Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Oncoimmunology ; 13(1): 2345859, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38686178

RESUMO

Immune checkpoint therapy (ICT) causes durable tumour responses in a subgroup of patients, but it is not well known how T cell receptor beta (TCRß) repertoire dynamics contribute to the therapeutic response. Using murine models that exclude variation in host genetics, environmental factors and tumour mutation burden, limiting variation between animals to naturally diverse TCRß repertoires, we applied TCRseq, single cell RNAseq and flow cytometry to study TCRß repertoire dynamics in ICT responders and non-responders. Increased oligoclonal expansion of TCRß clonotypes was observed in responding tumours. Machine learning identified TCRß CDR3 signatures unique to each tumour model, and signatures associated with ICT response at various timepoints before or during ICT. Clonally expanded CD8+ T cells in responding tumours post ICT displayed effector T cell gene signatures and phenotype. An early burst of clonal expansion during ICT is associated with response, and we report unique dynamics in TCRß signatures associated with ICT response.


Assuntos
Inibidores de Checkpoint Imunológico , Linfócitos do Interstício Tumoral , Receptores de Antígenos de Linfócitos T alfa-beta , Animais , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Camundongos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Feminino
2.
HGG Adv ; 5(1): 100257, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38007613

RESUMO

An estimated 3.5%-5.9% of the global population live with rare diseases, and approximately 80% of these diseases have a genetic cause. Rare genetic diseases are difficult to diagnose, with some affected individuals experiencing diagnostic delays of 5-30 years. Next-generation sequencing has improved clinical diagnostic rates to 33%-48%. In a majority of cases, novel variants potentially causing the disease are discovered. These variants require functional validation in specialist laboratories, resulting in a diagnostic delay. In the interim, the finding is classified as a genetic variant of uncertain significance (VUS) and the affected individual remains undiagnosed. A VUS (PTCHD1 c. 2489T>G) was identified in a child with autistic behavior, global developmental delay, and hypotonia. Loss of function mutations in PTCHD1 are associated with autism spectrum disorder and intellectual disability; however, the molecular function of PTCHD1 and its role in neurodevelopmental disease is unknown. Here, we apply CRISPR gene editing and induced pluripotent stem cell (iPSC) neural disease modeling to assess the variant. During differentiation from iPSCs to neural progenitors, we detect subtle but significant gene signatures in synaptic transmission and muscle contraction pathways. Our work supports the causal link between the genetic variant and the child's phenotype, providing evidence for the variant to be considered a pathogenic variant according to the American College of Medical Genetics and Genomics guidelines. In addition, our study provides molecular data on the role of PTCHD1 in the context of other neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista , Criança , Humanos , Transtorno do Espectro Autista/diagnóstico , Sistemas CRISPR-Cas/genética , Diagnóstico Tardio , Fenótipo , Células-Tronco/metabolismo , Proteínas de Membrana/genética
3.
Stem Cell Res Ther ; 14(1): 345, 2023 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-38049901

RESUMO

BACKGROUND: Genomic sequencing in congenital heart disease (CHD) patients often discovers novel genetic variants, which are classified as variants of uncertain significance (VUS). Functional analysis of each VUS is required in specialised laboratories, to determine whether the VUS is disease causative or not, leading to lengthy diagnostic delays. We investigated stem cell cardiac disease modelling and transcriptomics for the purpose of genetic variant classification using a GATA4 (p.Arg283Cys) VUS in a patient with CHD. METHODS: We performed high efficiency CRISPR gene editing with homology directed repair in induced pluripotent stem cells (iPSCs), followed by rapid clonal selection with amplicon sequencing. Genetic variant and healthy matched control cells were compared using cardiomyocyte disease modelling and transcriptomics. RESULTS: Genetic variant and healthy cardiomyocytes similarly expressed Troponin T (cTNNT), and GATA4. Transcriptomics analysis of cardiomyocyte differentiation identified changes consistent with the patient's clinical human phenotype ontology terms. Further, transcriptomics revealed changes in calcium signalling, and cardiomyocyte adrenergic signalling in the variant cells. Functional testing demonstrated, altered action potentials in GATA4 genetic variant cardiomyocytes were consistent with patient cardiac abnormalities. CONCLUSIONS: This work provides in vivo functional studies supportive of a damaging effect on the gene or gene product. Furthermore, we demonstrate the utility of iPSCs, CRISPR gene editing and cardiac disease modelling for genetic variant interpretation. The method can readily be applied to other genetic variants in GATA4 or other genes in cardiac disease, providing a centralised assessment pathway for patient genetic variant interpretation.


Assuntos
Edição de Genes , Cardiopatias Congênitas , Humanos , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Miócitos Cardíacos/metabolismo , Sequência de Bases , Transdução de Sinais
4.
Cell Rep Med ; 4(7): 101113, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37467718

RESUMO

Recurrences frequently occur following surgical removal of primary tumors. In many cancers, adjuvant therapies have limited efficacy. Surgery provides access to the tumor microenvironment, creating an opportunity for local therapy, in particular immunotherapy, which can induce local and systemic anti-cancer effects. Here, we develop a surgically optimized biodegradable hyaluronic acid-based hydrogel for sustained intraoperative delivery of Toll-like receptor 3 agonist poly(I:C) and demonstrate that it significantly reduces tumor recurrence after surgery in multiple mouse models. Mechanistically, poly(I:C) induces a transient interferon alpha (IFNα) response, reshaping the tumor/wound microenvironment by attracting inflammatory monocytes and depleting regulatory T cells. We demonstrate that a pre-existing IFN signature predicts response to the poly(I:C) hydrogel, which sensitizes tumors to immune checkpoint therapy. The safety, immunogenicity, and surgical feasibility are confirmed in a veterinary trial in canine soft tissue tumors. The surgically optimized poly(I:C)-loaded hydrogel provides a safe and effective approach to prevent cancer recurrence.


Assuntos
Hidrogéis , Recidiva Local de Neoplasia , Camundongos , Animais , Cães , Hidrogéis/uso terapêutico , Recidiva Local de Neoplasia/prevenção & controle , Imunoterapia , Modelos Animais de Doenças , Microambiente Tumoral
5.
Gene ; 821: 146287, 2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35176430

RESUMO

There are an estimated > 400 million people living with a rare disease globally, with genetic variants the cause of approximately 80% of cases. Next Generation Sequencing (NGS) rapidly identifies genetic variants however they are often of unknown significance. Low throughput functional validation in specialist laboratories is the current ad hoc approach for functional validation of genetic variants, which creating major bottlenecks in patient diagnosis. This study investigates the application of CRISPR gene editing followed by genome wide transcriptomic profiling to facilitate patient diagnosis. As proof-of-concept, we introduced a variant in the Euchromatin histone methyl transferase (EHMT1) gene into HEK293T cells. We identified changes in the regulation of the cell cycle, neural gene expression and suppression of gene expression changes on chromosome 19 and chromosome X, that are in keeping with Kleefstra syndrome clinical phenotype and/or provide insight into disease mechanism. This study demonstrates the utility of genome editing followed by functional readouts to rapidly and systematically validating the function of variants of unknown significance in patients suffering from rare diseases.


Assuntos
Anormalidades Craniofaciais/diagnóstico , Edição de Genes/métodos , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Cardiopatias Congênitas/diagnóstico , Histona-Lisina N-Metiltransferase/genética , Deficiência Intelectual/diagnóstico , Sistemas CRISPR-Cas , Deleção Cromossômica , Cromossomos Humanos Par 19/genética , Cromossomos Humanos Par 9/genética , Cromossomos Humanos X/genética , Anormalidades Craniofaciais/genética , Diagnóstico Precoce , Regulação da Expressão Gênica , Variação Genética , Células HEK293 , Cardiopatias Congênitas/genética , Humanos , Deficiência Intelectual/genética , Estudo de Prova de Conceito , Análise de Sequência de RNA
6.
Stem Cell Res Ther ; 13(1): 69, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35139903

RESUMO

BACKGROUND: Over 400 million people worldwide are living with a rare disease. Next Generation Sequencing (NGS) identifies potential disease causative genetic variants. However, many are identified as variants of uncertain significance (VUS) and require functional laboratory validation to determine pathogenicity, and this creates major diagnostic delays. METHODS: In this study we test a rapid genetic variant assessment pipeline using CRISPR homology directed repair to introduce single nucleotide variants into inducible pluripotent stem cells (iPSCs), followed by neuronal disease modelling, and functional genomics on amplicon and RNA sequencing, to determine cellular changes to support patient diagnosis and identify disease mechanism. RESULTS: As proof-of-principle, we investigated an EHMT1 (Euchromatin histone methyltransferase 1; EHMT1 c.3430C > T; p.Gln1144*) genetic variant pathogenic for Kleefstra syndrome and determined changes in gene expression during neuronal progenitor cell differentiation. This pipeline rapidly identified Kleefstra syndrome in genetic variant cells compared to healthy cells, and revealed novel findings potentially implicating the key transcription factors REST and SP1 in disease pathogenesis. CONCLUSION: The study pipeline is a rapid, robust method for genetic variant assessment that will support rare diseases patient diagnosis. The results also provide valuable information on genome wide perturbations key to disease mechanism that can be targeted for drug treatments.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Anormalidades Craniofaciais , Deleção Cromossômica , Cromossomos Humanos Par 9 , Anormalidades Craniofaciais/genética , Genômica , Haploinsuficiência/genética , Cardiopatias Congênitas , Histona-Lisina N-Metiltransferase/genética , Humanos , Deficiência Intelectual
8.
Cancer Immunol Immunother ; 70(11): 3249-3258, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33835222

RESUMO

Surgical resection of cancer remains the frontline therapy for millions of patients annually, but post-operative recurrence is common, with a relapse rate of around 45% for non-small cell lung cancer. The tumour draining lymph nodes (dLN) are resected at the time of surgery for staging purposes, and this cannot be a null event for patient survival and future response to immune checkpoint blockade treatment. This project investigates cancer surgery, lymphadenectomy, onset of metastatic disease, and response to immunotherapy in a novel model that closely reflects the clinical setting. In a murine metastatic lung cancer model, primary subcutaneous tumours were resected with associated dLNs remaining intact, completely resected or partially resected. Median survival after surgery was significantly shorter with complete dLN resection at the time of surgery (49 days (95%CI)) compared to when lymph nodes remained intact (> 88 days; p < 0.05). Survival was partially restored with incomplete lymph node resection and CD8 T cell dependent. Treatment with aCTLA4 whilst effective against the primary tumour was ineffective for metastatic lung disease. Conversely, aPD-1/aCD40 treatment was effective in both the primary and metastatic disease settings and restored the detrimental effects of complete dLN resection on survival. In this pre-clinical lung metastatic disease model that closely reflects the clinical setting, we observe decreased frequency of survival after complete lymphadenectomy, which was ameliorated with partial lymph node removal or with early administration of aPD-1/aCD40 therapy. These findings have direct relevance to surgical lymph node resection and adjuvant immunotherapy in lung cancer, and perhaps other cancer, patients.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Excisão de Linfonodo , Metástase Neoplásica/imunologia , Animais , Quimioterapia Adjuvante/métodos , Inibidores de Checkpoint Imunológico/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos , Metástase Neoplásica/patologia , Recidiva Local de Neoplasia/patologia
9.
Front Immunol ; 11: 584423, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262762

RESUMO

Immune checkpoint therapy (ICT) results in durable responses in individuals with some cancers, but not all patients respond to treatment. ICT improves CD8+ cytotoxic T lymphocyte (CTL) function, but changes in tumor antigen-specific CTLs post-ICT that correlate with successful responses have not been well characterized. Here, we studied murine tumor models with dichotomous responses to ICT. We tracked tumor antigen-specific CTL frequencies and phenotype before and after ICT in responding and non-responding animals. Tumor antigen-specific CTLs increased within tumor and draining lymph nodes after ICT, and exhibited an effector memory-like phenotype, expressing IL-7R (CD127), KLRG1, T-bet, and granzyme B. Responding tumors exhibited higher infiltration of effector memory tumor antigen-specific CTLs, but lower frequencies of regulatory T cells compared to non-responders. Tumor antigen-specific CTLs persisted in responding animals and formed memory responses against tumor antigens. Our results suggest that increased effector memory tumor antigen-specific CTLs, in the presence of reduced immunosuppression within tumors is part of a successful ICT response. Temporal and nuanced analysis of T cell subsets provides a potential new source of immune based biomarkers for response to ICT.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Inibidores de Checkpoint Imunológico/imunologia , Memória Imunológica/imunologia , Animais , Antígenos de Neoplasias/imunologia , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Granzimas/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia
10.
Oncoimmunology ; 9(1): 1684714, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002299

RESUMO

Immune checkpoint blockade (ICPB) is a powerfully effective cancer therapy in some patients. Tumor neo-antigens are likely main targets for attack but it is not clear which and how many tumor mutations in individual cancers are actually antigenic, with or without ICPB therapy and their role as neo-antigen vaccines or as predictors of ICPB responses. To examine this, we interrogated the immune response to tumor neo-antigens in a murine model in which the tumor is induced by a natural human carcinogen (i.e. asbestos) and mimics its human counterpart (i.e. mesothelioma). We identified and screened 33 candidate neo-antigens, and found T cell responses against one candidate in tumor-bearing animals, mutant UQCRC2. Interestingly, we found a high degree of inter-animal variation in the magnitude of neo-antigen responses in otherwise identical mice. ICPB therapy with Cytotoxic T-lymphocyte-associated protein (CTLA-4) and α-glucocorticoid-induced TNFR family related gene (GITR) in doses that induced tumor regression, increased the magnitude of responses and unmasked functional T cell responses against another neo-antigen, UNC45a. Importantly, the magnitude of the pre-treatment draining lymph node (dLN) response to UNC45a closely corresponded to ICPB therapy outcomes. Surprisingly however, boosting pre-treatment UNC45a-specific T cell numbers did not improve response rates to ICPB. These observations suggest a novel biomarker approach to the clinical prediction of ICPB response and have important implications for the development of neo-antigen vaccines.


Assuntos
Vacinas Anticâncer , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias , Animais , Antígenos de Neoplasias/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Linfonodos , Camundongos , Neoplasias/genética , Neoplasias/terapia , Linfócitos T Citotóxicos
11.
Epigenetics ; 15(1-2): 134-144, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31423932

RESUMO

Activation of naïve CD8+ T cells stimulates proliferation and differentiation into cytotoxic T-lymphocytes (CTLs). Adoptive T Cell Therapy (ACT) involves multiple rounds of ex vivo activation to generate enough CTLs for reinfusion into patients, but this drives differentiation into terminal effector T cells. Less differentiated CTL populations, such as stem cell memory T cells, are more ideal candidates for ACT because of increased self-renewal and persistent properties. Ex vivo targeting of T cell differentiation with epigenetic modifiers is a potential strategy to improve cytotoxic T-lymphocyte (CTL) generation for ACT. We established a pipeline to assess the effects of epigenetic modifiers on CD8+ T cell proliferation, differentiation, and efficacy in a preclinical melanoma model. Single treatment with epigenetic modifiers inhibited T cell proliferation in vitro, producing CD44hiCD62Lhi effector-like T cells rather than a stem cell memory T cell phenotype. Most epigenetic modifying agents had no significant effect on ACT efficacy with the notable exception of the bromodomain and extraterminal (BET)-inhibitor JQ1 which was associated with a decrease in efficacy compared to unmodified T cells. These findings reveal the complexity of epigenetic targeting of T cell differentiation, highlighting the need to precisely define the epigenetic targeting strategies to improve CTL generation for ACT.


Assuntos
Proliferação de Células , Epigênese Genética , Imunoterapia Adotiva/métodos , Melanoma Experimental/terapia , Linfócitos T/efeitos dos fármacos , Animais , Azepinas/farmacologia , Benzodiazepinas/farmacologia , Diferenciação Celular , Linhagem Celular Tumoral , Células Cultivadas , Indolizinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Sulfonas/farmacologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/fisiologia , Triazóis/farmacologia
12.
Oncoimmunology ; 8(8): 1609874, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413920

RESUMO

Immunotherapies such as adoptive cell therapy (ACT) are promising treatments for solid cancers. However, relapsing disease remains a problem and the molecular mechanisms underlying resistance are poorly defined. We postulated that the deregulated epigenetic landscape in cancer cells could underpin the acquisition of resistance to immunotherapy. To address this question, two preclinical models of ACT were employed to study transcriptional and epigenetic regulatory processes within ACT-treated cancer cells. In these models ACT consistently causes robust tumor regression, but resistance develops and tumors relapse. We identified down-regulated expression of immunogenic antigens at the mRNA level correlated with escape from immune control. To determine whether this down-regulation was under epigenetic control, we treated escaped tumor cells with DNA demethylating agents, azacytidine (AZA) and decitabine (DEC). AZA or DEC treatment restored antigen expression in a proportion of the tumor population. To explore the importance of other epigenetic modifications we isolated tumor cells refractory to DNA demethylation and screened clones against a panel of 19 different epigenetic modifying agents (EMAs). The library of EMAs included inhibitors of a range of chromosomal and transcription regulatory protein complexes, however, when tested as single agents none restored further antigen expression. These findings suggest that tumor cells employ multiple epigenetic and genetic mechanisms to evade immune control, and a combinatorial approach employing several EMAs targeting transcription and genome stability may be required to overcome tumor resistance to immunotherapy.

13.
Oncoimmunology ; 7(10): e1494111, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30288361

RESUMO

Mesothelioma is an aggressive asbestos induced cancer with extremely poor prognosis and limited treatment options. Immune checkpoint blockade (ICPB) has demonstrated effective therapy in melanoma and is now being applied to other cancers, including mesothelioma. However, the efficacy of ICPB and which immune checkpoint combinations constitute the best therapeutic option for mesothelioma have yet to be fully elucidated. Here, we used our well characterised mesothelioma tumour model to investigate the efficacy of different ICBP treatments to generate effective therapy for mesothelioma. We show that tumour resident regulatory T cell co-express high levels of CTLA-4, OX40 and GITR relative to T effector subsets and that these receptors are co-expressed on a large proportion of cells. Targeting any of CTLA-4, OX40 or GITR individually generated effective responses against mesothelioma. Furthermore, the combination of αCTLA-4 and αOX40 was synergistic, with an increase in complete tumour regressions from 20% to 80%. Other combinations did not synergise to enhance treatment outcomes. Finally, an early pattern in T cell response was predictive of response, with activation status and ICP receptor expression profile of T effector cells harvested from tumour and dLN correlating with response to immunotherapy. Taken together, these data demonstrate that combination ICPB can work synergistically to induce strong, durable immunity against mesothelioma in an animal model.

14.
J Immunol ; 197(1): 128-40, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27233959

RESUMO

NK cells possess inhibitory receptors that are responsible for self-MHC class I recognition; beyond their inhibitory function, accumulating evidence indicates that such receptors confer NK cell functional competence through an unclear process termed "licensing." Ly49C is the main self-specific inhibitory Ly49 receptor in H-2(b) C57BL/6 (B6) mice. We used B6 Ly49C-transgenic and B6 ß2 microglobulin (ß2m)-knockout Ly49C-transgenic mice to investigate the impact of licensing through this inhibitory receptor in precursor and mature NK cells. We found that self-specific inhibitory receptors affected NK cell precursor survival and proliferation at particular developmental stages in an MHC class I-dependent manner. The presence of Ly49C impacted the NK cell repertoire in a ß2m-dependent manner, with reduced Ly49A(+), Ly49G2(+), and Ly49D(+) subsets, an increased DNAM-1(+) subset, and higher NKG2D expression. Licensed NK cells displayed a skewed distribution of the maturation stages, which was characterized by differential CD27 and CD11b expression, toward the mature phenotypes. We found that Ly49C-mediated licensing induced a split effect on NK cell functions, with increased cytokine-production capabilities following engagement of various activating receptors while cytotoxicity remained unchanged. Analysis of licensed NK cell functions in vivo, in a system of mouse CMV infection, indicated that licensing did not play a major role in the NK cell antiviral response during acute infection, but it strongly impaired the generation and/or persistence of memory NK cells. This study unravels multifaceted effects of licensing on NK cell populations and their functions.


Assuntos
Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/imunologia , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Animais , Antígenos de Diferenciação de Linfócitos T/metabolismo , Diferenciação Celular , Células Cultivadas , Citotoxicidade Imunológica , Antígenos de Histocompatibilidade Classe I/genética , Imunidade Inata , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Subfamília A de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo
15.
PLoS Pathog ; 10(5): e1004161, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24873973

RESUMO

Natural Killer (NK) cells are crucial in early resistance to murine cytomegalovirus (MCMV) infection. In B6 mice, the activating Ly49H receptor recognizes the viral m157 glycoprotein on infected cells. We previously identified a mutant strain (MCMVG1F) whose variant m157 also binds the inhibitory Ly49C receptor. Here we show that simultaneous binding of m157 to the two receptors hampers Ly49H-dependent NK cell activation as Ly49C-mediated inhibition destabilizes NK cell conjugation with their targets and prevents the cytoskeleton reorganization that precedes killing. In B6 mice, as most Ly49H+ NK cells do not co-express Ly49C, the overall NK cell response remains able to control MCMVm157G1F infection. However, in B6 Ly49C transgenic mice where all NK cells express the inhibitory receptor, MCMV infection results in altered NK cell activation associated with increased viral replication. Ly49C-mediated inhibition also regulates Ly49H-independent NK cell activation. Most interestingly, MHC class I regulates Ly49C function through cis-interactions that mask the receptor and restricts m157 binding. B6 Ly49C Tg, ß2m ko mice, whose Ly49C receptors are unmasked due to MHC class I deficient expression, are highly susceptible to MCMVm157G1F and are unable to control a low-dose infection. Our study provides novel insights into the mechanisms that regulate NK cell activation during viral infection.


Assuntos
Infecções por Citomegalovirus/virologia , Antígenos de Histocompatibilidade Classe I/imunologia , Células Matadoras Naturais/virologia , Muromegalovirus , Subfamília A de Receptores Semelhantes a Lectina de Células NK/genética , Animais , Infecções por Citomegalovirus/metabolismo , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia
16.
Nat Immunol ; 14(7): 699-705, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23666294

RESUMO

Activating and inhibitory receptors on natural killer (NK) cells have a crucial role in innate immunity, although the basis of the engagement of activating NK cell receptors is unclear. The activating receptor Ly49H confers resistance to infection with murine cytomegalovirus by binding to the 'immunoevasin' m157. We found that m157 bound to the helical stalk of Ly49H, whereby two m157 monomers engaged the Ly49H dimer. The helical stalks of Ly49H lay centrally across the m157 platform, whereas its lectin domain was not required for recognition. Instead, m157 targeted an 'aromatic peg motif' present in stalks of both activating and inhibitory receptors of the Ly49 family, and substitution of this motif abrogated binding. Furthermore, ligation of m157 to Ly49H or Ly49C resulted in intracellular signaling. Accordingly, m157 has evolved to 'tackle the legs' of a family of NK cell receptors.


Assuntos
Infecções por Herpesviridae/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Motivos de Aminoácidos/imunologia , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Transdução de Sinais/imunologia , Organismos Livres de Patógenos Específicos , Ressonância de Plasmônio de Superfície
17.
J Immunol ; 186(3): 1713-22, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21187440

RESUMO

The Ly49H activating receptor on C57BL/6 (B6) NK cells plays a key role in early resistance to murine cytomegalovirus (MCMV) infection through specific recognition of the MCMV-encoded MHC class I-like molecule m157 expressed on infected cells. The m157 molecule is also recognized by the Ly49I inhibitory receptor from the 129/J mouse strain. The m157 gene is highly sequence variable among MCMV isolates, with many m157 variants unable to bind Ly49H(B6). In this study, we have sought to define if m157 variability leads to a wider spectrum of interactions with other Ly49 molecules and if this modifies host susceptibility to MCMV. We have identified novel m157-Ly49 receptor interactions, involving Ly49C inhibitory receptors from B6, BALB/c, and NZB mice, as well as the Ly49H(NZB) activation receptor. Using an MCMV recombinant virus in which m157(K181) was replaced with m157(G1F), which interacts with both Ly49H(B6) and Ly49C(B6), we show that the m157(G1F)-Ly49C interactions cause no apparent attenuating effect on viral clearance in B6 mice. Hence, when m157 can bind both inhibitory and activation NK cell receptors, the outcome is still activation. Thus, these data indicate that whereas m157 variants predominately interact with inhibitory Ly49 receptors, these interactions do not profoundly interfere with early NK cell responses.


Assuntos
Epitopos/imunologia , Variação Genética/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Muromegalovirus/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Epitopos/metabolismo , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Modelos Imunológicos , Muromegalovirus/genética , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Especificidade da Espécie
18.
Eur J Immunol ; 40(9): 2618-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20662096

RESUMO

NK cell-mediated resistance to murine cytomegalovirus (MCMV) is controlled by allelic Ly49 receptors, including activating Ly49H (C57BL/6 strain) and inhibitory Ly49I (129 strain), which specifically recognize MCMV m157, a glycosylphosphatidylinositol-linked protein with homology to MHC class I. Although the Ly49 receptors retain significant homology to classic carbohydrate-binding lectins, the role of glycosylation in ligand binding is unclear. Herein, we show that m157 is expressed in multiple, differentially N-glycosylated isoforms in m157-transduced or MCMV-infected cells. We used site-directed mutagenesis to express single and combinatorial asparagine (N)-to-glutamine (Q) mutations at N178, N187, N213, and N267 in myeloid and fibroblast cell lines. Progressive loss of N-linked glycans led to a significant reduction of total cellular m157 abundance, although all variably glycosylated m157 isoforms were expressed at the cell surface and retained the capacity to activate Ly49H(B6) and Ly49I(129) reporter cells and Ly49H(+) NK cells. However, the complete lack of N-linked glycans on m157 destabilized the m157-Ly49H interaction and prevented physical transfer of m157 to Ly49H-expressing cells. Thus, glycosylation on m157 enhances expression and binding to Ly49H, factors that may impact the interaction between NK cells and MCMV in vivo where receptor-ligand interactions are more limiting.


Assuntos
Fibroblastos/metabolismo , Infecções por Herpesviridae/metabolismo , Muromegalovirus/imunologia , Células Mieloides/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Fibroblastos/imunologia , Fibroblastos/patologia , Glicosilação , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Ativação Linfocitária/genética , Camundongos , Muromegalovirus/patogenicidade , Mutagênese Sítio-Dirigida , Mutação/genética , Células Mieloides/imunologia , Células Mieloides/patologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília A de Receptores Semelhantes a Lectina de Células NK/metabolismo , Ligação Proteica/genética , Isoformas de Proteínas/genética , Transgenes/genética , Proteínas Virais/genética , Proteínas Virais/imunologia
19.
Arch Virol ; 154(1): 65-75, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19066712

RESUMO

Previous analysis of the M73-to-m74/M75 intergenic region of murine cytomegalovirus (MCMV) identified a family of 3'-co-terminal spliced transcripts that includes M73 and M73.5. The current study investigated whether similar families of spliced genes also exist in the human CMV (HCMV) and rat CMV (RCMV) genomes. Northern blot, RT-PCR and RACE-PCR analysis of HCMV transcripts showed that while mRNAs from HCMV UL73 and a putative UL73.5 homologue were spliced and 3'-co-terminal, they were not 5'-co-terminal. In contrast, the spliced RCMV R73 and R73.5 transcripts were arranged in a similar manner to those in MCMV and found to be both 5' and 3'-co-terminal. In both the HCMV and RCMV genomes, additional non-coding spliced transcripts were found to originate from these regions. These results highlight that families of spliced transcripts coding for structural glycoproteins are likely to be a conserved feature of this region of betaherpesviral genomes.


Assuntos
Citomegalovirus/genética , DNA Recombinante/genética , Regulação Viral da Expressão Gênica , Muromegalovirus/genética , Proteínas Virais/genética , Animais , Northern Blotting , Células Cultivadas , DNA Intergênico/genética , Fibroblastos , Perfilação da Expressão Gênica , Humanos , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar
20.
J Gen Virol ; 88(Pt 3): 758-769, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17325348

RESUMO

Murine cytomegalovirus (MCMV) is a widely used model for human cytomegalovirus (HCMV) and has facilitated many important discoveries about the biology of CMVs. Most of these studies are conducted using the laboratory MCMV strains Smith and K181. However, wild-derived isolates of MCMV, like HCMV clinical isolates, exhibit genetic variation from laboratory strains, particularly at the ends of their genomes in areas containing known or putative immune-evasion and tropism genes. This study analysed the nucleotide sequence of the m02-m05 region, within the m02 gene family, of a number of laboratory and wild-derived MCMV isolates, and found a large degree of variation in both the sequence and arrangement of genes. A new open reading frame (ORF), designated m03.5, was found to be present in a number of wild isolates of MCMV in place of m03. Two distinct isolates, W8 and W8211, were found to possess both m03 and m03.5. Both m03 and m03.5 had early transcription kinetics and the encoded proteins could be detected on the cell surface, consistent with a possible role in immune evasion through binding to host-cell proteins. These data show that gene duplication and sequence variation occur within different isolates of MCMV found in the wild. As this variation among strains may alter the function of genes, these findings should be considered when analysing gene function or host-virus interactions in laboratory models.


Assuntos
DNA Viral/genética , Genes Virais , Variação Genética , Muromegalovirus/genética , Polimorfismo Genético , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , DNA Viral/química , Citometria de Fluxo , Expressão Gênica , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Muromegalovirus/isolamento & purificação , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Sintenia , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...