Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
MedComm (2020) ; 5(4): e528, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38606362

RESUMO

Lipid metabolic reprogramming is closely related to tumor progression with the mechanism not fully elucidated. Here, we report the immune-regulated role of lanosterol synthase (LSS), an essential enzyme in cholesterol synthesis. Database analysis and clinical sample experiments suggest that LSS was lowly expressed in colon and breast cancer tissues, which indicates poor prognosis. The biological activity of tumor cell lines and tumor progression in NOD scid gamma (NSG) mice were not affected after LSS knockdown, whereas LSS deficiency obviously aggravated tumor burden in fully immunized mice. Flow cytometry analysis showed that LSS knockdown significantly promoted the formation of tumor immunosuppressive microenvironment, characterized by the increase in M2 macrophages and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), as well as the decrease in anti-tumoral T lymphocytes. With the inhibition of myeloid infiltration or loss function of T lymphocytes, the propulsive effect of LSS knockdown on tumor progression disappeared. Mechanistically, LSS knockdown increased programmed death ligand 1 (PDL1) protein stability by 2,3-oxidosqualene (OS) binding to PDL1 protein. Anti-PDL1 therapy abolished LSS deficiency-induced immunosuppressive microenvironment and cancer progression. In conclusion, our results show that LSS deficiency promotes tumor progression by establishing an OS-PDL1 axis-dependent immunosuppressive microenvironment, indicative of LSS or OS as a potential hallmark of response to immune checkpoint blockade.

3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(2): 335-341, 2022 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-35332739

RESUMO

Cholesterol, an important lipid molecule of organisms, is involved in the formation of cell membrane structure, bile acid metabolism and steroid hormone synthesis, playing an important role in the regulation of cell structure and functions. In recent years, a large number of studies have shown that cholesterol metabolism is reprogrammed during tumor formation and development. In addition to directly affecting the biological behavior of tumor cells, cholesterol metabolic reprogramming also regulates the antitumor activity of immune cells in the tumor microenvironment. We reviewed herein the cholesterol metabolism reprogramming of and interactions among immune cells including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), dendritic cells (DCs), and T cells in the tumor microenvironment. However, the relationship between cholesterol metabolism and tumor immunity in tumor microenvironment is complex and diversified. The differences and similarities of cholesterol metabolism reprogramming in tumor microenvironment in regulating immune cell activity and the specific regulatory mechanism are still unresolved issues. Targeted intervention of the cholesterol metabolism pathway of immune cells is expected to become a new strategy of cholesterol metabolism in tumor immunotherapy.


Assuntos
Células Supressoras Mieloides , Neoplasias , Humanos , Imunoterapia , Metabolismo dos Lipídeos , Células Supressoras Mieloides/metabolismo , Células Supressoras Mieloides/patologia , Microambiente Tumoral
4.
Theranostics ; 12(2): 875-890, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976218

RESUMO

Background: 5-aza-2'-deoxycytidine (5Aza), a DNA methyltransferase (DNMT) inhibitor, could activate tumor adaptive immunity to inhibit tumor progression. However, the molecular mechanisms by which 5Aza regulates tumor immune microenvironment are still not fully understood. Methods: The role of 5Aza in immune microenvironment of peritoneal carcinomatosis (PC) of colorectal cancer (CRC) was investigated. The effects of 5Aza on macrophage activation were studied by flow cytometry, real-time PCR, Western blotting assays, and Drug Affinity Responsive Target Stability (DARTS). The effects of 5Aza on tumor immunity were validated in stromal macrophages and T cells from CRC patients. Results: 5Aza could stimulate the activation of macrophages toward an M1-like phenotype and subsequent activation of T cells in premetastatic fat tissues, and ultimately suppress CRC-PC in immune-competent mouse models. Mechanistically, 5Aza stimulated primary mouse macrophages toward to a M1-like phenotype characterized by the increase of p65 phosphorylation and IL-6 expression. Furthermore, we screened and identified ATP-binding cassette transporter A9 (ABC A9) as a binding target of 5Aza. 5Aza induced cholesterol accumulation, p65 phosphorylation and IL-6 expression in an ABC A9-dependent manner. Pharmacological inhibition of NF-κB, or genetic depletion of IL-6 abolished the antitumor effect of 5Aza in mice. In addition, the antitumor effect of 5Aza was synergistically potentiated by conventional chemotherapeutic drugs 5-Fu or OXP. Finally, we validated the reprogramming role of 5Aza in antitumor immunity in stromal macrophages and T cells from CRC patients. Conclusions: Taken together, our findings showed for the first time that 5Aza suppressed CRC-PC by regulating macrophage-dependent T cell activation in premetastatic microenvironment, meanwhile uncovered a DNA methylation-independent mechanism of 5Aza in regulating ABC A9-associated cholesterol metabolism and macrophage activation.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Colesterol/metabolismo , Neoplasias Colorretais/imunologia , Decitabina/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Metástase Neoplásica/imunologia , Neoplasias Peritoneais/imunologia , Animais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Humanos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Peritoneais/dietoterapia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 52(1): 45-49, 2021 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-33474888

RESUMO

Tumor associated macrophages (TAMs) are one of the most common types of stromal cells in solid tumors. They are closely related to the immunosuppressive status of tumor microenvironment and potentiate the malignant progress of tumors. Studies have shown that metabolism in tumor associated macrophages has been reprogrammed and involved in the regulation of their own polarization and corresponding functions and phenotypes. Metabolic reprogramming refers to the alteration of key enzymes activity, substrate and its associated metabolites' concentration in a certain metabolic pathway, which accounts for the disorder of original metabolic states. In this paper, we mainly concentrated on the lipid metabolic reprogramming of TAMs, including triglycerides, fatty acids and their derivatives, cholesterol, phospholipids, and their regulations on tumor progression. However, the metabolism of tumor and tumor microenvironment cells is highly heterogeneous. It is worthy of further exploration on the similarities and differences of lipid metabolism reprogramming between stromal cells and tumor cells, and the mechanism of how reprogramming modulates cell activity. It will be a new strategy for immunotherapy of tumor with metabolic intervention to accurately target the lipid metabolism reprogramming of TAMs, so as to promote the polarization of TAMs to M1 like macrophages, when synthetically considering the diverse types of tumors and different stages of development.


Assuntos
Metabolismo dos Lipídeos , Neoplasias , Humanos , Macrófagos , Microambiente Tumoral , Macrófagos Associados a Tumor
6.
Signal Transduct Target Ther ; 5(1): 239, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060562

RESUMO

Peritoneal carcinomatosis (PC) of colorectal cancer (CRC) is a terminal phase of malignancy with no effective strategies for the prevention of this condition. Here we established PC models in mice by intraperitoneal engraftment of CRC cells and revealed an unexpected role for a high-fat diet (HFD) in preventing metastatic seeding in the visceral fat. Mechanistically, the HFD stimulated the activation of adipose tissue macrophages (ATMs) toward an M1-like phenotype and enhanced ATM tumor phagocytosis in a TLR4-dependent manner. Furthermore, the TLR4-Cxcl10 axis in ATMs promoted T cell recruitment, and M1-like macrophages stimulated T cell activation in tumor-seeded fats. The inhibitory effect of the HFD on tumor seeding was abolished with the ablation of macrophages, inactivation of T cells, or blockade of the TLR4-Cxcl10 axis in macrophages. Finally, we showed that a HFD and conventional chemotherapeutic agents (oxaliplatin or 5-fluorouracil) synergistically improved the survival of tumor-seeded mice. Collectively, our findings demonstrate that peritoneal seeding of CRC can be suppressed by short-term treatment with a HFD in the early phase, providing a novel concept for the management of these patients in the clinic.


Assuntos
Tecido Adiposo/imunologia , Quimiocina CXCL10/imunologia , Neoplasias Colorretais/imunologia , Gorduras na Dieta/farmacologia , Macrófagos/imunologia , Neoplasias Peritoneais/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Metástase Neoplásica , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/prevenção & controle , Neoplasias Peritoneais/secundário , Fagocitose , Receptor 4 Toll-Like/genética
8.
Signal Transduct Target Ther ; 5(1): 24, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32296017

RESUMO

Lipid metabolic reprogramming plays an essential role in regulating the progression of colorectal cancer (CRC). However, the effect of lysophosphatidic acid (LPA) metabolism on CRC development is incompletely characterized. Here, we compared the mRNA levels of human CRC tissues to those of paracarcinoma tissues and focused on the notably enriched LPA metabolic pathways. We identified and verified that 1-acylglycerol-3-phosphate O-acyltransferase 4 (Agpat4) was aberrantly expressed in CRC tissues and predicted poor survival in CRC patients. Manipulating Agpat4 expression in CRC cells did not affect the growth or migration of CRC cells in vitro, whereas Agpat4 silencing suppressed CRC cell growth in subcutaneous and peritoneal xenograft models. Mechanistically, Agpat4 silencing-induced LPA release from CRC cells and polarized macrophages to an M1-like phenotype through LPA receptors 1 and 3. This M1 activation, characterized by elevated p38/p65 signaling and increased proinflammatory cytokines, promoted the infiltration and activation of CD4+ and CD8+ T cells in the tumor microenvironment. Modulation of the Agpat4/LPA/p38/p65 axis regulated macrophage polarization, T-cell activity and CRC progression. Notably, combined therapy with LPA and regular chemotherapy drugs synergistically suppressed CRC development. Taken together, our results showed that the Agpat4/LPA axis in CRC cells regulated p38/p65 signaling-dependent macrophage polarization, T-cell activation, and CRC progression. The Agpat4/LPA/p38/p65 axis might represent a potential target for therapy in the clinic.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Neoplasias Colorretais/genética , Receptores de Ácidos Lisofosfatídicos/genética , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Lisofosfolipídeos/genética , Lisofosfolipídeos/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , RNA Mensageiro/genética , Transdução de Sinais/genética , Linfócitos T/metabolismo , Linfócitos T/patologia , Microambiente Tumoral/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
9.
MedComm (2020) ; 1(1): 47-68, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34766109

RESUMO

Tumor microenvironment is a special environment for tumor survival, which is characterized by hypoxia, acidity, nutrient deficiency, and immunosuppression. The environment consists of the vasculature, immune cells, extracellular matrix, and proteins or metabolic molecules. A large number of recent studies have shown that not only tumor cells but also the immune cells in the tumor microenvironment have undergone metabolic reprogramming, which is closely related to tumor drug resistance and malignant progression. Tumor immunotherapy based on T cells gives patients new hope, but faces the dilemma of low response rate. New strategies sensitizing cancer immunotherapy are urgently needed. Metabolic reprogramming can directly affect the biological activity of tumor cells and also regulate the differentiation and activation of immune cells. The authors aim to review the characteristics of tumor microenvironment, the metabolic changes of tumor-associated immune cells, and the regulatory role of metabolic reprogramming in cancer immunotherapy.

10.
Cancer Res ; 79(21): 5513-5526, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31439546

RESUMO

Metabolic reprogramming in tumor-associated macrophages (TAM) is associated with cancer development, however, the role of macrophage triglyceride metabolism in cancer metastasis is unclear. Here, we showed that TAMs exhibited heterogeneous expression of abhydrolase domain containing 5 (ABHD5), an activator of triglyceride hydrolysis, with migratory TAMs expressing lower levels of ABHD5 compared with the nonmigratory TAMs. ABHD5 expression in macrophages inhibited cancer cell migration in vitro in xenograft models and in genetic cancer models. The effects of macrophage ABHD5 on cancer cell migration were dissociated from its metabolic function as neither triglycerides nor ABHD5-regulated metabolites from macrophages affected cancer cell migration. Instead, ABHD5 deficiency in migrating macrophages promoted NFκB p65-dependent production of matrix metalloproteinases (MMP). ABHD5 expression negatively correlated with MMP expression in TAMs and was associated with better survival in patients with colorectal cancer. Taken together, our findings show that macrophage ABHD5 suppresses NFκB-dependent MMP production and cancer metastasis and may serve as a prognostic marker in colorectal cancer. SIGNIFICANCE: These findings highlight the mechanism by which reduced expression of the metabolic enzyme ABHD5 in macrophages promotes cancer metastasis.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/21/5513/F1.large.jpg.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Metaloproteinases da Matriz/metabolismo , NF-kappa B/metabolismo , Metástase Neoplásica/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos/metabolismo , Xenoenxertos/patologia , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Microambiente Tumoral/fisiologia
11.
J Cancer ; 10(2): 367-377, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30719130

RESUMO

Colorectal cancer (CRC) usually gives rise to transcoelomic spread and ultimately causes peritoneal carcinomatosis (PC). However, mechanism studies, especially the immunological basis of colorectal PC, are rarely revealed due to lack of a suitable PC model. Here we selected a mouse colorectal cancer cell line MC-38 for intraperitoneal inoculation in the C57BL/6 mice to mimic the development of colorectal PC. We demonstrated that the injected CRC cells preferentially and rapidly migrated and colonized in the visceral fat tissues, but not in other visceral organs. With flow cytometric analysis, we found the proportions of spleen T cells and B cells were not affected by PC progression, while the ratios of blood CD4+ and CD8+ T cells were largely influenced. Especially, the quantity or activity of CD4+ and CD8+ T cells in visceral fats were intimately regulated by PC development. Taken together, we successfully constructed a colorectal PC model in immune-competent mice and revealed the alteration of adaptive immunity in PC development. Our study might potentiate the research and therapy strategies of colorectal PC.

12.
Nat Commun ; 9(1): 2574, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29968710

RESUMO

Metabolic reprogramming greatly contributes to the regulation of macrophage activation. However, the mechanism of lipid accumulation and the corresponding function in tumor-associated macrophages (TAMs) remain unclear. With primary investigation in colon cancer and confirmation in other cancer models, here we determine that deficiency of monoacylglycerol lipase (MGLL) results in lipid overload in TAMs. Functionally, macrophage MGLL inhibits CB2 cannabinoid receptor-dependent tumor progression in inoculated and genetic cancer models. Mechanistically, MGLL deficiency promotes CB2/TLR4-dependent macrophage activation, which further suppresses the function of tumor-associated CD8+ T cells. Treatment with CB2 antagonists delays tumor progression in inoculated and genetic cancer models. Finally, we verify that expression of macrophage MGLL is decreased in cancer tissues and positively correlated with the survival of cancer patients. Taken together, our findings identify MGLL as a switch for CB2/TLR4-dependent macrophage activation and provide potential targets for cancer therapy.


Assuntos
Macrófagos/imunologia , Monoacilglicerol Lipases/metabolismo , Neoplasias/patologia , Receptor CB2 de Canabinoide/metabolismo , Idoso , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Humanos , Metabolismo dos Lipídeos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Monoacilglicerol Lipases/genética , Neoplasias/imunologia , Cultura Primária de Células , Células RAW 264.7 , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/imunologia , Receptor 4 Toll-Like/metabolismo
13.
Cell Commun Signal ; 15(1): 39, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29017500

RESUMO

BACKGROUND: Chronic inflammation is causally linked to the carcinogenesis and progression of most solid tumors. LPTS is a well-identified tumor suppressor by inhibiting telomerase activity and cancer cell growth. However, whether and how LPTS is regulated by inflammation signaling is still incompletely elucidated. METHODS: Real-time PCR and western blotting were used to determine the expression of p65 and LPTS. Reporter gene assay, electrophoretic mobility shift assay and chromatin immunoprecipitation were performed to decipher the regulatory mechanism between p65 and LPTS. Cell counting kit-8 assays and xenograt models were used to detect p65-LPTS-regulated cancer cell growth in vitro and in vivo, respectively. RESULTS: Here we for the first time demonstrated that NF-κB could inhibit LPTS expression in the mRNA and protein levels in multiple cancer cells (e.g. cervical cancer and colon cancer cells). Mechanistically, NF-κB p65 could bind to two consensus response elements locating at -1143/-1136 and -888/-881 in the promoter region of human LPTS gene according to EMSA and ChIP assays. Mutation of those two binding sites rescued p65-suppressed LPTS promoter activity. Functionally, NF-κB regulated LPTS-dependent cell growth of cervical and colon cancers in vitro and in xenograft models. In translation studies, we verified that increased p65 expression was associated with decreased LPTS level in multiple solid cancers. CONCLUSIONS: Taken together, we revealed that NF-κB p65 potentiated tumor growth via suppressing a novel target LPTS. Modulation of NF-κB-LPTS axis represented a potential strategy for treatment of those inflammation-associated malignancies.


Assuntos
Terapia de Alvo Molecular , Fator de Transcrição RelA/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Proteínas Oncogênicas Virais/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
14.
Diagn Pathol ; 11(1): 72, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27506546

RESUMO

BACKGROUND: Aryl Hydrocarbon Receptor (AhR) is a ligand-activated transcription factor with multiple functions operating in a variety of organs, including the brain. Recent studies have revealed that AhR played a functional role in traumatic injuries. This paper aims to study the expression of AhR during the early phase following a traumatic brain injury (TBI) in rat brains by immunohistochemistry. METHODS: Weight-drop induced TBI was performed in rats. The expression of AhR in brain of TBI rats were examined by immunohistochemistry. RESULTS: Neuron expression of AhR in the rat brains of experiment group had been upregulated since day 3 in lesional hemisphere compared to that of the control group and mainly located in the cytoplasm, indicating an inactivated state. Interestingly, the accumulation of AhR(+) non-neuron cells became significant as early as 18 h after injury, which had kept increasing until 24 h post injury and then decreased slowly. For AhR(+) non-neuron cells, the AhR mainly located in cell nucleus, indicating a reactive status. Furthermore, double staining showed that most AhR(+) non-neuron cells co-localized with W3/13, a marker for T lymphocytes, but not with ED-1 (for activated microglia/macrophages) or GFAP (for activated astrocytes), suggesting that most AhR(+) non-neuron cells were T lymphocytes. CONCLUSION: This is the first study concerning AhR expression in brains following TBI, and our data demonstrated that AhR was upregulated and activated in T lymphocytes following TBI. More research is needed to make a more conclusive conclusion.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Regulação da Expressão Gênica , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Núcleo Celular/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Macrófagos/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Ratos , Linfócitos T/metabolismo , Regulação para Cima
15.
Nat Commun ; 7: 12177, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27397585

RESUMO

Inflammation resolution is an active process, the failure of which causes uncontrolled inflammation which underlies many chronic diseases. Therefore, endogenous pathways that regulate inflammation resolution are fundamental and of wide interest. Here, we demonstrate that phagocyte respiratory burst-induced hypoxia activates macrophage erythropoietin signalling to promote acute inflammation resolution. This signalling is activated following acute but not chronic inflammation. Pharmacological or genetical inhibition of the respiratory burst suppresses hypoxia and macrophage erythropoietin signalling. Macrophage-specific erythropoietin receptor-deficient mice and chronic granulomatous disease (CGD) mice, which lack the capacity for respiratory burst, display impaired inflammation resolution, and exogenous erythropoietin enhances this resolution in WT and CGD mice. Mechanistically, erythropoietin increases macrophage engulfment of apoptotic neutrophils via PPARγ, promotes macrophage removal of debris and enhances macrophage migration to draining lymph nodes. Together, our results provide evidences of an endogenous pathway that regulates inflammation resolution, with important implications for treating inflammatory conditions.


Assuntos
Eritropoetina/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Fagocitose , Explosão Respiratória , Animais , Movimento Celular , Hipóxia/metabolismo , Inflamação/imunologia , Linfonodos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama/metabolismo , Receptores da Eritropoetina/metabolismo , Transdução de Sinais
16.
Immunity ; 44(2): 287-302, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26872696

RESUMO

The failure of apoptotic cell clearance is linked to autoimmune diseases, nonresolving inflammation, and developmental abnormalities; however, pathways that regulate phagocytes for efficient apoptotic cell clearance remain poorly known. Apoptotic cells release find-me signals to recruit phagocytes to initiate their clearance. Here we found that find-me signal sphingosine 1-phosphate (S1P) activated macrophage erythropoietin (EPO) signaling promoted apoptotic cell clearance and immune tolerance. Dying cell-released S1P activated macrophage EPO signaling. Erythropoietin receptor (EPOR)-deficient macrophages exhibited impaired apoptotic cell phagocytosis. EPO enhanced apoptotic cell clearance through peroxisome proliferator activated receptor-γ (PPARγ). Moreover, macrophage-specific Epor(-/-) mice developed lupus-like symptoms, and interference in EPO signaling ameliorated the disease progression in lupus-like mice. Thus, we have identified a pathway that regulates macrophages to clear dying cells, uncovered the priming function of find-me signal S1P, and found a role of the erythropoiesis regulator EPO in apoptotic cell disposal, with implications for harnessing dying cell clearance.


Assuntos
Eritropoetina/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lisofosfolipídeos/metabolismo , Macrófagos/fisiologia , Receptores da Eritropoetina/metabolismo , Esfingosina/análogos & derivados , Animais , Apoptose , Linhagem Celular , Feminino , Tolerância Imunológica/genética , Lisofosfolipídeos/genética , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama/genética , PPAR gama/metabolismo , Comunicação Parácrina , Fagocitose/genética , Receptores da Eritropoetina/genética , Transdução de Sinais , Esfingosina/genética , Esfingosina/metabolismo
17.
Neurol Sci ; 37(2): 199-203, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26410085

RESUMO

Experimental autoimmune neuritis (EAN) is a well-known animal model of human demyelinating polyneuropathies. Macrophages are the major immune cells in peripheral nerves and may exert tissue-damage or tissue-protective activity during EAN. While considered to define a subpopulation of T lymphocytes, CD8 expression has been found on certain macrophages that show cytotoxic effects. Here we have studied the spatiotemporal accumulation of CD8(+) cells in sciatic nerves of EAN rats. A robust accumulation of CD8(+) cells was observed in the sciatic nerves of EAN rats, which was positively correlated with the severity of neurological signs in EAN. Moreover, double-labelling experiments showed that the major cellular sources of CD8 were reactive macrophages. Therefore, our data here suggest a pathological role of CD8(+) macrophages in EAN, which makes CD8(+) macrophage a potential therapeutic target for EAN.


Assuntos
Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Macrófagos/metabolismo , Neurite Autoimune Experimental/imunologia , Nervo Isquiático/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Macrófagos/patologia , Masculino , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
18.
Sci Rep ; 5: 15134, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26459940

RESUMO

Erythropoietin (EPO) has been identified as being crucial for obesity modulation; however, its erythropoietic activity may limit its clinical application. EPO-derived Helix B-surface peptide (pHBSP) is nonerythrogenic but has been reported to retain other functions of EPO. The current study aimed to evaluate the effects and potential mechanisms of pHBSP in obesity modulation. We found that pHBSP suppressed adipogenesis, adipokine expression and peroxisome proliferator-activated receptor γ (PPARγ) levels during 3T3-L1 preadipocyte maturation through the EPO receptor (EPOR). In addition, also through EPOR, pHBSP attenuated macrophage inflammatory activation and promoted PPARγ expression. Furthermore, PPARγ deficiency partly ablated the anti-inflammatory activity of pHBSP in macrophages. Correspondingly, pHBSP administration to high-fat diet (HFD)-fed mice significantly improved obesity, insulin resistance (IR) and adipose tissue inflammation without stimulating hematopoiesis. Therefore, pHBSP can significantly protect against obesity and IR partly by inhibiting adipogenesis and inflammation. These findings have therapeutic implications for metabolic disorders, such as obesity and diabetes.


Assuntos
Adipogenia/efeitos dos fármacos , Eritropoetina/química , Inflamação/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Peptídeos/farmacologia , Células 3T3-L1 , Adipocinas/genética , Adipocinas/metabolismo , Animais , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica , Regulação da Expressão Gênica/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Inflamação/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Obesidade/tratamento farmacológico , Obesidade/etiologia , PPAR gama/metabolismo
19.
J Neuroimmunol ; 268(1-2): 64-70, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24518674

RESUMO

ARA290 is a nonerythropoietic analog of erythropoietin (EPO) containing 11 amino acids which provides the anti-inflammatory and neuroprotective effects of EPO without stimulating hematopoiesis. Here we studied the therapeutic effects of ARA290 in experimental autoimmune encephalomyelitis (EAE) Lewis rats. Therapeutic (from Day 7 to Day 18 or from Day 9 to Day 19) administration of ARA290 (35, 70 µg/kg, intra-peritoneal) to EAE rats once daily significantly reduced the severity and shortened the duration of clinical score, reduced the accumulation of inflammatory cells in EAE spinal cords and suppressed mRNA levels of interleukin-1ß (IL-1ß), IL-17, tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), inducible nitric oxide synthase (iNOS), matrix metalloproteinase 9 (MMP9) and transcription factor T-bet in spinal cords of EAE rats. Furthermore, ARA290 treatment reduced the helper T cell number in lymph nodes and circulation in EAE. In vitro study showed that ARA290 dose-dependently inhibited antigen specific- and antigen non-specific-lymphocyte proliferation as well. In addition, ARA290 altered the cytokine milieu to favor the polarization of Th2 and regulatory T (Treg) cells but suppressed the polarization of Th1 and Th17 cells in EAE lymph nodes. In summary, our study here showed that ARA290 could alter T cell function to suppress inflammation to ameliorate EAE, suggesting that ARA290 may be a new therapeutic candidate for multiple sclerosis.


Assuntos
Anti-Inflamatórios/farmacologia , Diferenciação Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/imunologia , Oligopeptídeos/farmacologia , Animais , Encefalomielite Autoimune Experimental/patologia , Citometria de Fluxo , Imuno-Histoquímica , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/patologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos
20.
J Neurosci Res ; 92(6): 743-50, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24482305

RESUMO

Experimental autoimmune neuritis (EAN) is a helper T cell-mediated autoimmune demyelinating inflammatory disease of the peripheral nervous system that serves as an animal model for human Guillain-Barre syndrome. Curcumin, a naturally occurring polyphenolic phytochemical isolated from the medicinal plant Curcuma longa, has anti-inflammatory activities. Here we investigated the therapeutic effects and potential mechanisms of curcumin in EAN rats. Exogenous curcumin treatment (100 mg/kg/day) significantly delayed the onset of EAN neurological signs, ameliorated EAN neurological severity, and reduced body weight loss of EAN rats. In EAN sciatic nerves, curcumin treatment suppressed the inflammatory cell accumulation and the expression of interferon (IFN)-γ, tumor necrosis factor-α, interleukin (IL)-1ß, and IL-17. Furthermore, curcumin treatment significantly decreased the percentage of CD4(+) T helper cells in EAN spleen and suppressed concanavalin A-induced lymphocyte proliferation in vitro. In addition, curcumin altered helper T cell differentiation by decreasing IFN-γ(+) CD4(+) Th1 cells in EAN lymph node and spleen. In summary, our data demonstrate that curcumin could effectively suppress EAN by attenuating inflammation, indicating that curcumin might be a candidate for treatment of autoimmune neuropathies.


Assuntos
Curcumina/farmacologia , Inibidores Enzimáticos/farmacologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Citometria de Fluxo , Inflamação/imunologia , Inflamação/patologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...