Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Evol Biol ; 36(6): 847-873, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37255207

RESUMEN

Driven by co-evolution with pathogens, host immunity continuously adapts to optimize defence against pathogens within a given environment. Recent advances in genetics, genomics and transcriptomics have enabled a more detailed investigation into how immunogenetic variation shapes the diversity of immune responses seen across domestic and wild animal species. However, a deeper understanding of the diverse molecular mechanisms that shape immunity within and among species is still needed to gain insight into-and generate evolutionary hypotheses on-the ultimate drivers of immunological differences. Here, we discuss current advances in our understanding of molecular evolution underpinning jawed vertebrate immunity. First, we introduce the immunome concept, a framework for characterizing genes involved in immune defence from a comparative perspective, then we outline how immune genes of interest can be identified. Second, we focus on how different selection modes are observed acting across groups of immune genes and propose hypotheses to explain these differences. We then provide an overview of the approaches used so far to study the evolutionary heterogeneity of immune genes on macro and microevolutionary scales. Finally, we discuss some of the current evidence as to how specific pathogens affect the evolution of different groups of immune genes. This review results from the collective discussion on the current key challenges in evolutionary immunology conducted at the ESEB 2021 Online Satellite Symposium: Molecular evolution of the vertebrate immune system, from the lab to natural populations.


Asunto(s)
Inmunidad Adaptativa , Evolución Biológica , Animales , Inmunidad Adaptativa/genética , Vertebrados/genética , Evolución Molecular , Inmunidad Innata/genética
2.
Immunol Invest ; 52(6): 661-680, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37267050

RESUMEN

The wild Tasmanian devil (Sarcophilus harrisii) population has suffered a devastating decline due to two clonal transmissible cancers. The first devil facial tumor 1 (DFT1) was observed in 1996, followed by a second genetically distinct transmissible tumor, the devil facial tumor 2 (DFT2), in 2014. DFT1/2 frequently metastasize, with lymph nodes being common metastatic sites. MHC-I downregulation by DFT1 cells is a primary means of evading allograft immunity aimed at polymorphic MHC-I proteins. DFT2 cells constitutively express MHC-I, and MHC-I is upregulated on DFT1/2 cells by interferon gamma, suggesting other immune evasion mechanisms may contribute to overcoming allograft and anti-tumor immunity. Human clinical trials have demonstrated PD1/PDL1 blockade effectively treats patients showing increased expression of PD1 in tumor draining lymph nodes, and PDL1 on peritumoral immune cells and tumor cells. The effects of DFT1/2 on systemic immunity remain largely uncharacterized. This study applied the open-access software QuPath to develop a semiautomated pipeline for whole slide analysis of stained tissue sections to quantify PD1/PDL1 expression in devil lymph nodes. The QuPath protocol provided strong correlations to manual counting. PD-1 expression was approximately 10-fold higher than PD-L1 expression in lymph nodes and was primarily expressed in germinal centers, whereas PD-L1 expression was more widely distributed throughout the lymph nodes. The density of PD1 positive cells was increased in lymph nodes containing DFT2 metastases, compared to DFT1. This suggests PD1/PDL1 exploitation may contribute to the poorly immunogenic nature of transmissible tumors in some devils and could be targeted in therapeutic or prophylactic treatments.Abbreviations: PD1: programmed cell death protein 1; PDL1: programmed death ligand 1; DFT1: devil facial tumor 1; DFT2: devil facial tumor 2; DFTD: devil facial tumor disease; MCC: Matthew's correlation coefficient; DAB: diaminobenzidine; ROI: region of interest.


Asunto(s)
Antígeno B7-H1 , Neoplasias Faciales , Humanos , Antígeno B7-H1/genética , Receptor de Muerte Celular Programada 1/genética , Ganglios Linfáticos/patología , Microambiente Tumoral
3.
Curr Oncol Rep ; 23(9): 103, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34269916

RESUMEN

PURPOSE OF REVIEW: Immune checkpoint immunotherapies (ICI) are now approved for over 20 types of cancer and there are almost 6000 ongoing clinical trials investigating immuno-modulators as cancer therapies. This review investigated the effect of monoclonal antibody-based immune checkpoint immunotherapies when combined with cytokine therapy. We reviewed published clinical trial results from 2005 to 2020 for studies that used approved monoclonal antibody ICI in combination with the cytokines. Studies that met the search criteria were assessed for treatment efficacy and immunological changes associated with treatment. RECENT FINDING: ICI often fails to result in improved clinical outcomes for patients and lasting protection from cancer recurrence. The use of pro-inflammatory cytokines alongside ICI has been shown to enhance the efficacy of these therapies in vitro and in animal studies. However, the results in human clinical trials are less clear and many clinical trials do not publish results at the end of the trial. A deeper understanding of the molecular interactions between cytokines, tumors, and immune cells is needed to improve overall ICI outcomes and design combination trials. Critical examination of the design and characteristics of previous clinical trials can provide insight into the lack of effective clinical translation for many immunotherapeutic drugs.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citocinas/uso terapéutico , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Terapia Combinada , Humanos , Interleucina-2/uso terapéutico , Ipilimumab/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
4.
Cell Mol Life Sci ; 77(13): 2507-2525, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31900624

RESUMEN

The Tasmanian devil (Sarcophilus harrisii) is the only mammalian species known to be affected by multiple transmissible cancers. Devil facial tumours 1 and 2 (DFT1 and DFT2) are independent neoplastic cell lineages that produce large, disfiguring cancers known as devil facial tumour disease (DFTD). The long-term persistence of wild Tasmanian devils is threatened due to the ability of DFTD cells to propagate as contagious allografts and the high mortality rate of DFTD. Recent studies have demonstrated that both DFT1 and DFT2 cancers originated from founder cells of the Schwann cell lineage, an uncommon origin of malignant cancer in humans. This unprecedented finding has revealed a potential predisposition of Tasmanian devils to transmissible cancers of the Schwann cell lineage. In this review, we compare the molecular nature of human Schwann cells and nerve sheath tumours with DFT1 and DFT2 to gain insights into the emergence of transmissible cancers in the Tasmanian devil. We discuss a potential mechanism, whereby Schwann cell plasticity and frequent wounding in Tasmanian devils combine with an inherent cancer predisposition and low genetic diversity to give rise to transmissible Schwann cell cancers in devils on rare occasions.


Asunto(s)
Neoplasias Faciales/veterinaria , Marsupiales , Animales , Neoplasias Faciales/genética , Neoplasias Faciales/patología , Humanos , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/veterinaria , Células de Schwann/fisiología
5.
Cell Mol Life Sci ; 77(9): 1847-1858, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31375869

RESUMEN

Devil facial tumour disease (DFTD) comprises two genetically distinct transmissible cancers (DFT1 and DFT2) endangering the survival of the Tasmanian devil (Sarcophilus harrisii) in the wild. DFT1 first arose from a cell of the Schwann cell lineage; however, the tissue-of-origin of the recently discovered DFT2 cancer is unknown. In this study, we compared the transcriptome and proteome of DFT2 tumours to DFT1 and normal Tasmanian devil tissues to determine the tissue-of-origin of the DFT2 cancer. Our findings demonstrate that DFT2 expresses a range of Schwann cell markers and exhibits expression patterns consistent with a similar origin to the DFT1 cancer. Furthermore, DFT2 cells express genes associated with the repair response to peripheral nerve damage. These findings suggest that devils may be predisposed to transmissible cancers of Schwann cell origin. The combined effect of factors such as frequent nerve damage from biting, Schwann cell plasticity and low genetic diversity may allow these cancers to develop on rare occasions. The emergence of two independent transmissible cancers from the same tissue in the Tasmanian devil presents an unprecedented opportunity to gain insight into cancer development, evolution and immune evasion in mammalian species.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Faciales/veterinaria , Marsupiales/fisiología , Proteoma/análisis , Células de Schwann/patología , Transcriptoma , Animales , Biomarcadores de Tumor/genética , Neoplasias Faciales/genética , Neoplasias Faciales/metabolismo , Neoplasias Faciales/patología , Humanos , Células de Schwann/metabolismo
6.
mBio ; 14(4): e0071523, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37439571

RESUMEN

Wildlife is the source of many emerging infectious diseases. Several viruses from the order Nidovirales have recently emerged in wildlife, sometimes with severe consequences for endangered species. The order Nidovirales is currently classified into eight suborders, three of which contain viruses of vertebrates. Vertebrate coronaviruses (suborder Cornidovirineae) have been extensively studied, yet the other major suborders have received less attention. The aim of this minireview was to summarize the key findings from the published literature on nidoviruses of vertebrate wildlife from two suborders: Arnidovirineae and Tornidovirineae. These viruses were identified either during investigations of disease outbreaks or through molecular surveys of wildlife viromes, and include pathogens of reptiles and mammals. The available data on key biological features, disease associations, and pathology are presented, in addition to data on the frequency of infections among various host populations, and putative routes of transmission. While nidoviruses discussed here appear to have a restricted in vivo host range, little is known about their natural life cycle. Observational field-based studies outside of the mortality events are needed to facilitate an understanding of the virus-host-environment interactions that lead to the outbreaks. Laboratory-based studies are needed to understand the pathogenesis of diseases caused by novel nidoviruses and their evolutionary histories. Barriers preventing research progress include limited funding and the unavailability of virus- and host-specific reagents. To reduce mortalities in wildlife and further population declines, proactive development of expertise, technologies, and networks should be developed. These steps would enable effective management of future outbreaks and support wildlife conservation.

7.
Lett Biomath ; 9(1): 121-140, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-37655234

RESUMEN

Tasmanian Devils facial tumor disease (DFTD) is severely impacting the population of this wild animal. We developed a computational model of the population of Tasmanian Devils, and the change induced by DFTD. We use this model to test possible intervention strategies Tasmanian conservationists could do. We investigate bait drop vaccination programs, diseased animal removals programs, and evolution of natural immunity. We conclude that a combination of intervention strategies gives the most favorable outcome. An additional goal of this paper is reproducibility of our results. Our StochSS software platform features the ability to share and reproduce the computational notebooks that created all of the results in the paper. We endeavor that all readers should be able to reproduce our results with minimum effort.

8.
Open Biol ; 12(10): 220208, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36259237

RESUMEN

MHC-I and MHC-II molecules are critical components of antigen presentation and T cell immunity to pathogens and cancer. The two monoclonal transmissible devil facial tumours (DFT1, DFT2) exploit MHC-I pathways to overcome immunological anti-tumour and allogeneic barriers. This exploitation underpins the ongoing transmission of DFT cells across the wild Tasmanian devil population. We have previously shown that the overexpression of NLRC5 in DFT1 and DFT2 cells can regulate components of the MHC-I pathway but not MHC-II, establishing the stable upregulation of MHC-I on the cell surface. As MHC-II molecules are crucial for CD4+ T cell activation, MHC-II expression in tumour cells is beginning to gain traction in the field of immunotherapy and cancer vaccines. The overexpression of Class II transactivator in transfected DFT1 and DFT2 cells induced the transcription of several genes of the MHC-I and MHC-II pathways. This was further supported by the upregulation of MHC-I protein on DFT1 and DFT2 cells, but interestingly MHC-II protein was upregulated only in DFT1 cells. This new insight into the regulation of MHC-I and MHC-II pathways in cells that naturally overcome allogeneic barriers can inform vaccine, immunotherapy and tissue transplant strategies for human and veterinary medicine.


Asunto(s)
Neoplasias Faciales , Marsupiales , Animales , Neoplasias Faciales/genética , Neoplasias Faciales/veterinaria , Neoplasias Faciales/patología , Antígenos de Histocompatibilidad Clase II , Péptidos y Proteínas de Señalización Intracelular , Marsupiales/genética
9.
Blood ; 113(8): 1759-67, 2009 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-19109567

RESUMEN

B7-H4 is an immunoglobulin superfamily molecule and shown to be inhibitory for T-cell responses. To explore physiologic roles of B7-H4, we created B7-H4-deficient (KO) mice by genetic targeting. B7-H4KO mice are healthy and their T- and B-cell responses to polyclonal antigens are in normal range. However, B7-H4KO mice are more resistant to infection by Listeria monocytogenes than their littermates. Within 3 days after infection, bacterial colonies in livers and spleens are significantly lower than the controls, suggesting a role of B7-H4 in enhancing innate immunity. Further studies demonstrate that neutrophils increase in peripheral organs of B7-H4KO mice more so than their littermates but their bactericidal functions remain unchanged. Augmented innate resistance is completely dependent on neutrophils, even in the absence of adaptive immunity. In vitro B7-H4 inhibits the growth of bone marrow-derived neutrophil progenitors, suggesting an inhibitory function of B7-H4 in neutrophil expansion. Our results identify B7-H4 as a negative regulator of the neutrophil response to infection and provide a new target for manipulation of innate immunity.


Asunto(s)
Antígeno B7-1/genética , Antígeno B7-1/inmunología , Listeriosis/inmunología , Neutrófilos/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/microbiología , Células de la Médula Ósea/citología , Antígeno CD11b/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Neutrófilos/microbiología , Fagocitosis/inmunología , Receptores de Quimiocina/metabolismo , Estallido Respiratorio/inmunología , Linfocitos T/inmunología , Linfocitos T/microbiología , Inhibidor 1 de la Activación de Células T con Dominio V-Set
10.
J Cancer Res Clin Oncol ; 147(7): 1973-1991, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33797607

RESUMEN

PURPOSE: Downregulation of MHC class I (MHC-I) is a common immune evasion strategy of many cancers. Similarly, two allogeneic clonal transmissible cancers have killed thousands of wild Tasmanian devils (Sarcophilus harrisii) and also modulate MHC-I expression to evade anti-cancer and allograft responses. IFNG treatment restores MHC-I expression on devil facial tumor (DFT) cells but is insufficient to control tumor growth. Transcriptional co-activator NLRC5 is a master regulator of MHC-I in humans and mice but its role in transmissible cancers remains unknown. In this study, we explored the regulation and role of MHC-I in these unique genetically mis-matched tumors. METHODS: We used transcriptome and flow cytometric analyses to determine how MHC-I shapes allogeneic and anti-tumor responses. Cell lines that overexpress NLRC5 to drive antigen presentation, and B2M-knockout cell lines incapable of presenting antigen on MHC-I were used to probe the role of MHC-I in rare cases of tumor regressions. RESULTS: Transcriptomic results suggest that NLRC5 plays a major role in MHC-I regulation in devils. NLRC5 was shown to drive the expression of many components of the antigen presentation pathway but did not upregulate PDL1. Serum from devils with tumor regressions showed strong binding to IFNG-treated and NLRC5 cell lines; antibody binding to IFNG-treated and NRLC5 transgenic tumor cells was diminished or absent following B2M knockout. CONCLUSION: MHC-I could be identified as a target for anti-tumor and allogeneic immunity. Consequently, NLRC5 could be a promising target for immunotherapy and vaccines to protect devils from transmissible cancers and inform development of transplant and cancer therapies for humans.


Asunto(s)
Presentación de Antígeno/inmunología , Biomarcadores de Tumor/metabolismo , Neoplasias Faciales/inmunología , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias Faciales/genética , Neoplasias Faciales/metabolismo , Neoplasias Faciales/patología , Antígenos de Histocompatibilidad Clase I/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Marsupiales , Transcriptoma , Células Tumorales Cultivadas
11.
Dev Comp Immunol ; 115: 103882, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33039410

RESUMEN

Immune checkpoint immunotherapy is a pillar of human oncology treatment with potential for non-human species. The first checkpoint immunotherapy approved for human cancers targeted the CTLA4 protein. CTLA4 can inhibit T cell activation by capturing and internalizing CD80 and CD86 from antigen presenting cells, a process called trans-endocytosis. Similarly, CD28 can capture CD80 and CD86 via trogocytosis and retain the captured ligands on the surface of the CD28-expressing cells. The wild Tasmanian devil (Sarcophilus harrisii) population has declined by 77% due to transmissible cancers that evade immune defenses despite genetic mismatches between the host and tumors. We used a live cell-based assay to demonstrate that devil CTLA4 and CD28 can capture CD80 and CD86. Mutation of evolutionarily conserved motifs in CTLA4 altered functional interactions with CD80 and CD86 in accordance with patterns observed in other species. These results suggest that checkpoint immunotherapies can be translated to evolutionarily divergent species.


Asunto(s)
Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Antígeno CTLA-4/metabolismo , Marsupiales/inmunología , Secuencias de Aminoácidos/genética , Animales , Antígenos CD28/antagonistas & inhibidores , Células CHO , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/genética , Células Cultivadas , Clonación Molecular , Cricetulus , Especies en Peligro de Extinción , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Microscopía Intravital , Marsupiales/metabolismo , Mutación , Trogocitosis
12.
Bio Protoc ; 10(13): e3696, 2020 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-33659348

RESUMEN

This protocol provides a step-by-step method to create recombinant fluorescent fusion proteins that can be secreted from mammalian cell lines. This builds on many other recombinant protein and fluorescent protein techniques, but is among the first to harness fluorescent fusion proteins secreted directly into cell culture supernatant. This opens new possibilities that are not achievable with proteins produced in bacteria or yeast, such as direct use of the fluorescent protein-secreting cells in live co-culture assays. The Fluorescent Adaptable Simple Theranostic (FAST) protein system includes a histidine purification tag and a tobacco etch virus (TEV) cleavage site, allowing the purification tag and fluorescent protein to be removed for therapeutic use. This protocol is split into five parts: (A) In silico characterization of the gene-of-interest (GOI) and protein-of-interest (POI); (B) design of the expression vector; (C) assembly of the expression vector; (D) transfection of a eukaryotic cell line with the expression vector; (E) testing of the recombinant protein. This extensive protocol can be completed with only polymerase chain reaction (PCR) and cell culture training. Additionally, each part of the protocol can be used independently.

13.
Sci Adv ; 6(27)2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32937435

RESUMEN

Around 40% of humans and Tasmanian devils (Sarcophilus harrisii) develop cancer in their lifetime, compared to less than 10% for most species. In addition, devils are affected by two of the three known transmissible cancers in mammals. Immune checkpoint immunotherapy has transformed human medicine, but a lack of species-specific reagents has limited checkpoint immunology in most species. We developed a cut-and-paste reagent development system and used the fluorescent fusion protein system to show that immune checkpoint interactions are conserved across 160,000,000 years of evolution, CD200 is highly expressed on transmissible tumor cells, and coexpression of CD200R1 can block CD200 surface expression. The system's versatility across species was demonstrated by fusing a fluorescent reporter to a camelid-derived nanobody that binds human programmed death ligand 1. The evolutionarily conserved pathways suggest that naturally occurring cancers in devils and other species can be used to advance our understanding of cancer and immunological tolerance.


Asunto(s)
Neoplasias Faciales , Marsupiales , Animales , Neoplasias Faciales/metabolismo , Neoplasias Faciales/patología , Evasión Inmune , Inmunoterapia
14.
Expert Rev Vaccines ; 19(1): 1-10, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31971036

RESUMEN

Introduction: The Tasmanian devil (Sarcophilus harrisii) is the largest extant carnivorous marsupial. Since 1996, its population has declined by 77% primarily due to a clonal transmissible tumor, known as devil facial tumor (DFT1) disease. In 2014, a second transmissible devil facial tumor (DFT2) was discovered. DFT1 and DFT2 are nearly 100% fatal.Areas covered: We review DFT control approaches and propose a rabies-style oral bait vaccine (OBV) platform for DFTs. This approach has an extensive safety record and was a primary tool in large-scale rabies virus elimination from wild carnivores across diverse landscapes. Like rabies virus, DFTs are transmitted by oral contact, so immunizing the oral cavity and stimulating resident memory cells could be advantageous. Additionally, exposing infected devils that already have tumors to OBVs could serve as an oncolytic virus immunotherapy. The primary challenges may be identifying appropriate DFT-specific antigens and optimization of field delivery methods.Expert opinion: DFT2 is currently found on a peninsula in southern Tasmania, so an OBV that could eliminate DFT2 should be the priority for this vaccine approach. Translation of an OBV approach to control DFTs will be challenging, but the approach is feasible for combatting ongoing and future disease threats.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Neoplasias Faciales/prevención & control , Vacunación/métodos , Administración Oral , Animales , Vacunas contra el Cáncer/inmunología , Neoplasias Faciales/inmunología , Neoplasias Faciales/veterinaria , Humanos , Inmunoterapia/métodos , Marsupiales/inmunología , Viroterapia Oncolítica/métodos , Tasmania , Vacunación/veterinaria
15.
J Clin Invest ; 116(4): 1045-51, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16557300

RESUMEN

LIGHT is an important costimulatory molecule for T cell immunity. Recent studies have further implicated its role in innate immunity and inflammatory diseases, but its cellular and molecular mechanisms remain elusive. We report here that LIGHT is upregulated and functions as a proinflammatory cytokine in 2 independent experimental hepatitis models, induced by concanavalin A and Listeria monocytogenes. Molecular mutagenesis studies suggest that soluble LIGHT protein produced by cleavage from the cell membrane plays an important role in this effect through the interaction with the lymphotoxin-beta receptor (LTbetaR) but not herpes virus entry mediator. NK1.1+ T cells contribute to the production, but not the cleavage or effector functions, of soluble LIGHT. Importantly, treatment with a mAb that specifically interferes with the LIGHT-LTbetaR interaction protects mice from lethal hepatitis. Our studies thus identify a what we believe to be a novel function of soluble LIGHT in vivo and offer a potential target for therapeutic interventions in hepatic inflammatory diseases.


Asunto(s)
Citocinas/metabolismo , Hepatitis/etiología , Proteínas de la Membrana/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígenos Ly , Antígenos de Superficie/metabolismo , Concanavalina A/metabolismo , Concanavalina A/farmacología , Hepatitis/metabolismo , Inflamación/metabolismo , Lectinas Tipo C/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Receptor beta de Linfotoxina , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Receptores Virales/inmunología , Receptores Virales/metabolismo , Solubilidad , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
16.
Trends Ecol Evol ; 34(4): 303-314, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30704782

RESUMEN

Individual hosts differ extensively in their competence for parasites, but traditional research has discounted this variation, partly because modeling such heterogeneity is difficult. This discounting has diminished as tools have improved and recognition has grown that some hosts, the extremely competent, can have exceptional impacts on disease dynamics. Most prominent among these hosts are the superspreaders, but other forms of extreme competence (EC) exist and others await discovery; each with potentially strong but distinct implications for disease emergence and spread. Here, we propose a framework for the study and discovery of EC, suitable for different host-parasite systems, which we hope enhances our understanding of how parasites circulate and evolve in host communities.


Asunto(s)
Parásitos , Animales , Interacciones Huésped-Parásitos
17.
Front Immunol ; 9: 3117, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30692995

RESUMEN

The Tasmanian devil facial tumor (DFT) disease has led to an 80% reduction in the wild Tasmanian devil (Sarcophilus harrisii) population since 1996. The limited genetic diversity of wild devils and the lack of MHC-I expression on DFT cells have been implicated in the lack of immunity against the original DFT clonal cell line (DFT1). Recently, a second transmissible tumor of independent origin (DFT2) was discovered. Surprisingly, DFT2 cells do express MHC-I, but DFT2 cells appear to be on a trajectory for reduced MHC-I expression in vivo. Thus, much of the ongoing vaccine-development efforts and conservation plans have focused on MHC-I. A major limitation in conservation efforts is the lack of species-specific tools to understand Tasmanian devil gene function and immunology. To help fill this gap, we developed an all-in-one Tet-Off vector system to regulate expression of IFN-γ in DFT cells (DFT1.Tet/IFN-γ). IFN-γ can have negative effects on cell proliferation and viability; thus, doxycycline was used to suppress IFN-γ production whilst DFT1.Tet/IFN-γ cells were expanded in cell culture. Induction of IFN-γ following removal of doxycycline led to upregulation of MHC-I but also the inhibitory checkpoint molecule PD-L1. Additionally, DFT1.Tet/IFN-γ cells were capable of stimulating MHC-I upregulation on bystander wild type DFT cells in co-culture assays in vitro. This system represents a major step forward in DFT disease immunotherapy and vaccine development efforts, and ability to understand gene function in devils. Importantly, the techniques are readily transferable for testing gene function in DFT2 cells and other non-traditional species.


Asunto(s)
Neoplasias Faciales/veterinaria , Antígenos de Histocompatibilidad Clase I/metabolismo , Interferón gamma/metabolismo , Marsupiales/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/inmunología , Línea Celular Tumoral/metabolismo , Clonación Molecular , Doxiciclina/administración & dosificación , Cara , Neoplasias Faciales/genética , Neoplasias Faciales/inmunología , Neoplasias Faciales/patología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Interferón gamma/genética , Interferón gamma/inmunología , Marsupiales/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Regulación hacia Arriba
18.
Integr Comp Biol ; 58(6): 1043-1054, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30252058

RESUMEN

The Tasmanian devil, a marsupial carnivore, has been restricted to the island state of Tasmania since its extinction on the Australian mainland about 3000 years ago. In the past two decades, this species has experienced severe population decline due to the emergence of devil facial tumor disease (DFTD), a transmissible cancer. During these 20 years, scientists have puzzled over the immunological and evolutionary responses by the Tasmanian devil to this transmissible cancer. Targeted strategies in population management and disease control have been developed as well as comparative processes to identify variation in tumor and host genetics. A multi-disciplinary approach with multi-institutional teams has produced considerable advances over the last decade. This has led to a greater understanding of the molecular pathogenesis and genomic classification of this cancer. New and promising developments in the Tasmanian devil's story include evidence that most immunized, and some wild devils, can produce an immune response to DFTD. Furthermore, epidemiology combined with genomic studies suggest a rapid evolution to the disease and that DFTD will become an endemic disease. Since 1998 there have been more than 350 publications, distributed over 37 Web of Science categories. A unique endemic island species has become an international curiosity that is in the spotlight of integrative and comparative biology research.


Asunto(s)
Transmisión de Enfermedad Infecciosa/veterinaria , Neoplasias Faciales/veterinaria , Animales , Transmisión de Enfermedad Infecciosa/prevención & control , Neoplasias Faciales/inmunología , Neoplasias Faciales/patología , Neoplasias Faciales/prevención & control , Marsupiales , Tasmania
19.
Front Immunol ; 8: 513, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28515726

RESUMEN

Immune checkpoint molecules function as a system of checks and balances that enhance or inhibit immune responses to infectious agents, foreign tissues, and cancerous cells. Immunotherapies that target immune checkpoint molecules, particularly the inhibitory molecules programmed cell death 1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), have revolutionized human oncology in recent years, yet little is known about these key immune signaling molecules in species other than primates and rodents. The Tasmanian devil facial tumor disease is caused by transmissible cancers that have resulted in a massive decline in the wild Tasmanian devil population. We have recently demonstrated that the inhibitory checkpoint molecule PD-L1 is upregulated on Tasmanian devil (Sarcophilus harrisii) facial tumor cells in response to the interferon-gamma cytokine. As this could play a role in immune evasion by tumor cells, we performed a thorough comparative analysis of checkpoint molecule protein sequences among Tasmanian devils and eight other species. We report that many of the key signaling motifs and ligand-binding sites in the checkpoint molecules are highly conserved across the estimated 162 million years of evolution since the last common ancestor of placental and non-placental mammals. Specifically, we discovered that the CTLA-4 (MYPPPY) ligand-binding motif and the CTLA-4 (GVYVKM) inhibitory domain are completely conserved across all nine species used in our comparative analysis, suggesting that the function of CTLA-4 is likely conserved in these species. We also found that cysteine residues for intra- and intermolecular disulfide bonds were also highly conserved. For instance, all 20 cysteine residues involved in disulfide bonds in the human 4-1BB molecule were also present in devil 4-1BB. Although many key sequences were conserved, we have also identified immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and immunoreceptor tyrosine-based switch motifs (ITSMs) in genes and protein domains that have not been previously reported in any species. This checkpoint molecule analysis and review of salient features for each of the molecules presented here can serve as road map for the development of a Tasmanian devil facial tumor disease immunotherapy. Finally, the strategies can be used as a guide for veterinarians, ecologists, and other researchers willing to venture into the nascent field of wild immunology.

20.
J Med Entomol ; 53(4): 902-910, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27113100

RESUMEN

Ross River virus (RRV) is responsible for the most notifications of human arboviral infection in Australia. Seroprevalence and experimental infection studies have implicated macropods (e.g., kangaroos) as the major reservoir hosts. However, transmission ecology varies spatially, and infections in urban areas have prompted the question of what animals serve as reservoirs in regions where macropods are scarce. In South Australia (SA), human infection rates for RRV vary greatly by region as do vector and reservoir abundance. We hypothesized that mosquito abundance and feeding patterns would vary among ecoregions of SA and could help explain divergent human case rates. To test our hypothesis, we amplified and sequenced a 457 base pair region of the cytochrome B segment of mitochondrial DNA from blood fed mosquitoes collected in three main ecoregions of SA and identified sequences using a BLAST search in NCBI. Domestic livestock made up the vast majority of bloodmeals from the region with the highest human infection rate. Livestock are generally not considered to be important reservoir hosts for RRV, but our results suggest they may have a role in transmission ecology in some places. Surprisingly, none of the 199 bloodmeal samples were identified as macropod in origin. In the context of these findings, we consider the possible RRV vectors and reservoir hosts in these regions and propose that diverse spatial and temporal transmission ecologies occur in SA, depending on vector and reservoir availability.


Asunto(s)
Infecciones por Alphavirus/transmisión , Culicidae/fisiología , Insectos Vectores/fisiología , Virus del Río Ross/aislamiento & purificación , Infecciones por Alphavirus/virología , Animales , Aves/fisiología , Aves/virología , Reservorios de Enfermedades , Conducta Alimentaria , Humanos , Mamíferos/fisiología , Mamíferos/virología , Densidad de Población , Australia del Sur
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA