Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.262
Filtrar
1.
Methods Mol Biol ; 2854: 237-251, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192134

RESUMO

The innate immune system is the first line of host defense against infection by pathogenic microorganisms, among which macrophages are important innate immune cells. Macrophages are widely distributed throughout the body and recognize and eliminate viruses through pattern recognition receptors (PRRs) to sense pathogen-associated molecular patterns (PAMPs). In the present chapter, we provide detailed protocols for vesicular stomatitis virus (VSV) amplification, VSV titer detection, isolation of mouse primary peritoneal macrophages, in vitro and in vivo VSV infection, detection of interferon-beta (IFN-ß) expression, and lung injury. These protocols provide efficient and typical methods to evaluate virus-induced innate immunity in vitro and in vivo.


Assuntos
Imunidade Inata , Interferon beta , Macrófagos Peritoneais , Vesiculovirus , Animais , Camundongos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Macrófagos Peritoneais/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Interferon beta/genética , Vesiculovirus/imunologia , Vesiculovirus/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Receptores de Reconhecimento de Padrão/imunologia
2.
BMC Infect Dis ; 24(1): 915, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39232642

RESUMO

BACKGROUND: This study aimed to investigate the differential expression levels of the cGAS-STING pathway in peripheral blood mononuclear cells (PBMCs) of spinal tuberculosis (TB) patients with different progression and its feasibility as a diagnostic marker. METHODS: Peripheral blood and medical records of 25 patients with spinal TB and 10 healthy individuals, were prospectively collected and analyzed. PBMCs and serum were extracted from peripheral blood and the expression levels of the cGAS-STING pathway in PBMCs were measured by real-time PCR (RT-PCR) and serum interferon ß (IFN-ß) expression levels were measured by enzyme-linked immunosorbent assay (ELISA). The expression of Interferon regulatory Factor 3 (IRF3) in PBMCs was measured using western blot. Statistical analysis was performed using the SPSS 26.0 statistical package. RESULTS: The results showed that the expression level of the TANK-binding kinase 1 (TBK1) and IRF3 was significantly higher in PBMCs (P < 0.05), in patients with active lesions than in patients with stable lesions. The serum concentration of IFN-ß was significantly higher in patients with active lesions (P = 0.028). Compared with healthy individuals, the expression level of the cGAS-STING pathway was elevated in PBMCs of TB patients (P < 0.05), and the difference in the expression level of IFN-ß was not statistically significant (P > 0.05), and the serum IFN-ß concentration was elevated (P < 0.05). The calculated AUC values for TBK1 and IRF3 in PBMCs, IFN-ß in serum and erythrocyte sedimentation rate (ESR) to distinguish between patients with active and stable lesions were 0.732, 0.714, 0.839, and 0.714 respectively. CONCLUSIONS: The expression level of TBK1 and IRF3 in PBMCs, and IFN-ß in the serum of patients with spinal TB is positively correlated with disease activity. TBK1 has higher specificity and IFN-ß in serum has higher sensitivity when used to differentiate between patients with active and stable lesions.


Assuntos
Fator Regulador 3 de Interferon , Leucócitos Mononucleares , Proteínas de Membrana , Nucleotidiltransferases , Tuberculose da Coluna Vertebral , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Feminino , Adulto , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Nucleotidiltransferases/genética , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/sangue , Tuberculose da Coluna Vertebral/sangue , Tuberculose da Coluna Vertebral/genética , Interferon beta/sangue , Transdução de Sinais , Proteínas Serina-Treonina Quinases/genética , Biomarcadores/sangue , Estudos Prospectivos , Adulto Jovem , Idoso
3.
Mol Biol Rep ; 51(1): 974, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39259342

RESUMO

BACKGROUND: One of the causes of tubulointerstitial nephritis is viral infection, with innate immune responses affecting its pathogenesis. Toll-like receptor 3 (TLR3) recognizes viral infections and acts antivirally by activating signaling to produce inflammatory cytokines/chemokines, including C-C motif chemokine ligand 5 (CCL5) and interferon-ß (IFN-ß). Although cylindromatosis lysine 63 deubiquitinase (CYLD) is known to be associated with tubulointerstitial nephritis and renal function, its role in the antiviral innate immune response in tubular epithelial cells remains unknown. In this study, we investigated the association between CYLD and TLR3-mediated CCL5 production in cultured human renal proximal tubular epithelial cells (hRPTECs). METHODS AND RESULTS: Polyinosinic-polycytidylic acid (poly IC), a synthetic TLR3 ligand, was used to stimulate hRPTECs. mRNA expression was measured using reverse transcription-quantitative polymerase chain reaction. Protein expression was assayed using western blotting or an enzyme-linked immunosorbent assay. Knockdown of IFN-ß, nuclear factor-kappa B (NF-κB) p65, and CYLD was performed by transfecting cells with specific small interfering RNAs. The intracellular localization of CYLD in hRPTECs was analyzed using immunofluorescence. Poly IC induced CCL5 expression in a time- and concentration-dependent manner, and knockdown of either IFN-ß or p65 reduced poly IC-induced CCL5 expression. CYLD knockdown increased the poly IC-induced CCL5, phosphorylated IκB kinase α/ß (IKK complex), and phosphorylated p65 expression. The CYLD protein was localized in the cytoplasm, and poly IC did not alter its expression. CONCLUSION: CYLD may prevent excessive inflammation due to an antiviral innate immune response by suppressing IKK complex and NF-κB activation downstream of TLR3 in hRPTECs.


Assuntos
Quimiocina CCL5 , Enzima Desubiquitinante CYLD , Células Epiteliais , Túbulos Renais Proximais , Poli I-C , Receptor 3 Toll-Like , Humanos , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/genética , Enzima Desubiquitinante CYLD/metabolismo , Enzima Desubiquitinante CYLD/genética , Quimiocina CCL5/metabolismo , Quimiocina CCL5/genética , Túbulos Renais Proximais/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Poli I-C/farmacologia , Interferon beta/metabolismo , Interferon beta/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Imunidade Inata , NF-kappa B/metabolismo , Linhagem Celular
4.
Mult Scler Relat Disord ; 90: 105839, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39217809

RESUMO

BACKGROUND: Multiple sclerosis (MS) is a prevalent, disabling, inflammatory, neurodegenerative disease that typically manifests during a highly productive stage of life. Interferon beta-1a was among the first approved disease-modifying therapies for MS and remains among the first-line treatment options. Pegylation of the interferon beta-1a molecule prolongs its half-life while maintaining its efficacy and safety profile. In PEGINTEGRITY study, we aimed to compare peginterferon beta-1a with interferon beta-1a in terms of efficacy and safety in relapsing-remitting multiple sclerosis (RRMS) patients. METHODS: This study was a randomized, active-controlled, parallel-group, multi-center Phase 3 trial conducted in Iran in participants with RRMS. Participants received 125 µg of subcutaneous peginterferon beta-1a every two weeks or 30 µg of intramuscular interferon beta-1a once a week for up to 96 weeks. The primary outcome was the non-inferiority of peginterferon beta-1a to interferon beta-1a in reducing annualized relapse rate (ARR). Other outcomes included the number of patients with 12-week confirmed disability progression, the number of new or newly-enlarging T2 hyperintense lesions, the number of gadolinium-enhancing lesions, the number of new T1 hypointense lesions, the volume of new or newly-enlarging T2 hyperintense lesions, changes in brain volume, immunogenicity, and safety assessments. RESULTS: A total of 168 patients who met the eligibility criteria were enrolled and assigned to two arms of the study, each consisting of 84 participants. Totally, 41 participants (24 patients in the peginterferon beta-1a group and 17 patients in the interferon beta-1a group) were withdrawn from the study. The withdrawn patients were included in the per-protocol analysis for the period of time they were in the study. In 96 weeks, in the per-protocol population, the ARR was 0.05 in the peginterferon beta-1a group versus 0.11 in the interferon beta-1a group, which does not reflect a statistically significant difference (p=0.09; 95 % CI, 0.18-1.14). Considering the upper limit of the one-sided 95 % CI of the rate ratio of peginterferon beta-1a compared to interferon beta-1a, as well as the non-inferiority margin, it can be concluded that the primary outcome was met. The results were also comparable for other efficacy and safety outcomes. CONCLUSION: The results demonstrate the non-inferiority of peginterferon beta-1a to interferon beta-1a with similar efficacy in 96-week ARR in RRMS patients. Both arms were also comparable in other efficacy outcomes and safety profiles with no statistically significant differences. These findings support considering peginterferon beta-1a as a safe and efficient option in patients with RRMS. This study was registered on Iranian Registry of Clinical Trials (IRCT201612306135N8) and clinicaltrials.gov (NCT05242133).


Assuntos
Interferon beta-1a , Esclerose Múltipla Recidivante-Remitente , Polietilenoglicóis , Humanos , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Adulto , Masculino , Feminino , Interferon beta-1a/administração & dosagem , Interferon beta-1a/farmacologia , Interferon beta-1a/efeitos adversos , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/efeitos adversos , Polietilenoglicóis/farmacologia , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/farmacologia , Pessoa de Meia-Idade , Interferon beta/administração & dosagem , Interferon beta/efeitos adversos , Interferon beta/farmacologia , Adulto Jovem
5.
Cell Mol Biol (Noisy-le-grand) ; 70(8): 90-95, 2024 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-39262259

RESUMO

Multi-drug resistance (MDR) might be acquired by the cancer cells during chemotherapy, and ATP-binding cassette (ABC) transporters play a significant role in MDR. Interferon-γ (IFN-γ) and IFN-ß can inhibit cancer cell proliferation; however, the effects and mechanism of these cytokines on the growth and MDR are still unclear. To investigate the effects of IFN-γ and IFN-ß, alone or in combination, on viability, resistance, and the expression of ABC transporters of the MDA-MB-231 breast cancer cell line. Using the MDA-MB-231 cell line, we assessed the effects of 20, 100, and 500 IU/ml of IFN-γ and IFN-ß, alone or in combination, on cell viability by methyl thiazolyl tetrazolium (MTT) assay; and then we performed the Uptake and Efflux experiment to evaluate the effect of these IFNs on the cell resistance. Then, using quantitative real-time PCR, we evaluated changes in the expression of ABCB1, ABCC1, and ABCG2 mRNA levels. We discovered that IFN-γ and IFN-ß might both reduce viability, either alone or in combination. The combination of IFNs also displayed synergistic responses, particularly when utilizing equivalent dosages of 500 or 100 IU/ml. The combination of IFN-γ and IFN-ß resulted in a significant increase in Doxorubicin accumulation and down-regulation of the ABCC1 gene at the mRNA level. Our study suggested that equal doses of IFN-γ and IFN-ß in combination might result in potentiated responses against cancer, especially, along with chemotherapy agents.


Assuntos
Neoplasias da Mama , Proliferação de Células , Sobrevivência Celular , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Interferon beta , Interferon gama , Humanos , Interferon gama/farmacologia , Interferon gama/metabolismo , Interferon beta/farmacologia , Interferon beta/metabolismo , Linhagem Celular Tumoral , Neoplasias da Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Proliferação de Células/efeitos dos fármacos , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Doxorrubicina/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética
6.
Viruses ; 16(8)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39205214

RESUMO

Arteriviruses can establish persistent infections in animals such as equids, pigs, nonhuman primates, rodents, and possums. Some Arteriviruses can even cause overt and severe diseases such as Equine Arteritis in horses and Porcine Reproductive and Respiratory Syndrome in pigs, leading to huge economic losses. Arteriviruses have evolved viral proteins to antagonize the host cell's innate immune responses by inhibiting type I interferon (IFN) signaling, assisting viral evasion and persistent infection. So far, the role of the Arterivirus glycoprotein 5 (GP5) protein in IFN signaling inhibition remains unclear. Here, we investigated the inhibitory activity of 47 Arterivirus GP5 proteins derived from various hosts. We demonstrated that all GP5 proteins showed conserved activity for antagonizing TIR-domain-containing adapter proteins inducing interferon-ß (TRIF)-mediated IFN-ß signaling through TRIF degradation. In addition, Arterivirus GP5 proteins showed a conserved inhibitory activity against IFN-ß signaling, induced by either pig or human TRIF. Furthermore, certain Arterivirus GP5 proteins could inhibit the induction of IFN-stimulated genes. These findings highlight the role of Arterivirus GP5 proteins in supporting persistent infection.


Assuntos
Arterivirus , Interferon Tipo I , Transdução de Sinais , Animais , Arterivirus/genética , Arterivirus/metabolismo , Humanos , Suínos , Interferon Tipo I/metabolismo , Infecções por Arterivirus/veterinária , Infecções por Arterivirus/virologia , Infecções por Arterivirus/imunologia , Interferon beta/metabolismo , Interferon beta/genética , Imunidade Inata , Proteínas Virais/metabolismo , Proteínas Virais/genética , Células HEK293
7.
Neurology ; 103(6): e209752, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39197111

RESUMO

BACKGROUND AND OBJECTIVES: Isolated value of MRI metrics in relapsing multiple sclerosis (RMS) as a surrogate marker of response to disease-modifying treatment (DMT) and, thus, as decision criteria for DMT escalation in the absence of clinical signs of disease activity is still a matter of debate. The aim of this study was to investigate whether DMT escalation based on isolated MRI activity affects clinical outcome. METHODS: Combining data from 5 MS centers in Austria and Switzerland, we included patients with RMS aged at least 18 years who (1) had initiated first-line, low-to-moderate-efficacy DMT (interferon ß, glatiramer acetate, teriflunomide, or dimethyl fumarate) continued for ≥12 months, (2) were clinically stable (no relapses or disability progression) on DMT for 12 months, (3) had MRI at baseline and after 12 months on DMT, and (4) had available clinical follow-up for ≥2 years after the second MRI. The primary endpoint was occurrence of relapse during follow-up. The number of new T2 lesions (T2L) and DMT strategy (continuing low-/moderate-efficacy DMT vs escalating DMT) were used as covariates in regression analyses. RESULTS: A total of 131 patients with RMS, median age of 36 (25th-75th percentiles: 29-43) years, 73% women, were included and observed over a median period of 6 (5-9) years after second MRI. Sixty-two (47%) patients had relapse. Patients who continued first-line DMT had a 3-fold increased risk of relapse given 2 new T2L (hazard ratio [HR] 3.2, lower limit [LL] of 95% CI: 1.5) and a 4-fold increased risk given ≥3 new T2L (HR 4.0, LL-CI: 2.1). Escalation of DMT lowered the risk of relapse in patients with 2 new T2L by approximately 80% (HR 0.2, upper limit [UL] of 95% CI: 1.3) and with ≥3 new T2L by 70% (HR 0.3, UL-CI: 0.8). In case of only 1 new T2L, the increased risk of relapse and the treatment effect did not reach statistical significance of 5%. DISCUSSION: In our real-world cohort of patients clinically stable under low-to-moderate-efficacy DMT, escalation of DMT based on isolated MRI activity decreased risk of further relapse when at least 2 new T2L had occurred. CLASSIFICATION OF EVIDENCE: This study provides Class III evidence that clinically stable patients with MS on low-/moderate-efficacy DMT with ≥3 new T2L on MRI who escalate DMT have a reduced risk of relapse and Expanded Disability Status Scale progression.


Assuntos
Imageamento por Ressonância Magnética , Esclerose Múltipla Recidivante-Remitente , Humanos , Feminino , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/diagnóstico por imagem , Masculino , Adulto , Crotonatos/uso terapêutico , Resultado do Tratamento , Nitrilas/uso terapêutico , Toluidinas/uso terapêutico , Hidroxibutiratos , Fumarato de Dimetilo/uso terapêutico , Pessoa de Meia-Idade , Acetato de Glatiramer/uso terapêutico , Interferon beta/uso terapêutico , Áustria , Suíça , Fatores Imunológicos/uso terapêutico , Seguimentos , Imunossupressores/uso terapêutico , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos
8.
Int J Mol Sci ; 25(15)2024 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-39126076

RESUMO

Alcoholic liver disease (ALD) is a form of hepatic inflammation. ALD is mediated by gut leakiness. This study evaluates the anti-inflammatory effects of ASCs overexpressing interferon-beta (ASC-IFN-ß) on binge alcohol-induced liver injury and intestinal permeability. In vitro, ASCs were transfected with a non-viral vector carrying the human IFN-ß gene, which promoted hepatocyte growth factor (HGF) secretion in the cells. To assess the potential effects of ASC-IFN-ß, C57BL/6 mice were treated with three oral doses of binge alcohol and were administered intraperitoneal injections of ASC-IFN-ß. Mice treated with binge alcohol and administered ASC-IFN-ß showed reduced liver injury and inflammation compared to those administered a control ASC. Analysis of intestinal tissue from ethanol-treated mice administered ASC-IFN-ß also indicated decreased inflammation. Additionally, fecal albumin, blood endotoxin, and bacterial colony levels were reduced, indicating less gut leakiness in the binge alcohol-exposed mice. Treatment with HGF, but not IFN-ß or TRAIL, mitigated the ethanol-induced down-regulation of cell death and permeability in Caco-2 cells. These results demonstrate that ASCs transfected with a non-viral vector to induce IFN-ß overexpression have protective effects against binge alcohol-mediated liver injury and gut leakiness via HGF.


Assuntos
Etanol , Interferon beta , Hepatopatias Alcoólicas , Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Permeabilidade , Animais , Humanos , Interferon beta/metabolismo , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/patologia , Hepatopatias Alcoólicas/genética , Camundongos , Células-Tronco Mesenquimais/metabolismo , Etanol/efeitos adversos , Células CACO-2 , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/genética , Masculino , Tecido Adiposo/metabolismo , Fígado/metabolismo , Fígado/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia
9.
Cell Death Dis ; 15(8): 574, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39117638

RESUMO

Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNß, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNß-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNß, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.


Assuntos
Citocinas , Peptídeos e Proteínas de Sinalização Intracelular , Necroptose , Proteínas Serina-Treonina Quinases , Tristetraprolina , Tristetraprolina/metabolismo , Tristetraprolina/genética , Animais , Citocinas/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Macrófagos/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Inflamação/genética , Células RAW 264.7 , Regulação para Cima/efeitos dos fármacos , Interferon beta/metabolismo , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica
10.
Clin Immunol ; 267: 110339, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39137826

RESUMO

Anti-drug antibodies (ADA) reduce the efficacy of immunotherapies in multiple sclerosis (MS) and are associated with increased disease progression risk. Blood biomarkers predicting immunogenicity to biopharmaceuticals represent an unmet clinical need. Patients with relapsing remitting (RR)MS were recruited before (baseline), three, and 12 (M12) months after commencing interferon-beta treatment. Neutralising ADA-status was determined at M12, and patients were stratified at baseline according to their M12 ADA-status (ADA-positive/ADA-negative). Patients stratified as ADA-positive were characterised by an early dampened response to interferon-beta (prior to serum ADA detection) and distinct proinflammatory transcriptomic/proteomic peripheral blood signatures enriched for 'immune response activation' including phosphoinositide 3-kinase-γ and NFκB-signalling pathways both at baseline and throughout the treatment course, compared to ADA-negative patients. These immunogenicity-associated proinflammatory signatures significantly overlapped with signatures of MS disease severity. Thus, whole blood molecular profiling is a promising tool for prediction of ADA-development in RRMS and could provide insight into mechanisms of immunogenicity.


Assuntos
Interferon beta , Esclerose Múltipla Recidivante-Remitente , Proteômica , Transcriptoma , Humanos , Interferon beta/uso terapêutico , Interferon beta/imunologia , Feminino , Masculino , Adulto , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/imunologia , Esclerose Múltipla Recidivante-Remitente/sangue , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Biomarcadores/sangue , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Esclerose Múltipla/sangue
11.
Nat Commun ; 15(1): 7165, 2024 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-39187481

RESUMO

Programmed cell death 1 (PD-1) is a premier cancer drug target for immune checkpoint blockade (ICB). Because PD-1 receptor inhibition activates tumor-specific T-cell immunity, research has predominantly focused on T-cell-PD-1 expression and its immunobiology. In contrast, cancer cell-intrinsic PD-1 functional regulation is not well understood. Here, we demonstrate induction of PD-1 in melanoma cells via type I interferon receptor (IFNAR) signaling and reversal of ICB efficacy through IFNAR pathway inhibition. Treatment of melanoma cells with IFN-α or IFN-ß triggers IFNAR-mediated Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling, increases chromatin accessibility and resultant STAT1/2 and IFN regulatory factor 9 (IRF9) binding within a PD-1 gene enhancer, and leads to PD-1 induction. IFNAR1 or JAK/STAT inhibition suppresses melanoma-PD-1 expression and disrupts ICB efficacy in preclinical models. Our results uncover type I IFN-dependent regulation of cancer cell-PD-1 and provide mechanistic insight into the potential unintended ICB-neutralizing effects of widely used IFNAR1 and JAK inhibitors.


Assuntos
Inibidores de Checkpoint Imunológico , Interferon Tipo I , Melanoma , Receptor de Morte Celular Programada 1 , Receptor de Interferon alfa e beta , Transdução de Sinais , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/genética , Melanoma/metabolismo , Humanos , Receptor de Interferon alfa e beta/metabolismo , Receptor de Interferon alfa e beta/genética , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Camundongos , Interferon Tipo I/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Interferon beta/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Janus Quinases/metabolismo , Camundongos Endogâmicos C57BL , Interferon-alfa/farmacologia , Interferon-alfa/metabolismo , Feminino
12.
Nat Commun ; 15(1): 7226, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191748

RESUMO

mRNA vaccines against SARS-CoV-2 have revolutionized vaccine development, but their immunological mechanisms are not fully understood. Here, we investigate injection site responses of mRNA vaccines by generating a comprehensive single-cell transcriptome profile upon lipid nanoparticle (LNP) or LNP-mRNA challenge in female BALB/c mice. We show that LNP-induced stromal pro-inflammatory responses and mRNA-elicited type I interferon responses dominate the initial injection site responses. By tracking the fate of delivered mRNA, we discover that injection site fibroblasts are highly enriched with the delivered mRNA and that they express IFN-ß specifically in response to the mRNA component, not to the LNP component of mRNA vaccines. Moreover, the mRNA-LNP, but not LNP alone, induces migratory dendritic cells highly expressing IFN-stimulated genes (mDC_ISGs) at the injection site and draining lymph nodes. When co-injected with LNP-subunit vaccine, IFN-ß induces mDC_ISGs at the injection site, and importantly, it substantially enhances antigen-specific cellular immune responses. Furthermore, blocking IFN-ß signaling at the injection site significantly decreases mRNA vaccine-induced cellular immune responses. Collectively, these data highlight the importance of injection site fibroblasts and IFN-ß signaling during early immune responses against the mRNA vaccine and provide detailed information on the initial chain of immune reactions elicited by mRNA vaccine injection.


Assuntos
Células Dendríticas , Fibroblastos , Imunidade Celular , Imunidade Inata , Interferon beta , Camundongos Endogâmicos BALB C , Nanopartículas , Vacinas de mRNA , Animais , Interferon beta/imunologia , Interferon beta/metabolismo , Feminino , Imunidade Inata/imunologia , Imunidade Inata/efeitos dos fármacos , Camundongos , Vacinas de mRNA/imunologia , Células Dendríticas/imunologia , Nanopartículas/química , Fibroblastos/imunologia , Fibroblastos/metabolismo , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , SARS-CoV-2/imunologia , Lipídeos/química , COVID-19/prevenção & controle , COVID-19/imunologia , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Análise de Célula Única , Lipossomos
13.
Cell Rep ; 43(8): 114608, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39120972

RESUMO

Ubiquitination is essential for the proteasomal turnover of IRF3, the central factor mediating the antiviral innate immune response. However, the spatiotemporal regulation of IRF3 ubiquitination for the precise activation and timely resolution of innate immunity remains unclear. Here, we identified BRCA1-associated protein-1 (BAP1) and ubiquitin-protein ligase E3C (UBE3C) as the key deubiquitinase and ubiquitinase for temporal control of IRF3 stability during viral infection. In the early stage, BAP1 dominates and removes K48-linked ubiquitination of IRF3 in the nucleus, preventing its proteasomal degradation and facilitating efficient interferon (IFN)-ß production. In the late stage, E3 ligase UBE3C, induced by IFN-ß, specifically mediates IRF3 ubiquitination and promotes its proteasomal degradation. Overall, the sequential interactions with BAP1 and UBE3C govern IRF3 stability during innate response, ensuring effective viral clearance and inflammation resolution. Our findings provide insights into the temporal control of innate signaling and suggest potential interventions in viral infection.


Assuntos
Imunidade Inata , Fator Regulador 3 de Interferon , Proteínas Supressoras de Tumor , Ubiquitina Tiolesterase , Ubiquitina-Proteína Ligases , Ubiquitinação , Fator Regulador 3 de Interferon/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Humanos , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Células HEK293 , Camundongos , Interferon beta/metabolismo , Proteólise , Camundongos Endogâmicos C57BL , Transdução de Sinais , Complexo de Endopeptidases do Proteassoma/metabolismo
14.
J Clin Neurosci ; 128: 110811, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39197331

RESUMO

BACKGROUND: Although Interferon-beta (IFNß) has long been approved as a disease-modifying therapy (DMT) for Multiple sclerosis (MS), flu-like syndrome (FLS) persists as a common adverse effect of interferon therapy. Given the importance of circadian rhythm in regulating physiological processes, we aimed to assess the relationship between patient's chronotype and time of interferon injection with FLS score in MS patients receiving IFNß. METHODS: A cross-sectional study was conducted on 118 MS patients who were referred to the clinic of neurology of Zanjan Vali-e-Asr Hospital for interferon injection. The included were invited to complete a morningness-eveningness questionnaire (MEQ) assessing patients' chronotype. The following data were extracted from patients' record: age, gender, duration of interferon treatment, type of interferon taken, time of interferon injection (morning/evening), FLS score, MS subtype, and usage of pain killers. All data found were imported and statistically analyzed in SPSS ver.26. RESULTS: According to the patients' record, 114 (96.6%) patients had experienced post-interferon injection FLS with different severities. Statistical analysis revealed no significant relationship between the patient's chronotype and FLS score. Nevertheless, the FLS score was significantly higher in those who had evening injections. CONCLUSIONS: Time of interferon injection was significantly associated with FLS score, with higher FLS score following evening injection. However, no significant relationship was found between the FLS score and the patient's chronotype. It is recommended that further studies assessing circadian rhythm using laboratory tests such as melatonin measurement need to be undertaken to investigate the association of circadian rhythm with post-interferon injection FLS.


Assuntos
Ritmo Circadiano , Interferon beta , Esclerose Múltipla , Humanos , Feminino , Masculino , Adulto , Estudos Transversais , Interferon beta/efeitos adversos , Interferon beta/administração & dosagem , Esclerose Múltipla/tratamento farmacológico , Ritmo Circadiano/fisiologia , Ritmo Circadiano/efeitos dos fármacos , Pessoa de Meia-Idade , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/efeitos adversos , Influenza Humana/complicações , Influenza Humana/tratamento farmacológico , Adulto Jovem , Índice de Gravidade de Doença , Cronotipo
15.
J Biotechnol ; 394: 24-33, 2024 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-39103019

RESUMO

The production of therapeutic glycoproteins is primarily expensive due to the necessity of culturing mammalian cells. These systems often require complex and costly culture media and typically yield low amounts of protein. Leishmania tarentolae, a non-pathogenic protozoan to mammals, has emerged as a cost-effective alternative system for heterologous glycoprotein expression due to its suitability for large-scale production using low-cost culture media, and its ability to perform mammalian-like post-translational modifications, including glycosylation. Nevertheless, differences in the carbohydrate residues at the end of N-glycan chains are observed in Leishmania compared to mammalian cells due to the absence of biosynthetic enzymes in Leishmania that are required for the incorporation of terminal sialic acid. In this study, a genetically optimized L. tarentolae cell line was engineered for the production of recombinant interferon-ß (IFN-ß) featuring a complete mammalian N-glycosylation profile. Genomic and metabolomic analyses revealed that heterologous expression of the sialyltransferase enzyme and cultivation in a medium containing sialic acid were sufficient to generate mammalian-like protein N-glycosylation. N-glycan mass spectrometry analysis demonstrated a glycosylation pattern compatible with the incorporation of sialic acid into the glycan structure. In vitro IFN-ß activity indicated that the expressed protein exhibited reduced inflammatory effects compared to IFN-beta produced by other platforms, such as bacteria, non-optimized L. tarentolae, and mammalian cells.


Assuntos
Interferon beta , Leishmania , Proteínas Recombinantes , Sialiltransferases , Glicosilação , Leishmania/genética , Leishmania/metabolismo , Leishmania/enzimologia , Humanos , Interferon beta/metabolismo , Interferon beta/genética , Sialiltransferases/metabolismo , Sialiltransferases/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Animais , Polissacarídeos/metabolismo , Camundongos
16.
J Immunol ; 213(6): 808-822, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39109927

RESUMO

Type I IFNs play a pivotal role in immune response modulation, yet dysregulation is implicated in various disorders. Therefore, it is crucial to develop tools that facilitate the understanding of their mechanism of action and enable the development of more effective anti-IFN therapeutic strategies. In this study, we isolated, cloned, and characterized anti-IFN-α and anti-IFN-ß Abs from PBMCs of individuals treated with IFN-α or IFN-ß, harboring confirmed neutralizing Abs. Clones AH07856 and AH07857 were identified as neutralizing anti-IFN-α-specific with inhibition against IFN-α2a, -α2b, and -αK subtypes. Clones AH07859 and AH07866 were identified as neutralizing anti-IFN-ß1a-specific signaling and able to block lipopolysaccharide or S100 calcium-binding protein A14-induced IFN-ß signaling effects. Cloned Abs bind rhesus but not murine IFNs. The specificity of inhibition between IFN-α and IFN-ß suggests potential for diverse research and clinical applications.


Assuntos
Anticorpos Neutralizantes , Clonagem Molecular , Interferon-alfa , Interferon beta , Humanos , Interferon-alfa/imunologia , Interferon-alfa/genética , Anticorpos Neutralizantes/imunologia , Interferon beta/imunologia , Interferon beta/genética , Animais , Camundongos , Leucócitos Mononucleares/imunologia , Transdução de Sinais/imunologia
17.
Eur J Pharmacol ; 981: 176882, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39128808

RESUMO

Interferon beta (IFNß) is a member of the type-1 interferon family and has various immunomodulatory functions in neuropathological conditions. Although the level of IFNß is low under healthy conditions, it is increased during inflammatory processes to protect the central nervous system (CNS). In particular, microglia and astrocytes are the main sources of IFNß upon inflammatory insult in the CNS. The protective effects of IFNß are well characterized in reducing the progression of multiple sclerosis (MS); however, little is understood about its effects in other neurological/neurodegenerative diseases. In this review, different types of IFNs and their signaling pathways will be described. Then we will focus on the potential role and therapeutic effect of IFNß in several CNS-related diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury, prion disease and spinocerebellar ataxia 7.


Assuntos
Interferon beta , Humanos , Animais , Interferon beta/uso terapêutico , Interferon beta/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/metabolismo , Transdução de Sinais/efeitos dos fármacos
18.
Genes (Basel) ; 15(8)2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39202404

RESUMO

As a founding member of the Src family of kinases, Src has been confirmed to participate in the regulation of immune responses, integrin signaling, and motility. Ducks are usually asymptomatic carriers of RNA viruses such as Newcastle disease virus and avian influenza virus, which can be deadly to chickens. The beneficial role of Src in modulating the immune response remains largely unknown in ducks. Here, we characterized the duck Src and found that it contains a 192-base-pair 5' untranslated region, a 1602-base-pair coding region, and a 2541-base-pair 3' untranslated region, encoding 533 amino acid residues. Additionally, duSrc transcripts were significantly activated in duck tissues infected by Newcastle disease virus compared to controls. The duSrc transcripts were notably widespread in all tissues examined, and the expression level was higher in liver, blood, lung, pancreas, and thymus. Moreover, we found the expression levels of IFN-ß, NF-κB, IRF3, and Src were significantly increased in DEFs after infection with 5'ppp dsRNA, but there was no significant difference before and after treatment in DF1 cells. Furthermore, overexpression of duSrc followed by stimulation with 5'ppp dsRNA led to an elevation of IFN-ß levels. The SH3 and PTKc domains of duSrc contributed to promoting the activity of IFN-ß and NF-κB in DEFs stimulated by 5'ppp dsRNA.


Assuntos
Clonagem Molecular , Patos , Animais , Patos/genética , Patos/imunologia , Patos/virologia , Quinases da Família src/genética , Quinases da Família src/metabolismo , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/genética , Proteínas Aviárias/genética , Proteínas Aviárias/imunologia , Proteínas Aviárias/metabolismo , Doença de Newcastle/imunologia , Doença de Newcastle/virologia , Doença de Newcastle/genética , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Distribuição Tecidual , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/genética
19.
Front Immunol ; 15: 1358462, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39100663

RESUMO

The double-stranded DNA (dsDNA) sensor STING has been increasingly implicated in responses to "sterile" endogenous threats and pathogens without nominal DNA or cyclic di-nucleotide stimuli. Previous work showed an endoplasmic reticulum (ER) stress response, known as the unfolded protein response (UPR), activates STING. Herein, we sought to determine if ER stress generated a STING ligand, and to identify the UPR pathways involved. Induction of IFN-ß expression following stimulation with the UPR inducer thapsigargin (TPG) or oxygen glucose deprivation required both STING and the dsDNA-sensing cyclic GMP-AMP synthase (cGAS). Furthermore, TPG increased cytosolic mitochondrial DNA, and immunofluorescence visualized dsDNA punctae in murine and human cells, providing a cGAS stimulus. N-acetylcysteine decreased IFN-ß induction by TPG, implicating reactive oxygen species (ROS). However, mitoTEMPO, a mitochondrial oxidative stress inhibitor did not impact TPG-induced IFN. On the other hand, inhibiting the inositol requiring enzyme 1 (IRE1) ER stress sensor and its target transcription factor XBP1 decreased the generation of cytosolic dsDNA. iNOS upregulation was XBP1-dependent, and an iNOS inhibitor decreased cytosolic dsDNA and IFN-ß, implicating ROS downstream of the IRE1-XBP1 pathway. Inhibition of the PKR-like ER kinase (PERK) pathway also attenuated cytoplasmic dsDNA release. The PERK-regulated apoptotic factor Bim was required for both dsDNA release and IFN-ß mRNA induction. Finally, XBP1 and PERK pathways contributed to cytosolic dsDNA release and IFN-induction by the RNA virus, Vesicular Stomatitis Virus (VSV). Together, our findings suggest that ER stressors, including viral pathogens without nominal STING or cGAS ligands such as RNA viruses, trigger multiple canonical UPR pathways that cooperate to activate STING and downstream IFN-ß via mitochondrial dsDNA release.


Assuntos
Citosol , Estresse do Retículo Endoplasmático , Interferon beta , Proteínas de Membrana , Nucleotidiltransferases , Resposta a Proteínas não Dobradas , Humanos , Animais , Camundongos , Nucleotidiltransferases/metabolismo , Citosol/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Interferon beta/metabolismo , DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , eIF-2 Quinase/metabolismo , Endorribonucleases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Proteína 1 de Ligação a X-Box/genética , Tapsigargina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ativação Transcricional , DNA Mitocondrial/metabolismo
20.
Drug Dev Res ; 85(5): e22246, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39135358

RESUMO

Tilorone dihydrochloride (tilorone) is an orally active interferon inducer with anticancer effects. The present study aimed to evaluate the anticancer effects of tilorone in breast cancer. MTT assay was done to measure the proliferation of MCF-7 and MDA-MB-231 breast cancer cells after treatment with tilorone. Mammary carcinogenesis was induced by subcutaneous injection (35 mg/kg, 0.5 mL) of dimethylbenz[a]anthracene (DMBA) in mammary pads of Sprague Dawley (SD) rats. Tumors were allowed to grow for 16 weeks till their sizes reached to 550-700 mm3, and then treated with 10 and 20 mg/kg of tilorone and standard drug doxorubicin (4 mg/kg) twice a week for 3 weeks. Normal and disease-control animals received normal saline. Tumor volumes and body weights were measured. Tumors were isolated to measure the levels of interferon-ß (IFN-ß), vascular endothelial growth factor-A (VEGF-A), P53 and inflammatory markers by enzyme-linked immunosorbent assay (ELISA). Serum biochemistry, lipid peroxidation (LPO) and antioxidant enzymes were measured by standard methods. Histopathology and immunohistochemistry (IHC) of P53 was done in tumor sections. Tilorone reduced the proliferation of MCF-7 and MDA-MB-231 cells with IC50 concentrations at 34.08 µM and 14.27 µM, respectively. Tilorone treatment showed reduced tumor volume, and increased survival with no significant changes in the body weights. Tilorone treatment also decreased levels of inflammatory markers and VEGF-A and increased IFN-ß and P53 levels. Further, treatment with tilorone also decreased LPO and increased antioxidants levels. Histopathology of tumor sections showed normalizing morphology of treated animals. IHC of tumor sections showed increased levels of P53. In conclusion, tilorone has potential anticancer effects against breast cancer.


Assuntos
Citocinas , Estresse Oxidativo , Ratos Sprague-Dawley , Tilorona , Animais , Feminino , Estresse Oxidativo/efeitos dos fármacos , Humanos , Citocinas/metabolismo , Tilorona/farmacologia , Ratos , Células MCF-7 , Regulação para Baixo/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Interferon beta , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Doxorrubicina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...