATIONS
SEARCH DETAIL