Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
PLoS Genet ; 17(5): e1009578, 2021 05.
Article in English | MEDLINE | ID: mdl-34014925

ABSTRACT

The Receptor Transporter Protein (RTP) family is present in most, if not all jawed vertebrates. Most of our knowledge of this protein family comes from studies on mammalian RTPs, which are multi-function proteins that regulate cell-surface G-protein coupled receptor levels, influence olfactory system development, regulate immune signaling, and directly inhibit viral infection. However, mammals comprise less than one-tenth of extant vertebrate species, and our knowledge about the expression, function, and evolution of non-mammalian RTPs is limited. Here, we explore the evolutionary history of RTPs in vertebrates. We identify signatures of positive selection in many vertebrate RTP clades and characterize multiple, independent expansions of the RTP family outside of what has been described in mammals. We find a striking expansion of RTPs in the African clawed frog, Xenopus laevis, with 11 RTPs in this species as opposed to 1 to 4 in most other species. RNA sequencing revealed that most X. laevis RTPs are upregulated following immune stimulation. In functional assays, we demonstrate that at least three of these X. laevis RTPs inhibit infection by RNA viruses, suggesting that RTP homologs may serve as antiviral effectors outside of Mammalia.


Subject(s)
Antiviral Agents , Evolution, Molecular , Genomics , Membrane Transport Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Animals , Antiviral Agents/immunology , Membrane Transport Proteins/immunology , Poly I-C/immunology , Synteny , Xenopus Proteins/immunology , Xenopus laevis/immunology , Xenopus laevis/metabolism
2.
Cold Spring Harb Protoc ; 2021(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-33782096

ABSTRACT

Problems of cell biology and the molecular controls underpinning them have been studied in the remarkably versatile Xenopus systems for many years. This versatility is showcased in several accompanying protocols, which are introduced here. One protocol demonstrates how the Xenopus embryonic ectoderm can be used to study the effects of mechanical cell deformation; another illustrates how the developing eye can be used as a platform for determining cell-cycle length. Two protocols show how extracts from Xenopus embryos can be exploited to characterize the behavior of specific intracellular proteins-specifically, to determine protein phosphorylation status and the ability to bind to chromatin. Finally, because specific antibodies to Xenopus proteins are pivotal reagents for cell biology and biochemistry applications, four protocols describing how to generate, purify, and assay the specificity of antibodies raised against Xenopus proteins are included in hopes of stimulating the expansion of these critical resources across the Xenopus community.


Subject(s)
Cell Biology , Cytological Techniques/methods , Ectoderm/metabolism , Embryo, Nonmammalian/metabolism , Xenopus Proteins/metabolism , Animals , Antibodies/immunology , Antibodies/metabolism , Biochemistry/methods , Chromatin/metabolism , Ectoderm/embryology , Embryo, Nonmammalian/embryology , Humans , Immunity/immunology , Models, Animal , Phosphorylation , Protein Binding , Xenopus Proteins/immunology , Xenopus laevis
3.
Dev Biol ; 470: 74-83, 2021 02.
Article in English | MEDLINE | ID: mdl-33159936

ABSTRACT

We previously identified the protein Lbh as necessary for cranial neural crest (CNC) cell migration in Xenopus through the use of morpholinos. However, Lbh is a maternally deposited protein and morpholinos achieve knockdowns through prevention of translation. In order to investigate the role of Lbh in earlier embryonic events, we employed the new technique "Trim-Away" to degrade this maternally deposited protein. Trim-Away utilizes the E3 ubiquitin ligase trim21 to degrade proteins targeted with an antibody and was developed in mammalian systems. Our results show that Xenopus is amenable to the Trim-Away technique. We also show that early knockdown of Lbh in Xenopus results in defects in gastrulation that present with a decrease in fibronectin matrix assembly, an increased in mesodermal cell migration and decrease in endodermal cell cohesion. We further show that the technique is also effective on a second abundant maternal protein PACSIN2. We discuss potential advantages and limit of the technique in Xenopus embryos as well as the mechanism of gastrulation inhibition.


Subject(s)
Gastrulation , Xenopus Proteins/physiology , Xenopus laevis/embryology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibodies, Monoclonal/immunology , Cell Movement , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/pathology , Embryonic Induction , Endoderm/cytology , Endoderm/embryology , Endoderm/physiology , Fibronectins/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/physiology , Morpholinos , Neural Crest/cytology , Neural Crest/embryology , Proteolysis , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/immunology , Xenopus Proteins/metabolism
4.
Cytokine ; 136: 155254, 2020 12.
Article in English | MEDLINE | ID: mdl-32836028

ABSTRACT

The D1 domain of the CD4 co-receptor interacts with MHC class II during Helper CD4+ Th-cell activation and effector function in all gnathostomes but the sequence and structure of this region are not well conserved through phylogeny. Conversely, the proximal D4 domain of CD4 is the binding site of the cytokine IL-16 and is highly conserved, allowing for promiscuous binding of IL-16 to CD4 between disparate gnathostomes. We report here that recombinant human IL-16 (rhIL-16) bound to Xenopus lymphocytes to allow separation on a magnetic column. Incubation with rhIL-16 resulted in an increased expression of MHC class II mRNA by Xenopus CD8- cells more than by CD8+ cells. An in vivo assay demonstrated that rhIL-16 can recruit lymphocytes of Xenopus frogs. Our data suggest that a subset of Xenopus laevis lymphocytes express a CD4 homolog on their surface that is capable of binding IL-16. These results imply that CD4 most likely arose from a primordial cytokine receptor.


Subject(s)
CD4 Antigens/immunology , Evolution, Molecular , Interleukin-16/pharmacology , Lymphocytes/immunology , Xenopus Proteins/immunology , Animals , Humans , Interleukin-16/immunology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Xenopus laevis
5.
J Immunol ; 203(7): 1882-1896, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31492741

ABSTRACT

Comparative analyses suggest that the MHC was derived from a prevertebrate "primordial immune complex" (PIC). PIC duplicated twice in the well-studied two rounds of genome-wide duplications (2R) early in vertebrate evolution, generating four MHC paralogous regions (predominantly on human chromosomes [chr] 1, 6, 9, 19). Examining chiefly the amphibian Xenopus laevis, but also other vertebrates, we identified their MHC paralogues and mapped MHC class I, AgR, and "framework" genes. Most class I genes mapped to MHC paralogues, but a cluster of Xenopus MHC class Ib genes (xnc), which previously was mapped outside of the MHC paralogues, was surrounded by genes syntenic to mammalian CD1 genes, a region previously proposed as an MHC paralogue on human chr 1. Thus, this gene block is instead the result of a translocation that we call the translocated part of the MHC paralogous region (MHCtrans) Analyses of Xenopus class I genes, as well as MHCtrans, suggest that class I arose at 1R on the chr 6/19 ancestor. Of great interest are nonrearranging AgR-like genes mapping to three MHC paralogues; thus, PIC clearly contained several AgR precursor loci, predating MHC class I/II. However, all rearranging AgR genes were found on paralogues derived from the chr 19 precursor, suggesting that invasion of a variable (V) exon by the RAG transposon occurred after 2R. We propose models for the evolutionary history of MHC/TCR/Ig and speculate on the dichotomy between the jawless (lamprey and hagfish) and jawed vertebrate adaptive immune systems, as we found genes related to variable lymphocyte receptors also map to MHC paralogues.


Subject(s)
Antigens, CD1/genetics , Databases, Genetic , Histocompatibility Antigens Class I/genetics , Xenopus Proteins/genetics , Animals , Antigens, CD1/immunology , Histocompatibility Antigens Class I/immunology , Xenopus Proteins/immunology , Xenopus laevis
6.
Carcinogenesis ; 40(7): 924-935, 2019 07 20.
Article in English | MEDLINE | ID: mdl-31155639

ABSTRACT

The conditions that lead to antitumor or protumor functions of natural killer T (NKT) cells against mammalian tumors are only partially understood. Therefore, insights into the evolutionary conservation of NKT and their analogs-innate-like T (iT) cells-may reveal factors that contribute to tumor eradication. As such, we investigated the amphibian Xenopus laevis iT cells and interacting MHC class I-like (XNC or mhc1b.L) genes against ff-2 thymic lymphoid tumors. Upon ff-2 intraperitoneal transplantation into syngeneic tadpoles, two iT cell subsets iVα6 and iVα22, characterized by an invariant T-cell receptor α chain rearrangement (Vα6-Jα1.43 and Vα22-Jα1.32 respectively), were recruited to the peritoneum, concomitant with a decreased level of these transcripts in the spleen and thymus. To address the hypothesize that different iT cell subsets have distinct, possibly opposing, roles upon ff-2 tumor challenge, we determined whether ff-2 tumor growth could be manipulated by impairing Vα6 iT cells or by deleting their restricting element, the XNC gene, XNC10 (mhc1b10.1.L), on ff-2 tumors. Accordingly, the in vivo depletion of Vα6 iT cells using XNC10-tetramers enhanced tumor growth, indicating Vα6 iT cell-mediated antitumor activities. However, XNC10-deficient transgenic tadpoles that also lack Vα6 iT cells were resistant to ff-2 tumors, uncovering a potential new function of XNC10 besides Vα6 iT cell development. Furthermore, the CRISPR/Cas9-mediated knockout of XNC10 in ff-2 tumors broke the immune tolerance. Together, our findings demonstrate the relevance of XNC10/iT cell axis in controlling Xenopus tumor tolerance or rejection.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , Thymus Neoplasms/immunology , Tumor Escape/immunology , Xenopus Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor/transplantation , Disease Models, Animal , Gene Knockout Techniques , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Larva , Natural Killer T-Cells/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Neoplasms/pathology , Xenopus Proteins/immunology , Xenopus laevis
7.
PLoS One ; 14(5): e0216083, 2019.
Article in English | MEDLINE | ID: mdl-31048885

ABSTRACT

The use of Xenopus laevis as a model for vertebrate developmental biology is limited by a lack of antibodies specific for embryonic antigens. This study evaluated the use of immune and non-immune phage display libraries for the isolation of single domain antibodies, or nanobodies, with specificities for Xenopus embryonic antigens. The immune nanobody library was derived from peripheral blood lymphocyte RNA obtained from a llama immunized with Xenopus gastrula homogenates. Screening this library by immunostaining of embryonic tissues with pooled periplasmic material and sib-selection led to the isolation of several monoclonal phages reactive with the cytoplasm and nuclei of gastrula cells. One antigen recognized by a group of nanobodies was identified using a reverse proteomics approach as nucleoplasmin, an abundant histone chaperone. As an alternative strategy, a semi-synthetic non-immune llama nanobody phage display library was panned on highly purified Xenopus proteins. This proof-of-principle approach isolated monoclonal nanobodies that specifically bind Nuclear distribution element-like 1 (Ndel1) in multiple immunoassays. Our results suggest that immune and non-immune phage display screens on crude and purified embryonic antigens can efficiently identify nanobodies useful to the Xenopus developmental biology community.


Subject(s)
Embryonic Development/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/isolation & purification , Amino Acid Sequence , Animals , Antibodies/isolation & purification , Antibodies/metabolism , Antigens/immunology , Cell Surface Display Techniques/methods , Cytoskeletal Proteins/immunology , Gastrula , Peptide Library , Stage-Specific Embryonic Antigens/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/immunology , Xenopus laevis/embryology , Xenopus laevis/immunology , Xenopus laevis/metabolism
8.
Dev Comp Immunol ; 98: 89-97, 2019 09.
Article in English | MEDLINE | ID: mdl-31029710

ABSTRACT

Pathogens such as the Frog Virus 3 (FV3) ranavirus are contributing to the worldwide amphibian declines. While amphibian macrophages (Mϕs) are central to the immune defenses against these viruses, the pathogen recognition capacities of disparate amphibian Mϕ subsets remain unexplored. In turn, Mϕ differentiation and functionality are interdependent on the colony-stimulating factor-1 receptor (CSF-1R), which is ligated by colony-stimulating factor-1 (CSF-1) and the unrelated interleukin-34 (IL-34) cytokines. Notably, the Xenopus laevis frog CSF-1- and IL-34-derived Mϕs are functionally distinct, and while the CSF-1-Mϕs are more susceptible to FV3, the IL-34-Mϕs are highly resistant to this pathogen. Here, we elucidate the pathogen recognition capacities of CSF-1- and IL-34-differentiated Mϕs by evaluating their baseline transcript levels of key pathogen pattern recognition receptors (PRRs). Compared to the frog CSF-1-Mϕs, their IL-34-Mϕs exhibited greater expression of PRR genes associated with viral recognition as well as PRR genes known for recognizing bacterial pathogen-associated molecular patterns (PAMPs). By contrast, the CSF-1-Mϕs displayed greater expression of toll-like receptors (TLRs) that are absent in humans. Moreover, although the two Mϕ types possessed similar expression of most downstream PRR signaling components, they exhibited distinct outcomes upon stimulation with hallmark PAMPs, as measured by their tumor necrosis factor-alpha and interferon-7 gene expression. Remarkably, stimulation with a TLR2/6 agonist conferred FV3 resistance to the otherwise susceptible CSF-1-Mϕs while treatment with a TLR9 agonist significantly ablated the IL-34-Mϕ resistance to FV3. These changes in Mϕ-FV3 susceptibility and resistance appeared to be linked to changes in their expression of key immune genes. Greater understanding of the amphibian macrophage pathogen-recognition capacities will lend to further insights into the pathogen-associated causes of the amphibian declines.


Subject(s)
Cell Differentiation/immunology , Interleukins/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Ranavirus/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Xenopus Proteins/immunology , Animals , Host-Pathogen Interactions/immunology , Humans , Interleukins/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Macrophages/virology , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Ranavirus/physiology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Xenopus Proteins/metabolism
9.
Front Immunol ; 9: 2058, 2018.
Article in English | MEDLINE | ID: mdl-30258441

ABSTRACT

The glutamic acid-leucine-arginine (ELR) motif is a hallmark feature shared by mammalian inflammatory CXC chemokines such the granulocyte chemo-attractant CXCL8 (interleukin-8, IL-8). By contrast, most teleost fish inflammatory chemokines lack this motif. Interestingly, the amphibian Xenopus laevis encodes multiple isoforms of CXCL8, one of which (CXCL8a) possesses an ELR motif, while another (CXCL8b) does not. These CXCL8 isoforms exhibit distinct expression patterns during frog development and following immune challenge of animals and primary myeloid cultures. To define potential functional differences between these X. laevis CXCL8 chemokines, we produced them in recombinant form (rCXCL8a and rCXCL8b) and performed dose-response chemotaxis assays. Our results indicate that compared to rCXCL8b, rCXCL8a is a significantly more potent chemo-attractant of in vivo-derived tadpole granulocytes and of in vitro-differentiated frog bone marrow granulocytes. The mammalian CXCL8 mediates its effects through two distinct chemokine receptors, CXCR1 and CXCR2 and our pharmacological inhibition of these receptors in frog granulocytes indicates that the X. laevis CXCL8a and CXCL8b both chemoattract tadpole and adult frog granulocytes by engaging CXCR1 and CXCR2. To delineate which frog cells are recruited by CXCL8a and CXCL8b in vivo, we injected tadpoles and adult frogs intraperitoneally with rCXCL8a or rCXCL8b and recovered the accumulated cells by lavage. Our transcriptional and cytological analyses of these tadpole and adult frog peritoneal exudates indicate that they are comprised predominantly of granulocytes. Interestingly, the granulocytes recruited into the tadpole, but not adult frog peritonea by rCXCL8b, express significantly greater levels of several pan immunosuppressive genes.


Subject(s)
Chemotaxis/immunology , Evolution, Molecular , Granulocytes/immunology , Interleukin-8/immunology , Xenopus Proteins/immunology , Animals , Granulocytes/cytology , Interleukin-8/genetics , Xenopus Proteins/genetics , Xenopus laevis
10.
Proc Natl Acad Sci U S A ; 115(17): E4023-E4031, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29610296

ABSTRACT

The amphibian Xenopus laevis is to date the only species outside of mammals where a MHC class I-like (MHC-like) restricted innate-like (i) T cell subset (iVα6 T cells) reminiscent of CD1d-restricted iNKT cells has been identified and functionally characterized. This provides an attractive in vivo model to study the biological analogies and differences between mammalian iT cells and the evolutionarily antecedent Xenopus iT cell defense system. Here, we report the identification of a unique iT cell subset (Vα45-Jα1.14) requiring a distinct MHC-like molecule (mhc1b4.L or XNC4) for its development and function. We used two complementary reverse genetic approaches: RNA interference by transgenesis to impair expression of either XNC4 or the Vα45-Jα1.14 rearrangement, and CRISPR/Cas9-mediated disruption of the Jα1.14 gene segment. Both XNC4 deficiency that ablates iVα45T cell development and the direct disruption of the iVα45-Jα1.14 T cell receptor dramatically impairs tadpole resistance to Mycobacterium marinum (Mm) infection. The higher mortality of Mm-infected tadpoles deficient for iVα45T cells correlates with dysregulated expression responses of several immune genes. In contrast, iVα45-Jα1.14-deficient tadpoles remain fully competent against infection by the ranavirus FV3, which indicates a specialization of this unique iT cell subset toward mycobacterial rather than viral pathogens that involve iVα6 T cells. These data suggest that amphibians, which are evolutionarily separated from mammals by more than 350 My, have independently diversified a prominent and convergent immune surveillance system based on MHC-like interacting innate-like T cells.


Subject(s)
Histocompatibility Antigens Class I/immunology , Immunity, Cellular , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Xenopus Proteins/immunology , Animals , Histocompatibility Antigens Class I/genetics , Larva/genetics , Larva/immunology , Mycobacterium Infections, Nontuberculous/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Xenopus Proteins/genetics , Xenopus laevis
11.
J Biol Chem ; 293(5): 1736-1744, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29259133

ABSTRACT

Infections by ranaviruses such as Frog virus 3 (Fv3), are significantly contributing to worldwide amphibian population declines. Notably, amphibian macrophages (Mφs) are important to both the Fv3 infection strategies and the immune defense against this pathogen. However, the mechanisms underlying amphibian Mφ Fv3 susceptibility and resistance remain unknown. Mφ differentiation is mediated by signaling through the colony-stimulating factor-1 receptor (CSF-1R) which is now known to be bound not only by CSF-1, but also by the unrelated interleukin-34 (IL-34) cytokine. Pertinently, amphibian (Xenopus laevis) Mφs differentiated by CSF-1 and IL-34 are highly susceptible and resistant to Fv3, respectively. Accordingly, in the present work, we elucidate the facets of this Mφ Fv3 susceptibility and resistance. Because cellular resistance to viral replication is marked by expression of antiviral restriction factors, it was intuitive to find that IL-34-Mφs possess significantly greater mRNA levels of select restriction factor genes than CSF-1-Mφs. Xenopodinae amphibians have highly expanded repertoires of antiviral interferon (IFN) cytokine gene families, and our results indicated that in comparison with the X. laevis CSF-1-Mφs, the IL-34-Mφs express substantially greater transcripts of representative IFN genes, belonging to distinct gene family clades, as well as their cognate receptor genes. Finally, we demonstrate that IL-34-Mφ-conditioned supernatants confer IFN-mediated anti-Fv3 protection to the virally susceptible X. laevis kidney (A6) cell line. Together, this work underlines the differentiation pathways leading to Fv3-susceptible and -resistant amphibian Mφ populations and defines the molecular mechanisms responsible for these differences.


Subject(s)
Cell Differentiation/immunology , DNA Virus Infections/immunology , Immunity, Innate , Macrophages/immunology , Ranavirus/immunology , Animals , Interferons/immunology , Interleukins/immunology , Macrophages/virology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Xenopus Proteins/immunology , Xenopus laevis
12.
Science ; 355(6331)2017 03 24.
Article in English | MEDLINE | ID: mdl-28232581

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are heterotetrameric ion channels assembled as diheteromeric or triheteromeric complexes. Here, we report structures of the triheteromeric GluN1/GluN2A/GluN2B receptor in the absence or presence of the GluN2B-specific allosteric modulator Ro 25-6981 (Ro), determined by cryogenic electron microscopy (cryo-EM). In the absence of Ro, the GluN2A and GluN2B amino-terminal domains (ATDs) adopt "closed" and "open" clefts, respectively. Upon binding Ro, the GluN2B ATD clamshell transitions from an open to a closed conformation. Consistent with a predominance of the GluN2A subunit in ion channel gating, the GluN2A subunit interacts more extensively with GluN1 subunits throughout the receptor, in comparison with the GluN2B subunit. Differences in the conformation of the pseudo-2-fold-related GluN1 subunits further reflect receptor asymmetry. The triheteromeric NMDAR structures provide the first view of the most common NMDA receptor assembly and show how incorporation of two different GluN2 subunits modifies receptor symmetry and subunit interactions, allowing each subunit to uniquely influence receptor structure and function, thus increasing receptor complexity.


Subject(s)
Protein Multimerization , Receptors, Glutamate/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Xenopus Proteins/chemistry , Allosteric Regulation , Animals , Antibodies, Monoclonal , Cryoelectron Microscopy , Models, Molecular , Neuronal Plasticity , Protein Domains , Receptors, Glutamate/immunology , Receptors, Glutamate/ultrastructure , Receptors, N-Methyl-D-Aspartate/immunology , Receptors, N-Methyl-D-Aspartate/ultrastructure , Xenopus Proteins/immunology , Xenopus Proteins/ultrastructure , Xenopus laevis
13.
Virology ; 503: 12-20, 2017 03.
Article in English | MEDLINE | ID: mdl-28081430

ABSTRACT

Infections of amphibians by Frog Virus 3 (FV3) and other ranavirus genus members are significantly contributing to the amphibian declines, yet much remains unknown regarding amphibian antiviral immunity. Notably, amphibians represent an important step in the evolution of antiviral interferon (IFN) cytokines as they are amongst the first vertebrates to possess both type I and type III IFNs. Accordingly, we examined the roles of type I and III IFNs in the skin of FV3-challenged amphibian Xenopus laevis) tadpoles and adult frogs. Interestingly, FV3-infected tadpoles mounted type III IFN responses, whereas adult frogs relied on type I IFN immunity. Subcutaneous administration of type I or type III IFNs offered short-term protection of tadpoles against FV3 and these type I and type III IFNs induced the expression of distinct antiviral genes in the tadpole skin. Moreover, subcutaneous injection of tadpoles with type III IFN significantly extended their survival and reduced FV3 dissemination.


Subject(s)
DNA Virus Infections/immunology , Interferon Type I/immunology , Interferons/immunology , Larva/immunology , Ranavirus/immunology , Xenopus Proteins/immunology , Xenopus laevis/immunology , Xenopus laevis/virology , Animals , Azetidines/pharmacology , Cytokines/pharmacology , DNA Virus Infections/virology , Interferon Type I/pharmacology , Interferons/pharmacology , Larva/virology , Purines , Pyrazoles , Skin/immunology , Sulfonamides/pharmacology , Viral Load/immunology , Xenopus Proteins/pharmacology
14.
Dev Biol ; 426(2): 261-269, 2017 06 15.
Article in English | MEDLINE | ID: mdl-27318386

ABSTRACT

A large family of highly related and clustered Xenopus nonclassical MHC class Ib (XNC) genes influences Xenopus laevis immunity and potentially other physiological functions. Using RNA interference (RNAi) technology, we previously demonstrated that one of XNC genes, XNC10.1, is critical for the development and function of a specialized innate T (iT) cell population. However, RNAi limitation such as a variable and unstable degree of gene silencing in F0 and F1 generations is hampering a thorough functional analysis of XNC10.1 and other XNC genes. To overcome this obstacle, we adapted the CRISPR/Cas9-mediated gene editing technique for XNC genes. We efficiently and specifically generated single gene knockouts of XNC10.1, XNC11, and XNC1 as well as double gene knockouts of XNC10.1 and XNC11 in X. laevis. In single XNC10.1 knockout X. laevis tadpoles, the absence of XNC10.1 and Vα6-Jα1.43 invariant T cell receptor rearrangement transcripts indicated XNC10.1 loss-of-function and deficiency in Vα6-Jα1.43 iT cells. Notably, targeting XNC10.1 did not affect neighboring XNC genes exhibiting high sequence similarity. Furthermore, XNC1 gene disruption induced mortality during developmental stage 47, suggesting some non-immune but essential function of this gene. These data demonstrate that the CRISPR/Cas9 system can be successfully adapted for genetic analysis in F0 generation of X. laevis.


Subject(s)
CRISPR-Cas Systems , Genes, MHC Class I , Histocompatibility Antigens Class I/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Animals , Animals, Inbred Strains , Base Sequence , Chromosome Mapping , Embryo, Nonmammalian , Gene Knockout Techniques , Histocompatibility Antigens Class I/immunology , Larva , Microinjections , Multigene Family , Mutation , Protein Domains , RNA, Guide, Kinetoplastida/genetics , Reverse Genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Xenopus/genetics , Xenopus/immunology , Xenopus Proteins/immunology , Xenopus laevis/growth & development , Xenopus laevis/immunology
15.
Dev Comp Immunol ; 59: 229-39, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26855011

ABSTRACT

The intelectin (Intl) family is a group of secretory lectins in chordates that serve multiple functions, including innate immunity, through Ca(2+)-dependent recognition of carbohydrate chains. Although six Intl family lectins have so far been reported in Xenopus laevis, none have been identified in the intestine. Using a monoclonal antibody to the Xenopus embryonic epidermal lectin (XEEL or Intl-1), I identified cross-reactive proteins in the intestines. The proteins were purified by affinity chromatography on a galactose-Sepharose column and found to be oligomers consisting of N-glycosylated 39 kDa and 40.5 kDa subunit peptides. N-terminal amino acid sequencing of these peptides, followed by cDNA cloning, identified two novel Intls (designated Intl-3 and Intl-4) that showed 59-79% amino acid identities with known Xenopus Intl family proteins. From the amino acid sequence, immunoreactivity, and properties of the recombinant protein, Intl-3 was considered the intestinal lectin identified by the anti-XEEL antibody. The purified Intl-3 protein could potentially bind to Escherichia coli and its lipopolysaccharides (LPS), and to Staphylococcus aureus and its peptidoglycans, depending on Ca(2+). In addition, the Intl-3 protein agglutinated E. coli cells in the presence of Ca(2+). Intraperitoneal injection of LPS increased the intestinal and rectal contents of Intl-3 and XCL-1 (or 35K serum lectin) proteins within three days; however, unlike XCL-1, Intl-3 was detectable in neither the sera nor the other tissues regardless of LPS stimulation. Immunohistochemical analyses revealed accumulation of the Intl-3 protein in mucus secretory granules of intestinal goblet cells. The results of this study suggest that Xenopus Intl-3 is involved in the innate immune protection of the digestive tract against bacterial infections.


Subject(s)
Cytokines/immunology , Gastrointestinal Tract/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Lectins/immunology , Xenopus Proteins/immunology , Xenopus laevis/immunology , Amino Acid Sequence , Animals , Calcium/metabolism , Cloning, Molecular , Cytokines/genetics , Escherichia coli/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Intestinal Mucosa/metabolism , Lectins/genetics , Lipopolysaccharides/metabolism , Peptidoglycan/immunology , Protein Binding/immunology , Protein Structure, Tertiary , Staphylococcus aureus/immunology
16.
Biochemistry ; 55(4): 715-23, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26719910

ABSTRACT

Photolyases (PHRs) repair the UV-induced photoproducts, cyclobutane pyrimidine dimer (CPD) or pyrimidine-pyrimidone (6-4) photoproduct [(6-4) PP], restoring normal bases to maintain genetic integrity. CPD and (6-4) PP are repaired by substrate-specific PHRs, CPD PHR and (6-4) PHR, respectively. Flavin adenine dinucleotide (FAD) is the chromophore of both PHRs, and the resting oxidized form (FAD(ox)), at least under in vitro purified conditions, is first photoconverted to the neutral semiquinoid radical (FADH(•)) form, followed by photoconversion into the enzymatically active fully reduced (FADH(-)) form. Previously, we reported light-induced difference Fourier transform infrared (FTIR) spectra corresponding to the photoactivation process of Xenopus (6-4) PHR. Spectral differences between the absence and presence of (6-4) PP were observed in the photoactivation process. To identify the FTIR signals where these differences appeared, we compared the FTIR spectra of photoactivation (i) in the presence and absence of (6-4) PP, (ii) of (13)C labeling, (15)N labeling, and [(14)N]His/(15)N labeling, and (iii) of H354A and H358A mutants. We successfully assigned the vibrational bands for (6-4) PP, the α-helix and neutral His residue(s). In particular, we assigned three bands to the C ═ O groups of (6-4) PP in the three different redox states of FAD. Furthermore, the changed hydrogen bonding environments of C ═ O groups of (6-4) PP suggested restructuring of the binding pocket of the DNA lesion in the process of photoactivation.


Subject(s)
Deoxyribodipyrimidine Photo-Lyase/chemistry , Flavin-Adenine Dinucleotide/chemistry , Pyrimidine Dimers/chemistry , Xenopus Proteins/chemistry , Amino Acid Substitution , Animals , Catalytic Domain , Deoxyribodipyrimidine Photo-Lyase/genetics , Deoxyribodipyrimidine Photo-Lyase/metabolism , Flavin-Adenine Dinucleotide/genetics , Flavin-Adenine Dinucleotide/metabolism , Mutation, Missense , Pyrimidine Dimers/genetics , Pyrimidine Dimers/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/immunology , Xenopus laevis
17.
Immunogenetics ; 66(6): 411-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24771209

ABSTRACT

Nonclassical MHC class Ib (class Ib) genes are a family of highly diverse and rapidly evolving genes wherein gene numbers, organization, and expression markedly differ even among closely related species rendering class Ib phylogeny difficult to establish. Whereas among mammals there are few unambiguous class Ib gene orthologs, different amphibian species belonging to the anuran subfamily Xenopodinae exhibit an unusually high degree of conservation among multiple class Ib gene lineages. Comparative genomic analysis of class Ib gene loci of two divergent (~65 million years) Xenopodinae subfamily members Xenopus laevis (allotetraploid) and Xenopus tropicalis (diploid) shows that both species possess a large cluster of class Ib genes denoted as Xenopus/Silurana nonclassical (XNC/SNC). Our study reveals two distinct phylogenetic patterns among these genes: some gene lineages display a high degree of flexibility, as demonstrated by species-specific expansion and contractions, whereas other class Ib gene lineages have been maintained as monogenic subfamilies with very few changes in their nucleotide sequence across divergent species. In this second category, we further investigated the XNC/SNC10 gene lineage that in X. laevis is required for the development of a distinct semi-invariant T cell population. We report compelling evidence of the remarkable high degree of conservation of this gene lineage that is present in all 12 species of the Xenopodinae examined, including species with different degrees of ploidy ranging from 2, 4, 8 to 12 N. This suggests that the critical role of XNC10 during early T cell development is conserved in amphibians.


Subject(s)
Genome , Histocompatibility Antigens Class I/genetics , Phylogeny , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus/genetics , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Amino Acid Sequence , Animals , Biological Evolution , Conserved Sequence , Histocompatibility Antigens Class I/classification , Histocompatibility Antigens Class I/immunology , Molecular Sequence Data , Ploidies , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Species Specificity , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Xenopus/classification , Xenopus/immunology , Xenopus Proteins/classification , Xenopus Proteins/immunology , Xenopus laevis/immunology
18.
Carcinogenesis ; 35(8): 1807-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24776220

ABSTRACT

Non-classical class Ib (class Ib) genes are found in all jawed vertebrates, including the amphibian Xenopus, which possesses at least 20 distinct Xenopus non-classical class Ib genes (XNCs). As an immune evasion strategy, tumors often downregulate surface expression of classical major histocompatibility complex class Ia molecules. In contrast, cancers commonly express class Ib molecules, presenting an alternative for tumor immune recognition. We characterized a novel XNC, XNC10, functionally similar to CD1d from a class Ia-deficient thymic lymphoid tumor (15/0), which grows aggressively in Xenopus LG-15 cloned animals. To investigate the roles of XNC10 in antitumor immunity, we generated stable 15/0-transfectants with silenced XNC10 mRNA and protein expression. Notably, XNC10 silencing resulted in acute tumor rejection by naturally class Ia-deficient syngeneic tadpoles, with greater potency of rejection in tumors with more efficient XNC10 knockdown. In vivo killing assays shows that the rejection of XNC10-deficient tumors is due to a cell-mediated cytotoxic immune response elicited by the tadpole host. Importantly, priming enhances XNC10-deficient tumor rejection. Flow cytometry reveals that XNC10-deficient tumor rejection is associated with an accumulation of XNC10-restricted invariant T cells and conventional CD8 T cells as well as other leukocytes. Similarly, semisolid tumor grafts in tadpoles also exhibit leukocytes infiltration. These findings suggest that XNC10 allows the 15/0-tumor to escape immune recognition and class Ia-independent cytotoxicity, thus emphasizing the critical roles of class Ibs in tumor immunity.


Subject(s)
Histocompatibility Antigens Class I/immunology , Larva/immunology , Lymphoid Tissue/immunology , Thymus Neoplasms/immunology , Tumor Escape/immunology , Xenopus Proteins/immunology , Xenopus laevis/immunology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Female , Flow Cytometry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Immunoenzyme Techniques , Larva/metabolism , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/growth & development , Xenopus laevis/metabolism
19.
OMICS ; 17(7): 384-92, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23829578

ABSTRACT

TNFSF13 is one of the tumor necrosis factor (TNF) superfamily members that plays important roles in immune homeostasis and proliferation or apoptosis of certain tumor cell lines. This report describes the development of Xenopus laevis TNFSF13 as a model to study its important role in relation to immunological diseases. In brief, TNFSF13 from Xenopus laevis (designated XlTNFSF13) was first amplified by RT-PCR and rapid amplification of cDNA end (RACE) techniques. Bioinformatics analyses revealed the gene structure, three-dimensional structure, and evolutionary relationships. Real-time quantitative PCR (QPCR) analysis identified the tissue distribution of XlTNFSF13 in the major visceral organs. The recombinant plasmid SUMO-XsTNFSF13 was expressed in E. coli Rosseta (DE3). Subsequently, the recombinant protein purified through Ni-NTA affinity chromatography was analyzed by SDS-PAGE and confirmed by Western blot analysis. Laser scanning confocal microscopy analysis revealed the binding activity of pSUMO-XsTNFSF13 to the surface of B cells. WST-8 assays further indicated that purified XsTNFSF13 could cause the survival/proliferation of B cells. In conclusion, we underscore that as a model organism for human disease, Xenopus laevis has been widely used in molecular biology research. Yet while TNFSF13 research in mammalian, fish (e.g., zebrafish), mouse, and human is widely available, studies in the amphibian species are limited. The latter area of OMICS and integrative biology scholarship is directly informed with the present study, with a view to implications for the future study of human immunological diseases.


Subject(s)
B-Cell Activating Factor/genetics , B-Cell Activating Factor/immunology , Immune System Diseases/genetics , Xenopus Proteins/genetics , Xenopus Proteins/immunology , Amino Acid Sequence , Animals , B-Cell Activating Factor/biosynthesis , B-Lymphocytes/immunology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Cloning, Molecular/methods , Computational Biology/methods , DNA, Complementary/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Immune System Diseases/immunology , Mice , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Conformation , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Tissue Distribution , Xenopus Proteins/biosynthesis , Xenopus laevis
20.
Cancer Immun ; 13: 4, 2013.
Article in English | MEDLINE | ID: mdl-23390375

ABSTRACT

The heat shock proteins (HSPs) gp96 and HSP70 mediate potent antigen-dependent anti-tumor T cell responses in both mammals and Xenopus laevis. We have shown that frogs immunized with total HSP70 generate CD8+ T cell responses against the Xenopus thymic lymphoid tumor 15/0 that expresses several non-classical MHC class Ib (class Ib) genes, but no classical MHC class Ia (class Ia). In the absence of class Ia, we hypothesized that hsp72 can prime class Ib-mediated anti-tumor unconventional CD8+ T cells in an antigen-dependent manner. To test this, we produced Xenopus recombinant HSP70 proteins (both the cognate hsc73 and the inducible hsp72) from stable 15/0 tumor transfectants. We used an in vivo cross-presentation assay to prime animals by adoptive transfer of HSP-pulsed antigen-presenting cells (APCs) and showed that both hsp72-and hsc73-Ag complexes have a similar potential to elicit class Ia-mediated T cell responses against minor histocompatibility (H) Ag skin grafts. In contrast, our in vivo cross-presentation assay revealed that hsp72 was more potent than hsc73 in generating protective immune responses against the class Ia-negative 15/0 tumors in an Ag-dependent and class Ib-mediated manner. These results suggest that hsp72 can stimulate class Ib-mediated immune responses and represents a promising candidate for immunotherapy against malignancies with downregulated class Ia expression.


Subject(s)
HSC70 Heat-Shock Proteins/immunology , HSP72 Heat-Shock Proteins/immunology , Histocompatibility Antigens Class I/immunology , Neoplasms/immunology , Xenopus laevis/immunology , Animals , Cross-Priming/immunology , Graft Rejection/immunology , HSC70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/isolation & purification , HSP72 Heat-Shock Proteins/metabolism , Immunity/immunology , Leukocytes/metabolism , Neoplasms/pathology , Recombinant Proteins/isolation & purification , Skin Transplantation/immunology , Xenopus Proteins/immunology , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...