Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
Add more filters

Publication year range
1.
Cell ; 147(2): 436-46, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-22000020

ABSTRACT

STAT6 plays a prominent role in adaptive immunity by transducing signals from extracellular cytokines. We now show that STAT6 is required for innate immune signaling in response to virus infection. Viruses or cytoplasmic nucleic acids trigger STING (also named MITA/ERIS) to recruit STAT6 to the endoplasmic reticulum, leading to STAT6 phosphorylation on Ser(407) by TBK1 and Tyr(641), independent of JAKs. Phosphorylated STAT6 then dimerizes and translocates to the nucleus to induce specific target genes responsible for immune cell homing. Virus-induced STAT6 activation is detected in all cell-types tested, in contrast to the cell-type specific role of STAT6 in cytokine signaling, and Stat6(-/-) mice are susceptible to virus infection. Thus, STAT6 mediates immune signaling in response to both cytokines at the plasma membrane, and virus infection at the endoplasmic reticulum.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , RNA Virus Infections/immunology , RNA Viruses , STAT6 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , STAT6 Transcription Factor/genetics
2.
J Transl Med ; 22(1): 672, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033271

ABSTRACT

BACKGROUND: T cells play a pivotal role in chemotherapy-triggered anti-tumor effects. Emerging evidence underscores the link between impaired anti-tumor immune responses and resistance to paclitaxel therapy in triple-negative breast cancer (TNBC). Tumor-related endothelial cells (ECs) have potential immunoregulatory activity. However, how ECs regulate T cell activity during TNBC chemotherapy remains poorly understood. METHODS: Single-cell analysis of ECs in patients with TNBC receiving paclitaxel therapy was performed using an accessible single-cell RNA sequencing (scRNA-seq) dataset to identify key EC subtypes and their immune characteristics. An integrated analysis of a tumor-bearing mouse model, immunofluorescence, and a spatial transcriptome dataset revealed the spatial relationship between ECs, especially Tumor necrosis factor receptor (TNFR) 2+ ECs, and CD8+ T cells. RNA sequencing, CD8+ T cell proliferation assays, flow cytometry, and bioinformatic analyses were performed to explore the immunosuppressive function of TNFR2 in ECs. The downstream metabolic mechanism of TNFR2 was further investigated using RNA sequencing, cellular glycolysis assays, and western blotting. RESULTS: In this study, we identified an immunoregulatory EC subtype, characterized by enhanced TNFR2 expression in non-responders. By a mouse model of TNBC, we revealed a dynamic reduction in the proportion of the CD8+ T cell-contacting tumor vessels that could co-localize spatially with CD8+ T cells during chemotherapy and an increased expression of TNFR2 by ECs. TNFR2 suppresses glycolytic activity in ECs by activating NF-κB signaling in vitro. Tuning endothelial glycolysis enhances programmed death-ligand (PD-L) 1-dependent inhibitory capacity, thereby inducing CD8+ T cell suppression. In addition, TNFR2+ ECs showed a greater spatial affinity for exhausted CD8+ T cells than for non-exhausted CD8+ T cells. TNFR2 blockade restores impaired anti-tumor immunity in vivo, leading to the loss of PD-L1 expression by ECs and enhancement of CD8+ T cell infiltration into the tumors. CONCLUSIONS: These findings reveal the suppression of CD8+ T cells by ECs in chemoresistance and indicate the critical role of TNFR2 in driving the immunosuppressive capacity of ECs via tuning glycolysis. Targeting endothelial TNFR2 may serve as a potent strategy for treating TNBC with paclitaxel.


Subject(s)
CD8-Positive T-Lymphocytes , Drug Resistance, Neoplasm , Endothelial Cells , Glycolysis , Receptors, Tumor Necrosis Factor, Type II , Triple Negative Breast Neoplasms , Receptors, Tumor Necrosis Factor, Type II/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Glycolysis/drug effects , Animals , Humans , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Female , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Mice , Signal Transduction/drug effects
3.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891789

ABSTRACT

This review addresses the role of tight junction proteins at the blood-brain barrier (BBB). Their expression is described, and their role in physiological and pathological processes at the BBB is discussed. Based on this, new approaches are depicted for paracellular drug delivery and diagnostics in the treatment of cerebral diseases. Recent data provide convincing evidence that, in addition to its impairment in the course of diseases, the BBB could be involved in the aetiology of CNS disorders. Further progress will be expected based on new insights in tight junction protein structure and in their involvement in signalling pathways.


Subject(s)
Blood-Brain Barrier , Tight Junction Proteins , Tight Junctions , Blood-Brain Barrier/metabolism , Humans , Tight Junction Proteins/metabolism , Animals , Tight Junctions/metabolism , Central Nervous System Diseases/metabolism , Signal Transduction
4.
Angiogenesis ; 26(1): 19-36, 2023 02.
Article in English | MEDLINE | ID: mdl-35829860

ABSTRACT

Tumor-induced lymphangiogenesis promotes the formation of new lymphatic vessels, contributing to lymph nodes (LNs) metastasis of tumor cells in both mice and humans. Vessel sprouting appears to be a critical step in this process. However, how lymphatic vessels sprout during tumor lymphangiogenesis is not well-established. Here, we report that S100A4 expressed in lymphatic endothelial cells (LECs) promotes lymphatic vessel sprouting in a growing tumor by regulating glycolysis. In mice, the loss of S100A4 in a whole body (S100A4-/-), or specifically in LECs (S100A4ΔLYVE1) leads to impaired tumor lymphangiogenesis and disrupted metastasis of tumor cells to sentinel LNs. Using a 3D spheroid sprouting assay, we found that S100A4 in LECs was required for the lymphatic vessel sprouting. Further investigations revealed that S100A4 was essential for the position and motility of tip cells, where it activated AMPK-dependent glycolysis during lymphatic sprouting. In addition, the expression of S100A4 in LECs was upregulated under hypoxic conditions. These results suggest that S100A4 is a novel regulator of tumor-induced lymphangiogenesis. Targeting S100A4 in LECs may be a potential therapeutic strategy for lymphatic tumor metastasis.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Mice , Humans , Animals , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Lymphangiogenesis/physiology , Lymphatic Metastasis/pathology , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism
5.
Eur J Immunol ; 52(6): 978-993, 2022 06.
Article in English | MEDLINE | ID: mdl-35340022

ABSTRACT

Progressive loss of effector functions, especially IFN-γ secreting capability, in effector memory CD8+ T (CD8+ TEM ) cells plays a crucial role in asthma worsening. However, the mechanisms of CD8+ TEM cell dysfunction remain elusive. Here, we report that S100A4 drives CD8+ TEM cell dysfunction, impairing their protective memory response and promoting asthma worsening in an ovalbumin (OVA)-induced asthmatic murine model. We find that CD8+ TEM cells contain two subsets based on S100A4 expression. S100A4+ subsets exhibit dysfunctional effector phenotypes with increased proliferative capability, whereas S100A4- subsets retain effector function but are more inclined to apoptosis, giving rise to a dysfunctional CD8+ TEM cell pool. Mechanistically, S100A4 upregulation of mitochondrial metabolism results in a decrease of acetyl-CoA levels, which impair the transcription of effector genes, especially ifn-γ, facilitating cell survival, tolerance, and memory potential. Our findings thus reveal general insights into how S100A4+ CD8+ TEM cells reprogram into dysfunctional and less protective phenotypes to aggravate asthma.


Subject(s)
Asthma , CD8-Positive T-Lymphocytes , Animals , Asthma/metabolism , CD8-Positive T-Lymphocytes/metabolism , Immune Tolerance , Immunologic Memory/genetics , Interferon-gamma/metabolism , Mice , Ovalbumin/metabolism
6.
Eur J Immunol ; 52(5): 737-752, 2022 05.
Article in English | MEDLINE | ID: mdl-35245389

ABSTRACT

Resident memory T lymphocytes (TRM ) of epithelial tissues and the Bm protect their host tissue. To what extent these cells are mobilized and contribute to systemic immune reactions is less clear. Here, we show that in secondary immune reactions to the measles-mumps-rubella (MMR) vaccine, CD4+ TRM are mobilized into the blood within 16 to 48 h after immunization in humans. This mobilization of TRM is cognate: TRM recognizing other antigens are not mobilized, unless they cross-react with the vaccine. We also demonstrate through methylome analyses that TRM are mobilized from the Bm. These mobilized cells make significant contribution to the systemic immune reaction, as evidenced by their T-cell receptor Vß clonotypes represented among the newly generated circulating memory T-cells, 14 days after vaccination. Thus, TRM of the Bm confer not only local, but also systemic immune memory.


Subject(s)
Immunologic Memory , Vaccines , Bone Marrow , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Humans
7.
Biochem Biophys Res Commun ; 668: 8-18, 2023 08 06.
Article in English | MEDLINE | ID: mdl-37230046

ABSTRACT

PURPOSE: MicroRNAs (miRNAs) are dominant cargo in exosomes and act as master regulators of cell function, inhibiting mRNA translation and affecting gene silencing. Some aspects of tissue-specific miRNA transport in bladder cancer (BC) and its role in cancer progression are not fully understood. MATERIALS AND METHODS: A microarray was used to identify miRNAs in mouse bladder carcinoma cell line MB49 exosomes. Real-time reverse transcription polymerase chain reaction was used to examine the expression of miRNAs in BC and healthy donor serum. Western blotting and immunohistochemical staining were used to examine the expression of dexamethasone-induced protein (DEXI) in patients with BC. CRISPR-Cas 9 was used to knock out Dexi in MB49, and flow cytometry was performed to test cell proliferation ability and apoptosis under chemotherapy. Human BC organoid culture, miR-3960 transfection, and 293T-exosome-loaded miR-3960 delivery were used to analyze the effect of miR-3960 on BC progression. RESULTS: The results showed that miR-3960 levels in BC tissue were positively correlated with patient survival time. Dexi was a major target of miR-3960. Dexi knockout inhibited MB49 cell proliferation and promoted cisplatin- and gemcitabine-induced apoptosis. Transfection of miR-3960 mimic inhibited DEXI expression and organoid growth. In parallel, 293T-exosome-loaded miR-3960 delivery and Dexi knockout significantly inhibited subcutaneous growth of MB49 cells in vivo. CONCLUSION: Our results demonstrate the potential role of miR-3960-mediated inhibition of DEXI as a therapeutic strategy against BC.


Subject(s)
MicroRNAs , Urinary Bladder Neoplasms , Animals , Humans , Mice , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
8.
Phytother Res ; 37(12): 6006-6020, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37726983

ABSTRACT

Cytokine storms are the cause of complications in patients with severe COVID-19, and it becomes the target of therapy. Several natural compounds were selected to screen the inhibitory effect on T-cell proliferation by Fluorescence-Activated Cell Sorting (FACS) and cytokine production by enzyme-linked immunosorbent assay (ELISA). Open reading frame 3a (ORF3a) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulates the specific T-cell activation model in vivo and in vitro. The coculture system included the macrophage cell line RAW264.7 and splenocytes. Reactive oxygen species (ROS) levels and glycolysis in T cells were evaluated. Cinnamaldehyde effectively inhibits cytokine storms both in vitro and in vivo. It decreased inflammatory cytokine (such as IFN-γ, TNF-α, IL-6, and IL-2) production by murine peripheral blood cells upon direct stimulation with ConA, after immunization with the MHV-A59 virus or ORF3a peptide from SARS-CoV-2. Cinnamaldehyde restored the percentage of T cells, which was originally decreased in the peripheral blood and splenocytes of ORF3a-immunized mice. In a coculture system, cinnamaldehyde reduced the secretion of inflammatory cytokines from macrophages in a T-cell dependent manner. Furthermore, cinnamaldehyde decreased the ROS level in activated T cells, which in turn reduced glycolysis and the activation of T cells. Cinnamaldehyde can be used as a candidate molecule for COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Animals , Mice , Cytokine Release Syndrome/drug therapy , Reactive Oxygen Species , Open Reading Frames , Cytokines/metabolism
9.
Cancer Immunol Immunother ; 71(11): 2717-2730, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35428909

ABSTRACT

BACKGROUND: Since the lung is one of the most common sites for cancer metastasis, it could provide a suitable microenvironment for pre-metastatic niche (PMN) formation to facilitate tumor cell colonization. Regulatory T cells (Tregs) are an immunosuppressive cell type found ubiquitously in tumors and may play a crucial role in PNM formation. In this study, we investigated tumor-derived exosome (TDE)-induced Treg differentiation in the lung PMN as well as the underlying mechanisms. METHODS: TDEs were isolated from the Lewis lung carcinoma cell line (LLC-exo) and their effects on mouse pulmonary fibroblasts was investigated in vitro as well as on lung tumor formation and metastasis in a pre-injected mouse model. Immune cell populations in the lung were analyzed by flow cytometry. Expression of CCL1 and CCR8 was evaluated by immunofluorescence staining, qRT-PCR and Western blot analyses. Cytokine expression was measured using mouse cytokine arrays and ELISA. RESULTS: The number of CD4+ FoxP3+ Tregs was significantly increased in lungs in a LLC-exo pre-injected mouse model. Lung fibroblasts secreted increased amounts of CCL1 after co-culture with LLC-exo, which induced Treg differentiation by activating its specific receptor CCR8, ultimately contributing to the establishment of an immunologically tolerant PMN. Moreover, inhibiting the release of LLC-exo by GW4869, or blocking the CCL1-CCR8 axis using AZ084, suppressed Tregs differentiation and tumor metastasis in the lung. CONCLUSIONS: Collectively, our study provides a novel mechanism by which Tregs are activated to form an immunologically tolerant PMN and demonstrates a critical link among lung fibroblasts, Tregs and metastatic tumor cells.


Subject(s)
Exosomes , Neoplasms , Animals , Mice , Cell Communication , Chemokine CCL1/metabolism , Cytokines/metabolism , Exosomes/metabolism , Fibroblasts/metabolism , Forkhead Transcription Factors/metabolism , Lung/metabolism , Neoplasms/metabolism , Receptors, CCR8 , T-Lymphocytes, Regulatory , Tumor Microenvironment
10.
PLoS Pathog ; 16(12): e1009019, 2020 12.
Article in English | MEDLINE | ID: mdl-33315931

ABSTRACT

Testicular invasion and persistence are features of Zika virus (ZIKV), but their mechanisms are still unknown. Here, we showed that S100A4+ macrophages, a myeloid macrophage subpopulation with susceptibility to ZIKV infection, facilitated ZIKV invasion and persistence in the seminiferous tubules. In ZIKV-infected mice, S100A4+ macrophages were specifically recruited into the interstitial space of testes and differentiated into interferon-γ-expressing M1 macrophages. With interferon-γ mediation, S100A4+ macrophages down-regulated Claudin-1 expression and induced its redistribution from the cytosol to nucleus, thus increasing the permeability of the blood-testis barrier which facilitated S100A4+ macrophages invasion into the seminiferous tubules. Intraluminal S100A4+ macrophages were segregated from CD8+ T cells and consequently helped ZIKV evade cellular immunity. As a result, ZIKV continued to replicate in intraluminal S100A4+ macrophages even when the spermatogenic cells disappeared. Deficiencies in S100A4 or interferon-γ signaling both reduced ZIKV infection in the seminiferous tubules. These results demonstrated crucial roles of S100A4+ macrophages in ZIKV infection in testes.


Subject(s)
Macrophages/metabolism , S100 Calcium-Binding Protein A4/immunology , Zika Virus Infection/immunology , Animals , Claudin-1/genetics , Claudin-1/metabolism , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Viral , S100 Calcium-Binding Protein A4/metabolism , Seminiferous Tubules/virology , Testis/immunology , Testis/virology , Virus Replication/immunology , Virus Replication/physiology , Zika Virus/immunology , Zika Virus Infection/virology
11.
J Cell Mol Med ; 25(12): 5457-5469, 2021 06.
Article in English | MEDLINE | ID: mdl-33943003

ABSTRACT

Cancer-associated fibroblasts (CAFs) activation is crucial for the establishment of a tumour promoting microenvironment, but our understanding of CAFs activation is still limited. In this study, we found that hypoxia-inducible factor-1α (HIF-1α) was highly expressed in CAFs of human lung cancer tissues and mouse spontaneous lung tumour. Accordingly, enhancing the expression of HIF-1α in fibroblasts via hypoxia induced the conversion of normal fibroblasts into CAFs. HIF-1α-specific inhibitor or HIF-1α knockout (KO) significantly attenuated CAFs activation, which was manifested by the decreased expression of COL1A2 and α-SMA. In vivo, during tumour formation, the expression of Ki-67 and proliferating cell nuclear antigen (PCNA) in the tumour tissue with HIF-1α KO fibroblasts was significantly lower than that of normal fibroblasts. Moreover, HIF-1α in fibroblasts could activate the NF-κB signalling pathway and enhance a subsequent secretion of CCL5, thus promoting the tumour growth. In conclusion, our results suggest that HIF-1α is essential for the activation and tumour-promotion function of CAFs in lung cancer (LC). And targeting HIF-1α expression on CAFs may be a promising strategy for LC therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Lewis Lung/pathology , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cell Proliferation , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 148(6): 1519-1528, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33152113

ABSTRACT

To investigate the clinical value of Tie2-expressing monocytes (TEMs) in the early diagnosis of lung cancer and assess its correlation with angiogenesis, a total of 184 patients with non-small cell lung cancer (NSCLC), 101 patients with benign pulmonary disease (BPD), and 77 healthy controls were enrolled in our study. The distribution of TEMs in lung tissue was determined by immunofluorescence staining. Lung microvascular density was assessed by immunohistochemical staining. Receiver-operating characteristic (ROC) curve analysis was performed to assess the diagnostic value of TEM frequency. Patients with NSCLC were followed up for 26 months. We found that the TEM frequency in peripheral blood monocytes of patients with NSCLC was significantly greater than that in patients with BPD and healthy controls. TEM frequency showed a correlation with NSCLC recurrence. The majority of TEMs in tumor tissues were localized around blood vessels; tumoral TEM frequency showed a positive correlation with microvascular density. High percentage of TEMs in the peripheral blood was associated with poor overall survival. ROC curve analysis revealed the potential diagnostic value of circulating TEM frequency in NSCLC. Thus, we believe that TEM frequency is related to angiogenesis in tumor tissues and may serve as a diagnostic marker for NSCLC.


Subject(s)
Biomarkers/analysis , Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/diagnosis , Monocytes/pathology , Receptor, TIE-2/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neovascularization, Pathologic/pathology
13.
Arterioscler Thromb Vasc Biol ; 40(4): 958-972, 2020 04.
Article in English | MEDLINE | ID: mdl-32078339

ABSTRACT

OBJECTIVE: Angiocrine factors, mediating the endothelial-mural cell interaction in vascular wall construction as well as maintenance, are incompletely characterized. This study aims to investigate the role of endothelial cell-derived FSTL1 (follistatin-like protein 1) in vascular homeostasis. Approach and Results: Using conditional knockout mouse models, we show that loss of FSTL1 in endothelial cells (Fstl1ECKO) led to an increase of pulmonary vascular resistance, resulting in the heart regurgitation especially with tricuspid valves. However, this abnormality was not detected in mutant mice with Fstl1 knockout in smooth muscle cells or hematopoietic cells. We further showed that there was excessive αSMA (α-smooth muscle actin) associated with atrial endocardia, heart valves, veins, and microvessels after the endothelial FSTL1 deletion. There was also an increase in collagen deposition, as demonstrated in livers of Fstl1ECKO mutants. The SMAD3 (mothers against decapentaplegic homolog 3) phosphorylation (pSMAD3) was significantly enhanced, and pSMAD3 staining was colocalized with αSMA in vein walls, suggesting the activation of TGFß (transforming growth factor ß) signaling in vascular mural cells of Fstl1ECKO mice. Consistently, treatment with a TGFß pathway inhibitor reduced the abnormal association of αSMA with the atria and blood vessels in Fstl1ECKO mutant mice. CONCLUSIONS: The findings imply that endothelial FSTL1 is critical for the homeostasis of vascular walls, and its insufficiency may favor cardiovascular fibrosis leading to heart failure.


Subject(s)
Endothelium, Vascular/physiopathology , Fibrosis/physiopathology , Follistatin-Related Proteins/physiology , Smad3 Protein/physiology , Actins/metabolism , Animals , Disease Models, Animal , Endothelial Cells/physiology , Follistatin-Related Proteins/metabolism , Homeostasis , Humans , Mice, Knockout , Phosphorylation , Smad3 Protein/metabolism , Transforming Growth Factor beta/physiology , Tricuspid Valve Insufficiency/physiopathology , Vascular Resistance
14.
J Cell Mol Med ; 24(11): 5973-5983, 2020 06.
Article in English | MEDLINE | ID: mdl-32307910

ABSTRACT

Fibrosis is characterized by fibroblast activation, extracellular matrix (ECM) accumulation and infiltration of inflammatory cells that sometimes leads to irreversible organ dysfunction. Considerable evidence now indicates that inflammation plays a critical role in the initiation and progression of organ fibrosis. S100A4 protein, a ubiquitous member of the S100 family, has recently been discovered as a potential factor implicated in fibrotic diseases. S100A4 protein is released at inflammatory site and has a certain biological function to promote cell motility, invasion, ECM remodelling, autophagy and angiogenesis. In addition, extracellular S100A4 is also a potential causation of inflammatory processes and induces the release of cytokines and growth factors under different pathological conditions. Elevated S100A4 level in patients' serum closely correlates with disease activity in several fibrotic diseases and serves as a useful biomarker for diagnosis and monitoring disease progression. Analyses of knockout mouse models have identified a functional role of extracellular S100A4 protein in fibrotic diseases, suggesting that suppressing its expression, release or function might be a promising therapeutic strategy. This review will focus on the role of extracellular S100A4 as a key regulator of pro-inflammatory signalling pathways and its relative biological processes involved in the pathogenesis of fibrosis.


Subject(s)
Disease , Extracellular Space/metabolism , S100 Calcium-Binding Protein A4/metabolism , Animals , Fibrosis , Humans , Models, Biological , Molecular Targeted Therapy
15.
J Cell Mol Med ; 24(14): 7802-7813, 2020 07.
Article in English | MEDLINE | ID: mdl-32449597

ABSTRACT

Tumour-derived exosomes have been shown to induce pre-metastatic niche formation, favoring metastatic colonization of tumour cells, but the underlying molecular mechanism is still not fully understood. In this study, we showed that exosomes derived from the LLC cells could indeed significantly enhance their intrapulmonary colonization. Circulating LLC-derived exosomes were mainly engulfed by lung fibroblasts and led to the NF-κB signalling activation. Further studies indicated that the exosomal miR-3473b was responsible for that by hindering the NFKB inhibitor delta's (NFKBID) function. Blocking miR-3473b could reverse the exosome-mediated NF-κB activation of fibroblasts and decrease intrapulmonary colonization of lung tumour cells. Together, this study demonstrated that the miR-3473b in exosomes could mediate the interaction of lung tumour cells and local fibroblasts in metastatic sites and, therefore, enhance the metastasis of lung tumour cells.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Exosomes/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MicroRNAs/genetics , NF-kappa B/metabolism , Animals , Biological Transport , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Exosomes/ultrastructure , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Immunophenotyping , Inflammation Mediators/metabolism , Lung Neoplasms/pathology , Mice
16.
J Cell Physiol ; 235(2): 1588-1600, 2020 02.
Article in English | MEDLINE | ID: mdl-31309555

ABSTRACT

Prostate cancer (PCa) is one of the major health problems of the aging male. The roles of dysregulated microRNAs in PCa remain unclear. In this study, we mined the public published data and found that miR-487a-3p was significantly downregulated in 38 pairs of clinical prostate tumor tissues compared with the normal tissues. We further verified this result by in situ hybridization on tissue chip and quantitative real-time polymerase chain reaction (qRT-PCR) in PCa/normal cells. miR-487a-3p targeting of cyclin D1 (CCND1) was identified using bioinformatics, qRT-PCR and western blot analyses. The cellular proliferation, cell cycle, migration, and invasion were assessed by cell counting kit-8, flow cytometry analysis and transwell assay. We discovered that overexpression of miR-487a-3p suppressed PCa cell growth, migration, invasion by directly targeting CCND1. Knockdown of CCND1 in PCa cells showed similar results. Meanwhile, the expression level of CCND1 was significantly upregulated in the PCa tissues and cell lines, which presented negative correlation with the expression of miR-487a-3p. More important, we demonstrated significantly reduced growth of xenograft tumors of stable miR-487a-3p-overexpressed human PCa cells in nude mice. Taken together, for the first time, our results revealed that miR-487a-3p as a tumor suppressor of PCa could target CCND1. Our finding might reveal miR-487a-3p could be potentially contributed to the pathogenesis and a clinical biomarker or the new potential therapeutic target of PCa.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Heterografts , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Prostatic Neoplasms/pathology
17.
Arch Biochem Biophys ; 694: 108613, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33010228

ABSTRACT

Cancer-associated fibroblasts (CAFs) play an important role in tumorigenesis, development, and migration. Eliminating CAFs or reducing their tumor-promoting activity is beneficial for tumor immunotherapy. Curcumin is a natural polyphenol derived from turmeric, which has been shown to inhibit the growth of many types of tumor. In this study, we explored the effect of curcumin on prostate-CAFs and its underlying molecular mechanism. The effect of curcumin on CAFs was measured using MTT assay and plate colony formation assay. Flow cytometry was used to detect cell apoptosis, ROS, Cell cycle, and mitochondrial membrane potential (ΔΨm) changes after curcumin treatment. Western Blot was used to detect changes in expression levels of related proteins in CAFs after curcumin stimulation. Colorimetry was used to detect the change of caspase 3 activity. The mRNA levels of Bims, Puma, ATF4 and CHOP were determined by qRT-PCR. We found that curcumin induced the apoptosis and cell cycle arrest of CAFs, which is mainly caused by the ROS-mediated endoplasmic reticulum stress pathway. For mechanism, the up-regulation of ROS caused by curcumin triggers endoplasmic reticulum stress of CAFs through the PERK-eIF2α-ATF4 axis. Our study suggests that curcumin selectively inhibits prostate-CAFs by inducing apoptosis and cell cycle arrest in G2-M phase, indicating a novel application of curcumin in tumor therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cancer-Associated Fibroblasts/drug effects , Curcumin/pharmacology , Endoplasmic Reticulum Stress/drug effects , Reactive Oxygen Species/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , PC-3 Cells
18.
J Pathol ; 247(3): 320-332, 2019 03.
Article in English | MEDLINE | ID: mdl-30426505

ABSTRACT

Immune responses contribute to a large extent to heart diseases. However, it is still not clear how the key inflammatory mediator interferon-γ (IFNγ) plays a role in doxorubicin (DOX)-induced cardiomyopathy. We report here that DOX-induced heart dysfunction involves IFNγ signaling in mice. The IFNγ receptor was found to be highly expressed on cardiomyocytes, and its downstream signaling was activated in heart tissues upon DOX treatment. In vitro, IFNγ strongly aggravated the injury of cardiomyocytes exposed to DOX. Although not affecting DOX-induced cell death, IFNγ disrupted mitochondrial respiration and fatty acid oxidation in DOX-exposed cardiomyocytes. IFNγ extended the suppression of the AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) axis by DOX to a p38-dependent branch. Activation of AMPK or inhibition of p38 inhibited the enhancing effect of IFNγ on the DOX-induced cardiotoxicity and prolonged the survival time in DOX-treated mice. Taken together, our results indicate that reprogramming of cardiac metabolism by IFNγ represents a previously unidentified key step for DOX-induced cardiomyopathy. This unavoidable impact of IFNγ on cardiomyocyte metabolism during chemotherapy redirects our attention to the balance between beneficial immunosurveillance of cancer cells and unwanted toxic side-effects. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiotoxicity/immunology , Doxorubicin/toxicity , Interferon-gamma/immunology , Myocytes, Cardiac/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Cell Respiration/physiology , Cells, Cultured , Cellular Reprogramming/drug effects , Cellular Reprogramming/immunology , Doxorubicin/pharmacology , Fatty Acids/metabolism , Female , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Myocytes, Cardiac/immunology , Oxidation-Reduction , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/immunology
19.
Biochem Biophys Res Commun ; 512(3): 544-551, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30914199

ABSTRACT

Tumor metastasis accounts for most tumor-associated mortality and is closely related with stromal fibroblasts in the tumor microenvironment. It was reported that fibroblasts promoted tumor metastasis through directly leading tumor cell invasion; however, inflammatory microenvironment in the growing tumor may influence the outcome. Here, we found that the cytokine IFNγ, a key immune mediator secreted by T cells, could alter mouse lung tumor associated fibroblast-leading LLC tumor cell invasion in Matrigel. The motility of fibroblasts and adhesion with tumor cells were dramatically impaired upon IFNγ stimulation. We further found that IFNγ reduced the expression of N-cadherin on the surface of fibroblasts through upregulating SMAD7 and suppressing the downstream SMAD2 phosphorylation. N-cadherin was essential for fibroblast motility and adhesions with tumor cells. Moreover, fibroblasts could promote tumor progression and the deficiency of IFNγR signaling in fibroblasts reduced liver metastasis of LLC tumor in vivo. Collectively, our results demonstrate that IFNγ inhibits fibroblast-leading tumor cell invasion by inhibiting the motility of fibroblasts and their adhesion with tumor cells. The findings indicate that inflammatory cytokines in the tumor microenvironment may regulate the fibroblast-associated tumor metastasis.


Subject(s)
Cadherins/immunology , Fibroblasts/pathology , Interferon-gamma/immunology , Lung Neoplasms/pathology , Neoplasm Invasiveness/pathology , Animals , Cadherins/analysis , Cell Line, Tumor , Cell Movement , Cells, Cultured , Female , Fibroblasts/immunology , Lung Neoplasms/immunology , Mice, Inbred C57BL , Neoplasm Invasiveness/immunology
20.
J Neurosci ; 37(4): 871-881, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28123022

ABSTRACT

Clinical studies show that chronic pain is accompanied by memory deficits and reduction in hippocampal volume. Experimental studies show that spared nerve injury (SNI) of the sciatic nerve induces long-term potentiation (LTP) at C-fiber synapses in spinal dorsal horn, but impairs LTP in the hippocampus. The opposite changes may contribute to neuropathic pain and memory deficits, respectively. However, the cellular and molecular mechanisms underlying the functional synaptic changes are unclear. Here, we show that the dendrite lengths and spine densities are reduced significantly in hippocampal CA1 pyramidal neurons, but increased in spinal neurokinin-1-positive neurons in mice after SNI, indicating that the excitatory synaptic connectivity is reduced in hippocampus but enhanced in spinal dorsal horn in this neuropathic pain model. Mechanistically, tumor necrosis factor-alpha (TNF-α) is upregulated in bilateral hippocampus and in ipsilateral spinal dorsal horn, whereas brain-derived neurotrophic factor (BDNF) is decreased in the hippocampus but increased in the ipsilateral spinal dorsal horn after SNI. Importantly, the SNI-induced opposite changes in synaptic connectivity and BDNF expression are prevented by genetic deletion of TNF receptor 1 in vivo and are mimicked by TNF-α in cultured slices. Furthermore, SNI activated microglia in both spinal dorsal horn and hippocampus; pharmacological inhibition or genetic ablation of microglia prevented the region-dependent synaptic changes, neuropathic pain, and memory deficits induced by SNI. The data suggest that neuropathic pain involves different structural synaptic alterations in spinal and hippocampal neurons that are mediated by overproduction of TNF-α and microglial activation and may underlie chronic pain and memory deficits. SIGNIFICANCE STATEMENT: Chronic pain is often accompanied by memory deficits. Previous studies have shown that peripheral nerve injury produces both neuropathic pain and memory deficits and induces long-term potentiation (LTP) at C-fiber synapses in spinal dorsal horn (SDH) but inhibits LTP in hippocampus. The opposite changes in synaptic plasticity may contribute to chronic pain and memory deficits, respectively. However, the structural and molecular bases of these alterations of synaptic plasticity are unclear. Here, we show that the complexity of excitatory synaptic connectivity and brain-derived neurotrophic factor (BDNF) expression are enhanced in SDH but reduced in the hippocampus in neuropathic pain and the opposite changes depend on tumor necrosis factor-alpha/tumor necrosis factor receptor 1 signaling and microglial activation. The region-dependent synaptic alterations may underlie chronic neuropathic pain and memory deficits induced by peripheral nerve injury.


Subject(s)
Hippocampus/metabolism , Microglia/metabolism , Neuronal Plasticity/physiology , Peripheral Nerve Injuries/metabolism , Spinal Cord/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Hippocampus/drug effects , Hippocampus/pathology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Neuralgia/metabolism , Neuralgia/pathology , Neuronal Plasticity/drug effects , Organ Culture Techniques , Pain Measurement/drug effects , Pain Measurement/methods , Peripheral Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL