Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.341
Filtrar
1.
Cell Mol Biol Lett ; 29(1): 77, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38769475

RESUMO

BACKGROUND: Tumor-associated macrophages (TAMs) significantly influence the progression, metastasis, and recurrence of esophageal squamous cell carcinoma (ESCC). The aberrant expression of long noncoding RNAs (lncRNAs) in ESCC has been established, yet the role of lncRNAs in TAM reprogramming during ESCC progression remains largely unexplored. METHODS: ESCC TAM-related lncRNAs were identified by intersecting differentially expressed lncRNAs with immune-related lncRNAs and performing immune cell infiltration analysis. The expression profile and clinical relevance of LINC00330 were examined using the TCGA database and clinical samples. The LINC00330 overexpression and interference sequences were constructed to evaluate the effect of LINC00330 on ESCC progression. Single-cell sequencing data, CIBERSORTx, and GEPIA were utilized to analyze immune cell infiltration within the ESCC tumor microenvironment and to assess the correlation between LINC00330 and TAM infiltration. ESCC-macrophage coculture experiments were conducted to investigate the influence of LINC00330 on TAM reprogramming and its subsequent effect on ESCC progression. The interaction between LINC00330 and C-C motif ligand 2 (CCL2) was confirmed through transcriptomic sequencing, subcellular localization analysis, RNA pulldown, silver staining, RNA immunoprecipitation, and other experiments. RESULTS: LINC00330 is significantly downregulated in ESCC tissues and strongly associated with poor patient outcomes. Overexpression of LINC00330 inhibits ESCC progression, including proliferation, invasion, epithelial-mesenchymal transition, and tumorigenicity in vivo. LINC00330 promotes TAM reprogramming, and LINC00330-mediated TAM reprogramming inhibits ESCC progression. LINC00330 binds to the CCL2 protein and inhibits the expression of CCL2 and downstream signaling pathways. CCL2 is critical for LINC00330-mediated TAM reprogramming and ESCC progression. CONCLUSIONS: LINC00330 inhibited ESCC progression by disrupting the CCL2/CCR2 axis and its downstream signaling pathways in an autocrine fashion; and by impeding CCL2-mediated TAM reprogramming in a paracrine manner. The new mechanism of TAM reprogramming mediated by the LINC00330/CCL2 axis may provide potential strategies for targeted and immunocombination therapies for patients with ESCC.


Assuntos
Quimiocina CCL2 , Progressão da Doença , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Regulação Neoplásica da Expressão Gênica , RNA Longo não Codificante , Microambiente Tumoral , Macrófagos Associados a Tumor , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Carcinoma de Células Escamosas do Esôfago/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral/genética , Macrófagos Associados a Tumor/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/metabolismo , Animais , Camundongos , Feminino , Proliferação de Células/genética
2.
Int J Mol Sci ; 25(9)2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38732210

RESUMO

Investigating the role of podocytes in proteinuric disease is imperative to address the increasing global burden of chronic kidney disease (CKD). Studies strongly implicate increased levels of monocyte chemoattractant protein-1 (MCP-1/CCL2) in proteinuric CKD. Since podocytes express the receptor for MCP-1 (i.e., CCR2), we hypothesized that podocyte-specific MCP-1 production in response to stimuli could activate its receptor in an autocrine manner, leading to further podocyte injury. To test this hypothesis, we generated podocyte-specific MCP-1 knockout mice (Podo-Mcp-1fl/fl) and exposed them to proteinuric injury induced by either angiotensin II (Ang II; 1.5 mg/kg/d, osmotic minipump) or Adriamycin (Adr; 18 mg/kg, intravenous bolus). At baseline, there were no between-group differences in body weight, histology, albuminuria, and podocyte markers. After 28 days, there were no between-group differences in survival, change in body weight, albuminuria, kidney function, glomerular injury, and tubulointerstitial fibrosis. The lack of protection in the knockout mice suggests that podocyte-specific MCP-1 production is not a major contributor to either Ang II- or Adr-induced glomerular disease, implicating that another cell type is the source of pathogenic MCP-1 production in CKD.


Assuntos
Angiotensina II , Quimiocina CCL2 , Doxorrubicina , Camundongos Knockout , Podócitos , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Podócitos/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Camundongos , Masculino , Insuficiência Renal Crônica/induzido quimicamente , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Deleção de Genes , Modelos Animais de Doenças
3.
Arch Dermatol Res ; 316(5): 176, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38758283

RESUMO

Psoriasis is a chronic immune mediated inflammatory skin disease with systemic manifestations. It has been reported that caloric restriction could improve severity of psoriasis patients. However, the mechanism of intermittent fasting effects on psoriasis has not been investigated. Caloric restriction is known to reduce the number of circulating inflammatory monocytes in a CCL2-dependent manner. However, it is still unknown whether caloric restriction can improve psoriasis by regulating monocytes through CCL2. In this study, we used imiquimod (IMQ)-induced psoriasis-like mouse model to explore the effects and the mechanisms of intermittent fasting on psoriasis-like dermatitis. We found that intermittent fasting could significantly improve IMQ-induced psoriasis-like dermatitis, and reduce the number of γδT17 cells and IL-17 production in draining lymph nodes and psoriatic lesion via inhibiting proliferation and increasing death of γδT17 cells. Furthermore, intermittent fasting could significantly decrease monocytes in blood, and this was associated with decreased monocytes, macrophages and DC in psoriasis-like skin inflammation. Reduced monocytes in circulation and increased monocytes in BM of fasting IMQ-induced psoriasis-like mice is through reducing the production of CCL2 from BM to inhibit monocyte egress to the periphery. Our above data shads light on the mechanisms of intermittent fasting on psoriasis.


Assuntos
Quimiocina CCL2 , Modelos Animais de Doenças , Jejum , Imiquimode , Monócitos , Psoríase , Animais , Psoríase/imunologia , Psoríase/induzido quimicamente , Psoríase/patologia , Monócitos/imunologia , Monócitos/metabolismo , Camundongos , Jejum/sangue , Quimiocina CCL2/metabolismo , Células Th17/imunologia , Interleucina-17/metabolismo , Pele/patologia , Pele/imunologia , Humanos , Camundongos Endogâmicos C57BL , Masculino , Proliferação de Células , Restrição Calórica , Jejum Intermitente
4.
J Transl Med ; 22(1): 421, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702780

RESUMO

INTRODUCTION: Immune checkpoint inhibitors (ICIs) induce acute interstitial nephritis (AIN) in 2-5% of patients, with a clearly higher incidence when they are combined with platinum derivatives. Unfortunately, suitable disease models and non-invasive biomarkers are lacking. To fill this gap in our understanding, we investigated the renal effects of cisplatin and anti-PD-L1 antibodies in mice, assessing PD-1 renal expression and cytokine levels in mice with AIN, and then we compared these findings with those in AIN-diagnosed cancer patients. METHODS: Twenty C57BL6J mice received 200 µg of anti-PD-L1 antibody and 5 mg/kg cisplatin intraperitoneally and were compared with those receiving cisplatin (n = 6), anti-PD-L1 (n = 7), or saline (n = 6). After 7 days, the mice were euthanized. Serum and urinary concentrations of TNFα, CXCL10, IL-6, and MCP-1 were measured by Luminex. The kidney sections were stained to determine PD-1 tissue expression. Thirty-nine cancer patients with AKI were enrolled (AIN n = 33, acute tubular necrosis (ATN) n = 6), urine MCP-1 (uMCP-1) was measured, and kidney sections were stained to assess PD-1 expression. RESULTS: Cisplatin and anti PD-L1 treatment led to 40% AIN development (p = 0.03) in mice, accompanied by elevated serum creatinine and uMCP1. AIN-diagnosed cancer patients also had higher uMCP1 levels than ATN-diagnosed patients, confirming our previous findings. Mice with AIN exhibited interstitial PD-1 staining and stronger glomerular PD-1 expression, especially with combination treatment. Conversely, human AIN patients only showed interstitial PD-1 positivity. CONCLUSIONS: Only mice receiving cisplatin and anti-PDL1 concomitantly developed AIN, accompanied with a more severe kidney injury. AIN induced by this drug combination was linked to elevated uMCP1, consistently with human AIN, suggesting that uMCP1 can be potentially used as an AIN biomarker.


Assuntos
Quimiocina CCL2 , Cisplatino , Inibidores de Checkpoint Imunológico , Camundongos Endogâmicos C57BL , Nefrite Intersticial , Receptor de Morte Celular Programada 1 , Animais , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/farmacologia , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Nefrite Intersticial/urina , Nefrite Intersticial/patologia , Nefrite Intersticial/induzido quimicamente , Quimiocina CCL2/urina , Quimiocina CCL2/metabolismo , Cisplatino/efeitos adversos , Humanos , Masculino , Feminino , Glomérulos Renais/patologia , Glomérulos Renais/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Camundongos , Pessoa de Meia-Idade , Idoso , Doença Aguda
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 644-651, 2024 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-38708496

RESUMO

OBJECTIVE: To observe the effect of Shenqi Chongcao (SQCC) Formula on the ASS1/src/STAT3 signaling pathway in a rat model of lung fibrosis and explore its therapeutic mechanism. METHODS: A total of 120 male SD rats were divided equally into 5 groups, including a blank control group with saline treatment and 4 groups of rat models of idiopathic pulmonary fibrosis induced by intratracheal instillation of bleomycin. One day after modeling, the rat models were treated with daily gavage of 10 mL/kg saline, SQCC decoction (0.423 g/kg), pirfenidone (10 mL/kg), or intraperitoneal injection of arginine deiminase (ADI; 2.25 mg/kg, every 3 days) for 28 days. After the treatments, the lung tissues of the rats were collected for calculating the lung/body weight ratio, observing histopathology using HE and Masson staining, and analyzing the inflammatory cells in BALF using Giemsa staining. Serum chemokine ligand 2 (CCL2) and transforming growth factor-ß1 (TGF-ß1) levels were measured with ELISA. The protein expressions of src, p-srcTry529, STAT3, and p-STAT3Try705 and the mRNA expressions of ASS1, src and STAT3 in the lung tissues were detected using Western blotting and RT-qPCR. RESULTS: The neutrophil, macrophage and lymphocyte counts and serum levels of CCL2 and TGF-ß1 were significantly lower in SQCC, pirfenidone and ADI treatment groups than in the model group at each time point of measurement (P < 0.05). P-srcTry529 and p-STAT3Try705 protein expression levels and ASS1, src, and STAT3 mRNA in the lung tissues were also significantly lower in the 3 treatment groups than in the model group (P < 0.05). CONCLUSION: SQCC Formula can alleviate lung fibrosis in rats possibly by activating the ASS1/src/STAT3 signaling pathway in the lung tissues.


Assuntos
Medicamentos de Ervas Chinesas , Fibrose Pulmonar , Ratos Sprague-Dawley , Fator de Transcrição STAT3 , Transdução de Sinais , Animais , Fator de Transcrição STAT3/metabolismo , Ratos , Masculino , Medicamentos de Ervas Chinesas/uso terapêutico , Medicamentos de Ervas Chinesas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Modelos Animais de Doenças , Bleomicina , Quimiocina CCL2/metabolismo , Quinases da Família src/metabolismo
6.
Int J Mol Sci ; 25(9)2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38732029

RESUMO

Neointimal hyperplasia is the main cause of vascular graft failure in the medium term. Vitamin D receptor activation modulates the biology of vascular smooth muscle cells and has been reported to protect from neointimal hyperplasia following endothelial injury. However, the molecular mechanisms are poorly understood. We have now explored the impact of the selective vitamin D receptor activator, paricalcitol, on neointimal hyperplasia, following guidewire-induced endothelial cell injury in rats, and we have assessed the impact of paricalcitol or vehicle on the expression of key cell stress factors. Guidewire-induced endothelial cell injury caused neointimal hyperplasia and luminal stenosis and upregulated the expression of the growth factor growth/differentiation factor-15 (GDF-15), the cytokine receptor CD74, NFκB-inducing kinase (NIK, an upstream regulator of the proinflammatory transcription factor NFκB) and the chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2). Immunohistochemistry confirmed the increased expression of the cellular proteins CD74 and NIK. Paricalcitol (administered in doses of 750 ng/kg of body weight, every other day) had a non-significant impact on neointimal hyperplasia and luminal stenosis. However, it significantly decreased GDF-15, CD74, NIK and MCP-1/CCL2 mRNA expression, which in paricalcitol-injured arteries remained within the levels found in control vehicle sham arteries. In conclusion, paricalcitol had a dramatic effect, suppressing the stress response to guidewire-induced endothelial cell injury, despite a limited impact on neointimal hyperplasia and luminal stenosis. This observation identifies novel molecular targets of paricalcitol in the vascular system, whose differential expression cannot be justified as a consequence of improved tissue injury.


Assuntos
Anti-Inflamatórios , Quimiocina CCL2 , Ergocalciferóis , Hiperplasia , Animais , Ratos , Ergocalciferóis/farmacologia , Masculino , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Anti-Inflamatórios/farmacologia , Neointima/metabolismo , Neointima/patologia , Neointima/tratamento farmacológico , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Túnica Íntima/patologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos de Diferenciação de Linfócitos B/genética , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II
7.
Adv Drug Deliv Rev ; 209: 115318, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38643840

RESUMO

The communication between cells and their microenvironment represents an intrinsic and essential attribute that takes place in several biological processes, including tissue homeostasis and tissue repair. Among these interactions, inflammation is certainly a central biological response that occurs through cytokines and the crosstalk with their respective receptors. In particular, the interaction between CCL2 and its main receptor, CCR2, plays a pivotal role in both harmful and protective inflammatory states, including cancer-mediated inflammation. The activation of the CCL2/CCR2 axis was shown to dictate the migration of macrophages with immune-suppressive phenotype and to aggravate the progression of different cancer types. In addition, this interaction mediates metastasis formation, further limiting the potential therapeutic outcome of anti-cancer drugs. Attempts to inhibit pharmacologically the CCL2/CCR2 axis have yet to show its anti-cancer efficacy as a single agent, but it sheds light on its role as a powerful tool to selectively alleviate pro-tumorigenic and anti-repair inflammation. In this review, we will elucidate the role of CCL2/CCR2 axis in promoting cancer inflammation by activating the host pro-tumorigenic phenotype. Moreover, we will provide some insight into the potential therapeutic benefit of targeting the CCL2/CCR2 axis for cancer and inflammation using novel delivery systems, aiming to sensitize non-responders to currently approved immunotherapies and offer new combinatory approaches.


Assuntos
Quimiocina CCL2 , Inflamação , Nanomedicina , Neoplasias , Receptores CCR2 , Humanos , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL2/antagonistas & inibidores , Animais , Microambiente Tumoral/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos
8.
EMBO Rep ; 25(5): 2375-2390, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38594391

RESUMO

Cancer patients undergoing treatment with antineoplastic drugs often experience chemotherapy-induced neuropathic pain (CINP), and the therapeutic options for managing CINP are limited. Here, we show that systemic paclitaxel administration upregulates the expression of neurotrophin-3 (Nt3) mRNA and NT3 protein in the neurons of dorsal root ganglia (DRG), but not in the spinal cord. Blocking NT3 upregulation attenuates paclitaxel-induced mechanical, heat, and cold nociceptive hypersensitivities and spontaneous pain without altering acute pain and locomotor activity in male and female mice. Conversely, mimicking this increase produces enhanced responses to mechanical, heat, and cold stimuli and spontaneous pain in naive male and female mice. Mechanistically, NT3 triggers tropomyosin receptor kinase C (TrkC) activation and participates in the paclitaxel-induced increases of C-C chemokine ligand 2 (Ccl2) mRNA and CCL2 protein in the DRG. Given that CCL2 is an endogenous initiator of CINP and that Nt3 mRNA co-expresses with TrkC and Ccl2 mRNAs in DRG neurons, NT3 likely contributes to CINP through TrkC-mediated activation of the Ccl2 gene in DRG neurons. NT3 may be thus a potential target for CINP treatment.


Assuntos
Quimiocina CCL2 , Gânglios Espinais , Neuralgia , Neurônios , Neurotrofina 3 , Paclitaxel , Receptor trkC , Animais , Gânglios Espinais/metabolismo , Gânglios Espinais/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Neuralgia/genética , Paclitaxel/efeitos adversos , Paclitaxel/farmacologia , Neurotrofina 3/metabolismo , Neurotrofina 3/genética , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Feminino , Receptor trkC/metabolismo , Receptor trkC/genética , Antineoplásicos/efeitos adversos , RNA Mensageiro/metabolismo , RNA Mensageiro/genética
9.
Cancer Cell ; 42(5): 885-903.e4, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38608702

RESUMO

With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.


Assuntos
Caquexia , Citocina TWEAK , Macrófagos , Neoplasias Pancreáticas , Caquexia/metabolismo , Caquexia/etiologia , Caquexia/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/complicações , Citocina TWEAK/metabolismo , Animais , Humanos , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral , Atrofia Muscular/metabolismo , Atrofia Muscular/etiologia , Atrofia Muscular/patologia , Quimiocina CCL5/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Fatores de Necrose Tumoral/metabolismo , Receptores CCR2/metabolismo , Quimiocina CCL2/metabolismo , Camundongos Endogâmicos C57BL
10.
Int Immunopharmacol ; 133: 111877, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38608440

RESUMO

The gut microbiome plays an important role in tumor growth by regulating immune cell function. However, the role of the gut microbiome-mediated monocytes in liver metastasis remains unclear. In this study, we found that fecal microbiome transplantation (FMT) from the stool of patients with liver metastasis (LM) significantly promoted liver metastasis compared with healthy donors (HD). Monocytes were upregulated in liver tissues by the CCL2/CCR2 axis in LM patients' stool transplanted mouse model. CCL2/CCR2 inhibition and monocyte depletion significantly suppress liver metastasis. FMT using LM patients' stool enhanced the plasma lipopolysaccharides (LPS) concentration. The LPS/TLR4 signaling pathway is crucial for gut microbiome-mediated liver metastasis. These results indicated that monocytes contribute to liver metastasis via the CCL2/CCR2 axis.


Assuntos
Quimiocina CCL2 , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Neoplasias Hepáticas , Monócitos , Receptores CCR2 , Receptor 4 Toll-Like , Microbioma Gastrointestinal/imunologia , Animais , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/imunologia , Monócitos/imunologia , Quimiocina CCL2/metabolismo , Camundongos , Receptores CCR2/metabolismo , Receptor 4 Toll-Like/metabolismo , Masculino , Lipopolissacarídeos/imunologia , Camundongos Endogâmicos C57BL , Feminino , Transdução de Sinais , Linhagem Celular Tumoral , Fígado/patologia , Fígado/imunologia , Fígado/metabolismo
11.
Cytokine ; 179: 156622, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38648681

RESUMO

Tuberculosis is caused by Mycobacterium tuberculosis (M tb), which is recognized by macrophages and produces inflammatory cytokines, and chemokines at the site of infection. The present study was proposed to understand the interaction of M tb antigens, cytokines, and chemokines. We have evaluated the chemokine MCP-1 levels and its expression in PBMCs stimulated with M tb antigens Ag85A, ESAT6 and recombinant cytokines rhTNF-α, rhIFN-γ, rhTGF-ß, and rhIL-10 in active pulmonary TB (APTB) patients, household contacts (HHC) at 0 months, 6 months and healthy controls (HC). We have observed low levels of MCP-1 with Ag85A, ESAT6, and rhTNF-α stimulations in APTB 0M compared to HHC and HC (p < 0.0067, p < 0.0001, p < 0.01, p < 0.005, p < 0.0065, p < 0.0001) and significantly increased after treatment with rhTNF-α. The MCP-1 levels with rhIFN-γ were high in APTB, HHC at 0 M and significant between APTB 0 M vs. 6 M, HHC vs. HC, and HHC 0M vs. 6M (p < 0.0352, p < 0.0252, p < 0.00062). The rhTGF-ß, rhIL-10 induced high MCP-1 levels in APTB, HHC compared to HC (p < 0.0414, p < 0.0312, p < 0.004, p < 0.0001) and significantly decreased after treatment with rhIL-10 (p < 0.0001). The MCP-1 expression was low with all the stimulations in APTB 0M when compared to HC and after treatment. Whereas, HHC shown low MCP-1 expression with rhTNF-α, rhIFN-γ and Ag85A and high with rhTGF-ß, rhIL-10 and ESAT6. In conclusion, the study determined the differential expression and production of MCP-1 with M tb antigens and recombinant cytokines. Further, cohort studies are required to study these interaction to identify the high risk individuals, which might help for TB control.


Assuntos
Antígenos de Bactérias , Quimiocina CCL2 , Citocinas , Mycobacterium tuberculosis , Proteínas Recombinantes , Humanos , Antígenos de Bactérias/imunologia , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Masculino , Mycobacterium tuberculosis/imunologia , Feminino , Proteínas Recombinantes/imunologia , Adulto , Citocinas/metabolismo , Proteínas de Bactérias/imunologia , Pessoa de Meia-Idade , Interferon gama/imunologia , Interferon gama/metabolismo , Tuberculose Pulmonar/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-10/metabolismo , Interleucina-10/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Tuberculose/imunologia , Fator de Crescimento Transformador beta/imunologia
12.
Transpl Int ; 37: 12556, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38650846

RESUMO

Macrophages contribute to post-transplant lung rejection. Disulfiram (DSF), an anti-alcoholic drug, has an anti-inflammatory effect and regulates macrophage chemotactic activity. Here, we investigated DSF efficacy in suppressing acute rejection post-lung transplantation. Male Lewis rats (280-300 g) received orthotopic left lung transplants from Fisher 344 rats (minor histocompatibility antigen-mismatched transplantation). DSF (0.75 mg/h) monotherapy or co-solvent only (50% hydroxypropyl-ß-cyclodextrin) as control was subcutaneously administered for 7 days (n = 10/group). No post-transplant immunosuppressant was administered. Grades of acute rejection, infiltration of immune cells positive for CD68, CD3, or CD79a, and gene expression of monocyte chemoattractant protein and pro-inflammatory cytokines in the grafts were assessed 7 days post-transplantation. The DSF-treated group had significantly milder lymphocytic bronchiolitis than the control group. The infiltration levels of CD68+ or CD3+ cells to the peribronchial area were significantly lower in the DSF than in the control groups. The normalized expression of chemokine ligand 2 and interleukin-6 mRNA in allografts was lower in the DSF than in the control groups. Validation assay revealed interleukin-6 expression to be significantly lower in the DSF than in the control groups. DSF can alleviate acute rejection post-lung transplantation by reducing macrophage accumulation around peripheral bronchi and suppressing pro-inflammatory cytokine expression.


Assuntos
Dissulfiram , Rejeição de Enxerto , Transplante de Pulmão , Macrófagos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Animais , Transplante de Pulmão/efeitos adversos , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/imunologia , Masculino , Dissulfiram/farmacologia , Dissulfiram/uso terapêutico , Ratos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Aloenxertos , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Quimiocina CCL2/metabolismo , Pulmão/patologia , Pulmão/efeitos dos fármacos
13.
Int J Mol Sci ; 25(8)2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38673763

RESUMO

Chronic ethanol exposure often triggers neuroinflammation in the brain's reward system, potentially promoting the drive for ethanol consumption. A main marker of neuroinflammation is the microglia-derived monocyte chemoattractant protein 1 (MCP1) in animal models of alcohol use disorder in which ethanol is forcefully given. However, there are conflicting findings on whether MCP1 is elevated when ethanol is taken voluntarily, which challenges its key role in promoting motivation for ethanol consumption. Here, we studied MCP1 mRNA levels in areas implicated in consumption motivation-specifically, the prefrontal cortex, hippocampus, and striatum-as well as in the cerebellum, a brain area highly sensitive to ethanol, of C57BL/6 mice subjected to intermittent and voluntary ethanol consumption for two months. We found a significant increase in MCP1 mRNA levels in the cerebellum of mice that consumed ethanol compared to controls, whereas no significant changes were observed in the prefrontal cortex, hippocampus, or striatum or in microglia isolated from the hippocampus and striatum. To further characterize cerebellar neuroinflammation, we measured the expression changes in other proinflammatory markers and chemokines, revealing a significant increase in the proinflammatory microRNA miR-155. Notably, other classical proinflammatory markers, such as TNFα, IL6, and IL-1ß, remained unaltered, suggesting mild neuroinflammation. These results suggest that the onset of neuroinflammation in motivation-related areas is not required for high voluntary consumption in C57BL/6 mice. In addition, cerebellar susceptibility to neuroinflammation may be a trigger to the cerebellar degeneration that occurs after chronic ethanol consumption in humans.


Assuntos
Consumo de Bebidas Alcoólicas , Cerebelo , Quimiocina CCL2 , Corpo Estriado , Etanol , Hipocampo , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal , Animais , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Camundongos , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Cerebelo/metabolismo , Cerebelo/efeitos dos fármacos , Cerebelo/patologia , Masculino , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Corpo Estriado/efeitos dos fármacos , Etanol/efeitos adversos , Consumo de Bebidas Alcoólicas/efeitos adversos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/patologia , Microglia/metabolismo , Microglia/efeitos dos fármacos , Microglia/patologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/induzido quimicamente
14.
Int J Mol Sci ; 25(8)2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38674048

RESUMO

Inflammation processes of the central nervous system (CNS) play a vital role in the pathogenesis of several neurological and psychiatric disorders like depression. These processes are characterized by the activation of glia cells, such as microglia. Clinical studies showed a decrease in symptoms associated with the mentioned diseases after the treatment with anti-inflammatory drugs. Therefore, the investigation of novel anti-inflammatory drugs could hold substantial potential in the treatment of disorders with a neuroinflammatory background. In this in vitro study, we report the anti-inflammatory effects of a novel hexacyclic peptide-peptoid hybrid in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. The macrocyclic compound X15856 significantly suppressed Interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), c-c motif chemokine ligand 2 (CCL2), CCL3, C-X-C motif chemokine ligand 2 (CXCL2), and CXCL10 expression and release in LPS-treated BV2 microglial cells. The anti-inflammatory effects of the compound are partially explained by the modulation of the phosphorylation of p38 mitogen-activated protein kinases (MAPK), p42/44 MAPK (ERK 1/2), protein kinase C (PKC), and the nuclear factor (NF)-κB, respectively. Due to its remarkable anti-inflammatory properties, this compound emerges as an encouraging option for additional research and potential utilization in disorders influenced by inflammation, such as depression.


Assuntos
Anti-Inflamatórios , Lipopolissacarídeos , Microglia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Animais , Camundongos , Anti-Inflamatórios/farmacologia , Linhagem Celular , Peptoides/farmacologia , Peptoides/química , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Peptídeos/farmacologia , Peptídeos/química , Fator de Necrose Tumoral alfa/metabolismo , Quimiocina CXCL2/metabolismo , Citocinas/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Quimiocina CCL3/metabolismo , Quimiocina CCL3/genética , Compostos Macrocíclicos/farmacologia , Compostos Macrocíclicos/química
15.
Int Immunopharmacol ; 132: 112016, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38593506

RESUMO

Osteoarthritis (OA) is a low-grade inflammatory joint illness in which monocytes migrate and infiltrate synovial tissue, differentiating into the pro-inflammatory M1 macrophage phenotype. IL-17 is a proinflammatory mediator principally generated by Th17 cells, which is elevated in OA patients; nevertheless, investigators have yet to elucidate the function of IL-17 in M1 polarization during OA development. Our analysis of clinical tissues and results from the open online dataset discovered that the level of M1 macrophage markers is elevated in human OA tissue samples than in normal tissue. High-throughput screening demonstrated that MCP-1 is a potential candidate factor after IL-17 treatment in OA synovial fibroblasts (OASFs). Immunohistochemistry data revealed that the level of MCP-1 is higher in humans and mice with OA than in normal tissues. IL-17 stimulation facilitates MCP-1-dependent macrophage polarization to the M1 phenotype. It also appears that IL-17 enhances MCP-1 synthesis in human OASFs, enhancing monocyte migration via the JAK and STAT3 signaling cascades. Our findings indicate the IL-17/MCP-1 axis as a novel strategy for the remedy of OA.


Assuntos
Movimento Celular , Quimiocina CCL2 , Interleucina-17 , Macrófagos , Monócitos , Osteoartrite , Animais , Humanos , Masculino , Camundongos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/imunologia , Interleucina-17/metabolismo , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Osteoartrite/imunologia , Transdução de Sinais , Fator de Transcrição STAT3/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/patologia
16.
Environ Pollut ; 349: 123929, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38582190

RESUMO

Microcystin-LR (MC-LR) is a reproductive toxin produced by cyanobacteria in the aquatic environment and can be ingested by humans through drinking water and the food chain, posing a threat to human reproductive health. However, the toxic mechanisms and prospective interventions for MC-LR-induced ovarian dysfunction at environmental doses are unknown. The mulberry fruit is a traditional natural product of plant origin, with various pharmacological effects, such as antioxidant and anti-inflammatory effects. Here, mice were exposed to MC-LR (10, 100 µg/L) in drinking water for 90 days, during which mice were gavage 600 mg/kg/week of mulberry fruit extract (MFE). It was found that MC-LR can accumulate in mouse ovaries, causing sexual hormone disturbance, inflammatory infiltration, and ovarian pathological damage. Results from RNA-seq were shown that CCL2, a chemokine associated with inflammatory response, was significantly increased in mouse ovary after MC-LR exposure. Further investigation revealed that MC-LR exposure aggravates apoptosis of granulosa cells via the CCL2-CCR10 axis-mediated Jak/Stat pathway. Importantly, MFE can significantly ameliorate these ovarian dysfunction phenotypes by inhibiting the activation of the CCL2-CCR10 axis. This study broadened new insights into the ovarian toxicity of MC-LR and clarified the pharmacological effects of mulberry fruit on ovarian function protection.


Assuntos
Toxinas Marinhas , Microcistinas , Morus , Animais , Feminino , Microcistinas/toxicidade , Camundongos , Morus/química , Ovário/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Extratos Vegetais/farmacologia , Células da Granulosa/efeitos dos fármacos
17.
Int J Mol Sci ; 25(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38673738

RESUMO

The high content of bioactive compounds in Aronia melanocarpa fruit offers health benefits. In this study, the anti-atherosclerotic effect of Aronia extracts was assessed. The impact on the level of adhesion molecules and the inflammatory response of human umbilical vein endothelial cells (HUVECs) was shown in relation to the chemical composition and the stage of ripening of the fruits. Samples were collected between May (green, unripe) and October (red, overripe) on two farms in Poland, which differed in climate. The content of chlorogenic acids, anthocyanins, and carbohydrates in the extracts was determined using HPLC-DAD/RI. The surface expression of ICAM-1 and VCAM-1 in HUVECs was determined by flow cytometry. The mRNA levels of VCAM-1, ICAM-1, IL-6, and MCP-1 were assessed using the quantitative real-time PCR method. The farms' geographical location was associated with the quantity of active compounds in berries and their anti-atherosclerotic properties. Confirmed activity for green fruits was linked to their high chlorogenic acid content.


Assuntos
Aterosclerose , Frutas , Células Endoteliais da Veia Umbilical Humana , Photinia , Extratos Vegetais , Photinia/química , Humanos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Frutas/química , Aterosclerose/tratamento farmacológico , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Molécula 1 de Adesão Intercelular/genética , Antocianinas/farmacologia , Antocianinas/química , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ácido Clorogênico/farmacologia , Ácido Clorogênico/química , Interleucina-6/metabolismo , Interleucina-6/genética
18.
Clin Cancer Res ; 30(10): 2245-2259, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38451486

RESUMO

PURPOSE: Emerging evidence underscores the critical role of extrinsic factors within the microenvironment in protecting leukemia cells from therapeutic interventions, driving disease progression, and promoting drug resistance in acute myeloid leukemia (AML). This finding emphasizes the need for the identification of targeted therapies that inhibit intrinsic and extrinsic signaling to overcome drug resistance in AML. EXPERIMENTAL DESIGN: We performed a comprehensive analysis utilizing a cohort of ∼300 AML patient samples. This analysis encompassed the evaluation of secreted cytokines/growth factors, gene expression, and ex vivo drug sensitivity to small molecules. Our investigation pinpointed a notable association between elevated levels of CCL2 and diminished sensitivity to the MEK inhibitors (MEKi). We validated this association through loss-of-function and pharmacologic inhibition studies. Further, we deployed global phosphoproteomics and CRISPR/Cas9 screening to identify the mechanism of CCR2-mediated MEKi resistance in AML. RESULTS: Our multifaceted analysis unveiled that CCL2 activates multiple prosurvival pathways, including MAPK and cell-cycle regulation in MEKi-resistant cells. Employing combination strategies to simultaneously target these pathways heightened growth inhibition in AML cells. Both genetic and pharmacologic inhibition of CCR2 sensitized AML cells to trametinib, suppressing proliferation while enhancing apoptosis. These findings underscore a new role for CCL2 in MEKi resistance, offering combination therapies as an avenue to circumvent this resistance. CONCLUSIONS: Our study demonstrates a compelling rationale for translating CCL2/CCR2 axis inhibitors in combination with MEK pathway-targeting therapies, as a potent strategy for combating drug resistance in AML. This approach has the potential to enhance the efficacy of treatments to improve AML patient outcomes.


Assuntos
Quimiocina CCL2 , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda , Inibidores de Proteínas Quinases , Receptores CCR2 , Transdução de Sinais , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Receptores CCR2/metabolismo , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/genética , Resistencia a Medicamentos Antineoplásicos/genética , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Animais , Piridonas/farmacologia , Piridonas/uso terapêutico , Camundongos
19.
Cell Tissue Res ; 396(2): 231-243, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38438567

RESUMO

C-C motif chemokine ligand 2 (CCL2) has been reported to be expressed in the bovine endometrium during pregnancy. However, the details of its functions involved in the implantation mechanism are still not clear. The purpose of this study is to analyze the functional properties of CCL2 in the bovine endometrium and embryos. The expression of CCR2 was not different between the luteal phase and implantation phase of their endometrial tissues, but was significantly high in IFNa treated bovine endometrial stromal (BES) cells in vitro. The expressions of PGES1, PGES2, AKR1C4, and AKR1C4 were high at the implantation stage compared with the luteal stage. On the other hand, PGES2 and AKR1B1 in BEE and PGES3 and AKR1A1 in BES were significantly increased by CCL2 treatment, respectively. The expressions of PCNA and IFNt were found significantly high in the bovine trophoblastic cells (BT) treated with CCL2 compared to the control. CCL2 significantly increased the attachment rate of BT vesicles to BEE in in vitro co-culture system. The expression of OPN and ICAM-1 increased in BEE, and ICAM-1 increased in BT by CCL2 treatment, respectively. The present results indicate that CCL2 has the potential to regulate the synthesis of PGs in the endometrium and the embryo growth. In addition, CCL2 has the possibility to regulate the process of bovine embryo attachment to the endometrium by modulation of binding molecules expression.


Assuntos
Quimiocina CCL2 , Implantação do Embrião , Endométrio , Interferon Tipo I , Proteínas da Gravidez , Prostaglandinas , Animais , Bovinos , Feminino , Endométrio/metabolismo , Quimiocina CCL2/metabolismo , Implantação do Embrião/fisiologia , Prostaglandinas/metabolismo , Gravidez , Trofoblastos/metabolismo , Trofoblastos/citologia , Células Estromais/metabolismo , Receptores CCR2/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo
20.
Cell Mol Immunol ; 21(5): 495-509, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38448555

RESUMO

The overexpression of sialic acids on glycans, called hypersialylation, is a common alteration found in cancer cells. Sialylated glycans can enhance immune evasion by interacting with sialic acid-binding immunoglobulin-like lectin (Siglec) receptors on tumor-infiltrating immune cells. Here, we investigated the effect of sialylated glycans and their interaction with Siglec receptors on myeloid-derived suppressor cells (MDSCs). We found that MDSCs derived from the blood of lung cancer patients and tumor-bearing mice strongly express inhibitory Siglec receptors and are highly sialylated. In murine cancer models of emergency myelopoiesis, Siglec-E knockout in myeloid cells resulted in prolonged survival and increased tumor infiltration of activated T cells. Targeting suppressive myeloid cells by blocking Siglec receptors or desialylation strongly reduced their suppressive potential. We further identified CCL2 as a mediator involved in T-cell suppression upon interaction between sialoglycans and Siglec receptors on MDSCs. Our results demonstrated that sialylated glycans inhibit anticancer immunity by modulating CCL2 expression.


Assuntos
Quimiocina CCL2 , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides , Polissacarídeos , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Animais , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/metabolismo , Polissacarídeos/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Humanos , Quimiocina CCL2/metabolismo , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linhagem Celular Tumoral , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Ácido N-Acetilneuramínico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA