Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 654
Filtrar
1.
J Immunother Cancer ; 10(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35580930

RESUMO

BACKGROUND: Melanoma is the most lethal form of skin cancer in humans. Conventional therapies have limited efficacy, and overall response is still unsatisfactory considering that immune checkpoint inhibitors induce lasting clinical responses only in a low percentage of patients. This has prompted us to develop a vaccination strategy employing the tumor antigen chondroitin sulfate proteoglycan (CSPG)4 as a target. METHODS: To overcome the host's unresponsiveness to the self-antigen CSPG4, we have taken advantage of the conservation of CSPG4 sequence through phylogenetic evolution, so we have used a vaccine, based on a chimeric DNA molecule encompassing both human (Hu) and dog (Do) portions of CSPG4 (HuDo-CSPG4). We have tested its safety and immunogenicity (primary objectives), along with its therapeutic efficacy (secondary outcome), in a prospective, non-randomized, veterinary clinical trial enrolling 80 client-owned dogs with surgically resected, CSPG4-positive, stage II-IV oral melanoma. RESULTS: Vaccinated dogs developed anti-Do-CSPG4 and Hu-CSPG4 immune response. Interestingly, the antibody titer in vaccinated dogs was significantly associated with the overall survival. Our data suggest that there may be a contribution of the HuDo-CSPG4 vaccination to the improvement of survival of vaccinated dogs as compared with controls treated with conventional therapies alone. CONCLUSIONS: HuDo-CSPG4 adjuvant vaccination was safe and immunogenic in dogs with oral melanoma, with potential beneficial effects on the course of the disease. Thanks to the power of naturally occurring canine tumors as predictive models for cancer immunotherapy response, these data may represent a basis for the translation of this approach to the treatment of human patients with CSPG4-positive melanoma subtypes.


Assuntos
Vacinas Anticâncer , Proteoglicanas de Sulfatos de Condroitina , Doenças do Cão , Melanoma , Proteínas de Membrana , Neoplasias Bucais , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Proteoglicanas de Sulfatos de Condroitina/imunologia , Doenças do Cão/tratamento farmacológico , Doenças do Cão/imunologia , Cães , Melanoma/tratamento farmacológico , Melanoma/veterinária , Proteínas de Membrana/imunologia , Mimetismo Molecular/imunologia , Neoplasias Bucais/terapia , Neoplasias Bucais/veterinária , Filogenia , Estudos Prospectivos
2.
J Autoimmun ; 125: 102741, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34717185

RESUMO

The COVID-19 pandemic is still raging across the world and vaccination is expected to lead us out of this pandemic. Although the efficacy of the vaccines is beyond doubt, safety still remains a concern. We report a case of a 65-year-old woman who experienced acute severe autoimmune hepatitis two weeks after receiving the first dose of Moderna-COVID-19 vaccine. Serum immunoglobulin G was elevated and antinuclear antibody was positive (1:100, speckled pattern). Liver histology showed a marked expansion of the portal tracts, severe interface hepatitis and multiple confluent foci of lobular necrosis. She started treatment with prednisolone, with a favorable clinical and analytical evolution. Some recent reports have been suggested that COVID-19 vaccination can lead to the development of autoimmune diseases. It is speculated that the vaccine can disturb self-tolerance and trigger autoimmune responses through cross-reactivity with host cells. Therefore, healthcare providers must remain vigilant during mass COVID-19 vaccination.


Assuntos
Vacina BNT162/efeitos adversos , COVID-19/prevenção & controle , Hepatite Autoimune/etiologia , Icterícia/etiologia , Vacinação/efeitos adversos , Anticorpos Antinucleares/sangue , Vacina BNT162/imunologia , Bilirrubina/sangue , Feminino , Fibrose/patologia , Hepatite Autoimune/imunologia , Humanos , Icterícia/diagnóstico , Fígado/enzimologia , Pessoa de Meia-Idade , Mimetismo Molecular/imunologia , Prednisolona/uso terapêutico , SARS-CoV-2/imunologia
3.
Front Immunol ; 12: 715893, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34594330

RESUMO

Allogeneic stem cell transplantation (alloSCT) is a curative therapy for hematopoietic malignancies. The therapeutic effect relies on donor T cells and NK cells to recognize and eliminate malignant cells, known as the graft-versus-leukemia (GVL) effect. However, off target immune pathology, known as graft-versus-host disease (GVHD) remains a major complication of alloSCT that limits the broad application of this therapy. The presentation of recipient-origin alloantigen to donor T cells is the primary process initiating GVHD and GVL. Therefore, the understanding of spatial and temporal characteristics of alloantigen presentation is pivotal to attempts to separate beneficial GVL effects from detrimental GVHD. In this review, we discuss mouse models and the tools therein, that permit the quantification of alloantigen presentation after alloSCT.


Assuntos
Apresentação de Antígeno/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Imunologia de Transplantes , Animais , Doença Enxerto-Hospedeiro/etiologia , Efeito Enxerto vs Leucemia/imunologia , Antígenos H-2/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Isoantígenos/imunologia , Camundongos , Antígenos de Histocompatibilidade Menor/imunologia , Mimetismo Molecular/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
4.
J Autoimmun ; 125: 102738, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34653776

RESUMO

Autoimmune diseases, including autoimmune endocrine diseases (AIED), are thought to develop following environmental exposure in patients with genetic predisposition. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and vaccines against it could represent new environmental triggers for AIED. We report a patient, with history of vitiligo vulgaris and 8 years of type 2 diabetes, who came to our institution because of fever, weight loss, asthenia and thyrotoxicosis occurred 4 weeks later the administration of BNT162B2 (Pfizer-BioNTech) SARS-CoV-2 vaccine. Clinical, biochemical and instrumental work-up demonstrated Graves' disease and autoimmune diabetes mellitus. The occurrence of these disorders could be explained through different mechanism such as autoimmune/inflammatory syndrome induced by adjuvants (ASIA syndrome), mRNA "self-adjuvant" effect, molecular mimicry between human and viral proteins and immune disruption from external stimuli. However further studies are needed to better understand the underlying pathogenesis of AIED following SARS-CoV-2 vaccine.


Assuntos
Vacina BNT162/efeitos adversos , COVID-19/prevenção & controle , Diabetes Mellitus Tipo 1/etiologia , Doença de Graves/etiologia , Mimetismo Molecular/imunologia , Adjuvantes Imunológicos/efeitos adversos , Autoanticorpos/sangue , Vacina BNT162/imunologia , Peptídeo C/sangue , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hemoglobinas Glicadas/análise , Controle Glicêmico , Humanos , Masculino , Pessoa de Meia-Idade , SARS-CoV-2/imunologia , Tireotoxicose/patologia , Vitiligo/patologia
6.
Mol Immunol ; 137: 105-113, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34242919

RESUMO

Underlying mechanisms of multi-organ manifestations and exacerbated inflammation in COVID-19 are yet to be delineated. The hypothesis of SARS-CoV-2 triggering autoimmunity is gaining attention and, in the present study, we have identified 28 human proteins harbouring regions homologous to SARS-CoV-2 peptides that could possibly be acting as autoantigens in COVID-19 patients displaying autoimmune conditions. Interestingly, these conserved regions are amongst the experimentally validated B cell epitopes of SARS-CoV-2 proteins. The reported human proteins have demonstrated presence of autoantibodies against them in typical autoimmune conditions which may explain the frequent occurrence of autoimmune conditions following SARS-CoV-2 infection. Moreover, the proposed autoantigens' widespread tissue distribution is suggestive of their involvement in multi-organ manifestations via molecular mimicry. We opine that our report may aid in directing subsequent necessary antigen-specific studies, results of which would be of long-term relevance in management of extrapulmonary symptoms of COVID-19.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/complicações , COVID-19/etiologia , Epitopos de Linfócito B/imunologia , SARS-CoV-2/imunologia , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/virologia , Autoimunidade/imunologia , COVID-19/imunologia , COVID-19/patologia , Humanos , Mimetismo Molecular/imunologia
8.
Circulation ; 144(6): 471-484, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34281357

RESUMO

Myocarditis has been recognized as a rare complication of coronavirus disease 2019 (COVID-19) mRNA vaccinations, especially in young adult and adolescent males. According to the US Centers for Disease Control and Prevention, myocarditis/pericarditis rates are ≈12.6 cases per million doses of second-dose mRNA vaccine among individuals 12 to 39 years of age. In reported cases, patients with myocarditis invariably presented with chest pain, usually 2 to 3 days after a second dose of mRNA vaccination, and had elevated cardiac troponin levels. ECG was abnormal with ST elevations in most, and cardiac MRI was suggestive of myocarditis in all tested patients. There was no evidence of acute COVID-19 or other viral infections. In 1 case, a cardiomyopathy gene panel was negative, but autoantibody levels against certain self-antigens and frequency of natural killer cells were increased. Although the mechanisms for development of myocarditis are not clear, molecular mimicry between the spike protein of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and self-antigens, trigger of preexisting dysregulated immune pathways in certain individuals, immune response to mRNA, and activation of immunologic pathways, and dysregulated cytokine expression have been proposed. The reasons for male predominance in myocarditis cases are unknown, but possible explanations relate to sex hormone differences in immune response and myocarditis, and also underdiagnosis of cardiac disease in women. Almost all patients had resolution of symptoms and signs and improvement in diagnostic markers and imaging with or without treatment. Despite rare cases of myocarditis, the benefit-risk assessment for COVID-19 vaccination shows a favorable balance for all age and sex groups; therefore, COVID-19 vaccination is recommended for everyone ≥12 years of age.


Assuntos
Autoantígenos/imunologia , Vacinas contra COVID-19/efeitos adversos , COVID-19/prevenção & controle , Miocardite/induzido quimicamente , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacina de mRNA-1273 contra 2019-nCoV , Biomarcadores , COVID-19/epidemiologia , COVID-19/imunologia , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/uso terapêutico , Feminino , Humanos , Masculino , Mimetismo Molecular/imunologia , Miocardite/imunologia , Fatores Sexuais
9.
Cancer Immunol Res ; 9(8): 981-993, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34103348

RESUMO

Molecular mimicry is one of the leading mechanisms by which infectious agents can induce autoimmunity. Whether a similar mechanism triggers an antitumor immune response is unexplored, and the role of antiviral T cells infiltrating the tumor has remained anecdotal. To address these questions, we first developed a bioinformatic tool to identify tumor peptides with high similarity to viral epitopes. Using peptides identified by this tool, we demonstrated that, in mice, preexisting immunity toward specific viral epitopes enhanced the efficacy of cancer immunotherapy via molecular mimicry in different settings. To understand whether this mechanism could partly explain immunotherapy responsiveness in humans, we analyzed a cohort of patients with melanoma undergoing anti-PD1 treatment who had a high IgG titer for cytomegalovirus (CMV). In this cohort of patients, we showed that high levels of CMV-specific antibodies were associated with prolonged progression-free survival and found that, in some cases, peripheral blood mononuclear cells (PBMC) could cross-react with both melanoma and CMV homologous peptides. Finally, T-cell receptor sequencing revealed expansion of the same CD8+ T-cell clones when PBMCs were expanded with tumor or homologous viral peptides. In conclusion, we have demonstrated that preexisting immunity and molecular mimicry could influence the response to immunotherapies. In addition, we have developed a free online tool that can identify tumor antigens and neoantigens highly similar to pathogen antigens to exploit molecular mimicry and cross-reactive T cells in cancer vaccine development.


Assuntos
Imunidade/imunologia , Imunoterapia/métodos , Melanoma/imunologia , Mimetismo Molecular/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos
10.
J Autoimmun ; 120: 102634, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33892348

RESUMO

Atopic dermatitis (AD) is an itchy, non-contagious relapsing and chronic inflammatory skin disease that usually develops in early childhood. This pathology is associated with food allergy, allergic asthma, allergic rhinitis and anaphylaxis which may persist in adulthood. The underlying mechanisms of AD (endotypes) are just beginning to be discovered and show a complex interaction of various pathways including skin barrier function and immune deviation. Immune reactions to self-proteins (autoantigens) of the skin have been identified in patients with inflammatory skin diseases, such as chronic spontaneous urticaria, connective tissue disease, pemphigus vulgaris and bullous pemphigoid. IgE antibodies and T cells directed against epitopes of the skin were observed in adult patients with severe and chronic AD as well. This was associated with disease severity and suggests a progression from allergic inflammation to severe autoimmune processes against the skin. IgE-mediated autoimmunity and self-reactive T cells might accelerate the ongoing skin inflammation or might contribute to the relapsing course of the disease. However, to date, the exact mechanisms of IgE-mediated autoimmunity and self-reactive T cells in the pathophysiology of AD are still unclear. The aim of this review is to evaluate the development of (autoreactive) T cells and their response to (auto)antigens, as well as the role of the peripheral tolerance in autoimmunity in the pathophysiology of AD, including the unmet needs and gaps.


Assuntos
Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Autoimunidade , Dermatite Atópica/etiologia , Dermatite Atópica/metabolismo , Suscetibilidade a Doenças/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/terapia , Reações Cruzadas/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Dermatite Atópica/diagnóstico , Dermatite Atópica/terapia , Gerenciamento Clínico , Humanos , Imunoglobulina E/imunologia , Memória Imunológica , Mimetismo Molecular/imunologia , Fatores de Risco , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
11.
J R Soc Interface ; 18(175): 20200689, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33622145

RESUMO

Mimicry is exhibited in multiple scales, ranging from molecular, to organismal, and then to human society. 'Batesian'-type mimicry entails a conflict of interest between sender and receiver, reflected in a deceptive mimic signal. 'Müllerian'-type mimicry occurs when there is perfect common interest between sender and receiver in a particular type of encounter, manifested by an honest co-mimic signal. Using a signalling games approach, simulations show that invasion by Batesian mimics will make Müllerian mimicry unstable, in a coevolutionary chase. We use these results to better understand the deceptive strategies of SARS-CoV-2 and their key role in the COVID-19 pandemic. At the biomolecular level, we explain how cellularization promotes Müllerian molecular mimicry, and discourages Batesian molecular mimicry. A wide range of processes analogous to cellularization are presented; these might represent a manner of reducing oscillatory instabilities. Lastly, we identify examples of mimicry in human society that might be addressed using a signalling game approach.


Assuntos
Modelos Imunológicos , Mimetismo Molecular/imunologia , Pandemias , SARS-CoV-2/imunologia , COVID-19/epidemiologia , COVID-19/imunologia , Humanos
12.
Methods Mol Biol ; 2273: 131-138, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33604849

RESUMO

The current coronavirus disease-19 (COVID-19) pandemic, caused by "severe acute respiratory syndrome coronavirus 2" (SARS-CoV-2), underscores the threat posed by newly emerging viruses. The understanding of the mechanisms driving early infection events, that are crucial for the exponential spread of the disease, is mandatory and can be significantly implemented generating 3D in vitro models as experimental platforms to investigate the infection substrates and how the virus invades and ravages the tissues.We here describe a protocol for the creation of a synthetic hydrogel-based 3D culture system that mimics in vitro the complex architectures and mechanical cues distinctive of the upper airway epithelia. We then expose the in vitro generated 3D nasal and tracheal epithelia to gold nanoparticles (AuNPs) that display the typical shape and size distinctive of SARS-CoV-2 and of the majority of Coronaviridae presently known.The infection platform here described provides an efficient and highly physiological in vitro model that reproduces the host-pathogen early interactions, using virus-mimicking nanoparticles, and offers a flexible tool to study virus entry into the cell. At the same time, it reduces the risk of accidental infection/spillovers for researchers, which represents a crucial aspect when dealing with a virus that is highly contagious, virulent, and even deadly.


Assuntos
COVID-19/metabolismo , COVID-19/virologia , Técnicas de Cultura de Células/métodos , Células Epiteliais/citologia , Nanopartículas/metabolismo , Mucosa Respiratória/citologia , Animais , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/virologia , Ouro , Humanos , Nanopartículas Metálicas/química , Mimetismo Molecular/imunologia , Nariz/virologia , Mucosa Respiratória/virologia , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Traqueia/virologia , Células Vero , Internalização do Vírus
13.
Clin Immunol ; 226: 108694, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33610741

RESUMO

The pandemic of Coronavirus disease 2019 (COVID-19), caused by a new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spotlighted the link between viral infection and autoimmunity. In this review, we focus on coronavirus-induced autoimmunity based on evidence from experimental animal models, SARS-CoV infection with in vitro studies of molecular mimicry and COVID-19 with several clinical reports of autoimmune manifestations of this disease. Further studies will be needed to better characterize the role of SARS-CoV-2 in the development of autoimmunity.


Assuntos
Autoimunidade , COVID-19/imunologia , SARS-CoV-2/imunologia , Animais , Modelos Animais de Doenças , Encefalomielite/imunologia , Encefalomielite/virologia , Humanos , Mimetismo Molecular/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Doenças Retinianas/imunologia , Doenças Retinianas/virologia
14.
mBio ; 12(1)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33468699

RESUMO

Surface expression of the common vertebrate sialic acid (Sia) N-acetylneuraminic acid (Neu5Ac) by commensal and pathogenic microbes appears structurally to represent "molecular mimicry" of host sialoglycans, facilitating multiple mechanisms of host immune evasion. In contrast, ketodeoxynonulosonic acid (Kdn) is a more ancestral Sia also present in prokaryotic glycoconjugates that are structurally quite distinct from vertebrate sialoglycans. We detected human antibodies against Kdn-terminated glycans, and sialoglycan microarray studies found these anti-Kdn antibodies to be directed against Kdn-sialoglycans structurally similar to those on human cell surface Neu5Ac-sialoglycans. Anti-Kdn-glycan antibodies appear during infancy in a pattern similar to those generated following incorporation of the nonhuman Sia N-glycolylneuraminic acid (Neu5Gc) onto the surface of nontypeable Haemophilus influenzae (NTHi), a human commensal and opportunistic pathogen. NTHi grown in the presence of free Kdn took up and incorporated the Sia into its lipooligosaccharide (LOS). Surface display of the Kdn within NTHi LOS blunted several virulence attributes of the pathogen, including Neu5Ac-mediated resistance to complement and whole blood killing, complement C3 deposition, IgM binding, and engagement of Siglec-9. Upper airway administration of Kdn reduced NTHi infection in human-like Cmah null (Neu5Gc-deficient) mice that express a Neu5Ac-rich sialome. We propose a mechanism for the induction of anti-Kdn antibodies in humans, suggesting that Kdn could be a natural and/or therapeutic "Trojan horse" that impairs colonization and virulence phenotypes of free Neu5Ac-assimilating human pathogens.IMPORTANCE All cells in vertebrates are coated with a dense array of glycans often capped with sugars called sialic acids. Sialic acids have many functions, including serving as a signal for recognition of "self" cells by the immune system, thereby guiding an appropriate immune response against foreign "nonself" and/or damaged cells. Several pathogenic bacteria have evolved mechanisms to cloak themselves with sialic acids and evade immune responses. Here we explore a type of sialic acid called "Kdn" (ketodeoxynonulosonic acid) that has not received much attention in the past and compare and contrast how it interacts with the immune system. Our results show potential for the use of Kdn as a natural intervention against pathogenic bacteria that take up and coat themselves with external sialic acid from the environment.


Assuntos
Antígenos CD/imunologia , Infecções por Haemophilus/imunologia , Haemophilus influenzae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Ácido N-Acetilneuramínico/química , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Ácidos Siálicos/imunologia , Animais , Anticorpos/química , Anticorpos/metabolismo , Antígenos CD/metabolismo , Transporte Biológico , Complemento C3/imunologia , Complemento C3/metabolismo , Feminino , Glicoconjugados/química , Glicoconjugados/imunologia , Infecções por Haemophilus/genética , Infecções por Haemophilus/microbiologia , Haemophilus influenzae/química , Interações Hospedeiro-Patógeno/genética , Humanos , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mimetismo Molecular/genética , Mimetismo Molecular/imunologia , Ácido N-Acetilneuramínico/imunologia , Ligação Proteica , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo , Ácidos Siálicos/química , Açúcares Ácidos/química , Açúcares Ácidos/imunologia
15.
Arthritis Rheumatol ; 73(2): 232-243, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33124780

RESUMO

OBJECTIVE: Changes in gut microbiota have been linked to systemic lupus erythematosus (SLE), but knowledge is limited. Our study aimed to provide an in-depth understanding of the contribution of gut microbiota to the immunopathogenesis of SLE. METHODS: Fecal metagenomes from 117 patients with untreated SLE and 52 SLE patients posttreatment were aligned with 115 matched healthy controls and analyzed by whole-genome profiling. For comparison, we assessed the fecal metagenome of MRL/lpr mice. The oral microbiota origin of the gut species that existed in SLE patients was documented by single-nucleotide polymorphism-based strain-level analyses. Functional validation assays were performed to demonstrate the molecular mimicry of newly found microbial peptides. RESULTS: Gut microbiota from individuals with SLE displayed significant differences in microbial composition and function compared to healthy controls. Certain species, including the Clostridium species ATCC BAA-442 as well as Atopobium rimae, Shuttleworthia satelles, Actinomyces massiliensis, Bacteroides fragilis, and Clostridium leptum, were enriched in SLE gut microbiota and reduced after treatment. Enhanced lipopolysaccharide biosynthesis aligned with reduced branched chain amino acid biosynthesis was observed in the gut of SLE patients. The findings in mice were consistent with our findings in human subjects. Interestingly, some species with an oral microbiota origin were enriched in the gut of SLE patients. Functional validation assays demonstrated the proinflammatory capacities of some microbial peptides derived from SLE-enriched species. CONCLUSION: This study provides detailed information on the microbiota of untreated patients with SLE, including their functional signatures, similarities with murine counterparts, oral origin, and the definition of autoantigen-mimicking peptides. Our data demonstrate that microbiome-altering approaches may offer valuable adjuvant therapies in SLE.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Microbioma Gastrointestinal/imunologia , Lúpus Eritematoso Sistêmico/microbiologia , Mimetismo Molecular/imunologia , Actinobacteria , Actinomyces , Adulto , Aminoácidos de Cadeia Ramificada/biossíntese , Animais , Antirreumáticos/uso terapêutico , Bacteroides fragilis , Estudos de Casos e Controles , Clostridiales , Clostridium , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/genética , Humanos , Lipopolissacarídeos/biossíntese , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Masculino , Metagenômica , Camundongos , Camundongos Endogâmicos MRL lpr , Boca/microbiologia , Polimorfismo de Nucleotídeo Único , Adulto Jovem
16.
Gut ; 70(10): 1904-1913, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32883872

RESUMO

OBJECTIVE: A comprehensive analysis of the immune landscape of pancreatic neuroendocrine tumours (PanNETs) was performed according to clinicopathological parameters and previously defined molecular subtypes to identify potential therapeutic vulnerabilities in this disease. DESIGN: Differential expression analysis of 600 immune-related genes was performed on 207 PanNET samples, comprising a training cohort (n=72) and two validation cohorts (n=135) from multiple transcriptome profiling platforms. Different immune-related and subtype-related phenotypes, cell types and pathways were investigated using different in silico methods and were further validated using spatial multiplex immunofluorescence. RESULTS: The study identified an immune signature of 132 genes segregating PanNETs (n=207) according to four previously defined molecular subtypes: metastasis-like primary (MLP)-1 and MLP-2, insulinoma-like and intermediate. The MLP-1 subtype (26%-31% samples across three cohorts) was strongly associated with elevated levels of immune-related genes, poor prognosis and a cascade of tumour evolutionary events: larger hypoxic and necroptotic tumours leading to increased damage-associated molecular patterns (viral mimicry), stimulator of interferon gene pathway, T cell-inflamed genes, immune checkpoint targets, and T cell-mediated and M1 macrophage-mediated immune escape mechanisms. Multiplex spatial profiling validated significantly increased macrophages in the MLP-1 subtype. CONCLUSION: This study provides novel data on the immune microenvironment of PanNETs and identifies MLP-1 subtype as an immune-high phenotype featuring a broad and robust activation of immune-related genes. This study, with further refinement, paves the way for future precision immunotherapy studies in PanNETs to potentially select a subset of MLP-1 patients who may be more likely to respond.


Assuntos
Genes Neoplásicos/imunologia , Mimetismo Molecular/imunologia , Tumores Neuroendócrinos/imunologia , Neoplasias Pancreáticas/imunologia , Microambiente Tumoral/imunologia , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Gradação de Tumores , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Fenótipo , Prognóstico , Carga Tumoral
17.
Science ; 371(6525)2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33214287

RESUMO

Neutralizing antibodies elicited by HIV-1 coevolve with viral envelope proteins (Env) in distinctive patterns, in some cases acquiring substantial breadth. We report that primary HIV-1 envelope proteins-when expressed by simian-human immunodeficiency viruses in rhesus macaques-elicited patterns of Env-antibody coevolution very similar to those in humans, including conserved immunogenetic, structural, and chemical solutions to epitope recognition and precise Env-amino acid substitutions, insertions, and deletions leading to virus persistence. The structure of one rhesus antibody, capable of neutralizing 49% of a 208-strain panel, revealed a V2 apex mode of recognition like that of human broadly neutralizing antibodies (bNAbs) PGT145 and PCT64-35S. Another rhesus antibody bound the CD4 binding site by CD4 mimicry, mirroring human bNAbs 8ANC131, CH235, and VRC01. Virus-antibody coevolution in macaques can thus recapitulate developmental features of human bNAbs, thereby guiding HIV-1 immunogen design.


Assuntos
Coevolução Biológica/imunologia , Anticorpos Amplamente Neutralizantes , Anticorpos Anti-HIV , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Sítios de Ligação , Anticorpos Amplamente Neutralizantes/química , Anticorpos Amplamente Neutralizantes/genética , Anticorpos Amplamente Neutralizantes/imunologia , Antígenos CD4/imunologia , Microscopia Crioeletrônica , Epitopos/imunologia , Anticorpos Anti-HIV/química , Anticorpos Anti-HIV/genética , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Humanos , Macaca mulatta , Mimetismo Molecular/imunologia , Vírus da Imunodeficiência Símia/genética , Replicação Viral
18.
Nature ; 588(7836): 169-173, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33087935

RESUMO

Cancer therapies that target epigenetic repressors can mediate their effects by activating retroelements within the human genome. Retroelement transcripts can form double-stranded RNA (dsRNA) that activates the MDA5 pattern recognition receptor1-6. This state of viral mimicry leads to loss of cancer cell fitness and stimulates innate and adaptive immune responses7,8. However, the clinical efficacy of epigenetic therapies has been limited. To find targets that would synergize with the viral mimicry response, we sought to identify the immunogenic retroelements that are activated by epigenetic therapies. Here we show that intronic and intergenic SINE elements, specifically inverted-repeat Alus, are the major source of drug-induced immunogenic dsRNA. These inverted-repeat Alus are frequently located downstream of 'orphan' CpG islands9. In mammals, the ADAR1 enzyme targets and destabilizes inverted-repeat Alu dsRNA10, which prevents activation of the MDA5 receptor11. We found that ADAR1 establishes a negative-feedback loop, restricting the viral mimicry response to epigenetic therapy. Depletion of ADAR1 in patient-derived cancer cells potentiates the efficacy of epigenetic therapy, restraining tumour growth and reducing cancer initiation. Therefore, epigenetic therapies trigger viral mimicry by inducing a subset of inverted-repeats Alus, leading to an ADAR1 dependency. Our findings suggest that combining epigenetic therapies with ADAR1 inhibitors represents a promising strategy for cancer treatment.


Assuntos
Adenosina Desaminase/metabolismo , Elementos Alu/efeitos dos fármacos , Elementos Alu/genética , Decitabina/farmacologia , Decitabina/uso terapêutico , Epigênese Genética/efeitos dos fármacos , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Adenosina Desaminase/deficiência , Elementos Alu/imunologia , Animais , Linhagem Celular Tumoral , Ilhas de CpG/efeitos dos fármacos , Ilhas de CpG/genética , DNA Intergênico/efeitos dos fármacos , DNA Intergênico/genética , DNA Intergênico/imunologia , DNA-Citosina Metilases/antagonistas & inibidores , Retroalimentação Fisiológica , Humanos , Imunidade Inata/efeitos dos fármacos , Helicase IFIH1 Induzida por Interferon/metabolismo , Íntrons/efeitos dos fármacos , Íntrons/genética , Íntrons/imunologia , Sequências Repetidas Invertidas/efeitos dos fármacos , Sequências Repetidas Invertidas/genética , Sequências Repetidas Invertidas/imunologia , Masculino , Camundongos , Mimetismo Molecular/efeitos dos fármacos , Mimetismo Molecular/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , RNA de Cadeia Dupla/efeitos dos fármacos , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Vírus/efeitos dos fármacos , Vírus/imunologia
19.
Nat Commun ; 11(1): 5066, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033255

RESUMO

The inducible co-stimulator (ICOS) is a member of the CD28/B7 superfamily, and delivers a positive co-stimulatory signal to activated T cells upon binding to its ligand (ICOS-L). Dysregulation of this pathway has been implicated in autoimmune diseases and cancer, and is currently under clinical investigation as an immune checkpoint blockade. Here, we describe the molecular interactions of the ICOS/ICOS-L immune complex at 3.3 Å resolution. A central FDPPPF motif and residues within the CC' loop of ICOS are responsible for the specificity of the interaction with ICOS-L, with a distinct receptor binding orientation in comparison to other family members. Furthermore, our structure and binding data reveal that the ICOS N110 N-linked glycan participates in ICOS-L binding. In addition, we report crystal structures of ICOS and ICOS-L in complex with monoclonal antibodies under clinical evaluation in immunotherapy. Strikingly, antibody paratopes closely mimic receptor-ligand binding core interactions, in addition to contacting peripheral residues to confer high binding affinities. Our results uncover key molecular interactions of an immune complex central to human adaptive immunity and have direct implications for the ongoing development of therapeutic interventions targeting immune checkpoint receptors.


Assuntos
Anticorpos/uso terapêutico , Complexo Antígeno-Anticorpo/química , Ligante Coestimulador de Linfócitos T Induzíveis/química , Proteína Coestimuladora de Linfócitos T Induzíveis/química , Mimetismo Molecular/imunologia , Sequência de Aminoácidos , Complexo Antígeno-Anticorpo/metabolismo , Antígenos CD28/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Cinética , Ligantes , Modelos Moleculares , Ligação Proteica , Multimerização Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...