Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 467
Filtrar
1.
Fish Shellfish Immunol ; 153: 109848, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39168293

RESUMO

The immune regulatory roles of microRNAs (miRNAs) have recently attracted considerable attention. Bioinformatics prediction revealed that both let-7 and miR-210 provide potential binding sites for the Akt (rac-alpha serine/threonine-protein kinase) gene sequence in the sea cucumber Apostichopus japonicus (termed AjAkt). In this study, we first used a dual-luciferase reporter assay and functional validation techniques to verify the interactions between these two miRNAs (let-7 and miR-210) and AjAkt, and then investigated the functions of the validated miRNA/mRNA pair as part of the innate immune response against Vibrio splendidus infection. We found that AjAkt interacts with miR-210 rather than let-7, and miR-210 negatively regulates the expression of AjAkt. From 8 to 48 h after infection with V. splendidus, opposite trends were observed in the expression levels of miR-210 and AjAkt (mRNA and protein) in coelomocytes, suggesting that the miR-210/AjAkt pair is involved in immune regulation during this period after infection. Both AjAkt silencing and miR-210 overexpression enhanced the phagocytic capacity and reduced the infectivity of A. japonicus after pathogen infection, suggesting that the miR-210/AjAkt pair may regulate the innate immune response of A. japonicus by altering phagocytic capacity. The findings of this study enrich our knowledge of the role of miRNA/mRNA pairs in immune regulation in sea cucumbers and provide insights into the molecular mechanisms of the innate immune response in marine echinoderms.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata , MicroRNAs , Proteínas Proto-Oncogênicas c-akt , Stichopus , Vibrio , Animais , Vibrio/fisiologia , MicroRNAs/genética , MicroRNAs/imunologia , Stichopus/imunologia , Stichopus/genética , Stichopus/microbiologia , Imunidade Inata/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação da Expressão Gênica/imunologia
2.
Mol Immunol ; 173: 30-39, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39018745

RESUMO

PURPOSE: The etiology of asthma remains elusive, with no known cure. Based on accumulating evidence, autophagy, a self-degradation process that maintains cellular metabolism and homeostasis, participates in the development of asthma. Mycobacterium vaccae vaccine (M. vaccae), an immunomodulatory agent, has previously been shown to effectively alleviate airway inflammation and airway remodeling. However, its therapeutic effect on asthma via the regulation of autophagy remains unknown. Therefore, this study aimed to investigate the impact of M. vaccae in attenuating asthma airway inflammation via autophagy-mediated pathways. METHODS: Balb/c mice were used to generate an ovalbumin (OVA)-immunized allergic airway model and were subsequently administered either M. vaccae or M. vaccae + rapamycin (an autophagy activator) prior to each challenge. Next, airway inflammation, mucus secretion, and airway remodeling in mouse lung tissue were assessed via histological analyses. Lastly, the expression level of autophagy proteins LC3B, Beclin1, p62, and autolysosome was determined both in vivo and in vitro, along with the expression level of p-PI3K, PI3K, p-Akt, and Akt in mouse lung tissue. RESULTS: The findings indicated that aerosol inhalation of M. vaccae in an asthma mouse model has the potential to decrease eosinophil counts, alleviate airway inflammation, mucus secretion, and airway remodeling through the inhibition of autophagy. Likewise, M. vaccae could reduce the levels of OVA-specific lgE, IL-5, IL-13, and TNF-α in asthma mouse models by inhibiting autophagy. Furthermore, this study revealed that M. vaccae also suppressed autophagy in IL-13-stimulated BEAS-2B cells. Moreover, M. vaccae may activate the PI3K/Akt signaling pathway in the lung tissue of asthmatic mice. CONCLUSION: In summary, the present study suggests that M. vaccae may contribute to alleviating airway inflammation and remodeling in allergic asthma by potentially modulating autophagy and the PI3K/Akt signaling pathway. These discoveries offer a promising avenue for the development of therapeutic interventions targeting allergic airway inflammation.


Assuntos
Asma , Autofagia , Inflamação , Mycobacteriaceae , Ovalbumina , Transdução de Sinais , Animais , Feminino , Camundongos , Remodelação das Vias Aéreas/imunologia , Asma/imunologia , Asma/terapia , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Inflamação/imunologia , Pulmão/patologia , Pulmão/imunologia , Camundongos Endogâmicos BALB C , Mycobacteriaceae/imunologia , Ovalbumina/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/imunologia
3.
Int Immunopharmacol ; 137: 112360, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-38852524

RESUMO

Sarcoidosis is a systemic granulomatous disease characterized by non-caseating epithelioid cell granulomas. One of its immunological hallmarks is the differentiation of CD4 + naïve T cells into Th1/Th17 cells, accompanied by the release of numerous pro-inflammatory cytokines. The TL1A/DR3 signaling pathway plays a crucial role in activating effector lymphocytes, thereby triggering pro-inflammatory responses. The primary aim of this investigation was to scrutinize the impact of anti-TL1A monoclonal antibody on the dysregulation of Th1/Th17 cells and granuloma formation in sarcoidosis. Initially, the abnormal activation of the TL1A/DR3 signaling pathway in pulmonary tissues of sarcoidosis patients was confirmed using qPCR and immunohistochemistry techniques. Subsequently, employing a murine model of sarcoidosis, the inhibitory effects of anti-TL1A monoclonal antibody on the TL1A/DR3 signaling pathway in sarcoidosis were investigated through qPCR, immunohistochemistry, and Western blot experiments. The influence of anti-TL1A monoclonal antibody on granulomas was assessed through HE staining, while their effects on sarcoidosis Th1/Th17 cells and associated cytokine mRNA levels were evaluated using flow cytometry and qPCR, respectively. Immunofluorescence and Western blot experiments corroborated the inhibitory effects of anti-TL1A monoclonal antibody on the aberrant activation of the PI3K/AKT signaling pathway in sarcoidosis. The findings of this study indicate that the TL1A/DR3 signaling pathway is excessively activated in sarcoidosis. Anti-TL1A monoclonal antibody effectively inhibit this abnormal activation in sarcoidosis, thereby alleviating the dysregulation of Th1/Th17 cells and reducing the formation of pulmonary granulomas. This effect may be associated with the inhibition of the downstream PI3K/AKT signaling pathway. Anti-TL1A monoclonal antibody hold promise as a potential novel therapeutic intervention for sarcoidosis.


Assuntos
Anticorpos Monoclonais , Granuloma , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Sarcoidose , Transdução de Sinais , Células Th1 , Células Th17 , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Animais , Células Th1/imunologia , Células Th17/imunologia , Transdução de Sinais/efeitos dos fármacos , Humanos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/imunologia , Granuloma/imunologia , Granuloma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/imunologia , Feminino , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Masculino , Sarcoidose/imunologia , Sarcoidose/tratamento farmacológico , Camundongos , Adulto , Pessoa de Meia-Idade , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Pulmão/imunologia , Pulmão/patologia , Citocinas/metabolismo , Citocinas/imunologia , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C
4.
Fish Shellfish Immunol ; 149: 109586, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38670410

RESUMO

Recent research has highlighted complex and close interaction between miRNAs, autophagy, and viral infection. In this study, we observed the autophagy status in CIK cells infected with GCRV at various time points. We found that GCRV consistently induced cellar autophagy from 0 h to 12 h post infection. Subsequently, we performed deep sequencing on CIK cells infected with GCRV at 0 h and 12 h respectively, identifying 38 DEMs and predicting 9581 target genes. With the functional enrichment analyses of GO and KEGG, we identified 35 autophagy-related target genes of these DEMs, among which akt3 was pinpointed as the most central hub gene using module assay of the PPI network. Then employing the miRanda and Targetscan programs for prediction, and verification through a double fluorescent enzyme system and qPCR method, we confirmed that miR-193 b-3p could target the 3'-UTR of grass carp akt3, reducing its gene expression. Ultimately, we illustrated that grass carp miR-193 b-3p could promote autophagy in CIK cells. Above results collectively indicated that miRNAs might play a critical role in autophagy of grass carp during GCRV infection and contributed significantly to antiviral immunity by targeting autophagy-related genes. This study may provide new insights into the intricate mechanisms involved in virus, autophagy, and miRNAs.


Assuntos
Autofagia , Carpas , Doenças dos Peixes , MicroRNAs , Proteínas Proto-Oncogênicas c-akt , Infecções por Reoviridae , Reoviridae , Animais , MicroRNAs/genética , MicroRNAs/imunologia , Carpas/imunologia , Carpas/genética , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/veterinária , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reoviridae/fisiologia , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Linhagem Celular , Regulação da Expressão Gênica/imunologia
5.
J Biol Chem ; 299(11): 105276, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37739035

RESUMO

Imbalanced immune responses are a prominent hallmark of cancer and autoimmunity. Myeloid cells can be overly suppressive, inhibiting protective immune responses or inactive not controlling autoreactive immune cells. Understanding the mechanisms that induce suppressive myeloid cells, such as myeloid-derived suppressor cells (MDSCs) and tolerogenic dendritic cells (TolDCs), can facilitate the development of immune-restoring therapeutic approaches. MDSCs are a major barrier for effective cancer immunotherapy by suppressing antitumor immune responses in cancer patients. TolDCs are administered to patients to promote immune tolerance with the intent to control autoimmune disease. Here, we investigated the development and suppressive/tolerogenic activity of human MDSCs and TolDCs to gain insight into signaling pathways that drive immunosuppression in these different myeloid subsets. Moreover, monocyte-derived MDSCs (M-MDSCs) generated in vitro were compared to M-MDSCs isolated from head-and-neck squamous cell carcinoma patients. PI3K-AKT signaling was identified as being crucial for the induction of human M-MDSCs. PI3K inhibition prevented the downregulation of HLA-DR and the upregulation of reactive oxygen species and MerTK. In addition, we show that the suppressive activity of dexamethasone-induced TolDCs is induced by ß-catenin-dependent Wnt signaling. The identification of PI3K-AKT and Wnt signal transduction pathways as respective inducers of the immunomodulatory capacity of M-MDSCs and TolDCs provides opportunities to overcome suppressive myeloid cells in cancer patients and optimize therapeutic application of TolDCs. Lastly, the observed similarities between generated- and patient-derived M-MDSCs support the use of in vitro-generated M-MDSCs as powerful model to investigate the functionality of human MDSCs.


Assuntos
Células Dendríticas , Células Supressoras Mieloides , Fosfatidilinositol 3-Quinases , Transdução de Sinais , Via de Sinalização Wnt , Humanos , Células Dendríticas/imunologia , Imunomodulação/imunologia , Imunoterapia , Células Supressoras Mieloides/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais/imunologia , Via de Sinalização Wnt/imunologia , Células Tumorais Cultivadas
6.
Int Immunopharmacol ; 121: 110452, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37302368

RESUMO

BACKGROUND: Immune checkpoint inhibitors evoke the immune system, which may cause immune-related adverse effects. The predictors and mechanisms of anti-PD-1-associated thyroid immune injury remain unclear. METHODS: A retrospective analysis is conducted on 518 patients treated with anti PD-1/PD-L1. Firstly, the differences between anti PD-1 and anti PD-L1 are compared on the risk of thyroid immune injury. Then, the predictors of the risk and thyroid function for anti PD-1 related thyroid immune injury are analyzed. Furthermore, the in vitro mechanism of normal thyroid cells (NTHY) is studied. First, the effect of anti PD-1 on the cell viability and immune sensitivity of thyroid cells is observed. Cell viability includes cell proliferation, apoptosis, cell cycle, T4 secretion, while immune sensitivity includes molecular expression and CD8 + T cell aggregation and killing towards NTHY. Then the differentially expressed proteins (DEPs) are screened by protein mass spectrometry. Enrichment of KEGG pathway and annotation of GO function on DEPs are conducted. Human protein-protein interactions are obtained from STRING database. The network is constructed and analyzed using Cytoscape software. In vitro, key proteins and their pathways are validated through overexpression plasmids or inhibitors. The recovery experiment and the immuno-coprecipitation experiment are designed to support the results. In vivo, the key proteins are detected in the thyroid tissue of mice fed with anti PD-1, as well as in the thyroid tissue of patients with Hashimoto's thyroiditis. RESULTS: Thyroid irAE is associated with female, IgG, FT4, TPOAb, TGAb, TSHI, TFQI, and TSH. Peripheral lymphocytes are associated with thyroid function. In vitro, the NIVO group shows prologed G1 phase, decreased FT4, downregulated PD-L1, upregulated IFN-γ, and more CD8 + T cell infiltration and cytotoxicity. AKT1-SKP2 is chosen as the key protein. AKT1 overexpression and SKP2 inhibitor replies to NIVO and AKT1 overexpression, respectively. Immunoprecipitation shows SKP2 and PD-L1 interaction. CONCLUSION: Female, impaired thyroid hormone sensitivity and IgG4 contribute to the risk of thyroid irAE, while peripheral blood lymphocyte characteristics affect thyroid function. Anti-PD-1 induces thyroid irAE by downregulating AKT1-SKP2 to enhance thyroid immunosensitivity.


Assuntos
Antígeno B7-H1 , Doença de Hashimoto , Inibidores de Checkpoint Imunológico , Glândula Tireoide , Animais , Feminino , Humanos , Camundongos , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos , Linfócitos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/imunologia , Estudos Retrospectivos , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Proteínas Quinases Associadas a Fase S/efeitos dos fármacos , Proteínas Quinases Associadas a Fase S/imunologia , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/patologia
7.
J Transl Med ; 20(1): 559, 2022 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463188

RESUMO

BACKGROUND: Cancer-associated fibroblasts (CAFs), one of the major components of the tumor stroma, contribute to an immunosuppressive tumor microenvironment (TME) through the induction and functional polarization of protumoral macrophages. We have herein investigated the contribution of CAFs to monocyte recruitment and macrophage polarization. We also sought to identify a possible paracrine mechanism by which CAF-educated monocytes affect breast cancer (BC) cell progression. METHODS: Monocytes were educated by primary CAFs and normal fibroblast (NF); the phenotypic alterations of CAF- or NF-educated monocytes were measured by flow cytometry. Exosomes isolated from the cultured conditioned media of the educated monocytes were characterized. An in vivo experiment using a subcutaneous transplantation tumor model in athymic nude mice was conducted to uncover the effect of exosomes derived from CAF- or NF-educated monocytes on breast tumor growth. Gain- and loss-of-function experiments were performed to explore the role of miR-181a in BC progression with the involvement of the AKT signaling pathway. Western blotting, enzyme-linked immunosorbent assay, RT-qPCR, flow cytometry staining, migration assay, immunohistochemical staining, and bioinformatics analysis were performed to reveal the underlying mechanisms. RESULTS: We illustrated that primary CAFs recruited monocytes and established pro-tumoral M2 macrophages. CAF may also differentiate human monocyte THP-1 cells into anti-inflammatory M2 macrophages. Besides, we revealed that CAFs increased reactive oxygen species (ROS) generation in THP-1 monocytes, as differentiating into M2 macrophages requires a level of ROS for proper polarization. Importantly, T-cell proliferation was suppressed by CAF-educated monocytes and their exosomes, resulting in an immunosuppressive TME. Interestingly, CAF-activated, polarized monocytes lost their tumoricidal abilities, and their derived exosomes promoted BC cell proliferation and migration. In turn, CAF-educated monocyte exosomes exhibited a significant promoting effect on BC tumorigenicity in vivo. Of clinical significance, we observed that up-regulation of circulating miR-181a in BC was positively correlated with tumor aggressiveness and found a high level of this miRNA in CAF-educated monocytes and their exosomes. We further clarified that the pro-oncogenic effect of CAF-educated monocytes may depend in part on the exosomal transfer of miR-181a through modulating the PTEN/Akt signaling axis in BC cells. CONCLUSIONS: Our findings established a connection between tumor stromal communication and tumor progression and demonstrated an inductive function for CAF-educated monocytes in BC cell progression. We also proposed a supporting model in which exosomal transfer of miR-181a from CAF-educated monocytes activates AKT signaling by regulating PTEN in BC cells.


Assuntos
Neoplasias da Mama , Fibroblastos Associados a Câncer , MicroRNAs , Monócitos , Microambiente Tumoral , Animais , Humanos , Camundongos , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Fibroblastos Associados a Câncer/imunologia , Macrófagos/imunologia , Camundongos Nus , MicroRNAs/genética , MicroRNAs/imunologia , Monócitos/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Espécies Reativas de Oxigênio , Transdução de Sinais , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
8.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-36463233

RESUMO

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Assuntos
Lesões Encefálicas Traumáticas , Animais , Masculino , Camundongos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/imunologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/imunologia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptores Imunológicos/agonistas , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Modelos Animais de Doenças , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/imunologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/imunologia , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/imunologia
9.
Can Respir J ; 2022: 8437348, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091328

RESUMO

Introduction: Vascular smooth muscle cells (VSMCs) are highly involved in airway vascular remodeling in asthma. Objectives: This study aimed to investigate the mechanisms underlying the effects of a disintegrin and metalloproteinase-33 (ADAM33) gene on the migration capacity and inflammatory cytokine secretion of VSMCs. Methods: Human aortic smooth muscle cells (HASMCs) were transfected with lentiviral vectors carrying short hairpin RNA (shRNA) targeting ADAM33 or negative control vectors. The migration capacity of HASMCs was evaluated by a transwell assay. The levels of secreted inflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA) kits. Reverse transcription-quantitative polymerase chain reaction and Western blot assays were performed to detect mRNA and protein expression levels. Results: Silencing of ADAM33 significantly inhibited the migration of HASMCs. The expression of tumor necrosis factor alpha (TNF-α) in the supernatant of HASMCs was decreased, while that of interferon gamma (IFN-γ) was increased after the transfection of shRNA targeting ADAM33. Insufficient ADAM33 expression also suppressed the expression levels of phosphatidylinositol 3-kinase (PI3K), phospho-protein kinase B (AKT), phospho-mammalian target of rapamycin (mTOR), Rho-associated protein kinases, phospho-forkhead box protein O1 (FOXO1), and cyclin D1, but it did not affect the levels of AKT, mTOR, or Rho. Conclusion: Silencing of the ADAM33 gene inhibited HASMC migration and regulated inflammatory cytokine secretion via targeting the PI3K/AKT/mTOR pathway and its downstream signaling. These data contribute to a better understanding of the regulatory mechanisms of airway vascular remodeling in asthma.


Assuntos
Proteínas ADAM , Remodelação das Vias Aéreas , Asma , Inativação Gênica , Músculo Liso Vascular , Remodelação Vascular , Proteínas ADAM/genética , Proteínas ADAM/imunologia , Remodelação das Vias Aéreas/genética , Remodelação das Vias Aéreas/imunologia , Asma/genética , Asma/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Inativação Gênica/fisiologia , Humanos , Músculo Liso Vascular/imunologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Remodelação Vascular/genética , Remodelação Vascular/imunologia
10.
Cell Rep ; 39(13): 111019, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35767961

RESUMO

Binding of BAFF to BAFFR activates in mature B cells PI3K/AKT signaling regulating protein synthesis, metabolic fitness, and survival. In humans, naive and memory B cells express the same levels of BAFFR, but only memory B cells seem to survive without BAFF. Here, we show that BAFF activates PI3K/AKT only in naive B cells and changes the expression of genes regulating migration, proliferation, growth, and survival. BAFF-induced PI3K/AKT activation requires direct interactions between BAFFR and the B cell antigen receptor (BCR) components CD79A and CD79B and is enhanced by the AKT coactivator TCL1A. Compared to memory B cells, naive B cells express more surface BCRs, which interact better with BAFFR than IgG or IgA, thus allowing stronger responses to BAFF. As ablation of BAFFR in naive and memory B cells causes cell death independent of BAFF-induced signaling, BAFFR seems to act also as an intrinsic factor for B cell survival.


Assuntos
Receptor do Fator Ativador de Células B , Células B de Memória , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Receptores de Antígenos de Linfócitos B , Fator Ativador de Células B/imunologia , Fator Ativador de Células B/metabolismo , Receptor do Fator Ativador de Células B/imunologia , Receptor do Fator Ativador de Células B/metabolismo , Humanos , Células B de Memória/imunologia , Células B de Memória/metabolismo , Fosfatidilinositol 3-Quinases/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo
11.
J Immunol ; 208(8): 1912-1923, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35379745

RESUMO

The mechanism regulating the life span of short-lived plasma cells (SLPCs) remains poorly understood. Here we demonstrated that the EP4-mediated activation of AKT by PGE2 was required for the proper control of inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) hyperactivation and hence the endoplasmic reticulum (ER) homeostasis in IgM-producing SLPCs. Disruption of the PGE2-EP4-AKT signaling pathway resulted in IRE1α-induced activation of JNK, leading to accelerated death of SLPCs. Consequently, Ptger4-deficient mice (C57BL/6) exhibited a markedly impaired IgM response to T-independent Ags and increased susceptibility to Streptococcus pneumoniae infection. This study reveals a highly selective impact of the PGE2-EP4 signal on the humoral immunity and provides a link between ER stress response and the life span of SLPCs.


Assuntos
Sobrevivência Celular , Dinoprostona , Estresse do Retículo Endoplasmático , Endorribonucleases , Plasmócitos , Proteínas Serina-Treonina Quinases , Animais , Sobrevivência Celular/imunologia , Dinoprostona/imunologia , Estresse do Retículo Endoplasmático/imunologia , Endorribonucleases/imunologia , Imunoglobulina M/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Prostaglandinas/imunologia , Prostaglandinas E/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia
12.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163412

RESUMO

Protein kinase B2 (AKT2) is involved in various cardiomyocyte signaling processes, including those important for survival and metabolism. Coxsackievirus B3 (CVB3) is one of the most common pathogens that cause myocarditis in humans. The role of AKT2 in CVB3 infection is not yet well understood. We used a cardiac-specific AKT2 knockout (KO) mouse to determine the role of AKT2 in CVB3-mediated myocarditis. CVB3 was injected intraperitoneally into wild-type (WT) and KO mice. The mice's survival rate was recorded: survival in KO mice was significantly decreased compared with WT mice (WT vs. KO: 73.3 vs. 27.1%). Myocardial damage and inflammation were significantly increased in the hearts of KO mice compared with those of WT mice. Moreover, from surface ECG, AKT2 KO mice showed a prolonged atria and ventricle conduction time (PR interval, WT vs. KO: 47.27 ± 1.17 vs. 64.79 ± 7.17 ms). AKT2 deletion induced severe myocarditis and cardiac dysfunction due to CVB3 infection. According to real-time PCR, the mRNA level of IL-1, IL-6, and TNF-α decreased significantly in KO mice compared with WT mice on Days 5 after infection. In addition, innate immune response antiviral effectors, Type I interferon (interferon-α and ß), and p62, were dramatically suppressed in the heart of KO mice. In particular, the adult cardiac myocytes isolated from the heart showed high induction of TLR4 protein in KO mice in comparison with WT. AKT2 deletion suppressed the activation of Type I interferon and p62 transcription in CVB3 infection. In cardiac myocytes, AKT2 is a key signaling molecule for the heart from damage through the activation of innate immunity during acute myocarditis.


Assuntos
Enterovirus Humano B/imunologia , Infecções por Enterovirus/imunologia , Imunidade Inata , Miocardite/imunologia , Miocárdio/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Doença Aguda , Animais , Enterovirus Humano B/genética , Infecções por Enterovirus/genética , Células HeLa , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/virologia , Camundongos , Camundongos Knockout , Miocardite/genética , Miocardite/virologia , Proteínas Proto-Oncogênicas c-akt/genética
13.
Int Immunopharmacol ; 102: 108389, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34920312

RESUMO

The G protein-coupled receptor MrgprX2 in mast cells is known to be a crucial receptor for pseudo-allergic reactions. MrgprX2 activation leads to elevated intracellular calcium levels and mast cell degranulation, but the underlying mechanism remains to be elucidated. Herein, we investigated the role of the phosphatidylinositol 3 kinase (PI3K)/serum-threonine kinase (AKT) signaling pathway and phospholipase C gamma (PLCγ) in mast cell degranulation mediated by MrgprX2 in LAD2 human-derived mast cells. The results showed that phosphorylated AKT (p-AKT) and PLCγ up-regulation were accompanied by an increase in intracellular calcium following activation of MrgprX2 by Compound 48/80, an inducer of mast cell degranulation. In contrast, p-AKT and PLCγ were down-regulated and intracellular calcium levels decreased after MrgprX2 knockdown. Mast cell degranulation was clearly suppressed; however, inhibiting PI3K and PLCγ phosphorylation did not influence MrgprX2 expression. The increase in calcium concentration was suppressed and mast cell degranulation was weakened. Furthermore, by inhibiting PI3K and PLCγ phosphorylation in animals, the allergic symptoms caused by C48/80 were obviously reduced. We deduced that during the mast cell degranulation observed in pseudoallergic reactions, MrgprX2 regulated intracellular calcium levels via the PI3K/AKT and PLCγ pathways.


Assuntos
Hipersensibilidade/imunologia , Mastócitos/imunologia , Proteínas do Tecido Nervoso/imunologia , Receptores Acoplados a Proteínas G/imunologia , Receptores de Neuropeptídeos/imunologia , Animais , Cálcio/metabolismo , Degranulação Celular , Linhagem Celular , Humanos , Hipersensibilidade/metabolismo , Mastócitos/metabolismo , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Fosfatidilinositol 3-Quinase/imunologia , Fosfolipase C gama/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/genética , Transdução de Sinais
14.
Pathol Res Pract ; 229: 153684, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34839095

RESUMO

BACKGROUND: The prognosis of squamous cell carcinoma of head and neck (HNSCC) patients remains relatively poor over the last years. Tobacco, alcohol and active human papillomavirus (HPV) infection are involved in HNSCC development. Akt is a serine-threonine protein kinase with main phosphorylation sites at Thr308 and Ser473, which are critical to generate a high level of Akt activity. MATERIALS AND METHODS: The aim of the study was to compare the expression and prognostic potential of total Akt and its 2 phosphorylated forms - pAkt(Ser473) and pAkt(Thr308) in relation to HPV status in HNSCC patients. The expression levels of proteins were assessed immunohistochemically. To select independent prognostic factors univariate and multivariate analyses with Cox proportional regression model were performed. RESULTS: Among HNSCC with active HPV16 infection significantly more tumors with high Akt (67.86%, p = 0.026) and low pAkt(Ser473) (64.29%, p = 0.000) expressions were found as compared to those with HPV negativity, while there was no significant difference in the pAkt(Thr308) expression level between HPV positive and negative tumors (p = 0.359). In the whole group of HNSCC patients independent favorable prognostic factors were low T stage, low pAkt(Thr308) expression, HPV16 active infection presence (for OS and DFS) and female gender (for OS only). CONCLUSIONS: Our results indicate an important role of pAkt(Thr308) as prognostic biomarker for HNSCC patients. There is a high probability that using Akt inhibitors would improve therapeutical benefits and treatment effectiveness, especially in HNSCC patients with high expression of pAkt.


Assuntos
Papillomaviridae/isolamento & purificação , Proteínas Proto-Oncogênicas c-akt/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/imunologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/virologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo
15.
Nat Immunol ; 23(2): 303-317, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34949833

RESUMO

Antigen-specific memory CD4+ T cells can persist and confer rapid and efficient protection from microbial reinfection. However, the mechanisms underlying the long-term maintenance of the memory CD4+ T cell pool remain largely unknown. Here, using a mouse model of acute infection with lymphocytic choriomeningitis virus (LCMV), we found that the serine/threonine kinase complex mammalian target of rapamycin complex 2 (mTORC2) is critical for the long-term persistence of virus-specific memory CD4+ T cells. The perturbation of mTORC2 signaling at memory phase led to an enormous loss of virus-specific memory CD4+ T cells by a unique form of regulated cell death (RCD), ferroptosis. Mechanistically, mTORC2 inactivation resulted in the impaired phosphorylation of downstream AKT and GSK3ß kinases, which induced aberrant mitochondrial reactive oxygen species (ROS) accumulation and ensuing ferroptosis-causative lipid peroxidation in virus-specific memory CD4+ T cells; furthermore, the disruption of this signaling cascade also inhibited glutathione peroxidase 4 (GPX4), a major scavenger of lipid peroxidation. Thus, the mTORC2-AKT-GSK3ß axis functions as a key signaling hub to promote the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Ferroptose/imunologia , Memória Imunológica/imunologia , Longevidade/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Alvo Mecanístico do Complexo 2 de Rapamicina/imunologia , Animais , Glicogênio Sintase Quinase 3 beta/imunologia , Peroxidação de Lipídeos/imunologia , Ativação Linfocitária/imunologia , Contagem de Linfócitos/métodos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/imunologia
16.
Acta Haematol ; 145(1): 9-17, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34515042

RESUMO

Immune thrombocytopenia (ITP) is an acquired autoimmune disorder characterized by persistent thrombocytopenia resulting from increased platelet destruction and a loss of autoimmune tolerance. The pathogenesis of ITP is highly complex. Although ITP may be effectively controlled with currently available medications in some patients, a subset of cases remain refractory. The application of mesenchymal stem cells (MSCs) for human hematopoietic stem cell transplantation has increasingly demonstrated that MSCs modulate innate or adaptive immunity, thus resulting in a tolerant microenvironment. Functional defects and immunomodulatory disorders have been observed after the use of bone marrow mesenchymal stem cells (BM-MSCs) from patients with ITP. Here, we summarize the underlying mechanisms and clinical applications of various derived MSCs for ITP treatment, focusing on the main mechanisms underlying the functional defects and immune dysfunction of BM-MSCs from patients with ITP. Functional effects associated with the activation of the p53 pathway include decreased activity of the phosphatidylinositol 3 kinase/Akt pathway and activation of the TNFAIP3/NF-κB/SMAD7 pathway. Immune dysfunction appears to be associated with an impaired ability of BM-MSCs to induce various types of immune cells in ITP. At present, research focusing on MSCs in ITP remains in preliminary stages. The application of autologous or exogenous MSCs in the clinical treatment of ITP has been attempted in only a small case study and must be validated in larger-scale clinical trials.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Mesenquimais/imunologia , Púrpura Trombocitopênica Idiopática/imunologia , Púrpura Trombocitopênica Idiopática/terapia , Transdução de Sinais/imunologia , Humanos , NF-kappa B/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/imunologia , Proteína Supressora de Tumor p53/imunologia
17.
Bioengineered ; 12(2): 12461-12469, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34931923

RESUMO

Severe mortality due to the COVID-19 pandemic resulted from the lack of effective treatment. Although COVID-19 vaccines are available, their side effects have become a challenge for clinical use in patients with chronic diseases, especially cancer patients. In the current report, we applied network pharmacology and systematic bioinformatics to explore the use of biochanin A in patients with colorectal cancer (CRC) and COVID-19 infection. Using the network pharmacology approach, we identified two clusters of genes involved in immune response (IL1A, IL2, and IL6R) and cell proliferation (CCND1, PPARG, and EGFR) mediated by biochanin A in CRC/COVID-19 condition. The functional analysis of these two gene clusters further illustrated the effects of biochanin A on interleukin-6 production and cytokine-cytokine receptor interaction in CRC/COVID-19 pathology. In addition, pathway analysis demonstrated the control of PI3K-Akt and JAK-STAT signaling pathways by biochanin A in the treatment of CRC/COVID-19. The findings of this study provide a therapeutic option for combination therapy against COVID-19 infection in CRC patients.


Assuntos
Anticarcinógenos/uso terapêutico , Antivirais/uso terapêutico , Tratamento Farmacológico da COVID-19 , Neoplasias Colorretais/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/uso terapêutico , Fitoestrógenos/uso terapêutico , Atlas como Assunto , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/virologia , Ciclina D1/genética , Ciclina D1/imunologia , Receptores ErbB/genética , Receptores ErbB/imunologia , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Janus Quinases/genética , Janus Quinases/imunologia , Redes e Vias Metabólicas/efeitos dos fármacos , Redes e Vias Metabólicas/genética , Terapia de Alvo Molecular/métodos , Família Multigênica , Farmacologia em Rede/métodos , PPAR gama/genética , PPAR gama/imunologia , Farmacogenética/métodos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/crescimento & desenvolvimento , SARS-CoV-2/patogenicidade , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais
18.
Front Immunol ; 12: 743354, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34764959

RESUMO

Macrophages are functionally plastic and can thus play different roles in various microenvironments. Testis is an immune privileged organ, and testicular macrophages (TMs) show special immunosuppressive phenotype and low response to various inflammatory stimuli. However, the underlying mechanism to maintain the immunosuppressive function of TMs remains unclear. S100A9, a small molecular Ca2+ binding protein, is associated with the immunosuppressive function of macrophages. However, no related research is available about S100A9 in mouse testis. In the present study, we explored the role of S100A9 in TMs. We found that S100A9 was expressed in TMs from postnatal to adulthood and contributed to maintaining the immunosuppressive phenotype of TMs, which is associated with the activation of PI3K/Akt pathway. S100A9 treatment promotes the polarization of bone marrow-derived macrophages from M0 to M2 in vitro. S100A9 was significantly increased in TMs following UPEC-infection and elevated S100A9 contributed to maintain the M2 polarization of TMs. Treatment with S100A9 and PI3K inhibitor decreased the proportion of M2-type TMs in control and UPEC-infected mouse. Our findings reveal a crucial role of S100A9 in maintaining the immunosuppressive function of TMs through the activation of PI3K/Akt pathway, and provide a reference for further understanding the mechanism of immunosuppressive function of TMs.


Assuntos
Calgranulina B/imunologia , Privilégio Imunológico/imunologia , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Testículo/imunologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/imunologia
19.
Inflamm Res ; 70(10-12): 1027-1042, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34652489

RESUMO

INTRODUCTION: Mitochondrial dysfunction is a common denominator of neuroinflammation recognized by neuronal oxidative stress-mediated apoptosis that is well recognized by common intracellular molecular pathway-interlinked neuroinflammation and mitochondrial oxidative stress, a feature of epileptogenesis. In addition, the neuronal damage in the epileptic brain corroborated the concept of brain injury-mediated neuroinflammation, further providing an interlink between inflammation, mitochondrial dysfunction, and oxidative stress in epilepsy. MATERIALS AND METHODS: A systematic literature review of Bentham, Scopus, PubMed, Medline, and EMBASE (Elsevier) databases was carried out to provide evidence of preclinical and clinically used drugs targeting such nuclear, cytosolic, and mitochondrial proteins suggesting that the correlation of mechanisms linked to neuroinflammation has been elucidated in the current review. Despite that, the evidence of elevated levels of inflammatory mediators and pro-apoptotic protein levels can provide the correlation of inflammatory responses often concerned with hyperexcitability attributing to the fact that mitochondrial redox mechanisms and higher susceptibilities to neuroinflammation result from repetitive recurring epileptic seizures. Therefore, providing an understanding of seizure-induced pathological changes read by activating neuroinflammatory cascades like NF-kB, RIPK, MAPK, ERK, JNK, and JAK-STAT signaling further related to mitochondrial damage promoting hyperexcitability. CONCLUSION: The current review highlights the further opportunity for establishing therapeutic interventions underlying the apparent correlation of neuroinflammation mediated mitochondrial oxidative stress might contribute to common intracellular mechanisms underlying a future prospective of drug treatment targeting mitochondrial dysfunction linked to the neuroinflammation in epilepsy.


Assuntos
Epilepsia/imunologia , Mitocôndrias/imunologia , Doenças Neuroinflamatórias/imunologia , Animais , Morte Celular , Humanos , Inflamassomos/imunologia , Neurônios/imunologia , PPAR gama/imunologia , Fosfatidilinositol 3-Quinase/imunologia , Proteínas Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Fatores de Transcrição STAT/imunologia , Proteína Desacopladora 2/imunologia
20.
Hum Immunol ; 82(12): 960-967, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34538530

RESUMO

OBJECTIVE: To explore the effect of LINC00323 on the polarization of M1 macrophages in diabetic nephropathy. To study the effect and biological mechanism of LINC00323 on the occurrence and development of diabetic nephropathy. METHODS: We used clinical samples to analyze the correlation between macrophage polarization and the occurrence and development of diabetic nephropathy. In addition, we used bioinformatics to analyze the key molecules of macrophage polarization. We then verified the key pathways that promote the M1 polarization of macrophages at the level of cell biology. And we verify the effectiveness of treatment against this target in animal experiments. RESULTS: We analyzed in clinical samples that the expression of inflammatory factors (TNF-α and IL-6) increased in patients with diabetic nephropathy. In addition, we found that the expression of M1 marker protein CD86 increased through PCR and western blot analysis. We found a key target (LINC00323) through bioinformatics. The expression of LINC00323 in patients' blood samples is also at a high level. We further explored the mechanism of LINC00323 involved in the polarization of M1 macrophages at the level of cellular molecular biology, and found that it is closely related to the PI3K/AKT signaling pathway. In animal models, we found that inhibiting the expression of LINC00323 can reduce the damage of diabetic nephropathy. CONCLUSION: We found that LINC00323 mediates the polarization of M1 macrophages through the PI3K/AKT signaling pathway. LINC00323 plays an important role in the occurrence and development of diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/imunologia , Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , RNA Longo não Codificante/imunologia , Transdução de Sinais/imunologia , Animais , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA