Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 354
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38612464

RESUMO

Immunodominant alloantigens in pig sperm membranes include 15 known gene products and a previously undiscovered Mr 20,000 sperm membrane-specific protein (SMA20). Here we characterize SMA20 and identify it as the unannotated pig ortholog of PMIS2. A composite SMA20 cDNA encoded a 126 amino acid polypeptide comprising two predicted transmembrane segments and an N-terminal alanine- and proline (AP)-rich region with no apparent signal peptide. The Northern blots showed that the composite SMA20 cDNA was derived from a 1.1 kb testis-specific transcript. A BLASTp search retrieved no SMA20 match from the pig genome, but it did retrieve a 99% match to the Pmis2 gene product in warthog. Sequence identity to predicted PMIS2 orthologs from other placental mammals ranged from no more than 80% overall in Cetartiodactyla to less than 60% in Primates, with the AP-rich region showing the highest divergence, including, in the extreme, its absence in most rodents, including the mouse. SMA20 immunoreactivity localized to the acrosome/apical head of methanol-fixed boar spermatozoa but not live, motile cells. Ultrastructurally, the SMA20 AP-rich domain immunolocalized to the inner leaflet of the plasma membrane, the outer acrosomal membrane, and the acrosomal contents of ejaculated spermatozoa. Gene name search failed to retrieve annotated Pmis2 from most mammalian genomes. Nevertheless, individual pairwise interrogation of loci spanning Atp4a-Haus5 identified Pmis2 in all placental mammals, but not in marsupials or monotremes. We conclude that the gene encoding sperm-specific SMA20/PMIS2 arose de novo in Eutheria after divergence from Metatheria, whereupon rapid molecular evolution likely drove the acquisition of a species-divergent function unique to fertilization in placental mammals.


Assuntos
Placenta , Sêmen , Masculino , Feminino , Gravidez , Suínos , Animais , Camundongos , DNA Complementar , Espermatozoides , Eutérios , Alanina , Isoantígenos/genética , Fertilização/genética
2.
PLoS One ; 16(12): e0260800, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34879110

RESUMO

The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.


Assuntos
Biomarcadores Tumorais/metabolismo , Fibroblastos Associados a Câncer/patologia , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Isoantígenos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Microambiente Tumoral , Apoptose , Biomarcadores Tumorais/genética , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/metabolismo , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Isoantígenos/genética , Glicoproteínas de Membrana/genética , Neutrófilos/imunologia , Neutrófilos/metabolismo , Prognóstico , Receptores de Superfície Celular/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
3.
Int J Infect Dis ; 105: 662-667, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33667695

RESUMO

OBJECTIVES: In this study, we aimed to develop a simple gene model to identify bacterial infection, which can be implemented in general clinical settings. METHODS: We used a clinically availablereal-time quantitative polymerase chain reaction platform to conduct focused gene expression assays on clinical blood samples. Samples were collected from 2 tertiary hospitals. RESULTS: We found that the 8 candidate genes for bacterial infection were significantly dysregulated in bacterial infection and displayed good performance in group classification, whereas the 2 genes for viral infection displayed poor performance. A two-gene model (S100A12 and CD177) displayed 93.0% sensitivity and 93.7% specificity in the modeling stage. In the independent validation stage, 87.8% sensitivity and 96.6% specificity were achieved in one set of case-control groups, and 93.6% sensitivity and 97.1% specificity in another set. CONCLUSIONS: We have validated the signature genes for bacterial infection and developed a two-gene model to identify bacterial infection in general clinical settings.


Assuntos
Infecções Bacterianas/diagnóstico , Infecções Bacterianas/genética , Modelos Genéticos , Biomarcadores/análise , Proteína C-Reativa/análise , Estudos de Casos e Controles , Feminino , Proteínas Ligadas por GPI/genética , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Isoantígenos/genética , Masculino , Pró-Calcitonina/análise , Reação em Cadeia da Polimerase em Tempo Real/métodos , Receptores de Superfície Celular/genética , Proteína S100A12/genética , Sensibilidade e Especificidade , Viroses/genética
4.
Arthritis Rheumatol ; 73(7): 1334-1340, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33452871

RESUMO

OBJECTIVE: Canakinumab is a human anti-interleukin-1ß (anti-IL-1ß) blocking agent that effectively neutralizes IL-1ß-mediated signaling for treatment of systemic juvenile idiopathic arthritis (JIA). While many patients have dramatic clinical response to IL-1 blockade, approximately one-third fail to respond, but there are currently no validated clinical or immunologic predictors of response. We undertook this study to characterize distinct gene signatures for treatment response and nonresponse to canakinumab in systemic JIA patients. METHODS: We performed a secondary analysis of whole-blood gene expression microarrays using blood samples obtained from healthy controls and systemic JIA patients at baseline and on day 3 after canakinumab treatment (GEO accession no. GSE80060). Patients were considered strong clinical responders if they met the ACR90 response (exhibited ≥90% improvement in the American College of Rheumatology [ACR] JIA response criteria; nonresponders were those who met ACR30 [exhibiting ≤30% improvement in the ACR JIA response criteria]). A random-effects model with patient identity as the random variable was used for differential expression analysis. RESULTS: We identified a distinct gene expression signature in patients with a strong clinical response to canakinumab treatment as compared to nonresponders, mediated by up-regulation of neutrophil- and IL-1-associated genes and characterized by increasing divergence from control transcriptomes with increasing clinical response. We also identified a signature including up-regulated CD163 expression that was associated with canakinumab nonresponse. Intriguingly, canakinumab treatment induced either up- or down-regulation of type I interferon (IFN) genes, independent of clinical response. CONCLUSION: Here, we identify a gene signature in systemic JIA patients prior to receiving treatment that distinguishes strong responders to canakinumab from nonresponders. Further prospective studies are needed to assess the utility of these insights for treatment decisions in systemic JIA and to track the association of up-regulated type I IFN signatures with systemic JIA complications.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Artrite Juvenil/tratamento farmacológico , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Artrite Juvenil/genética , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas de Ligação ao Cálcio/genética , Caspases/genética , Quimiocina CXCL1/genética , Criança , Proteínas de Ligação a DNA/genética , Proteínas Ligadas por GPI/genética , Ontologia Genética , Glicoproteínas/genética , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Proteína Acessória do Receptor de Interleucina-1/genética , Interleucina-1beta/genética , Isoantígenos/genética , Ativação de Neutrófilo/genética , Receptores de Superfície Celular/genética , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais , Receptores Toll-Like/genética , Transcriptoma , Resultado do Tratamento , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
5.
BMC Biotechnol ; 21(1): 9, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33514392

RESUMO

BACKGROUND: The ability of CRISPR/Cas9 to mutate any desired genomic locus is being increasingly explored in the emerging area of cancer immunotherapy. In this respect, current efforts are mostly focused on the use of autologous (i.e. patient-derived) T cells. The autologous approach, however, has drawbacks in terms of manufacturing time, cost, feasibility and scalability that can affect therapeutic outcome or wider clinical application. The use of allogeneic T cells from healthy donors may overcome these limitations. For this strategy to work, the endogenous T cell receptor (TCR) needs to be knocked out in order to reduce off-tumor, graft-versus-host-disease (GvHD). Furthermore, CD52 may be knocked out in the donor T cells, since this leaves them resistant to the commonly used anti-CD52 monoclonal antibody lymphodepletion regimen aiming to suppress rejection of the infused T cells by the recipient. Despite the great prospect, genetic manipulation of human T cells remains challenging, in particular how to deliver the engineering reagents: virus-mediated delivery entails the inherent risk of altering cancer gene expression by the genomically integrated CRISPR/Cas9. This is avoided by delivery of CRISPR/Cas9 as ribonucleoproteins, which, however, are fragile and technically demanding to produce. Electroporation of CRISPR/Cas9 expression plasmids would bypass the above issues, as this approach is simple, the reagents are robust and easily produced and delivery is transient. RESULTS: Here, we tested knockout of either TCR or CD52 in human primary T cells, using electroporation of CRISPR/Cas9 plasmids. After validating the CRISPR/Cas9 constructs in human 293 T cells by Tracking of Indels by Decomposition (TIDE) and Indel Detection by Amplicon Analysis (IDAA) on-target genomic analysis, we evaluated their efficacy in primary T cells. Four days after electroporation with the constructs, genomic analysis revealed a knockout rate of 12-14% for the two genes, which translated into 7-8% of cells showing complete loss of surface expression of TCR and CD52 proteins, as determined by flow cytometry analysis. CONCLUSION: Our results demonstrate that genomic knockout by electroporation of plasmids encoding CRISPR/Cas9 is technically feasible in human primary T cells, albeit at low efficiency.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Isoantígenos/genética , Linfócitos T/metabolismo , Antígeno CD52/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Eletroporação , Edição de Genes/métodos , Genômica , Células HEK293 , Humanos , Mutação INDEL , Plasmídeos
6.
Int J Immunogenet ; 48(2): 145-156, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32970372

RESUMO

Granulocytes are an essential part of both the innate and adaptive immune systems. Human neutrophil antigens (HNAs) are a family of epitopes that are located on glycoproteins that are mostly expressed on human granulocytes. Antibodies that recognize these epitopes have been associated with neutropenia, transfusion complications, haematopoietic stem cell transplant nonengraftment and renal transplant rejection. Currently, there are fourteen recognized HNA alleles across five antigen systems (HNA-1 through HNA-5), the molecular basis of which are located on the genes FCGR3B, CD177, SLC44A2, ITGAM and ITGAL, respectively. Elucidation of the associated genes has permitted the development of testing strategies for HNA typing and aided understanding of the associated epitopes. This review will outline the associated clinical conditions that require HNA investigation and how these are performed in specialized laboratories. Investigations provided are both reactive for patients with a variety of existing or suspected neutropenias and proactive in the testing of blood component donors in order to reduce the potential risk to patients who require transfusion.


Assuntos
Isoantígenos/imunologia , Neutrófilos/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Técnicas de Genotipagem , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Testes Imunológicos , Isoanticorpos/sangue , Isoanticorpos/imunologia , Isoantígenos/sangue , Isoantígenos/genética , Neutropenia/imunologia , Fenótipo , Reação Transfusional/imunologia , Imunologia de Transplantes
7.
J Clin Invest ; 130(12): 6317-6324, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32817591

RESUMO

Desmoglein 3 chimeric autoantibody receptor T cells (DSG3-CAART) expressing the pemphigus vulgaris (PV) autoantigen DSG3 fused to CD137-CD3ζ signaling domains, represent a precision cellular immunotherapy approach for antigen-specific B cell depletion. Here, we present definitive preclinical studies enabling a first-in-human trial of DSG3-CAART for mucosal PV. DSG3-CAART specifically lysed human anti-DSG3 B cells from PV patients and demonstrated activity consistent with a threshold dose in vivo, resulting in decreased target cell burden, decreased serum and tissue-bound autoantibodies, and increased DSG3-CAART engraftment. In a PV active immune model with physiologic anti-DSG3 IgG levels, DSG3-CAART inhibited antibody responses against pathogenic DSG3 epitopes and autoantibody binding to epithelial tissues, leading to clinical and histologic resolution of blisters. DSG3 autoantibodies stimulated DSG3-CAART IFN-γ secretion and homotypic clustering, consistent with an activated phenotype. Toxicology screens using primary human cells and high-throughput membrane proteome arrays did not identify off-target cytotoxic interactions. These preclinical data guided the trial design for DSG3-CAART and may help inform CAART preclinical development for other antibody-mediated diseases.


Assuntos
Transferência Adotiva , Linfócitos B/imunologia , Depleção Linfocítica , Pênfigo/terapia , Medicina de Precisão , Adulto , Animais , Autoanticorpos/imunologia , Linfócitos B/patologia , Desmogleína 3/genética , Desmogleína 3/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Interferon gama/imunologia , Isoantígenos/genética , Isoantígenos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pênfigo/genética , Pênfigo/imunologia , Pênfigo/patologia
8.
J Immunol ; 205(6): 1554-1563, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32796024

RESUMO

The genetic basis and mechanisms of disparate antitumor immune response was investigated in Diversity Outbred (DO) F1 mice that express human HER2. DO mouse stock samples nearly the entire genetic repertoire of the species. We crossed DO mice with syngeneic HER2 transgenic mice to study the genetics of an anti-self HER2 response in a healthy outbred population. Anti-HER2 IgG was induced by Ad/E2TM or naked pE2TM, both encoding HER2 extracellular and transmembrane domains. The response of DO F1 HER2 transgenic mice was remarkably variable. Still, immune sera inhibited HER2+ SKBR3 cell survival in a dose-dependent fashion. Using DO quantitative trait locus (QTL) analysis, we mapped the QTL that influences both total IgG and IgG2(a/b/c) Ab response to either Ad/E2TM or pE2TM. QTL from these four datasets identified a region in chromosome 17 that was responsible for regulating the response. A/J and NOD segments of genes in this region drove elevated HER2 Ig levels. This region is rich in MHC-IB genes, several of which interact with inhibitory receptors of NK cells. (B6xA/J)F1 and (B6xNOD)F1 HER2 transgenic mice received Ad/E2TM after NK cell depletion, and they produced less HER2 IgG, demonstrating positive regulatory function of NK cells. Depletion of regulatory T cells enhanced response. Using DO QTL analysis, we show that MHC-IB reactive NK cells exert positive influence on the immunity, countering negative regulation by regulatory T cells. This new, to our knowledge, DO F1 platform is a powerful tool for revealing novel immune regulatory mechanisms and for testing new interventional strategies.


Assuntos
Autoantígenos/metabolismo , Isoantígenos/metabolismo , Células Matadoras Naturais/fisiologia , Locos de Características Quantitativas/genética , Receptor ErbB-2/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Animais não Endogâmicos , Autoantígenos/genética , Autoantígenos/imunologia , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunidade , Imunoglobulina G/sangue , Isoantígenos/genética , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Domínios Proteicos/genética , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia
9.
Immunol Res ; 68(1): 63-70, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32096010

RESUMO

With several different CAR T cell therapies under advanced phases of clinical trials, and the first FDA-approved CAR treatments in 2017 (Yescarta and Kymriah), CAR T cell therapy has become one of the most promising therapies for the treatment of certain types of cancer. This success has bred an opportunity to optimize the production of CAR T cells for easier patient access. CAR T cell therapy is a rather expensive and personalized process that requires expensive measures to collect cells from patients, engineer those cells, and re-infuse the cells into the patient with adequate quality controls at each phase. With this in mind, significant attempts at creating a "universal" CAR T cell are underway in order to create an "off-the-shelf" product that would reduce the expense and time required for traditional CAR T cell treatment. The primary obstacle facing this endeavor is avoiding graft-versus-host disease that accompanies allogeneic transplants between genetically dissimilar individuals. With the advent of CRISPR and TALEN technology, editing the genome of allogeneic cells has become very possible, and several groups have provided initial data analyzing the effects of CAR T cells that have been edited to avoid host rejection and avoid endogenous TCR alloreactivity. These engineered cells not only have to avoid GVHD but also have to retain their anti-tumor efficacy in vivo. Here, we expand on the recent efforts and strides that have been made in the design and testing of universal allogeneic CAR T cells.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Imunoterapia Adotiva/métodos , Isoantígenos/metabolismo , Neoplasias/terapia , Linfócitos T/fisiologia , Animais , Sistemas CRISPR-Cas , Engenharia Genética , Humanos , Isoantígenos/genética , Isoantígenos/imunologia , Neoplasias/imunologia , Medicina de Precisão , Linfócitos T/transplante , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Transplante Homólogo
10.
Front Immunol ; 10: 2549, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31736968

RESUMO

Cytomegalovirus (CMV) infection is associated with allograft rejection but the mechanisms behind are poorly defined yet. Although cross-reactivity of T cells to alloantigen and CMV has been hypothesized, direct evidence in patients is lacking. In this observational cohort study, we tested the pre-transplant effector/memory T cell response to CMV peptide pools and alloantigen in 78 living donor/recipient pairs using the interferon-gamma Enzyme-Linked ImmunoSpot (ELISPOT) assay. To prove the hypothesis of cross-reactivity, we analyzed by applying next-generation sequencing the T cell receptor ß (TCR- ß) repertoire of CMV- and alloantigen-reactive T cells enriched from peripheral pre-transplant blood of 11 CMV-seropositive and HLA class I mismatched patients. Moreover, the TCR-repertoire was also analyzed in the allograft biopsies of those patients. There was a significant association between the presence of pre-transplant CMV immediate-early protein 1 (IE-1)-specific effector/memory T cells and acute renal allograft rejection and function (p = 0.01). Most importantly, we revealed shared TCR-ß sequences between CMV-IE1 and donor alloantigen-reactive T cells in all pre-transplant peripheral blood samples analyzed in CMV-seropositive patients who received HLA class I mismatched grafts. Identical TCR sequences were also found in particular in post-transplant allograft biopsies of patients with concomitant CMV infection and rejection. Our data show the presence of functional, cross-reactive T cells and their clonotypes in peripheral blood and in kidney allograft tissue. It is therefore likely that CMV-donor cross-reactivity as well as CMV specific T cell elicited inflammation is involved in the processes that affect allograft outcomes.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus/imunologia , Memória Imunológica , Transplante de Rim , Receptores de Antígenos de Linfócitos T alfa-beta , Linfócitos T , Adulto , Aloenxertos , Biópsia , Estudos de Coortes , Infecções por Citomegalovirus/etiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Feminino , Humanos , Isoantígenos/genética , Isoantígenos/imunologia , Masculino , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/microbiologia , Linfócitos T/patologia
11.
Int J Mol Sci ; 20(2)2019 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-30642095

RESUMO

The advancement of bioinformatics and machine learning has facilitated the discovery and validation of omics-based biomarkers. This study employed a novel approach combining multi-platform transcriptomics and cutting-edge algorithms to introduce novel signatures for accurate diagnosis of colorectal cancer (CRC). Different random forests (RF)-based feature selection methods including the area under the curve (AUC)-RF, Boruta, and Vita were used and the diagnostic performance of the proposed biosignatures was benchmarked using RF, logistic regression, naïve Bayes, and k-nearest neighbors models. All models showed satisfactory performance in which RF appeared to be the best. For instance, regarding the RF model, the following were observed: mean accuracy 0.998 (standard deviation (SD) < 0.003), mean specificity 0.999 (SD < 0.003), and mean sensitivity 0.998 (SD < 0.004). Moreover, proposed biomarker signatures were highly associated with multifaceted hallmarks in cancer. Some biomarkers were found to be enriched in epithelial cell signaling in Helicobacter pylori infection and inflammatory processes. The overexpression of TGFBI and S100A2 was associated with poor disease-free survival while the down-regulation of NR5A2, SLC4A4, and CD177 was linked to worse overall survival of the patients. In conclusion, novel transcriptome signatures to improve the diagnostic accuracy in CRC are introduced for further validations in various clinical settings.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/diagnóstico , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Algoritmos , Área Sob a Curva , Teorema de Bayes , Fatores Quimiotáticos/genética , Neoplasias Colorretais/genética , Feminino , Proteínas Ligadas por GPI/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Isoantígenos/genética , Modelos Logísticos , Aprendizado de Máquina , Prognóstico , Receptores de Superfície Celular/genética , Receptores Citoplasmáticos e Nucleares/genética , Proteínas S100/genética , Sensibilidade e Especificidade , Simportadores de Sódio-Bicarbonato/genética , Análise de Sobrevida , Fator de Crescimento Transformador beta1/genética
12.
Br J Cancer ; 120(1): 69-78, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30377339

RESUMO

BACKGROUND: Immune infiltration is implicated in the development of acquired resistance to anti-angiogenic cancer therapy. We therefore investigated the correlation between neutrophil infiltration in metastasis of colorectal cancer (CRC) patients and survival after treatment with bevacizumab. Our study identifies CD177+ tumour neutrophil infiltration as an adverse prognostic factor for bevacizumab treatment. We further demonstrate that a novel anti-VEGF/anti-Ang2 compound (BI-880) can overcome resistance to VEGF inhibition in experimental tumour models. METHODS: A total of 85 metastatic CRC patients were stratified into cohorts that had either received chemotherapy alone (n = 39) or combined with bevacizumab (n = 46). Tumour CD177+ neutrophil infiltration was correlated to clinical outcome. The impact of neutrophil infiltration on anti-VEGF or anti-VEGF/anti-Ang2 therapy was studied in both xenograft and syngeneic tumour models by immunohistochemistry. RESULTS: The survival of bevacizumab-treated CRC patients in the presence of CD177+ infiltrates was significantly reduced compared to patients harbouring CD177- metastases. BI-880 treatment reduced the development of hypoxia associated with bevacizumab treatment and improved vascular normalisation in xenografts. Furthermore, neutrophil depletion or BI-880 treatment restored treatment sensitivity in a syngeneic tumour model of anti-VEGF resistance. CONCLUSIONS: Our findings implicate CD177 as a biomarker for bevacizumab and suggest VEGF/Ang2 inhibition as a strategy to overcome neutrophil associated resistance to anti-angiogenic treatment.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Isoantígenos/genética , Neovascularização Patológica/tratamento farmacológico , Receptores de Superfície Celular/genética , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas de Transporte Vesicular/genética , Idoso , Inibidores da Angiogênese/administração & dosagem , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Bevacizumab/administração & dosagem , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Proteínas Ligadas por GPI/genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica , Células-Tronco Neoplásicas/efeitos dos fármacos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neutrófilos/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Front Immunol ; 9: 1819, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30166983

RESUMO

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is routinely used to treat hematopoietic malignancies. The eradication of residual tumor cells during engraftment is mediated by donor cytotoxic T lymphocytes reactive to alloantigens. In a HLA-matched transplantation context, alloantigens are encoded by various polymorphic genes situated outside the HLA locus, also called minor histocompatibility antigens (MiHAs). Recently, MiHAs have been recognized as promising targets for post-transplantation T-cell immunotherapy as they have several appealing advantages over tumor-associated antigens (TAAs) and neoantigens, i.e., they are more abundant than TAAs, which potentially facilitates multiple targeting; and unlike neoantigens, they are encoded by germline polymorphisms, some of which are common and thus, suitable for off-the-shelf therapy. The genetic sources of MiHAs are nonsynonymous polymorphisms that cause differences between the recipient and donor proteomes and subsequently, the immunopeptidomes. Systematic description of the alloantigen landscape in HLA-matched transplantation is still lacking as previous studies focused only on a few immunogenic and common MiHAs. Here, we perform a thorough in silico analysis of the public genomic data to classify genetic polymorphisms that lead to MiHA formation and estimate the number of potentially available MiHA mismatches. Our findings suggest that a donor/recipient pair is expected to have at least several dozen mismatched strong MHC-binding SNP-associated peptides per HLA allele (116 ± 26 and 65 ± 15 for non-related pairs and siblings respectively in European populations as predicted by two independent algorithms). Over 70% of them are encoded by relatively frequent polymorphisms (minor allele frequency > 0.1) and thus, may be targetable by off-the-shelf therapeutics. We showed that the most appealing targets (probability of mismatch over 20%) reside in the asymmetric allele frequency region, which spans from 0.15 to 0.47 and corresponds to an order of several hundred (213 ± 47) possible targets per HLA allele that can be considered for immunogenicity validation. Overall, these findings demonstrate the significant potential of MiHAs as targets for T-cell immunotherapy and emphasize the need for the systematic discovery of novel MiHAs.


Assuntos
Genoma Humano , Genômica , Antígenos de Histocompatibilidade Menor/genética , Alelos , Biologia Computacional/métodos , Mapeamento de Epitopos , Genômica/métodos , Genótipo , Transplante de Células-Tronco Hematopoéticas , Teste de Histocompatibilidade , Humanos , Epitopos Imunodominantes/química , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Isoantígenos/genética , Isoantígenos/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , Peptídeos/química , Peptídeos/genética , Peptídeos/imunologia , Polimorfismo de Nucleotídeo Único , Imunologia de Transplantes , Transplante Homólogo
14.
JCI Insight ; 3(15)2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30089715

RESUMO

Adeno-associated viral vector-mediated (AAV-mediated) expression of allogeneic major histocompatibility complex class I (MHC class I) in recipient liver induces donor-specific tolerance in mouse skin transplant models in which a class I allele (H-2Kb or H-2Kd) is mismatched between donor and recipient. Tolerance can be induced in mice primed by prior rejection of a donor-strain skin graft, as well as in naive recipients. Allogeneic MHC class I may be recognized by recipient T cells as an intact molecule (direct recognition) or may be processed and presented as an allogeneic peptide in the context of self-MHC (indirect recognition). The relative contributions of direct and indirect allorecognition to tolerance induction in this setting are unknown. Using hepatocyte-specific AAV vectors encoding WT allogeneic MHC class I molecules, or class I molecules containing a point mutation (D227K) that impedes direct recognition of intact allogeneic MHC class I by CD8+ T cells without hampering the presentation of processed peptides derived from allogeneic MHC class I, we show here that tolerance induction depends upon recognition of intact MHC class I. Indirect recognition alone yielded a modest prolongation of subsequent skin graft survival, attributable to the generation of CD4+ Tregs, but it was not sufficient to induce tolerance.


Assuntos
Rejeição de Enxerto/imunologia , Hepatócitos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Tolerância Imunológica , Isoantígenos/imunologia , Aloenxertos/citologia , Aloenxertos/imunologia , Aloenxertos/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Dependovirus/genética , Modelos Animais de Doenças , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Vetores Genéticos/genética , Sobrevivência de Enxerto/imunologia , Hepatócitos/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Isoantígenos/genética , Isoantígenos/metabolismo , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Transplante de Fígado/efeitos adversos , Masculino , Camundongos , Camundongos Transgênicos , Mutação Puntual , Linfócitos T Reguladores/imunologia , Transdução Genética
15.
Biochem Biophys Res Commun ; 501(1): 313-319, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29738769

RESUMO

CD177 is considered to represent neutrophils. We analyzed mRNA expression level of CD177 and clinical follow-up survey of PDAC to estimate overall survival (OS) from Gene Expression Omnibus (GEO) dataset (GSE21501, containing samples from 102 PDAC patients) by R2 platform (http://r2.amc.nl). We also analyzed correlated genes of CD177 by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis to predict the potential relationship between neutrophils and prognosis of PDAC. We then performed hematoxylin and eosin (H&E) staining and immunohistochemical staining of surgical specimens to verify infiltration of neutrophils in PDAC tissues. After analyzing mRNA expression data and clinical follow-up survey provided in the GEO dataset (GSE21501, containing samples from 102 PDAC patients) and clinicopathological data of 23 PDAC patients, we demonstrated that CD177 was correlated with poor prognosis. The univariate Kaplan-Meier survival analysis revealed that OS was inversely associated with increased expression of CD177 (P = 0.012). Expression of phosphodiesterase (PDE)4D was positively related to CD177 in gene correlation analysis (R = 0.413, P < 0.001) by R2 platform. H&E staining and immunohistochemistry of CD177 in 23 PDAC surgical samples showed accumulation of neutrophils in the stroma and blood vessels around the cancer cells. In addition, immunohistochemical staining showed that CD177 was highly expressed in the stroma and blood vessels around tumor tissues of PDAC, which was similar to H&E staining. Expression of CD177 can be used to represent infiltration of neutrophils, which may have potential prognostic value in PDAC.


Assuntos
Carcinoma Ductal Pancreático/patologia , Neutrófilos/patologia , Neoplasias Pancreáticas/patologia , Idoso , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Imuno-Histoquímica , Isoantígenos/genética , Isoantígenos/metabolismo , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Regulação para Cima
16.
Eur Cytokine Netw ; 29(4): 146-152, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30698159

RESUMO

Severe congenital neutropenia (SCN) is a primary immunodeficiency disease in which a number of underlying gene defects are responsible for abnormalities in neutrophil development. The HCLS1-associated protein X1 (HAX1) mutation is associated with an autosomal-recessive form of SCN. Considering the potential of gene therapy approaches for the treatment of monogenic disorders, in this study we aimed to develop retroviral vectors expressing coding sequences (CDS) to be used for the removal of the genetic blockade in deficient hematopoietic cells. Following amplification of CDS with primers containing appropriate restriction sites, HAX1 CDS was cloned into an intermediate vector using TA-cloning. The sequence was transferred into a retroviral vector, followed by retroviral packaging in Plat-A cells. To show HAX1 protein expression, HEK293T cells were exposed to 10 multiplicity of infection (MOI) of retroviral particles and HAX1 expression was confirmed in these cells, using indirect intracellular flow cytometry. This vector was applied for in vitro transduction of hematopoietic stem cell with HAX1 mutation; after 11 days, cultured cells were analyzed for CD66acde and CD177 (neutrophil surface markers) expression. Increased neutrophil production in HAX1 viral vector-expressing hematopoietic cells was observed as compared to control vector transduced cells. Hence, according to the results, this type of therapy could be considered a potential treatment protocol for the disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neutropenia/congênito , Retroviridae/genética , Antígenos CD/genética , Moléculas de Adesão Celular/genética , Linhagem Celular , Síndrome Congênita de Insuficiência da Medula Óssea , Terapia Genética/métodos , Células HEK293 , Células-Tronco Hematopoéticas/metabolismo , Humanos , Isoantígenos/genética , Mutação/genética , Neutropenia/genética , Neutrófilos/metabolismo , Transdução Genética/métodos
17.
J Immunol Res ; 2017: 8415205, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29159187

RESUMO

Pig-to-human xenotransplantation offers a potential bridge to the growing disparity between patients with end-stage organ failure and graft availability. Early studies attempting to overcome cross-species barriers demonstrated robust humoral immune responses to discordant xenoantigens. Recent advances have led to highly efficient and targeted genomic editing, drastically altering the playing field towards rapid production of less immunogenic porcine tissues and even the discussion of human xenotransplantation trials. However, as these humoral immune barriers to cross-species transplantation are overcome with advanced transgenics, cellular immunity to these novel xenografts remains an outstanding issue. Therefore, understanding and optimizing immunomodulation will be paramount for successful clinical xenotransplantation. Costimulation blockade agents have been introduced in xenotransplantation research in 2000 with anti-CD154mAb. Most recently, prolonged survival has been achieved in solid organ (kidney xenograft survival > 400 days with anti-CD154mAb, heart xenograft survival > 900 days, and liver xenograft survival 29 days with anti-CD40mAb) and islet xenotransplantation (>600 days with anti-CD154mAb) with the use of these potent experimental agents. As the development of novel genetic modifications and costimulation blocking agents converges, we review their impact thus far on preclinical xenotransplantation and the potential for future application.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Rejeição de Enxerto/imunologia , Imunoterapia/métodos , Transplante das Ilhotas Pancreáticas , Transplante de Órgãos , Animais , Ligante de CD40/imunologia , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Terapia Genética , Rejeição de Enxerto/prevenção & controle , Humanos , Imunidade Celular , Imunomodulação , Isoantígenos/genética , Isoantígenos/imunologia , Suínos , Transplante Heterólogo
18.
J Exp Med ; 214(7): 2089-2101, 2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-28559244

RESUMO

CD177 presents antigens in allo- and autoimmune diseases on the neutrophil surface. Individuals can be either CD177-deficient or harbor distinct CD177neg and CD177pos neutrophil subsets. We studied mechanisms controlling subset-restricted CD177 expression in bimodal individuals. CD177pos, but not CD177neg neutrophils, produced CD177 protein and mRNA. Haplotype analysis indicated a unique monoallelic CD177 expression pattern, where the offspring stably transcribed either the maternal or paternal allele. Hematopoietic stem cells expressed both CD177 alleles and silenced one copy during neutrophil differentiation. ChIP and reporter assays in HeLa cells with monoallelic CD177 expression showed that methylation reduced reporter activity, whereas demethylation caused biallelic CD177 expression. HeLa cell transfection with c-Jun and c-Fos increased CD177 mRNA. Importantly, CD177pos human neutrophils, but not CD177neg neutrophils, showed a euchromatic CD177 promoter, unmethylated CpGs, and c-Jun and c-Fos binding. We describe epigenetic mechanisms explaining the two distinct CD177 neutrophil subsets and a novel monoallelic CD177 expression pattern that does not follow classical random monoallelic expression or imprinting.


Assuntos
Perfilação da Expressão Gênica/métodos , Inativação Gênica , Isoantígenos/genética , Neutrófilos/metabolismo , Receptores de Superfície Celular/genética , Alelos , Sequência de Bases , Diferenciação Celular/genética , Ilhas de CpG/genética , Metilação de DNA , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Células HeLa , Células-Tronco Hematopoéticas/metabolismo , Humanos , Immunoblotting , Isoantígenos/metabolismo , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
19.
Childs Nerv Syst ; 33(3): 437-446, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28236065

RESUMO

PURPOSE: Diffuse intrinsic pontine glioma (DIPG) is universally fatal without proven therapy other than radiation therapy for palliation. Representative animal models will play an essential role in the preclinical stage of future therapy development. To address the shortage of representative models, we created a novel infiltrative brainstem glioma model in rats based on glioblastoma spheroids. METHODS: Cells dissociated from glioblastoma spheroids grown from surgical specimens were implanted into the brainstem of NIH nude rats. Animals were serially assessed clinically and radiographically with magnetic resonance imaging (MRI). Tumors were further characterized using histology, immunohistochemistry, and cytogenetics. RESULTS: Tumor generation was successful in all animals receiving glioblastoma spheroid cells. The rats survived 17-25 weeks before severe symptoms developed. The tumors showed as diffuse hyperintense lesions on T2-weighted images. Histologically, they demonstrated cellular heterogeneity, and infiltrative and invasive features, with cells engorging vascular structures. The tumors were shown to comprise immature human origin glial tumor cells, with human epidermal growth factor receptor (EGFR) gene amplification and gain. CONCLUSIONS: This study showed that cells from glioblastoma spheroids produced infiltrative gliomas in rat brainstem. The rat brainstem gliomas are radiographically and histologically accurate compared to DIPG. These tumors develop over several months that would allow sequential clinical and radiographic assessments of therapeutic interventions. This study demonstrated in principle the feasibility of developing patient-specific animal models based on putative cancer stem cells from biopsy or resection samples.


Assuntos
Neoplasias do Tronco Encefálico/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/patologia , Infiltração de Neutrófilos/fisiologia , Animais , Neoplasias do Tronco Encefálico/diagnóstico por imagem , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Glioblastoma/diagnóstico por imagem , Humanos , Isoantígenos/genética , Isoantígenos/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Transplante de Neoplasias , Ratos , Ratos Nus , Esferoides Celulares , Células Tumorais Cultivadas
20.
J Hepatol ; 66(4): 765-777, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27914923

RESUMO

BACKGROUND & AIMS: Induction of donor-specific immune tolerance is a good alternative to chronic life-long immunosuppression for transplant patients. Donor major histocompatibility complex (MHC) molecules represent the main targets of the allogeneic immune response of transplant recipients. Liver targeted gene transfer with viral vectors induces tolerance toward the encoded antigen. The aim of this work was to determine whether alloantigen gene transfer to hepatocytes induces tolerance and promotes graft acceptance. METHODS: C57BL/6 (H-2b) mice were treated with adeno-associated viral (AAV) vector targeting the expression of the MHC class I molecule H-2Kd to hepatocytes, before transplantation with fully allogeneic pancreatic islet from BALB/c mice (H-2d). RESULTS: AAV H-2Kd treated mice were tolerant to the alloantigen, as demonstrated by its long-term expression by the hepatocytes, even after a highly immunogenic challenge with an adenoviral vector. After chemical induction of diabetes, the AAV treated mice had significantly delayed rejection of fully allogeneic pancreatic islet grafts, with more than 40% of recipients tolerant (>100days). AAV-mediated expression of H-2Kd in the liver induced the local expansion of CD8+ T lymphocytes with allo-specific suppressive properties. The adoptive transfer of these liver-generated CD8+ Tregs into naive diabetic mice promoted the long-term survival of allogeneic pancreatic islet grafts. CONCLUSION: AAV-mediated long-term expression of a single MHC class I molecule in the liver induces the generation of a subset of allo-specific CD8+ Treg cells, which promote tolerance toward fully allogeneic graft. Liver gene transfer represents a promising strategy for in vivo induction of donor-specific tolerance. LAY SUMMARY: The liver has a special immune system, biased toward tolerance. In this study, we investigated the possibility of harnessing this property of the liver to induce tolerance to an allogeneic transplantation. We demonstrate for the first time that the in vivo gene transfer of an allogeneic antigen with an adeno-associated viral vector to mouse hepatocytes induces the expansion of a population of CD8+ regulatory T lymphocytes. These Tregs are then instrumental in preventing the rejection of allogeneic pancreatic islets transplanted in these animals. Allogeneic transplantation is the main treatment for the end-stage diseases of a number of organs. Life-long immunosuppressive treatments are still required to limit graft rejection, and these treatments exhibit serious side effects. Our present findings open a new avenue for promoting allo-specific tolerance via in vivo induction of CD8+ Treg expansion.


Assuntos
Hepatócitos/imunologia , Terapia de Imunossupressão/métodos , Transplante das Ilhotas Pancreáticas/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Dependovirus , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Sobrevivência de Enxerto/imunologia , Antígenos H-2/genética , Isoantígenos/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Parvovirinae/genética , Doadores de Tecidos , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA