Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 587
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 162(3): 505-15, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26213383

RESUMO

Exposure to maternal tissue during in utero development imprints tolerance to immunologically foreign non-inherited maternal antigens (NIMA) that persists into adulthood. The biological advantage of this tolerance, conserved across mammalian species, remains unclear. Here, we show maternal cells that establish microchimerism in female offspring during development promote systemic accumulation of immune suppressive regulatory T cells (Tregs) with NIMA specificity. NIMA-specific Tregs expand during pregnancies sired by males expressing alloantigens with overlapping NIMA specificity, thereby averting fetal wastage triggered by prenatal infection and non-infectious disruptions of fetal tolerance. Therefore, exposure to NIMA selectively enhances reproductive success in second-generation females carrying embryos with overlapping paternally inherited antigens. These findings demonstrate that genetic fitness, canonically thought to be restricted to Mendelian inheritance, is enhanced in female placental mammals through vertically transferred maternal cells that promote conservation of NIMA and enforce cross-generational reproductive benefits.


Assuntos
Feto/imunologia , Aptidão Genética , Tolerância Imunológica , Mamíferos/fisiologia , Gravidez/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos/imunologia , Quimerismo , Feminino , Humanos , Masculino , Mamíferos/imunologia , Camundongos , Placenta/imunologia
2.
Cell ; 157(1): 142-50, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24679532

RESUMO

The virome contains the most abundant and fastest mutating genetic elements on Earth. The mammalian virome is constituted of viruses that infect host cells, virus-derived elements in our chromosomes, and viruses that infect the broad array of other types of organisms that inhabit us. Virome interactions with the host cannot be encompassed by a monotheistic view of viruses as pathogens. Instead, the genetic and transcriptional identity of mammals is defined in part by our coevolved virome, a concept with profound implications for understanding health and disease.


Assuntos
Bacteriófagos/fisiologia , Mamíferos/virologia , Microbiota , Vírus/genética , Animais , Bactérias/virologia , Bacteriófagos/classificação , Bacteriófagos/genética , Interações Hospedeiro-Patógeno , Humanos , Mamíferos/genética , Mamíferos/imunologia , Vírus/classificação , Vírus/metabolismo
3.
Cell ; 159(4): 829-43, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25417159

RESUMO

Aging of immune organs, termed as immunosenescence, is suspected to promote systemic inflammation and age-associated disease. The cause of immunosenescence and how it promotes disease, however, has remained unclear. We report that the Drosophila fat body, a major immune organ, undergoes immunosenescence and mounts strong systemic inflammation that leads to deregulation of immune deficiency (IMD) signaling in the midgut of old animals. Inflamed old fat bodies secrete circulating peptidoglycan recognition proteins that repress IMD activity in the midgut, thereby promoting gut hyperplasia. Further, fat body immunosenecence is caused by age-associated lamin-B reduction specifically in fat body cells, which then contributes to heterochromatin loss and derepression of genes involved in immune responses. As lamin-associated heterochromatin domains are enriched for genes involved in immune response in both Drosophila and mammalian cells, our findings may provide insights into the cause and consequence of immunosenescence during mammalian aging. PAPERFLICK:


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Corpo Adiposo/imunologia , Lamina Tipo B/metabolismo , Envelhecimento , Animais , Proliferação de Células , Drosophila melanogaster/química , Drosophila melanogaster/imunologia , Corpo Adiposo/crescimento & desenvolvimento , Corpo Adiposo/metabolismo , Trato Gastrointestinal/crescimento & desenvolvimento , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Heterocromatina , Inflamação/imunologia , Mamíferos/imunologia , Modelos Animais , Transdução de Sinais
4.
Mol Cell ; 81(24): 5099-5111.e8, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34919820

RESUMO

The SARS-CoV-2 spike protein is a critical component of vaccines and a target for neutralizing monoclonal antibodies (nAbs). Spike is also undergoing immunogenic selection with variants that increase infectivity and partially escape convalescent plasma. Here, we describe Spike Display, a high-throughput platform to rapidly characterize glycosylated spike ectodomains across multiple coronavirus-family proteins. We assayed ∼200 variant SARS-CoV-2 spikes for their expression, ACE2 binding, and recognition by 13 nAbs. An alanine scan of all five N-terminal domain (NTD) loops highlights a public epitope in the N1, N3, and N5 loops recognized by most NTD-binding nAbs. NTD mutations in variants of concern B.1.1.7 (alpha), B.1.351 (beta), B.1.1.28 (gamma), B.1.427/B.1.429 (epsilon), and B.1.617.2 (delta) impact spike expression and escape most NTD-targeting nAbs. Finally, B.1.351 and B.1.1.28 completely escape a potent ACE2 mimic. We anticipate that Spike Display will accelerate antigen design, deep scanning mutagenesis, and antibody epitope mapping for SARS-CoV-2 and other emerging viral threats.


Assuntos
Mamíferos/virologia , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , COVID-19/imunologia , COVID-19/virologia , Linhagem Celular , Epitopos/genética , Epitopos/imunologia , Células HEK293 , Humanos , Mamíferos/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia
5.
Immunity ; 50(4): 1033-1042.e6, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30926232

RESUMO

Ancient organisms have a combined coagulation and immune system, and although links between inflammation and hemostasis exist in mammals, they are indirect and slower to act. Here we investigated direct links between mammalian immune and coagulation systems by examining cytokine proproteins for potential thrombin protease consensus sites. We found that interleukin (IL)-1α is directly activated by thrombin. Thrombin cleaved pro-IL-1α at a site perfectly conserved across disparate species, indicating functional importance. Surface pro-IL-1α on macrophages and activated platelets was cleaved and activated by thrombin, while tissue factor, a potent thrombin activator, colocalized with pro-IL-1α in the epidermis. Mice bearing a mutation in the IL-1α thrombin cleavage site (R114Q) exhibited defects in efficient wound healing and rapid thrombopoiesis after acute platelet loss. Thrombin-cleaved IL-1α was detected in humans during sepsis, pointing to the relevance of this pathway for normal physiology and the pathogenesis of inflammatory and thrombotic diseases.


Assuntos
Coagulação Sanguínea/fisiologia , Sistema Imunitário/imunologia , Interleucina-1alfa/fisiologia , Trombina/fisiologia , Imunidade Adaptativa , Sequência de Aminoácidos , Animais , Plaquetas/metabolismo , Humanos , Imunidade Inata , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Queratinócitos/metabolismo , Macrófagos/metabolismo , Mamíferos/imunologia , Camundongos , Precursores de Proteínas/metabolismo , Seleção Genética , Sepse/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Trombopoese/imunologia , Cicatrização/imunologia
6.
Immunity ; 49(3): 545-559.e5, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30193848

RESUMO

Although the mammalian microbiota is well contained within the intestine, it profoundly shapes development and metabolism of almost every host organ. We questioned the range and depth of microbial metabolite penetration into the host, and how this is modulated by intestinal immunity. Chemically identical microbial and host metabolites were distinguished by stable isotope tracing from 13C-labeled live non-replicating Escherichia coli, differentiating 12C host isotopes with high-resolution mass spectrometry. Hundreds of endogenous microbial compounds penetrated 23 host tissues and fluids after intestinal exposure: subsequent 12C host metabolome signatures included lipidemia, reduced glycolysis, and inflammation. Penetrant bacterial metabolites from the small intestine were rapidly cleared into the urine, whereas induced antibodies curtailed microbial metabolite exposure by accelerating intestinal bacterial transit into the colon where metabolite transport mechanisms are limiting. Pervasive penetration of microbial molecules can cause extensive host tissue responses: these are limited by immune and non-immune intestinal mucosal adaptations to the microbiota.


Assuntos
Anticorpos/metabolismo , Microbioma Gastrointestinal/fisiologia , Glicólise/imunologia , Hiperlipidemias/imunologia , Inflamação/imunologia , Mamíferos/imunologia , Animais , Anticorpos/imunologia , Radioisótopos de Carbono/análise , Interações Hospedeiro-Patógeno , Imunidade , Cadeias Pesadas de Imunoglobulinas/genética , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Cell ; 150(1): 29-38, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22770213

RESUMO

Regulatory T (Treg) cells, whose differentiation and function are controlled by X chromosome-encoded transcription factor Foxp3, are generated in the thymus (tTreg) and extrathymically (peripheral, pTreg), and their deficiency results in fatal autoimmunity. Here, we demonstrate that a Foxp3 enhancer, conserved noncoding sequence 1 (CNS1), essential for pTreg but dispensable for tTreg cell generation, is present only in placental mammals. CNS1 is largely composed of mammalian-wide interspersed repeats (MIR) that have undergone retrotransposition during early mammalian radiation. During pregnancy, pTreg cells specific to a model paternal alloantigen were generated in a CNS1-dependent manner and accumulated in the placenta. Furthermore, when mated with allogeneic, but not syngeneic, males, CNS1-deficient females showed increased fetal resorption accompanied by increased immune cell infiltration and defective remodeling of spiral arteries. Our results suggest that, during evolution, a CNS1-dependent mechanism of extrathymic differentiation of Treg cells emerged in placental animals to enforce maternal-fetal tolerance.


Assuntos
Tolerância Imunológica , Mamíferos/imunologia , Placenta/citologia , Placenta/imunologia , Gravidez/imunologia , Linfócitos T Reguladores/imunologia , Animais , Elementos Facilitadores Genéticos , Feminino , Feto/imunologia , Fatores de Transcrição Forkhead/genética , Humanos , Masculino , Mamíferos/genética , Camundongos , Gambás
8.
Trends Immunol ; 44(2): 129-145, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36623953

RESUMO

There are striking similarities between the sea urchin cavity macrophage-like phagocytes (coelomocytes) and mammalian cavity macrophages in not only their location, but also their behaviors. These cells are crucial for maintaining homeostasis within the cavity following a breach, filling the gap and functioning as a barrier between vital organs and the environment. In this review, we summarize the evolving literature regarding these Gata6+ large peritoneal macrophages (GLPMs), focusing on ontogeny, their responses to perturbations, including their rapid aggregation via coagulation, as well as scavenger receptor cysteine-rich domains and their potential roles in diseases, such as cancer. We challenge the 50-year old phenomenon of the 'macrophage disappearance reaction' (MDR) and propose the new term 'macrophage disturbance of homeostasis reaction' (MDHR), which may better describe this complex phenomenon.


Assuntos
Fator de Transcrição GATA6 , Macrófagos Peritoneais , Mamíferos , Animais , Fator de Transcrição GATA6/imunologia , Macrófagos Peritoneais/imunologia , Mamíferos/imunologia , Fagócitos/imunologia , Ouriços-do-Mar/imunologia
9.
Nature ; 586(7829): 424-428, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33029010

RESUMO

Since 1814, when rubella was first described, the origins of the disease and its causative agent, rubella virus (Matonaviridae: Rubivirus), have remained unclear1. Here we describe ruhugu virus and rustrela virus in Africa and Europe, respectively, which are, to our knowledge, the first known relatives of rubella virus. Ruhugu virus, which is the closest relative of rubella virus, was found in apparently healthy cyclops leaf-nosed bats (Hipposideros cyclops) in Uganda. Rustrela virus, which is an outgroup to the clade that comprises rubella and ruhugu viruses, was found in acutely encephalitic placental and marsupial animals at a zoo in Germany and in wild yellow-necked field mice (Apodemus flavicollis) at and near the zoo. Ruhugu and rustrela viruses share an identical genomic architecture with rubella virus2,3. The amino acid sequences of four putative B cell epitopes in the fusion (E1) protein of the rubella, ruhugu and rustrela viruses and two putative T cell epitopes in the capsid protein of the rubella and ruhugu viruses are moderately to highly conserved4-6. Modelling of E1 homotrimers in the post-fusion state predicts that ruhugu and rubella viruses have a similar capacity for fusion with the host-cell membrane5. Together, these findings show that some members of the family Matonaviridae can cross substantial barriers between host species and that rubella virus probably has a zoonotic origin. Our findings raise concerns about future zoonotic transmission of rubella-like viruses, but will facilitate comparative studies and animal models of rubella and congenital rubella syndrome.


Assuntos
Mamíferos/virologia , Filogenia , Vírus da Rubéola/classificação , Vírus da Rubéola/isolamento & purificação , Sequência de Aminoácidos , Animais , Animais de Zoológico/imunologia , Animais de Zoológico/virologia , Membrana Celular/virologia , Quirópteros/virologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Equidae/imunologia , Equidae/virologia , Evolução Molecular , Feminino , Mapeamento Geográfico , Alemanha , Especificidade de Hospedeiro , Humanos , Masculino , Mamíferos/imunologia , Marsupiais/imunologia , Marsupiais/virologia , Fusão de Membrana , Camundongos , Modelos Animais , Modelos Moleculares , Rubéola (Sarampo Alemão)/congênito , Rubéola (Sarampo Alemão)/virologia , Vírus da Rubéola/química , Vírus da Rubéola/imunologia , Alinhamento de Sequência , Uganda , Proteínas do Envelope Viral/química
10.
Nat Immunol ; 14(4): 320-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23507645

RESUMO

Recent years have witnessed a renaissance in the study of fish immune systems. Such studies have greatly expanded the knowledge of the evolution and diversification of vertebrate immune systems. Several findings in those studies have overturned old paradigms about the immune system and led to the discovery of novel aspects of mammalian immunity. Here I focus on how findings pertaining to immunity in teleost (bony) fish have led to major new insights about mammalian B cell function in innate and adaptive immunity. Additionally, I illustrate how the discovery of the most ancient mucosal immunoglobulin described thus far will help resolve unsettled paradigms of mammalian mucosal immunity.


Assuntos
Peixes/imunologia , Mamíferos/imunologia , Animais , Linfócitos B/imunologia , Imunidade nas Mucosas , Fagocitose/imunologia , Vertebrados/imunologia
11.
Allergy ; 79(6): 1440-1454, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38193233

RESUMO

The recent recognition of a syndrome of tick-acquired mammalian meat allergy has transformed the previously held view that mammalian meat is an uncommon allergen. The syndrome, mediated by IgE antibodies against the oligosaccharide galactose-alpha-1,3-galactose (alpha-gal), can also involve reactions to visceral organs, dairy, gelatin and other products, including medications sourced from non-primate mammals. Thus, fittingly, this allergic disorder is now called the alpha-gal syndrome (AGS). The syndrome is strikingly regional, reflecting the important role of tick bites in sensitization, and is more common in demographic groups at risk of tick exposure. Reactions in AGS are delayed, often by 2-6 h after ingestion of mammalian meat. In addition to classic allergic symptomatology such as urticaria and anaphylaxis, AGS is increasingly recognized as a cause of isolated gastrointestinal morbidity and alpha-gal sensitization has also been linked with cardiovascular disease. The unusual link with tick bites may be explained by the fact that allergic cells and mediators are mobilized to the site of tick bites and play a role in resistance against ticks and tick-borne infections. IgE directed to alpha-gal is likely an incidental consequence of what is otherwise an adaptive immune strategy for host defense against endo- and ectoparasites, including ticks.


Assuntos
Anafilaxia , Hipersensibilidade Alimentar , Imunoglobulina E , Picadas de Carrapatos , Doenças Transmitidas por Carrapatos , Urticária , Animais , Humanos , Alérgenos/imunologia , Anafilaxia/imunologia , Anafilaxia/etiologia , Anafilaxia/diagnóstico , Dissacarídeos/imunologia , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/etiologia , Imunoglobulina E/imunologia , Mamíferos/imunologia , Carne/efeitos adversos , Síndrome , Picadas de Carrapatos/imunologia , Picadas de Carrapatos/complicações , Carrapatos/imunologia , Urticária/imunologia , Urticária/etiologia , Doenças Transmitidas por Carrapatos/imunologia
12.
EMBO J ; 38(8)2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30872283

RESUMO

To protect against the harmful consequences of viral infections, organisms are equipped with sophisticated antiviral mechanisms, including cell-intrinsic means to restrict viral replication and propagation. Plant and invertebrate cells utilise mostly RNA interference (RNAi), an RNA-based mechanism, for cell-intrinsic immunity to viruses while vertebrates rely on the protein-based interferon (IFN)-driven innate immune system for the same purpose. The RNAi machinery is conserved in vertebrate cells, yet whether antiviral RNAi is still active in mammals and functionally relevant to mammalian antiviral defence is intensely debated. Here, we discuss cellular and viral factors that impact on antiviral RNAi and the contexts in which this system might be at play in mammalian resistance to viral infection.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Mamíferos/imunologia , Interferência de RNA , RNA Viral/genética , Viroses/imunologia , Vírus/imunologia , Animais , Antivirais/administração & dosagem , Interações Hospedeiro-Patógeno/genética , Mamíferos/genética , Mamíferos/virologia , Viroses/genética , Viroses/virologia , Replicação Viral , Vírus/isolamento & purificação
13.
Annu Rev Microbiol ; 72: 399-422, 2018 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-29927706

RESUMO

Mammalian immune systems evolved within a diverse world dominated by microbes, making interactions between these two life-forms inevitable. Adaptive immunity protects against microbes through antigen-specific responses. In classical studies, these responses were investigated in the context of pathogenicity; however, we now know that they have significant effects on our resident microbes. In turn, microbes employ an arsenal of mechanisms to influence development and specificity of host immunity. Understanding these complex reactions will be necessary to develop microbiota-based strategies to prevent or treat disease. Here we review the literature detailing the cross talk between resident microbes with a focus on the specificity of host responses and the microbial molecules that influence them.


Assuntos
Imunidade Adaptativa , Interações Hospedeiro-Patógeno , Imunidade Inata , Mamíferos/imunologia , Mamíferos/microbiologia , Microbiota/imunologia , Animais , Humanos
14.
J Immunol ; 207(2): 371-375, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34233911

RESUMO

The repertoire of Abs is generated by genomic rearrangements during B cell differentiation. Although V(D)J rearrangements lead to repertoires mostly different between individuals, recent studies have shown that they contain a substantial fraction of overrepresented and shared "public" clones. We previously reported a strong public IgHµ clonotypic response against the rhabdovirus viral hemorrhagic septicemia virus in a teleost fish. In this study, we identified an IgL chain associated with this public response that allowed us to characterize its functionality. We show that this public Ab response has a potent neutralizing capacity that is typically associated with host protection during rhabdovirus infections. We also demonstrate that the public response is not restricted to a particular trout isogenic line but expressed in multiple genetic backgrounds and may be used as a marker of successful vaccination. Our work reveals that public B cell responses producing generic Abs constitute a mechanism of protection against infection conserved across vertebrates.


Assuntos
Formação de Anticorpos/imunologia , Peixes/imunologia , Mamíferos/imunologia , Animais , Linfócitos B/imunologia , Células Clonais/imunologia , Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/imunologia , Recombinação V(D)J/imunologia , Vacinação/métodos
15.
J Immunol ; 206(5): 1046-1057, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33472906

RESUMO

The zebrafish (Danio rerio) is a powerful model organism for studies of the innate immune system. One apparent difference between human and zebrafish innate immunity is the cellular machinery for LPS sensing. In amniotes, the protein complex formed by TLR4 and myeloid differentiation factor 2 (Tlr4/Md-2) recognizes the bacterial molecule LPS and triggers an inflammatory response. It is believed that zebrafish have neither Md-2 nor Tlr4; Md-2 has not been identified outside of amniotes, whereas the zebrafish tlr4 genes appear to be paralogs, not orthologs, of amniote TLR4s We revisited these conclusions. We identified a zebrafish gene encoding Md-2, ly96 Using single-cell RNA sequencing, we found that ly96 is transcribed in cells that also transcribe genes diagnostic for innate immune cells, including the zebrafish tlr4-like genes. In larval zebrafish, ly96 is expressed in a small number of macrophage-like cells. In a functional assay, zebrafish Md-2 and Tlr4ba form a complex that activates NF-κB signaling in response to LPS. In larval zebrafish ly96 loss-of-function mutations perturbed LPS-induced cytokine production but gave little protection against LPS toxicity. Finally, by analyzing the genomic context of tlr4 genes in 11 jawed vertebrates, we found that tlr4 arose prior to the divergence of teleosts and tetrapods. Thus, an LPS-sensitive Tlr4/Md-2 complex is likely an ancestral feature shared by mammals and zebrafish, rather than a de novo invention on the tetrapod lineage. We hypothesize that zebrafish retain an ancestral, low-sensitivity Tlr4/Md-2 complex that confers LPS responsiveness to a specific subset of innate immune cells.


Assuntos
Antígeno 96 de Linfócito/genética , Receptor 4 Toll-Like/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Linhagem Celular , Células HEK293 , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/imunologia , Antígeno 96 de Linfócito/imunologia , Macrófagos/imunologia , Mamíferos/genética , Mamíferos/imunologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Peixe-Zebra/imunologia , Proteínas de Peixe-Zebra/imunologia
16.
Proc Natl Acad Sci U S A ; 117(11): 5977-5986, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123117

RESUMO

Understanding the molecular basis of adaptation to the environment is a central question in evolutionary biology, yet linking detected signatures of positive selection to molecular mechanisms remains challenging. Here we demonstrate that combining sequence-based phylogenetic methods with structural information assists in making such mechanistic interpretations on a genomic scale. Our integrative analysis shows that positively selected sites tend to colocalize on protein structures and that positively selected clusters are found in functionally important regions of proteins, indicating that positive selection can contravene the well-known principle of evolutionary conservation of functionally important regions. This unexpected finding, along with our discovery that positive selection acts on structural clusters, opens previously unexplored strategies for the development of better models of protein evolution. Remarkably, proteins where we detect the strongest evidence of clustering belong to just two functional groups: Components of immune response and metabolic enzymes. This gives a coherent picture of pathogens and xenobiotics as important drivers of adaptive evolution of mammals.


Assuntos
Adaptação Fisiológica , Evolução Molecular , Mamíferos/genética , Mamíferos/fisiologia , Seleção Genética , Animais , Meio Ambiente , Enzimas/química , Genômica , Imunidade , Mamíferos/imunologia , Modelos Moleculares , Filogenia , Conformação Proteica , Proteínas/química
17.
Proc Natl Acad Sci U S A ; 117(1): 610-618, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31843890

RESUMO

APOBEC3 (A3) genes are members of the AID/APOBEC gene family that are found exclusively in mammals. A3 genes encode antiviral proteins that restrict the replication of retroviruses by inducing G-to-A mutations in their genomes and have undergone extensive amplification and diversification during mammalian evolution. Endogenous retroviruses (ERVs) are sequences derived from ancient retroviruses that are widespread mammalian genomes. In this study we characterize the A3 repertoire and use the ERV fossil record to explore the long-term history of coevolutionary interaction between A3s and retroviruses. We examine the genomes of 160 mammalian species and identify 1,420 AID/APOBEC-related genes, including representatives of previously uncharacterized lineages. We show that A3 genes have been amplified in mammals and that amplification is positively correlated with the extent of germline colonization by ERVs. Moreover, we demonstrate that the signatures of A3-mediated mutation can be detected in ERVs found throughout mammalian genomes and show that in mammalian species with expanded A3 repertoires, ERVs are significantly enriched for G-to-A mutations. Finally, we show that A3 amplification occurred concurrently with prominent ERV invasions in primates. Our findings establish that conflict with retroviruses is a major driving force for the rapid evolution of mammalian A3 genes.


Assuntos
Desaminases APOBEC/genética , Retrovirus Endógenos/genética , Evolução Molecular , Interações Hospedeiro-Patógeno/genética , Mamíferos/genética , Desaminases APOBEC/metabolismo , Animais , Retrovirus Endógenos/imunologia , Fósseis/virologia , Interações Hospedeiro-Patógeno/imunologia , Mamíferos/imunologia , Mamíferos/virologia , Mutação , Filogenia , Edição de RNA/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/imunologia , RNA Viral/metabolismo
18.
J Zoo Wildl Med ; 53(4): 832-837, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36640087

RESUMO

Staphylococcal A and streptococcal G proteins are widely used in immunoassays when specific immunological reagents are unavailable, such as for wild animals. The affinity of bacterial proteins A and G to the immunoglobulins of seven Brazilian mammals were tested, including black-tufted marmoset (Callithrix penicillata, n = 5), golden-bellied capuchin (Sapajus xanthosternos, n = 13), woolly mouse opossum (Micoureus demerarae, n = 6), long-nosed armadillo (Dasypus novemcinctus, n = 5), collared anteater (Tamandua tetradactyla, n = 5), ocelot (Leopardus pardalis, n = 6), and vampire bat (Desmodus rotundus, n = 5). Blood samples were collected from animals that were rescued in peri-urban rainforest fragments. Sera pools of each species were tested by ELISA to determine the intensity of each bacterial protein affinity to the immunoglobulins. When comparing the affinity to both proteins, immunoglobulins from D. rotundus, S. xanthosternos, and T. tetradactyla presented a higher affinity to protein G, whereas a higher affinity to protein A was found for immunoglobulins of C. penicillata and L. pardalis. The only species that presented a very low affinity to both bacterial proteins was M. demerarae. This study can be used as a reference for further studies on the development of sensitive and specific immunodiagnostic assays to be used for the monitoring of the health of these wild mammals.


Assuntos
Proteínas de Bactérias , Imunoglobulinas , Mamíferos , Animais , Animais Selvagens/imunologia , Proteínas de Bactérias/imunologia , Brasil , Imunoglobulinas/imunologia , Mamíferos/imunologia
19.
PLoS Biol ; 17(11): e3000528, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31751331

RESUMO

The immune system comprises a complex network of specialized cells that protects against infection, eliminates cancerous cells, and regulates tissue repair, thus serving a critical role in homeostasis, health span, and life span. The subterranean-dwelling naked mole-rat (NM-R; Heterocephalus glaber) exhibits prolonged life span relative to its body size, is unusually cancer resistant, and manifests few physiological or molecular changes with advancing age. We therefore hypothesized that the immune system of NM-Rs evolved unique features that confer enhanced cancer immunosurveillance and prevent the age-associated decline in homeostasis. Using single-cell RNA-sequencing (scRNA-seq) we mapped the immune system of the NM-R and compared it to that of the short-lived, cancer-prone mouse. In contrast to the mouse, we find that the NM-R immune system is characterized by a high myeloid-to-lymphoid cell ratio that includes a novel, lipopolysaccharide (LPS)-responsive, granulocyte cell subset. Surprisingly, we also find that NM-Rs lack canonical natural killer (NK) cells. Our comparative genomics analyses support this finding, showing that the NM-R genome lacks an expanded gene family that controls NK cell function in several other species. Furthermore, we reconstructed the evolutionary history that likely led to this genomic state. The NM-R thus challenges our current understanding of mammalian immunity, favoring an atypical, myeloid-biased mode of innate immunosurveillance, which may contribute to its remarkable health span.


Assuntos
Ratos-Toupeira/genética , Ratos-Toupeira/imunologia , Animais , Evolução Biológica , Biologia Computacional/métodos , Genoma , Genômica/métodos , Longevidade/genética , Mamíferos/imunologia , Camundongos/imunologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma/genética
20.
J Immunol ; 205(4): 994-1008, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32641385

RESUMO

Mucosal surfaces such as fish gills interface between the organism and the external environment and as such are major sites of foreign Ag encounter. In the gills, the balance between inflammatory responses to waterborne pathogens and regulatory responses toward commensal microbes is critical for effective barrier function and overall fish health. In mammals, IL-4 and IL-13 in concert with IL-10 are essential for balancing immune responses to pathogens and suppressing inflammation. Although considerable progress has been made in the field of fish immunology in recent years, whether the fish counterparts of these key mammalian cytokines perform similar roles is still an open question. In this study, we have generated IL-4/13A and IL-4/13B mutant zebrafish (Danio rerio) and, together with an existing IL-10 mutant line, characterized the consequences of loss of function of these cytokines. We demonstrate that IL-4/13A and IL-4/13B are required for the maintenance of a Th2-like phenotype in the gills and the suppression of type 1 immune responses. As in mammals, IL-10 appears to have a more striking anti-inflammatory function than IL-4-like cytokines and is essential for gill homeostasis. Thus, both IL-4/13 and IL-10 paralogs in zebrafish exhibit aspects of conserved function with their mammalian counterparts.


Assuntos
Proteínas de Peixes/imunologia , Brânquias/imunologia , Homeostase/imunologia , Inflamação/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Peixe-Zebra/imunologia , Animais , Imunidade/imunologia , Interleucina-13/imunologia , Mamíferos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA