Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
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
2.
Front Immunol ; 13: 891220, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35967383

RESUMO

Ym1 is a rodent-specific chitinase-like protein (CLP) lacking catalytic activity, whose cellular origins are mainly macrophages, neutrophils and other cells. Although the detailed function of Ym1 remains poorly understood, Ym1 has been generally recognized as a fundamental feature of alternative activation of macrophages in mice and hence one of the prevalent detecting targets in macrophage phenotype distinguishment. Studies have pointed out that Ym1 may have regulatory effects, which are multifaceted and even contradictory, far more than just a mere marker. Allergic lung inflammation, parasite infection, autoimmune diseases, and central nervous system diseases have been found associations with Ym1 to varying degrees. Thus, insights into Ym1's role in diseases would help us understand the pathogenesis of different diseases and clarify the genuine roles of CLPs in mammals. This review summarizes the information on Ym1 from the gene to its expression and regulation and focuses on the association between Ym1 and diseases.


Assuntos
Doença , Lectinas , Macrófagos , beta-N-Acetil-Hexosaminidases , Animais , Quitinases/genética , Quitinases/imunologia , Doença/genética , Imunidade/genética , Imunidade/imunologia , Lectinas/genética , Lectinas/imunologia , Macrófagos/imunologia , Mamíferos/genética , Mamíferos/imunologia , Camundongos , Neutrófilos/imunologia , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/imunologia
3.
Sci Rep ; 11(1): 22098, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764369

RESUMO

Small antibody mimetics that contain high-affinity target-binding peptides can be lower cost alternatives to monoclonal antibodies (mAbs). We have recently developed a method to create small antibody mimetics called FLuctuation-regulated Affinity Proteins (FLAPs), which consist of a small protein scaffold with a structurally immobilized target-binding peptide. In this study, to further develop this method, we established a novel screening system for FLAPs called monoclonal antibody-guided peptide identification and engineering (MAGPIE), in which a mAb guides selection in two manners. First, antibody-guided design allows construction of a peptide library that is relatively small in size, but sufficient to identify high-affinity binders in a single selection round. Second, in antibody-guided screening, the fluorescently labeled mAb is used to select mammalian cells that display FLAP candidates with high affinity for the target using fluorescence-activated cell sorting. We demonstrate the reliability and efficacy of MAGPIE using daclizumab, a mAb against human interleukin-2 receptor alpha chain (CD25). Three FLAPs identified by MAGPIE bound CD25 with dissociation constants of approximately 30 nM as measured by biolayer interferometry without undergoing affinity maturation. MAGPIE can be broadly adapted to any mAb to develop small antibody mimetics.


Assuntos
Anticorpos Monoclonais/imunologia , Técnicas de Visualização da Superfície Celular/métodos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Mamíferos/imunologia , Ligação Proteica/imunologia , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo/métodos , Células HEK293 , Células HeLa , Humanos , Células K562 , Biblioteca de Peptídeos
4.
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
5.
J Immunol ; 205(11): 3071-3082, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33148715

RESUMO

Malaria is associated with complicated immunopathogenesis. In this study, we provide evidence for an unexpected role of TLR3 in promoting the establishment of Plasmodium yoelii infection through delayed clearance of parasitemia in wild type C57BL/6jRj (B6) compared with TLR3 knockout mice. In this study, we confirmed an increased expression of Tlr3, Trif, Tbk1, and Irf7/Irf3 in the liver 42 h postinfection and the initiation of an early burst of proinflammatory response such as Ifng, NF-kB, and Tnfa in B6 mice that may promote parasite fitness. Interestingly, in the absence of TLR3, we showed the involvement of high IFN-γ and lower type I IFN response in the early clearance of parasitemia. In parallel, we observed an increase in splenic NK and NKT cells expressing TLR3 in infected B6 mice, suggesting a role for TLR sensing in the innate immune response. Finally, we find evidence that the increase in the frequency of CD19+TLR3+ B cells along with reduced levels of total IgG in B6 mice possibly suggests the initiation of TLR3-dependent pathway early during P. yoelii infection. Our results thus reveal a new mechanism in which a parasite-activated TLR3 pathway promotes blood stage infection along with quantitative and qualitative differences in Ab responses.


Assuntos
Malária/imunologia , Mamíferos/imunologia , Mamíferos/parasitologia , Plasmodium yoelii/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Linfócitos B/imunologia , Imunidade Inata/imunologia , Imunoglobulina G/imunologia , Inflamação/imunologia , Inflamação/parasitologia , Interferon Tipo I/imunologia , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/parasitologia , Parasitemia/imunologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia
6.
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
7.
Exp Mol Med ; 52(7): 1004-1015, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32636442

RESUMO

Epigenetic regulation is mainly mediated by enzymes that can modify the structure of chromatin by altering the structure of DNA or histones. Proteins involved in epigenetic processes have been identified to study the detailed molecular mechanisms involved in the regulation of specific mRNA expression. Evolutionarily well-conserved polycomb group (PcG) proteins can function as transcriptional repressors by the trimethylation of histone H3 at the lysine 27 residue (H3K27me3) and the monoubiquitination of histone H2A at the lysine 119 residue (H2AK119ub). PcG proteins form two functionally distinct protein complexes: polycomb repressor complex 1 (PRC1) and PRC2. In mammals, the structural heterogeneity of each PRC complex is dramatically increased by several paralogs of its subunit proteins. Genetic studies with transgenic mice along with RNA-seq and chromatin immunoprecipitation (ChIP)-seq analyses might be helpful for defining the cell-specific functions of paralogs of PcG proteins. Here, we summarize current knowledge about the immune regulatory role of PcG proteins related to the compositional diversity of each PRC complex and introduce therapeutic drugs that target PcG proteins in hematopoietic malignancy.


Assuntos
Imunidade , Mamíferos/imunologia , Complexo Repressor Polycomb 1/química , Complexo Repressor Polycomb 1/metabolismo , Animais , Ensaios Clínicos como Assunto , Neoplasias Hematológicas/patologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Complexo Repressor Polycomb 1/antagonistas & inibidores
8.
Sci Rep ; 10(1): 12241, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32699244

RESUMO

Autophagy is a cellular homeostatic pathway with functions ranging from cytoplasmic protein turnover to immune defense. Therapeutic modulation of autophagy has been demonstrated to positively impact the outcome of autophagy-dysregulated diseases such as cancer or microbial infections. However, currently available agents lack specificity, and new candidates for drug development or potential cellular targets need to be identified. Here, we present an improved method to robustly detect changes in autophagy in a high-throughput manner on a single cell level, allowing effective screening. This method quantifies eGFP-LC3B positive vesicles to accurately monitor autophagy. We have significantly streamlined the protocol and optimized it for rapid quantification of large numbers of cells in little time, while retaining accuracy and sensitivity. Z scores up to 0.91 without a loss of sensitivity demonstrate the robustness and aptness of this approach. Three exemplary applications outline the value of our protocols and cell lines: (I) Examining autophagy modulating compounds on four different cell types. (II) Monitoring of autophagy upon infection with e.g. measles or influenza A virus. (III) CRISPR/Cas9 screening for autophagy modulating factors in T cells. In summary, we offer ready-to-use protocols to generate sensitive autophagy reporter cells and quantify autophagy in high-throughput assays.


Assuntos
Autofagia/imunologia , Ensaios de Triagem em Larga Escala/métodos , Mamíferos/imunologia , Animais , Sistemas CRISPR-Cas/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Infecções/imunologia , Células Jurkat , Linfócitos T/imunologia , Células THP-1
9.
Elife ; 92020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32255429

RESUMO

Multifunctional proteins are evolutionary puzzles: how do proteins evolve to satisfy multiple functional constraints? S100A9 is one such multifunctional protein. It potently amplifies inflammation via Toll-like receptor four and is antimicrobial as part of a heterocomplex with S100A8. These two functions are seemingly regulated by proteolysis: S100A9 is readily degraded, while S100A8/S100A9 is resistant. We take an evolutionary biochemical approach to show that S100A9 evolved both functions and lost proteolytic resistance from a weakly proinflammatory, proteolytically resistant amniote ancestor. We identify a historical substitution that has pleiotropic effects on S100A9 proinflammatory activity and proteolytic resistance but has little effect on S100A8/S100A9 antimicrobial activity. We thus propose that mammals evolved S100A8/S100A9 antimicrobial and S100A9 proinflammatory activities concomitantly with a proteolytic 'timer' to selectively regulate S100A9. This highlights how the same mutation can have pleiotropic effects on one functional state of a protein but not another, thus facilitating the evolution of multifunctionality.


A single protein sometimes does multiple jobs. For instance, our immune system uses a small number of multipurpose proteins to respond quickly to a large number of threats. One example is the protein S100A9. It acts as an antimicrobial by preventing microbes from getting the nutrients they need, while also stimulating inflammation by inducing the release of molecules that recruit white blood cells. S100A9, like all proteins, is made up of a chain of small building blocks. These building blocks interact with each other and with other molecules in the environment. The sequence of the building blocks thus determines what jobs the protein can do. Therefore, a single change to the sequence of building blocks can have a dramatic effect: one change might render the protein faulty, while another change might allow it to do a new job. Proteins face similar challenges humans do when trying to do several things at once. A person driving a car while using their phone will not do either task well. Likewise, a protein that does two jobs faces challenges a single-purpose protein does not. Harman et al. were interested in how S100A9 was able to evolve and maintain its dual functionality, despite this potential problem. They started by asking when S100A9 acquired its two purposes. They measured the antimicrobial and inflammatory activity of S100A9 proteins from humans, mice and opossums. The activities of S100A9 in these species was similar, suggesting that S100A9 acquired its different jobs in the ancestor of mammals, some 160 million years ago. Next, Harman et al. computationally reconstructed ancestral forms of S100A9 by comparing hundreds of similar proteins and building an evolutionary tree. They then measured the antimicrobial and inflammatory activity of these ancestral proteins. By comparing the last ancestor that did not have these activities to the first ancestor that did, they identified the sequence changes that gave S100A9 its dual activity. Importantly, these changes are located in separate regions of the protein, meaning they could occur independently, without affecting each other. Further, the same sequence change that converted S100A9 into an inflammatory signal also introduced a mechanism to regulate this activity. The results suggest that a small number of sequence changes ­ or even a single change ­ can make a protein more versatile. This means that evolving multipurpose proteins may not be as difficult as is often thought.


Assuntos
Calgranulina B/genética , Calgranulina B/imunologia , Evolução Molecular , Imunidade Inata/genética , Animais , Calgranulina A/genética , Calgranulina A/imunologia , Humanos , Inflamação , Mamíferos/imunologia , Proteólise , Receptor 4 Toll-Like/imunologia
10.
Rev. bras. parasitol. vet ; 29(4): e004720, 2020. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1138143

RESUMO

Abstract The objective of this study was to investigate the frequency of antibodies to Toxoplasma gondii present in wild mammals that were trap captured in forest fragments in the State of Bahia, northeastern Brazil. A total of 368 individuals (246 rodents, 104 marsupials and 18 bats) were captured using live catch traps. Serum samples were tested using the modified agglutination test, with a cut-off point at 1:25 dilution. The total occurrence of antibodies to T. gondii was 10.6% (39/368), being 16.3% (17/104) in marsupials, 8.5% (21/246) in rodents, and 5.5% (1/18) in bats. Antibody titers varied between 25 and 50 for rodents, between 25 and 400 for marsupials, and were 25 for bats. This is the first report on antibodies to T. gondii in certain rodent species (Thaptomys nigrita, Hylaeamys laticeps, and Cerradomys subflavus), marsupial species (Monodelphis americana, Gracilinanus microtarsus, Gracilinanus agilis and Marmosops incanus), and bats of the genus Rhynchonycteris. The presence of antibodies to T. gondii in wild mammals demonstrates the possibility of these animals as sentinels of toxoplasmosis, especially on regions under high anthropogenic effect.


Resumo O objetivo deste trabalho foi investigar a frequência de anticorpos anti-Toxoplasma gondii presentes em mamíferos selvagens, capturados em fragmentos florestais do Estado da Bahia, Nordeste do Brasil. Um total de 368 indivíduos (246 roedores, 104 marsupiais e 18 morcegos) foram capturados, usando-se armadilhas de captura viva. Os soros foram testados pelo teste de aglutinação modificada, com ponto de corte na diluição de 1:25. A ocorrência total de anticorpos anti-T. gondii foi de 10,6% (39/368), sendo 16,3% (17/104) em marsupiais, 8,5% (21/246) em roedores e 5,5% (1/18) em morcegos. Os títulos variaram de 25 a 50 e 25 a 400, respectivamente, para roedores e marsupiais, e o título máximo em morcegos foi de 25. Este é o primeiro relato de anticorpos para T. gondii em algumas espécies de roedores (Thaptomys nigrita, Hylaeamys laticeps e Cerradomys subflavus), em marsupiais (Monodelphis americana, Gracilinanus microtarsus, Gracilinanus agilis e Marmosops incanus) e em quiróptero do gênero Rhynchonycteris. A presença de anticorpos anti-T. gondii em mamíferos selvagens demonstra a possibilidade desses animais como sentinelas da toxoplasmose, principalmente em regiões com alto efeito antropogênico.


Assuntos
Animais , Anticorpos Antiprotozoários/sangue , Toxoplasmose Animal/diagnóstico , Toxoplasmose Animal/epidemiologia , Mamíferos/imunologia , Toxoplasma , Brasil/epidemiologia , Estudos Soroepidemiológicos , Florestas , Inquéritos e Questionários , Mamíferos/parasitologia
11.
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
12.
Front Immunol ; 10: 1220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191557

RESUMO

The term "acute phase response" (APR) is referred to a nonspecific and complex reaction of an organism that occurs shortly after any tissue damage, such as infection, trauma, neoplasia, inflammation, and stress. The APR can be identified and monitored with some laboratory tests, such as the concentration of several plasma proteins, the acute phase proteins (APPs). The APPs are components of the non-specific innate immune response, and their plasma concentration is proportional to the severity and/or the extent of tissue damage. The evaluation of health status of marine mammals is difficult because the classical clinical signs of illness used for human and domestic animals are difficult to recognize and understand. For this reason, in the past years, several efforts were done to identify laboratory markers of disease in these animals. The APPs have demonstrated their role as early markers of inflammation in veterinary medicine, thus several APPs were tested in marine mammals, such as C-reactive protein (CRP), serum amyloid-A (SAA), and Haptoglobin (Hp). However, the difficulty to extrapolate the knowledge about APPs in one species to another, the lack of specie-specific reagents, the absence of data about negative APPs have hampered their extent use in marine mammals. Herein, the state of art of APPs in marine mammals is reviewed, with particular attention to pre-analytical and analytical factors that should be taken into account in validation and interpretation of APPs assays. Moreover, the current application, potential utility and the future developments of APPs in marine mammals is highlighted and discussed.


Assuntos
Proteínas de Fase Aguda/imunologia , Proteínas de Fase Aguda/metabolismo , Organismos Aquáticos/imunologia , Organismos Aquáticos/metabolismo , Mamíferos/imunologia , Mamíferos/metabolismo , Proteínas de Fase Aguda/genética , Reação de Fase Aguda/genética , Reação de Fase Aguda/imunologia , Reação de Fase Aguda/metabolismo , Animais , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade Inata
13.
Cells ; 8(5)2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31052239

RESUMO

The Hippo pathway was originally identified as an evolutionarily-conserved signaling mechanism that contributes to the control of organ size. It was then rapidly expanded as a key pathway in the regulation of tissue development, regeneration, and cancer pathogenesis. The increasing amount of evidence in recent years has also connected this pathway to the regulation of innate and adaptive immune responses. Notably, the Hippo pathway has been revealed to play a pivotal role in adaptive immune cell lineages, as represented by the patients with T- and B-cell lymphopenia exhibiting defective expressions of the pathway component. The complex regulatory mechanisms of and by the Hippo pathway have also been evident as alternative signal transductions are employed in some immune cell types. In this review article, we summarize the current understanding of the emerging roles of the Hippo pathway in adaptive immune cell development and differentiation. We also highlight the recent findings concerning the dual functions of the Hippo pathway in autoimmunity and anti-cancer immune responses and discuss the key open questions in the interplay between the Hippo pathway and the mammalian immune system.


Assuntos
Imunidade Adaptativa/imunologia , Autoimunidade/imunologia , Linfopenia/imunologia , Mamíferos/imunologia , Neoplasias/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Transdução de Sinais , Animais , Diferenciação Celular/imunologia , Drosophila/metabolismo , Humanos , Linfócitos T/citologia , Linfócitos T/imunologia
14.
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
15.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651366

RESUMO

The development of a prophylactic vaccine for hepatitis C virus (HCV) remains a global health challenge. Cumulative evidence supports the importance of antibodies targeting the HCV E2 envelope glycoprotein to facilitate viral clearance. However, a significant challenge for a B cell-based vaccine is focusing the immune response on conserved E2 epitopes capable of eliciting neutralizing antibodies not associated with viral escape. We hypothesized that glycosylation might influence the antigenicity and immunogenicity of E2. Accordingly, we performed head-to-head molecular, antigenic, and immunogenic comparisons of soluble E2 (sE2) produced in (i) mammalian (HEK293) cells, which confer mostly complex- and high-mannose-type glycans; and (ii) insect (Sf9) cells, which impart mainly paucimannose-type glycans. Mass spectrometry demonstrated that all 11 predicted N-glycosylation sites were utilized in both HEK293- and Sf9-derived sE2, but that N-glycans in insect sE2 were on average smaller and less complex. Both proteins bound CD81 and were recognized by conformation-dependent antibodies. Mouse immunogenicity studies revealed that similar polyclonal antibody responses were generated against antigenic domains A to E of E2. Although neutralizing antibody titers showed that Sf9-derived sE2 induced moderately stronger responses than did HEK293-derived sE2 against the homologous HCV H77c isolate, the two proteins elicited comparable neutralization titers against heterologous isolates. Given that global alteration of HCV E2 glycosylation by expression in different hosts did not appreciably affect antigenicity or overall immunogenicity, a more productive approach to increasing the antibody response to neutralizing epitopes may be complete deletion, rather than just modification, of specific N-glycans proximal to these epitopes.IMPORTANCE The development of a vaccine for hepatitis C virus (HCV) remains a global health challenge. A major challenge for vaccine development is focusing the immune response on conserved regions of the HCV envelope protein, E2, capable of eliciting neutralizing antibodies. Modification of E2 by glycosylation might influence the immunogenicity of E2. Accordingly, we performed molecular and immunogenic comparisons of E2 produced in mammalian and insect cells. Mass spectrometry demonstrated that the predicted glycosylation sites were utilized in both mammalian and insect cell E2, although the glycan types in insect cell E2 were smaller and less complex. Mouse immunogenicity studies revealed similar polyclonal antibody responses. However, insect cell E2 induced stronger neutralizing antibody responses against the homologous isolate used in the vaccine, albeit the two proteins elicited comparable neutralization titers against heterologous isolates. A more productive approach for vaccine development may be complete deletion of specific glycans in the E2 protein.


Assuntos
Formação de Anticorpos/imunologia , Hepacivirus/imunologia , Insetos/imunologia , Mamíferos/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Linhagem Celular , Epitopos/imunologia , Feminino , Glicosilação , Células HEK293 , Hepatite C/imunologia , Hepatite C/virologia , Anticorpos Anti-Hepatite C/imunologia , Humanos , Insetos/virologia , Mamíferos/virologia , Camundongos , Polissacarídeos/imunologia , Células Sf9
16.
BMC Biol ; 16(1): 115, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30322384

RESUMO

The ability of certain tumor cells of mammals and molluscs to spread from the original host to others reopens the question of distinguishing self from non-self. It is part of a wider phenomenon of cellular parasitism and cell chimerism including germ cells.


Assuntos
Mamíferos/imunologia , Moluscos/imunologia , Células Neoplásicas Circulantes/imunologia , Animais , Neoplasias
17.
Virulence ; 9(1): 1625-1639, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30257608

RESUMO

The insect immune response demonstrates many similarities to the innate immune response of mammals and a wide range of insects is now employed to assess the virulence of pathogens and produce results comparable to those obtained using mammals. Many of the humoral responses in insects and mammals are similar (e.g. insect transglutaminases and human clotting factor XIIIa) however a number show distinct differences. For example in mammals, melanization plays a role in protection from solar radiation and in skin and hair pigmentation. In contrast, insect melanization acts as a defence mechanism in which the proPO system is activated upon pathogen invasion. Human and insect antimicrobial peptides share distinct structural and functional similarities, insects produce the majority of their AMPs from the fat body while mammals rely on production locally at the site of infection by epithelial/mucosal cells. Understanding the structure and function of the insect immune system and the similarities with the innate immune response of mammals will increase the attractiveness of using insects as in vivo models for studying host - pathogen interactions.


Assuntos
Imunidade Humoral , Imunidade Inata , Insetos/imunologia , Mamíferos/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/imunologia , Doenças Transmissíveis/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Corpo Adiposo/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Mucosa/imunologia , Virulência
18.
Vet Immunol Immunopathol ; 202: 63-69, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30078600

RESUMO

Polymeric immunoglobulin receptor (pIgR) functions in transporting polymeric immunoglobulin across epithelial cells into external secretion in animals. During animal evolution, fish was situated at a transition point on the phylogenetic spectrum between species possessing only innate immunity (i.e., invertebrates) and species depending heavily on adaptive immunity (i.e., mammals). Previous studies reported that fish and mammals significantly differ in pIgR. This review summarized the differences in pIgR structure, function, and transcriptional regulation between fish and mammals. A model of the transcriptional regulation of the pIgR gene was suggested. In this model, microbes could activate Toll-like receptor, trigger the cascade reactions in the signaling pathway, and then activate transcription factors that regulate pIgR expression through combining with the pIgR promoter. This review provides some suggestions for further studies on the function and regulatory mechanism of pIgR in fish and other animals.


Assuntos
Peixes/imunologia , Mamíferos/imunologia , Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/imunologia , Imunidade Adaptativa , Animais , Células Epiteliais/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Filogenia , Regiões Promotoras Genéticas , Transdução de Sinais , Receptores Toll-Like/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
19.
Front Immunol ; 9: 1880, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30150995

RESUMO

Tumor necrosis factor ligand superfamily members such as B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) have been identified in mammals as key regulators of B cell homeostasis and activation. However, the immune functions of APRIL are not as well defined as those of BAFF. Furthermore, while BAFF is present in all vertebrates, APRIL is missing in some animal groups, suggesting that BAFF has compensated the functions of APRIL in these species. In this context, we thought of great interest to explore the effects of APRIL on teleost B cells, given that APRIL appears for the first time in evolution in bony fish. Thus, in this study, we have performed an extensive analysis of the effect of APRIL on B cells using rainbow trout (Oncorhynchus mykiss) as a model species. Our results demonstrate that APRIL induces a specific proliferation of IgM+ B cells by itself and increases IgM secretion without promoting a terminal differentiation to plasma cells. APRIL also increased the levels of surface MHC II and augmented the capacity of these cells to process antigen, effects that were exclusively exerted on IgM+ B cells. Although our results point to a highly conserved role of APRIL on B cell homeostasis and activation throughout evolution, some specific differential effects have been observed in fish in comparison to the effects of APRIL previously described in mammals. Finally, the effects that APRIL induces on rainbow trout IgM+ B cells described in this paper have been compared with those previously reported in response to BAFF.


Assuntos
Linfócitos B/imunologia , Proteínas de Peixes/metabolismo , Mamíferos/imunologia , Oncorhynchus mykiss/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Fator Ativador de Células B/metabolismo , Evolução Biológica , Diferenciação Celular , Proliferação de Células , Autorrenovação Celular , Proteínas de Peixes/genética , Homeostase , Imunoglobulina M/metabolismo , Ativação Linfocitária , Filogenia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
20.
J Immunol ; 201(2): 465-480, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29866701

RESUMO

Despite teleost fish being the first animal group in which all elements of adaptive immunity are present, the lack of follicular structures, as well as the fact that systemic Ab responses rely exclusively on unswitched low-affinity IgM responses, strongly suggests that fish B cell responses resemble mammalian B1 cell responses rather than those of B2 cells. In line with this hypothesis, in the current study, we have identified a homolog of CD5 in teleost fish. This pan-T marker belonging to the scavenger receptor cysteine-rich family of receptors is commonly used in mammals to distinguish a subset of B1 cells. Subsequently, we have demonstrated that a very high percentage of teleost IgM+ B cells express this marker, in contrast to the limited population of CD5-expressing B1 cells found in most mammals. Furthermore, we demonstrate that fish IgM+ B cells share classical phenotypic features of mammalian B1 cells such as large size, high complexity, high surface IgM, and low surface IgD expression, regardless of CD5 expression. Additionally, fish IgM+ B cells, unlike murine B2 cells, also displayed extended survival in cell culture and did not proliferate after BCR engagement. Altogether, our results demonstrate that although fish are evolutionarily the first group in which all the elements of acquired immunity are present, in the absence of follicular structures, most teleost IgM+ B cells have retained phenotypical and functional characteristics of mammalian B1 cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Antígenos CD5/imunologia , Peixes/imunologia , Imunoglobulina M/imunologia , Mamíferos/imunologia , Imunidade Adaptativa/imunologia , Animais , Subpopulações de Linfócitos B/metabolismo , Biomarcadores/metabolismo , Feminino , Peixes/metabolismo , Imunoglobulina D/imunologia , Imunoglobulina D/metabolismo , Imunoglobulina M/metabolismo , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA