Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunogenetics ; 73(3): 263-275, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33544183

RESUMEN

Advances in genome sequencing technology have enabled genomes of extinct species to be sequenced. However, given the fragmented nature of these genome assemblies, it is not clear whether it is possible to comprehensively annotate highly variable and repetitive genes such as those involved in immunity. As such, immune genes have only been investigated in a handful of extinct genomes, mainly in human lineages. In 2018 the genome of the thylacine (Thylacinus cynocephalus), a carnivorous marsupial from Tasmania that went extinct in 1936, was sequenced. Here we attempt to characterise the immune repertoire of the thylacine and determine similarity to its closest relative with a genome available, the Tasmanian devil (Sarcophilus harrisii), as well as other marsupials. Members from all major immune gene families were identified. However, variable regions could not be characterised, and complex families such as the major histocompatibility complex (MHC) were highly fragmented and located across multiple small scaffolds. As such, at a gene level we were unable to reconstruct full-length coding sequences for the majority of thylacine immune genes. Despite this, we identified genes encoding functionally important receptors and immune effector molecules, which suggests the functional capacity of the thylacine immune system was similar to other mammals. However, the high number of partial immune gene sequences identified limits our ability to reconstruct an accurate picture of the thylacine immune repertoire.


Asunto(s)
Citocinas/genética , Extinción Biológica , Inmunoglobulinas/genética , Complejo Mayor de Histocompatibilidad/genética , Marsupiales/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores Toll-Like/genética , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Genoma , Sistema Inmunológico/inmunología , Inmunoglobulinas/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Marsupiales/inmunología , Anotación de Secuencia Molecular , Receptores de Antígenos de Linfocitos T/inmunología , Homología de Secuencia , Receptores Toll-Like/inmunología
2.
Immunology ; 163(2): 169-184, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33460454

RESUMEN

Transmissible cancers are malignant cells that can spread between individuals of a population, akin to both a parasite and a mobile graft. The survival of the Tasmanian devil, the largest remaining marsupial carnivore, is threatened by the remarkable emergence of two independent lineages of transmissible cancer, devil facial tumour (DFT) 1 and devil facial tumour 2 (DFT2). To aid the development of a vaccine and to interrogate how histocompatibility barriers can be overcome, we analysed the peptides bound to major histocompatibility complex class I (MHC-I) molecules from Tasmanian devil cells and representative cell lines of each transmissible cancer. Here, we show that DFT1 + IFN-γ and DFT2 cell lines express a restricted repertoire of MHC-I allotypes compared with fibroblast cells, potentially reducing the breadth of peptide presentation. Comparison of the peptidomes from DFT1 + IFNγ, DFT2 and host fibroblast cells demonstrates a dominant motif, despite differences in MHC-I allotypes between the cell lines, with preference for a hydrophobic leucine residue at position 3 and position Ω of peptides. DFT1 and DFT2 both present peptides derived from neural proteins, which reflects a shared cellular origin that could be exploited for vaccine design. These results suggest that polymorphisms in MHC-I molecules between tumours and host can be 'hidden' by a common peptide motif, providing the potential for permissive passage of infectious cells and demonstrating complexity in mammalian histocompatibility barriers.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Vacunas contra el Cáncer/inmunología , Neoplasias Faciales/inmunología , Inmunoterapia/métodos , Marsupiales/inmunología , Células Neoplásicas Circulantes/patología , Péptidos/metabolismo , Secuencias de Aminoácidos/genética , Animales , Presentación de Antígeno , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Histocompatibilidad , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/genética , Polimorfismo Genético , Unión Proteica
3.
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
4.
Nature ; 586(7829): 424-428, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33029010

RESUMEN

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.


Asunto(s)
Mamíferos/virología , Filogenia , Virus de la Rubéola/clasificación , Virus de la Rubéola/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Animales de Zoológico/inmunología , Animales de Zoológico/virología , Membrana Celular/virología , Quirópteros/virología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Equidae/inmunología , Equidae/virología , Evolución Molecular , Femenino , Mapeo Geográfico , Alemania , Especificidad del Huésped , Humanos , Masculino , Mamíferos/inmunología , Marsupiales/inmunología , Marsupiales/virología , Fusión de Membrana , Ratones , Modelos Animales , Modelos Moleculares , Rubéola (Sarampión Alemán)/congénito , Rubéola (Sarampión Alemán)/virología , Virus de la Rubéola/química , Virus de la Rubéola/inmunología , Alineación de Secuencia , Uganda , Proteínas del Envoltorio Viral/química
5.
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
6.
Mol Immunol ; 117: 1-11, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31726269

RESUMEN

Marsupials and eutherians are mammals that differ in their physiological traits, predominately their reproductive and developmental strategies; eutherians give birth to well-developed young, while marsupials are born highly altricial after a much shorter gestation. These developmental traits also result in differences in the development of the immune system of eutherian and marsupial species. In eutherians, B-cells are the key to humoral immunity as they are found in multiple lymphoid organs and have the unique ability to mediate the production of antigen-specific antibodies in the presence of extracellular pathogens. The development of B-cells in marsupials has been reported and hypothesised to be similar to that of eutherians, except that haematopoiesis occurs in the liver, postpartum, until the bone marrow fully matures. In eutherians, specific genes are linked to specific stages in B-cell development, maturation, and differentiation processes, and have been identified including immunoglobulins (heavy and light chains), cluster of differentiation markers (CD10, 19, 34 and CD79α/ß), signal transduction molecules (BTK, Lyn and Syk) and transcriptional regulators (EBF1, E2A, and Pax5). This review aims to discuss the known similarities and differences between marsupial and eutherian B-cells, in regards to their genetic presence, homology, and developmental stages, as well as to highlight the areas requiring further investigation. By enhancing our understanding of the genes that are involved with B-cells in the marsupial lineage, it will, in turn, aid our understanding of the marsupial immune system and support the development of specific immunological reagents for research and wildlife conservation purposes.


Asunto(s)
Linfocitos B/inmunología , Marsupiales/inmunología , Animales , Inmunogenética
7.
Monoclon Antib Immunodiagn Immunother ; 38(6): 261-265, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31621497

RESUMEN

The highly O-glycosylated membrane glycoprotein podoplanin (PDPN) is frequently overexpressed in several malignant cancers, such as oral cancer, lung cancer, germinal neoplasia, mesothelioma, and brain tumor. The expression of PDPN is strongly associated with cancer progression and poor prognosis. PDPN possesses three tandem repeats of platelet aggregation-stimulating (PLAG) domains (PLAG1, PLAG2, and PLAG3) and PLAG-like domain (PLD), and binds to C-type lectin-like receptor 2 (CLEC-2) on platelets, followed by PDPN-mediated platelet aggregation. We have previously established a novel anti-Tasmanian devil PDPN (tasPDPN) monoclonal antibody (mAb), PMab-233, which specifically detects tasPDPN using flow cytometry, Western blot, and immunohistochemical analyses. However, the specific binding epitope of tasPDPN for PMab-233 remains to be clarified. Herein, a series of deletion or point mutants of tasPDPN were utilized for investigating the binding epitopes of PMab-233 using flow cytometry. The findings of this study demonstrated that Asp30, Thr33, and Thr34 of tasPDPN, which are located in PLAG1, are responsible for the binding of PMab-233 to tasPDPN.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Lectinas Tipo C/inmunología , Marsupiales/inmunología , Glicoproteínas de Membrana/inmunología , Animales , Especificidad de Anticuerpos/inmunología , Células CHO , Cricetinae , Cricetulus/inmunología , Mapeo Epitopo/métodos , Citometría de Flujo , Glicosilación , Humanos , Agregación Plaquetaria/inmunología
8.
Commun Biol ; 2: 99, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30886908

RESUMEN

The Tasmanian devil (Sarcophilus harrisii) is threatened by a contagious cancer, known as Devil Facial Tumour Disease (DFTD). A highly diverse T-cell receptor (TCR) repertoire is crucial for successful host defence against cancers. By investigating TCR beta chain diversity in devils of different ages, we show that the T-cell repertoire in devils constricts in their second year of life, which may explain the higher DFTD prevalence in older devils. Unexpectedly, we also observed a pronounced decline in TCR diversity and T cell clonal expansion in devils after DFTD infection. These findings overturned the previous assumption that DFTD did not directly impact host immunity.


Asunto(s)
Marsupiales/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunosenescencia/genética , Masculino , Neoplasias/genética , Transcripción Genética
9.
BMC Res Notes ; 11(1): 712, 2018 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305168

RESUMEN

OBJECTIVE: Marsupials suffer from an increasing number of stressors in this changing world. Functional studies are thus needed to broaden our understanding of the marsupial immune system. The red-tailed phascogale (Phascogale calura) is a small Australian marsupial previously used in descriptive immunological studies. Here, we aimed to develop functional assays by isolating and stimulating blood and spleen mononuclear cells in vitro. RESULTS: While peripheral blood mononuclear cell (PBMC) were relatively easy to isolate, only 105 mononuclear cells (> 90% purity and > 75% viability) could be recovered from the spleen, independently of the sex and age of the animal or the centrifugation time and speed tested. The pores of the mesh sieve used for tissue homogenization might have been too big to yield a single cell suspension. Nevertheless, in spite of the overall low number of cells recovered, PBMC and splenic mononuclear cells were successfully activated in preliminary trials with phytohemaglutinin. This activation state was evidenced by a change in shape and the presence of small cell aggregations in the mitogen-stimulated cultures. A non-radioactive colorimetric assay was also performed to confirm cell proliferation in these wells. This work highlights the importance of developing and reporting detailed methodological protocols in non-traditional research species.


Asunto(s)
Separación Celular/métodos , Leucocitos Mononucleares/efectos de los fármacos , Marsupiales/inmunología , Fitohemaglutininas/farmacología , Animales , Australia , Femenino , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Masculino , Cultivo Primario de Células , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
10.
Mamm Genome ; 29(11-12): 731-738, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30225648

RESUMEN

Genetic and genomic technologies have facilitated a greater understanding of the Tasmanian devil immune system and the origins, evolution and spread of devil facial tumour disease (DFTD). DFTD is a contagious cancer that has caused significant declines in devil populations across Tasmania. Immune responses to DFTD are rarely detected, allowing the cancer to pass between individuals and proliferate unimpeded. Early immunosenscence in devils appears to decrease anti-tumour immunity in older animals compared to younger animals, which may increase susceptibility to DFTD and explain high DFTD prevalence in this age group. Devils also have extremely low major histocompatibility complex (MHC) diversity, and multiple alleles are shared with the tumour, lowering histocompatibility barriers which may have contributed to DFTD evolution. DFTD actively evades immune attack by down-regulating cell-surface MHC I molecules, making it effectively invisible to the immune system. Altered MHC I profiles should activate natural killer (NK) cell anti-tumour responses, but these are absent in DFTD infection. Recent immunisation and immunotherapy using modified DFTD cells has induced an anti-DFTD immune response and regression of DFTD in some devils. Knowledge gained from immune responses to a transmissible cancer in devils will ultimately reveal useful insights into immunity to cancer in humans and other species.


Asunto(s)
Neoplasias Faciales/genética , Sistema Inmunológico , Marsupiales/genética , Neoplasias/inmunología , Animales , Proliferación Celular/genética , Neoplasias Faciales/inmunología , Humanos , Marsupiales/inmunología , Neoplasias/genética
11.
PLoS One ; 13(4): e0196469, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29702669

RESUMEN

The Tasmanian devil (Sarcophilus harrisii), the largest extant carnivorous marsupial and endemic to Tasmania, is at the verge of extinction due to the emergence of a transmissible cancer known as devil facial tumour disease (DFTD). DFTD has spread over the distribution range of the species and has been responsible for a severe decline in the global devil population. To protect the Tasmanian devil from extinction in the wild, our group has focused on the development of a prophylactic vaccine. Although this work has shown that vaccine preparations using whole DFTD tumour cells supplemented with adjuvants can induce anti-DFTD immune responses, alternative strategies that induce stronger and more specific immune responses are required. In humans, heat shock proteins (HSPs) derived from tumour cells have been used instead of whole-tumour cell preparations as a source of antigens for cancer immunotherapy. As HSPs have not been studied in the Tasmanian devil, this study presents the first characterisation of HSPs in this marsupial and evaluates the suitability of these proteins as antigenic components for the enhancement of a DFTD vaccine. We show that tissues and cancer cells from the Tasmanian devil express constitutive and inducible HSP. Additionally, this study suggests that HSP derived from DFTD cancer cells are immunogenic supporting the future development of a HSP-based vaccine against DFTD.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Faciales/prevención & control , Proteínas de Choque Térmico/metabolismo , Marsupiales/inmunología , Adyuvantes Inmunológicos , Animales , Anticuerpos/inmunología , Formación de Anticuerpos , Vacunas contra el Cáncer/inmunología , Línea Celular , Extinción Biológica , Neoplasias Faciales/tratamiento farmacológico , Neoplasias Faciales/inmunología , Inmunoterapia , Espectrometría de Masas , Proteoma , Tasmania
12.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29695421

RESUMEN

The discovery of highly diverse nonprimate hepatoviruses illuminated the evolutionary origins of hepatitis A virus (HAV) ancestors in mammals other than primates. Marsupials are ancient mammals that diverged from other Eutheria during the Jurassic. Viruses from marsupials may thus provide important insight into virus evolution. To investigate Hepatovirus macroevolutionary patterns, we sampled 112 opossums in northeastern Brazil. A novel marsupial HAV (MHAV) in the Brazilian common opossum (Didelphis aurita) was detected by nested reverse transcription-PCR (RT-PCR). MHAV concentration in the liver was high, at 2.5 × 109 RNA copies/g, and at least 300-fold higher than those in other solid organs, suggesting hepatotropism. Hepatovirus seroprevalence in D. aurita was 26.6% as determined using an enzyme-linked immunosorbent assay (ELISA). Endpoint titers in confirmatory immunofluorescence assays were high, and marsupial antibodies colocalized with anti-HAV control sera, suggesting specificity of serological detection and considerable antigenic relatedness between HAV and MHAV. MHAV showed all genomic hallmarks defining hepatoviruses, including late-domain motifs likely involved in quasi-envelope acquisition, a predicted C-terminal pX extension of VP1, strong avoidance of CpG dinucleotides, and a type 3 internal ribosomal entry site. Translated polyprotein gene sequence distances of at least 23.7% from other hepatoviruses suggested that MHAV represents a novel Hepatovirus species. Conserved predicted cleavage sites suggested similarities in polyprotein processing between HAV and MHAV. MHAV was nested within rodent hepatoviruses in phylogenetic reconstructions, suggesting an ancestral hepatovirus host switch from rodents into marsupials. Cophylogenetic reconciliations of host and hepatovirus phylogenies confirmed that host-independent macroevolutionary patterns shaped the phylogenetic relationships of extant hepatoviruses. Although marsupials are synanthropic and consumed as wild game in Brazil, HAV community protective immunity may limit the zoonotic potential of MHAV.IMPORTANCE Hepatitis A virus (HAV) is a ubiquitous cause of acute hepatitis in humans. Recent findings revealed the evolutionary origins of HAV and the genus Hepatovirus defined by HAV in mammals other than primates in general and in small mammals in particular. The factors shaping the genealogy of extant hepatoviruses are unclear. We sampled marsupials, one of the most ancient mammalian lineages, and identified a novel marsupial HAV (MHAV). The novel MHAV shared specific features with HAV, including hepatotropism, antigenicity, genome structure, and a common ancestor in phylogenetic reconstructions. Coevolutionary analyses revealed that host-independent evolutionary patterns contributed most to the current phylogeny of hepatoviruses and that MHAV was the most drastic example of a cross-order host switch of any hepatovirus observed so far. The divergence of marsupials from other mammals offers unique opportunities to investigate HAV species barriers and whether mechanisms of HAV immune control are evolutionarily conserved.


Asunto(s)
Virus de la Hepatitis A/clasificación , Hígado/virología , Marsupiales/virología , Animales , Anticuerpos Antivirales/metabolismo , Brasil , Evolución Molecular , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/fisiología , Hígado/inmunología , Marsupiales/inmunología , Filogenia , Proteínas Virales/química , Proteínas Virales/genética , Tropismo Viral
13.
Front Immunol ; 9: 259, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29515577

RESUMEN

Devil facial tumor disease (DFTD) is renowned for its successful evasion of the host immune system. Down regulation of the major histocompatabilty complex class I molecule (MHC-I) on the DFTD cells is a primary mechanism of immune escape. Immunization trials on captive Tasmanian devils have previously demonstrated that an immune response against DFTD can be induced, and that immune-mediated tumor regression can occur. However, these trials were limited by their small sample sizes. Here, we describe the results of two DFTD immunization trials on cohorts of devils prior to their wild release as part of the Tasmanian Government's Wild Devil Recovery project. 95% of the devils developed anti-DFTD antibody responses. Given the relatively large sample sizes of the trials (N = 19 and N = 33), these responses are likely to reflect those of the general devil population. DFTD cells manipulated to express MHC-I were used as the antigenic basis of the immunizations in both trials. Although the adjuvant composition and number of immunizations differed between trials, similar anti-DFTD antibody levels were obtained. The first trial comprised DFTD cells and the adjuvant combination of ISCOMATRIX™, polyIC, and CpG with up to four immunizations given at monthly intervals. This compared to the second trial whereby two immunizations comprising DFTD cells and the adjuvant combination ISCOMATRIX™, polyICLC (Hiltonol®) and imiquimod were given a month apart, providing a shorter and, therefore, more practical protocol. Both trials incorporated a booster immunization given up to 5 months after the primary course. A key finding was that devils in the second trial responded more quickly and maintained their antibody levels for longer compared to devils in the first trial. The different adjuvant combination incorporating the RNAase resistant polyICLC and imiquimod used in the second trial is likely to be responsible. The seroconversion in the majority of devils in these anti-DFTD immunization trials was remarkable, especially as DFTD is hallmarked by its immune evasion mechanisms. Microsatellite analyzes of MHC revealed that some MHC-I microsatellites correlated to stronger immune responses. These trials signify the first step in the long-term objective of releasing devils with immunity to DFTD into the wild.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer/inmunología , Neoplasias Faciales/inmunología , Inmunoterapia/métodos , Marsupiales/inmunología , Animales , Carboximetilcelulosa de Sodio/análogos & derivados , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Imiquimod/inmunología , Inmunidad Humoral , Inmunización Secundaria , Inmunoglobulina G/sangre , Masculino , Poli I-C/inmunología , Polilisina/análogos & derivados , Polilisina/inmunología , Escape del Tumor
14.
Sci Rep ; 8(1): 4175, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520077

RESUMEN

Devil Facial Tumour Disease (DFTD), a highly contagious cancer, has decimated Tasmanian devil (Sarcophilus harrisii) numbers in the wild. To ensure its long-term survival, a captive breeding program was implemented but has not been as successful as envisaged at its launch in 2005. We therefore investigated the reproductive success of 65 captive devil pair combinations, of which 35 produced offspring (successful pairs) whereas the remaining 30 pairs, despite being observed mating, produced no offspring (unsuccessful pairs). The devils were screened at six MHC Class I-linked microsatellite loci. Our analyses revealed that younger females had a higher probability of being successful than older females. In the successful pairs we also observed a higher difference in total number of heterozygous loci, i.e. when one devil had a high total number of heterozygous loci, its partner had low numbers. Our results therefore suggest that devil reproductive success is subject to disruptive MHC selection, which to our knowledge has never been recorded in any vertebrate. In order to enhance the success of the captive breeding program the results from the present study show the importance of using young (2-year old) females as well as subjecting the devils to MHC genotyping.


Asunto(s)
Enfermedades de los Animales , Especies en Peligro de Extinción , Genes MHC Clase I/inmunología , Marsupiales , Repeticiones de Microsatélite/inmunología , Neoplasias , Envejecimiento/genética , Envejecimiento/inmunología , Enfermedades de los Animales/genética , Enfermedades de los Animales/inmunología , Animales , Australia , Femenino , Marsupiales/genética , Marsupiales/inmunología , Neoplasias/genética , Neoplasias/inmunología
15.
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
16.
J Parasitol ; 104(1): 31-38, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29119858

RESUMEN

A novel coccidian species was discovered in the prostate of an Antechinus flavipes (yellow-footed antechinus) in South Australia during the period of postmating male antechinus immunosuppression and mortality. This novel coccidian is unusual because it develops extraintestinally and sporulates endogenously within the prostate gland of its mammalian host. Histological examination of prostatic tissue revealed dense aggregations of spherical and thin-walled tetrasporocystic, dizoic, sporulated coccidian oocysts within tubular lumina, with unsporulated oocysts and gamogonic stages within the cytoplasm of glandular epithelial cells. This coccidian was observed occurring concurrently with dasyurid gammaherpesvirus 1 infection of the antechinus' prostate. Eimeria-specific 18S small-subunit ribosomal (r)DNA polymerase chain reaction amplification was used to obtain a partial 18S rDNA nucleotide sequence from the antechinus coccidian. Bayesian phylogenetic analysis based on 18S rDNA gene sequences revealed that the novel coccidian clusters with reptile-host coccidians, forming an ancestral basal lineage of the eimeriid clade. The species has been named Eimeria taggarti n. sp. on the basis of both sporulated oocyst morphology and molecular characterization. It is suspected that E. taggarti is sexually transmitted via excretion of sporulated oocysts or free sporocysts with prostatic secretions in semen.


Asunto(s)
Coccidiosis/veterinaria , Eimeria/aislamiento & purificación , Marsupiales/parasitología , Próstata/parasitología , Enfermedades de la Próstata/veterinaria , Animales , Secuencia de Bases , Coccidiosis/parasitología , ADN Protozoario/química , ADN Protozoario/aislamiento & purificación , ADN Ribosómico/química , ADN Ribosómico/aislamiento & purificación , Eimeria/clasificación , Eimeria/genética , Eimeria/ultraestructura , Tolerancia Inmunológica , Masculino , Marsupiales/inmunología , Microscopía Electrónica de Transmisión/veterinaria , Oocistos/crecimiento & desarrollo , Oocistos/ultraestructura , Filogenia , Enfermedades de la Próstata/parasitología , ARN Protozoario/genética , ARN Ribosómico 18S/genética , Alineación de Secuencia/veterinaria , Australia del Sur
17.
Immunogenetics ; 69(8-9): 537-545, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28695294

RESUMEN

The Tasmanian devil, a marsupial species endemic to the island of Tasmania, harbours two contagious cancers, Devil Facial Tumour 1 (DFT1) and Devil Facial Tumour 2 (DFT2). These cancers pass between individuals in the population via the direct transfer of tumour cells, resulting in the growth of large tumours around the face and neck of affected animals. While these cancers are rare, a contagious cancer also exists in dogs and five contagious cancers circulate in bivalves. The ability of tumour cells to emerge and transmit in mammals is surprising as these cells are an allograft and should be rejected due to incompatibility between Major Histocompatibility Complex (MHC) genes. As such, considerable research has focused on understanding how DFT1 cells evade the host immune system with particular reference to MHC molecules. This review evaluates the role that MHC class I expression and genotype plays in allowing DFT1 to circumvent histocompatibility barriers in Tasmanian devils. We also examine recent research that suggests that Tasmanian devils can mount an immune response to DFT1 and may form the basis of a protective vaccine against the tumour.


Asunto(s)
Genes MHC Clase I/fisiología , Marsupiales/inmunología , Neoplasias/veterinaria , Animales , Genotipo , Neoplasias/genética , Neoplasias/inmunología , Tasmania
18.
Dev Comp Immunol ; 76: 352-360, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28689773

RESUMEN

Devil facial tumour disease (DFTD) describes two genetically distinct transmissible tumours that pose a significant threat to the survival of the Tasmanian devil. A prophylactic vaccine could protect devils from DFTD transmission. For this vaccine to be effective, potent immune adjuvants will be required. Toll-like receptors (TLRs) promote robust immune responses in human cancer studies and are highly conserved across mammalian species. In this study, we investigated the proficiency of TLR ligands for immune activation in the Tasmanian devil using in vitro mononuclear cell stimulations and in vivo immunisation trials with a model antigen. We identified two such TLR ligands, polyICLC (Hiltonol®) (TLR3) and imiquimod (TLR7), that in combination induced significant IFNγ production from Tasmanian devil lymphocytes in vitro. Immunisation with these ligands and the model antigen keyhole limpet haemocyanin activated robust antigen-specific primary, secondary and long-term memory IgG responses. Our results support the conserved nature of TLR signaling across mammalian species. PolyICLC and imiquimod will be trialed as immune adjuvants in future DFTD vaccine formulations.


Asunto(s)
Aminoquinolinas/inmunología , Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Carboximetilcelulosa de Sodio/análogos & derivados , Neoplasias Faciales/inmunología , Leucocitos Mononucleares/inmunología , Marsupiales/inmunología , Poli I-C/inmunología , Polilisina/análogos & derivados , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Neoplasias Faciales/prevención & control , Hemocianinas/inmunología , Humanos , Imiquimod , Inmunidad , Inmunidad Innata , Inmunización , Inmunoglobulina G/metabolismo , Activación de Linfocitos , Polilisina/inmunología , Receptores Toll-Like/agonistas
19.
Sci Rep ; 7: 43827, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28276463

RESUMEN

Devil facial tumour disease (DFTD) is a transmissible cancer devastating the Tasmanian devil (Sarcophilus harrisii) population. The cancer cell is the 'infectious' agent transmitted as an allograft by biting. Animals usually die within a few months with no evidence of antibody or immune cell responses against the DFTD allograft. This lack of anti-tumour immunity is attributed to an absence of cell surface major histocompatibility complex (MHC)-I molecule expression. While the endangerment of the devil population precludes experimentation on large experimental groups, those examined in our study indicated that immunisation and immunotherapy with DFTD cells expressing surface MHC-I corresponded with effective anti-tumour responses. Tumour engraftment did not occur in one of the five immunised Tasmanian devils, and regression followed therapy of experimentally induced DFTD tumours in three Tasmanian devils. Regression correlated with immune cell infiltration and antibody responses against DFTD cells. These data support the concept that immunisation of devils with DFTD cancer cells can successfully induce humoral responses against DFTD and trigger immune-mediated regression of established tumours. Our findings support the feasibility of a protective DFTD vaccine and ultimately the preservation of the species.


Asunto(s)
Neoplasias Faciales/inmunología , Inmunización/métodos , Inmunoterapia/métodos , Marsupiales/inmunología , Animales , Formación de Anticuerpos/inmunología , Neoplasias Faciales/terapia , Neoplasias Faciales/veterinaria , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Humoral/inmunología , Masculino , Resultado del Tratamiento
20.
Sci Rep ; 7: 44716, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28300197

RESUMEN

Tasmanian devils (Sarcophilus harrisii) are at risk of extinction in the wild due to Devil Facial Tumour Disease (DFTD), a rare contagious cancer. The prevalence of DFTD differs by age class: higher disease prevalence is seen in adults (2-3 years) versus younger devils (<2 years). Here we propose that immunological changes during puberty may play a role in susceptibility to DFTD. We show that the second year of life is a key developmental period for Tasmanian devils, during which they undergo puberty and pronounced changes in the immune system. Puberty coincides with a significant decrease in lymphocyte abundance resulting in a much higher neutrophil:lymphocyte ratio in adults than subadults. Quantitative PCR analysis of gene expression of transcription factors T-bet and GATA-3 and cytokines interferon gamma (IFN-γ) and interleukin 4 (IL-4) revealed a drastic increase in GATA-3 and IL-4 expression during puberty. These changes led to a significantly lower IFN-γ:IL-4 ratio in 2-year-olds than <1 year olds (on average 1.3-fold difference in males and 4.0-fold in females), which reflects a major shift of the immune system towards Th2 responses. These results all indicate that adult devils are expected to have a lower anticancer immune capacity than subadults, which may explain the observed pattern of disease prevalence of DFTD in the wild.


Asunto(s)
Marsupiales/inmunología , Neoplasias/inmunología , Maduración Sexual , Animales , Peso Corporal , Femenino , Regulación de la Expresión Génica , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Recuento de Linfocitos , Linfocitos/metabolismo , Masculino , Neoplasias/sangre , Neoplasias/patología , Neutrófilos/metabolismo , Progesterona/sangre , Células TH1/metabolismo , Células Th2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...