Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Control Release ; 373: 447-462, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39038546

ABSTRACT

Immunosuppressive tumor-associated macrophages (TAMs) account for a high proportion of the tumor tissue and significantly impede immunoefficacy. Furthermore, the signal regulatory protein α (SIRPα) expressed in TAMs adversely correlates with macrophage activation and phagocytosis, resulting in immunosurveillance escape. To address these difficulties, a mannose-modified, pH-responsive nanoplatform with resiquimod (R848) and 2', 3'-cyclic GMP-AMP (cGAMP) co-encapsulation (named M-PNP@R@C) is designed to polarize TAMs and lower SIRPα expression. The co-delivery of R848 and cGAMP synergistically facilitates the polarization of TAMs from the anti-inflammatory M2 phenotype into the pro-inflammatory M1 phenotype, thereby enhancing antitumor immunotherapy. Remarkably, activation of the cGAMP-mediated stimulator of interferon genes (STING) in TAMs significantly downregulates the expression of SIRPα, which synergizes with the cluster of differentiation 47 (CD47) antibody for the dual blockade of the CD47-SIRPα axis. Further analysis of single-cell RNA sequencing indicates that STING activation downregulates SIRPα by regulating intracellular fatty acid oxidation metabolism. In vivo studies indicate that M-PNP@R@C significantly inhibits tumor growth with a potent antitumor immune response in melanoma graft tumor models. After synergy with anti-CD47, the double blockade strategies of the SIRPα/CD47 axis result in a notable inhibition of lung metastasis. A prolonged survival rate is observed after combination treatment with CD47 and programmed death ligand-1 antibodies for the triple immune checkpoint blockade. In summary, our study provides original insights into the potential role of the STING pathway in macrophage-based immunotherapy, thus offering a potential combinatorial strategy for cancer therapy.

2.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167314, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38936516

ABSTRACT

The integrity of the lymphatic system is critical for preventing the dissemination of tumor cells, such as melanoma, to distant parts of the body. IFN-γ is well studied as a negative regulator for lymphangiogenesis, which is strongly associated with cancer metastasis. However, the exact mechanisms underlying this process remain unclear. In the present study, we investigated whether IFN-γ signaling in lymphatic endothelial cells (LECs) affects tumor cell dissemination by regulating the barrier function of tumor-associated lymphatic vessels. Using LEC-specific IFN-γ receptor (IFN-γR) knockout mice, we found that the loss of IFN-γR in LECs increased the dissemination of melanoma cells into the draining lymph nodes. Notably, IFN-γ signaling in LECs inhibited trans-lymphatic endothelial cell migration of melanoma cells, indicating its regulation of lymphatic barrier function. Further investigations revealed that IFN-γ upregulated the expression of the tight junction protein Claudin-3 in LECs, while knockdown of Claudin-3 in LECs abolished IFN-γ-induced inhibition of trans-lymphatic endothelial migration activity. Mechanistically, IFN-γ inhibits AMPK signaling activation, which is involved in the regulation of fatty acid metabolism. Modulating fatty acid metabolism and AMPK activation in LECs also affected the lymphatic dissemination of melanoma cells, further confirming that this process is involved in IFN-γ-induced regulation of lymphatic barrier function. These results provide novel insights into how IFN-γ modulates tight junctions in LECs, inhibiting the dissemination of melanoma cells via the lymphatic vessels.


Subject(s)
AMP-Activated Protein Kinases , Endothelial Cells , Interferon-gamma , Melanoma , Mice, Knockout , Animals , Interferon-gamma/metabolism , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , AMP-Activated Protein Kinases/metabolism , Melanoma/pathology , Melanoma/metabolism , Cell Movement , Signal Transduction , Interferon gamma Receptor , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Lymphatic Metastasis , Cell Line, Tumor , Lymphangiogenesis , Humans , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Mice, Inbred C57BL
3.
Radiat Oncol ; 18(1): 183, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37936130

ABSTRACT

BACKGROUND: Radiotherapy is widely used in treating cervical cancer patients, however, radioresistance unavoidably occurs and seriously affects the treatment effect. It is well known that hypoxia plays an important role in promoting radioresistance in tumor microenvironment, yet our understanding of the effect of small extracellular vesicles miRNA on cervical cancer radiosensitivity in hypoxic environment is still limited. METHODS: Small extracellular vesicles extracted from hypoxic and normoxic cultured cervical cancer cells were evaluated for their effects on radioresistance. miR-152-3p was found to be a potential effector in hypoxia-derived extracellular vesicles by searching the GEO database. Its downstream substrate was confirmed by double luciferase report, which was KLF15. The role of miR-152-3p and KLF15 in regulating cervical cancer radioresistance was detected by cell activity assays. The findings were confirmed in vivo by animal models. The expression of miR-152-3p was quantified by qRT-PCR and its prognostic significance was evaluated. RESULTS: Hypoxic environment promoted the secretion of small extracellular vesicles, and reduced the apoptosis and DNA damage caused by radiation, accompanied by increased expression of small extracellular vesicles miR-152-3p from hypoxic cervical cancer cells. Furthermore, small extracellular vesicles miR-152-3p promoted Hela xenograft growth and reduced the radiosensitivity vivo. Mechanism studies revealed that KLF15 protein was the downstream target of miR-152-3p in regulating radioresistance. CONCLUSION: Our findings suggest that small extracellular vesicles miR-152-3p affects the therapeutic effect of radiotherapy and holds potential as a biomarker or therapeutic target for cervical cancer prognosis and improving radiotherapy.


Subject(s)
MicroRNAs , Uterine Cervical Neoplasms , Female , Animals , Humans , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/radiotherapy , Uterine Cervical Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , HeLa Cells , Hypoxia/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Tumor Microenvironment , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
4.
Cancer Biol Ther ; 24(1): 2287128, 2023 12 31.
Article in English | MEDLINE | ID: mdl-38010897

ABSTRACT

Radioresistance is the major obstacle that affects the efficacy of radiotherapy which is an important treatment for cervical cancer. By analyzing the databases, we found that aldolase A (ALDOA), which is a key enzyme in metabolic reprogramming, has a higher expression in cervical cancer patients and is associated with poor prognosis. We detected the expression of ALDOA in the constructed cervical cancer radioresistance (RR) cells by repetitive irradiation and found that it was upregulated compared to the control cells. Functional assays were conducted and the results showed that the knockdown of ALDOA in cervical cancer RR cells inhibited the proliferation, migration, and clonogenic abilities by regulating the cell glycolysis. In addition, downregulation of ALDOA enhanced radiation-induced apoptosis and DNA damage by causing G2/M phase arrest and further promoted radiosensitivity of cervical cancer cells. The functions of ALDOA in regulating tumor radiosensitivity were also verified by the mouse tumor transplantation model in vivo. Therefore, our study provides new insights into the functions of ALDOA in regulating the efficacy of radiotherapy and indicates that ALDOA might be a promising target for enhancing radiosensitivity in treating cervical cancer patients.


Subject(s)
Fructose-Bisphosphate Aldolase , Uterine Cervical Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Proliferation/genetics , DNA Damage , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Gene Expression Regulation, Neoplastic , Glycolysis , Radiation Tolerance/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/radiotherapy , Uterine Cervical Neoplasms/pathology
5.
Front Bioeng Biotechnol ; 11: 1264006, 2023.
Article in English | MEDLINE | ID: mdl-37720318

ABSTRACT

Intrauterine adhesion (IUA), also referred to as Asherman Syndrome (AS), results from uterine trauma in both pregnant and nonpregnant women. The IUA damages the endometrial bottom layer, causing partial or complete occlusion of the uterine cavity. This leads to irregular menstruation, infertility, or repeated abortions. Transcervical adhesion electroreception (TCRA) is frequently used to treat IUA, which greatly lowers the prevalence of adhesions and increases pregnancy rates. Although surgery aims to disentangle the adhesive tissue, it can exacerbate the development of IUA when the degree of adhesion is severer. Therefore, it is critical to develop innovative therapeutic approaches for the prevention of IUA. Endometrial fibrosis is the essence of IUA, and studies have found that the use of different types of mesenchymal stem cells (MSCs) can reduce the risk of endometrial fibrosis and increase the possibility of pregnancy. Recent research has suggested that exosomes derived from MSCs can overcome the limitations of MSCs, such as immunogenicity and tumorigenicity risks, thereby providing new directions for IUA treatment. Moreover, the hydrogel drug delivery system can significantly ameliorate the recurrence rate of adhesions and the intrauterine pregnancy rate of patients, and its potential mechanism in the treatment of IUA has also been studied. It has been shown that the combination of two or more therapeutic schemes has broader application prospects; therefore, this article reviews the pathophysiology of IUA and current treatment strategies, focusing on exosomes combined with hydrogels in the treatment of IUA. Although the use of exosomes and hydrogels has certain challenges in treating IUA, they still provide new promising directions in this field.

6.
Nanomedicine ; 50: 102669, 2023 06.
Article in English | MEDLINE | ID: mdl-36933756

ABSTRACT

Epidermal Growth Factor Receptor (EGFR) is a promising therapeutic target for triple-negative breast cancer (TNBC). Recently, specific EGFR-targeting peptide GE11-based delivery nano-system shows excellent potential because of its chemical versatility and good targeting ability. However, no further research focusing on the downstream of EGFR after binding with GE11 was explored. Hence, we tailor-designed a self-assembled nanoplatform named GENP using amphiphilic molecule of stearic acid-modified GE11. After loading doxorubicin (DOX), the resulted nanoplatform GENP@DOX demonstrated high loading efficiency and sustainable drug release. Importantly, our findings proved that GENP alone significantly suppressed the proliferation of MDA-MB-231 cells via EGFR-downstream PI3K/AKT signaling pathways, contributing to the synergistic treatment with its DOX release. Further work illustrated remarkable therapeutic efficacy both in orthotopic TNBC and its bone metastasis models with minimal biotoxicity. Together, the results highlight that our GENP-functionalized nanoplatform is a promising strategy for the synergistic therapeutic efficacy targeting EGFR-overexpressed cancer.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , Phosphatidylinositol 3-Kinases , Cell Line, Tumor , ErbB Receptors/chemistry , Doxorubicin/chemistry , Peptides/pharmacology , Peptides/chemistry
7.
Front Cell Infect Microbiol ; 13: 1145950, 2023.
Article in English | MEDLINE | ID: mdl-36909733

ABSTRACT

Cervical cancer (CC) is the fourth most frequent malignancy among women worldwide, and its prevention and treatment are evolving rapidly. The gut microbiota has been reported to play a crucial role both in the preservation of homeostasis and the development of cervical cancer. In this study, we collected fecal samples to investigate the microbial signatures in cervical cancer patients compared with healthy controls using 16S rRNA sequencing analysis and metagenomic next-generation sequencing (mNGS) testing. Our findings demonstrated a substantial difference in the gut microbiota composition of cervical cancer patients and healthy controls. The disease and stage were most significantly negatively correlated with Ruminococcus 2, which might be considered a potential clinically relevant biomarker. Functions of differential microbiomes were also analyzed, indicating significant differences in metabolisms and biosynthesis between the two groups. These findings demonstrate that patients with cervical cancer have certain species of gut microbiota that are exclusive to them and particular species have the potential to be used in the prognosis of cervical cancer.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Uterine Cervical Neoplasms , Humans , Female , RNA, Ribosomal, 16S/genetics , Gastrointestinal Microbiome/genetics , Feces
8.
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
9.
Front Oncol ; 12: 999643, 2022.
Article in English | MEDLINE | ID: mdl-36313645

ABSTRACT

Radiotherapy is widely used as an indispensable treatment option for cervical cancer patients. However, radioresistance always occurs and has become a big obstacle to treatment efficacy. The reason for radioresistance is mainly attributed to the high repair ability of tumor cells that overcome the DNA damage caused by radiotherapy, and the increased self-healing ability of cancer stem cells (CSCs). Accumulating findings have demonstrated that the tumor microenvironment (TME) is closely related to cervical cancer radioresistance in many aspects, especially in the metabolic processes. In this review, we discuss radiotherapy in cervical cancer radioresistance, and focus on recent research progress of the TME metabolism that affects radioresistance in cervical cancer. Understanding the mechanism of metabolism in cervical cancer radioresistance may help identify useful therapeutic targets for developing novel therapy, overcome radioresistance and improve the efficacy of radiotherapy in clinics and quality of life of patients.

10.
Sci Rep ; 12(1): 17440, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261482

ABSTRACT

Claudin-3 is a tight junction protein that has often been associated with the progression and metastasis of various tumors. Here, the role of claudin-3 in tumor-induced lymphangiogenesis is investigated. We found an increased lymphangiogenesis in the B16F10 tumor in claudin-3 knockout mice, accompanied by augmented melanoma cell metastasis into sentinel lymph nodes. In vitro, the overexpression of claudin-3 on lymphatic endothelial cells inhibited tube formation by suppressing cell migration, resulting in restricted lymphangiogenesis. Further experiments showed that claudin-3 inhibited lymphatic endothelial cell migration by regulating the PI3K signaling pathway. Interestingly, the expression of claudin-3 in lymphatic endothelial cells is down-regulated by vascular endothelial growth factor C that is often present in the tumor microenvironment. This study indicates that claudin-3 plays an important role as a signaling molecule in lymphatic endothelial cell activity associated with tumor lymphangiogenesis, which may further contribute to melanoma metastasis.


Subject(s)
Claudin-3 , Lymphatic Vessels , Melanoma , Animals , Mice , Claudin-3/genetics , Claudin-3/metabolism , Endothelial Cells/metabolism , Lymphangiogenesis , Lymphatic Metastasis/pathology , Lymphatic Vessels/metabolism , Melanoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Tumor Microenvironment , Vascular Endothelial Growth Factor C/metabolism
11.
Ther Adv Med Oncol ; 14: 17588359221118010, 2022.
Article in English | MEDLINE | ID: mdl-35983027

ABSTRACT

Ovarian cancer (OC) is the third most common gynecological malignancy with the highest mortality worldwide. OC is usually diagnosed at an advanced stage, and the standard treatment is surgery combined with platinum or paclitaxel chemotherapy. However, chemoresistance inevitably appears coupled with the easy recurrence and poor prognosis. Thus, early diagnosis, predicting prognosis, and reducing chemoresistance are of great significance for controlling the progression and improving treatment effects of OC. Recently, much insight has been gained into the non-coding RNA (ncRNA) that is employed for RNAs but does not encode a protein, and many types of ncRNAs have been characterized including long-chain non-coding RNAs, microRNAs, and circular RNAs. Accumulating evidence indicates these ncRNAs play very active roles in OC progression and metastasis. In this review, we briefly discuss the ncRNAs as biomarkers for OC prognosis. We focus on the recent advances of ncRNAs as therapeutic targets in preventing OC metastasis, chemoresistance, immune escape, and metabolism. The novel strategies for ncRNAs-targeted therapy are also exploited for improving the survival of OC patients.

12.
Sci Rep ; 12(1): 11632, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35804089

ABSTRACT

Aldolase A (ALDOA) is an enzyme that plays an important role in glycolysis and gluconeogenesis, which is closely related to tumor metabolism. In this study, the overall roles of ALDOA in pan-cancer have been investigated from several aspects using databases and online analysis tools. Using the ONCOMINE database, the expression of ALDOA in various cancers was analyzed. The prognostic role of ALDOA was explored by PrognoScan, GEPIA, and Kaplan-Meier Plotter. The immune-related role of ALDOA and its downstream substrates was decided by TIMER, cBioPortal and String. Our data indicate that ALDOA expression level in lung adenocarcinoma, liver hepatocellular carcinoma, head and neck squamous cell carcinoma is higher than that in normal tissues. Increased expression of ALDOA often indicates a poor prognosis for patients. The correlation between ALDOA and immune infiltration among different tumors is very different. We also investigate the relationship between ALDOA and its upstream/downstream proteins. Our results showed that ALDOA could be used as a biomarker for the tumor prognosis, and could be correlated with the infiltrating levels of macrophages, CD4+ T cells and CD8+ T cells.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Hepatocellular , Liver Neoplasms , Lung Neoplasms , Biomarkers, Tumor , Computational Biology , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Humans , Prognosis
13.
Front Oncol ; 12: 830570, 2022.
Article in English | MEDLINE | ID: mdl-35692779

ABSTRACT

Mesothelin (MSLN) is a glycoprotein with various expression degrees in different tumors including mesothelioma, ovarian cancer, pancreatic cancer, etc. MSLN is considered to play an important role in cell survival, proliferation, and tumor progression. Although the expression of MSLN in tumors makes it a potential therapeutic target, its mechanism of action is still unclear, especially its correlation with immune cells infiltration in the tumor microenvironment has not been investigated. In this study, we detected the overexpression of MSLN in ovarian cancer using database analysis and tissue-array staining. We further evaluated the diagnostic value of MSLN and found it was associated with poor overall survival in ovarian cancer. In addition, the high expression of MSLN was significantly related to the immune-related genes and chemoresistant genes. We confirmed the overexpression of MSLN in the chemoresistant ovarian cancer cell lines. Our research suggests that MSLN participates in a variety of pathways related to the suppression of immune activation and promotion of chemoresistance, leading to a poor prognosis in ovarian cancer.

14.
Cancer Res ; 82(13): 2472-2484, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35580275

ABSTRACT

Migration of myeloid-derived suppressor cells (MDSC) out of the circulation, across vascular walls, and into tumor is crucial for their immunosuppressive activity. A deeper understanding of critical junctional molecules and the regulatory mechanisms that mediate the extravasation of MDSCs could identify approaches to overcome cancer immunosuppression. In this study, we used mice deficient in tight junction protein Claudin-12 (Cldn12) compared with wild-type mice and found that loss of host Cldn12 inhibited the growth of transplanted tumors, reduced intratumoral accumulation of MDSCs, increased antitumor immune responses, and decreased tumor vascular density. Further studies revealed that Cldn12 expression on the cell surface of both MDSCs and endothelial cells (EC) is required for MDSCs transit across tumor vascular ECs. Importantly, expression of Cldn12 in MDSCs was modulated by GM-CSF in an AKT-dependent manner. Therefore, our results indicate that Cldn12 could serve as a promising target for restoring the antitumor response by interfering with MDSCs transendothelial migration. SIGNIFICANCE: Claudin-12-mediated homotypic interactions are critical for migration of myeloid-derived suppressor cells across vascular walls into tumor tissue, providing a potential therapeutic approach to overcome cancer immunosuppression.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Animals , Claudins/metabolism , Endothelial Cells , Mice , Neoplasms/genetics , Neoplasms/metabolism , Transendothelial and Transepithelial Migration
15.
Cell Death Dis ; 13(1): 64, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35042862

ABSTRACT

Chemoresistance and metastasis are the major challenges for the current ovarian cancer treatment. Understanding the mechanisms of ovarian cancer progression and metastasis is critically important for developing novel therapies. The advances in extracellular vesicles (EVs) research in recent years have attracted extensive attention. EVs contain a variety of proteins, RNAs, DNAs, and metabolites. Accumulating evidence indicates that ovarian cancer cells secrete a large amount of EVs, playing an important role in tumor progression and recurrence. In the microenvironment of ovarian tumor, EVs participate in the information transmission between stromal cells and immune cells, promoting the immune escape of ovarian cancer cells and facilitating cancer metastasis. Here, we review the recent advances of EVs in chemoresistance, mechanisms of metastasis, and immune evasion of ovarian cancer. Furthermore, we also discuss the challenges of EV research and future application of EVs as promising biomarker sources in response to therapy and in therapy-delivery approaches for ovarian cancer patients.


Subject(s)
Extracellular Vesicles , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial/metabolism , Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , Humans , Immune Evasion , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Microenvironment
16.
Life Sci ; 256: 117955, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32534038

ABSTRACT

AIMS: Cancer associated fibroblasts (CAFs) play a crucial role in lung tumor development, but the underlying mechanism is still not fully understood. MAIN METHODS: SCRIB expression in the CAFs of human lung cancer tissues was examined by immunohistochemistry (IHC). A coculture of mouse Lewis lung cancer cells (LLC) and fibroblasts was used to investigate SCRIB expression in cocultured fibroblasts. Proliferation, scratch wound, and transwell assays were used to examine the proliferation, migration and invasion ability of SCRIB knockdown fibroblasts and their effects on LLC. A 3D-coculture system and co-injection xenograft model were used to examine LLC invasion. RNA sequencing and transwell experiments were used to explore the molecules that may participate in LLC invasion. KEY FINDINGS: Herein, we found that the low expression of SCRIB in CAFs is correlated with advanced tumor stages and poor survival for human lung squamous cell carcinoma. SCRIB expression in fibroblasts is drastically downregulated by LLC cells. SCRIB knockdown fibroblasts not only enhance invasion but also facilitate LLC invasion in a 3D-coculture system and in an in vivo subcutaneous transplantation model. The upregulation of asporin in SCRIB knockdown fibroblasts is involved in LLC invasion in vitro. SIGNIFICANCE: Collectively, the results indicate that fibroblasts with low SCRIB expression promote lung cancer cell invasion, which suggests that the downregulated expression of SCRIB may represent one of the important characteristics of tumor-promoting CAFs in lung squamous cell cancer.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Membrane Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation/genetics , Extracellular Matrix Proteins/metabolism , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred C57BL , Models, Biological , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism
17.
Cancer Cell Int ; 20: 61, 2020.
Article in English | MEDLINE | ID: mdl-32123519

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) are implicated in many pathophysiological processes, including cancers. In particular, lncRNA DANCR is regarded as a cancer-associated lncRNA exerting various regulatory mechanisms. However, the expressions, functions, and mechanisms of action of DANCR in cervical cancer are still unclear. METHODS: The expressions of DANCR in cervical cancer tissues and cell lines were evaluated using qRT-PCR. Correlations between DANCR expression and clinicopathological features and prognosis were analyzed. The roles of DANCR in cervical cancer growth were evaluated by in vitro CCK-8 and EdU assay, and in vivo xenograft assay. The regulatory effects of DANCR on Wnt/ß-catenin signaling pathway were evaluated using nuclear proteins extraction, western blot, and qRT-PCR. RESULTS: DANCR is increased in cervical cancer tissues and cell lines. Increased expression of DANCR is associated with large tumor size, advanced FIGO stage, and poor overall survival of cervical cancer patients. Functional experiments showed that enhanced expression of DANCR promotes cervical cancer cell proliferation in vitro and xenograft growth in vivo. Conversely, DANCR knockdown inhibits cervical cancer cell proliferation in vitro and xenograft growth in vivo. Mechanistic investigation demonstrated that DANCR upregulates the expressions of FRAT1 and FRAT2 and activates the Wnt/ß-catenin signaling pathway. Blocking the Wnt/ß-catenin signaling pathway abolishes the pro-proliferative roles of DANCR overexpression and anti-proliferative roles of DANCR knockdown. CONCLUSIONS: Our findings suggest DANCR as an oncogenic lncRNA in cervical cancer through activating the Wnt/ß-catenin signaling pathway, and imply that DANCR may be a promising prognostic biomarker and therapeutic target for cervical cancer.

18.
Front Oncol ; 10: 547683, 2020.
Article in English | MEDLINE | ID: mdl-33643892

ABSTRACT

Polysaccharides from Lentinus edodes (L. edodes) have been successfully used as adjuvant chemotherapy drug to treat lymphatic metastasis in some malignancies, such as colorectal cancer (CRC), lung cancer and gastric cancer. The CRC could metastasize via lymphatic vessels. Lymphatic metastasis is commonly thought to be the cause of poor prognosis of CRC. The mechanism of polysaccharides from L. edodes inhibiting lymphatic metastasis of CRC is still unclear. In this study, we explored how MPSSS, a novel polysaccharide component of L. edodes, influences lymphangiogenesis and lymph node metastasis. The results show that MPSSS can reduce lymphangiogenesis and lymphatic metastasis of CRC in mouse model. And combined with in vitro study, a likely mechanism is that MPSSS reduce the secretion of VEGF-C by cancer associated fibroblasts (CAFs). This effect can be suppressed by a TLR4 inhibitor, which suggests that MPSSS plays a role in CAFs through the TLR4/JNK signaling pathway. In conclusion, MPSSS may reduce lymphangiogenesis by decreasing the VEGF-C secretion of CAFs, which may provide a new strategy for the comprehensive treatment of CRC.

19.
Biosci Rep ; 39(5)2019 05 31.
Article in English | MEDLINE | ID: mdl-30992392

ABSTRACT

The polysaccharides MPSSS was extracted from Lentinus edodes and has been reported to effectively inhibit tumor growth and eliminate the function of myeloid-derived immune suppressor cell-mediated T cell inhibition, thus improving the efficacy of cancer therapy. The exploration of how MPSSS affects the functions of cancer-associated fibroblasts (CAFs) will provide a new perspective for understanding the antitumor effects of MPSSS. In the present study, prostate CAFs were selected as target cells to study whether MPSSS affected cell proliferation and function. The results showed that MPSSS did not directly inhibit the growth of prostate CAFs but interfered with CAF-mediated T cell inhibition and affected the immunosuppressive function of prostate CAFs. Mechanistic studies were further performed and showed that MPSSS activated key node proteins in the NF-κB pathway that were dependent on MyD88, and a TLR4 inhibitor blocked the changes in these proteins and the effect of MPSSS. We hypothesize that MPSSS can activate the MyD88-dependent TLR4-NF-κB signaling pathway to change the function of CAFs. In conclusion, these results demonstrate that MPSSS can not only effectively inhibit the growth of prostate cancer as we previously reported but also alter the function of prostate CAFs by activating the TLR4-NF-κB pathway, providing a new strategy for the comprehensive treatment of tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Fungal Polysaccharides/pharmacology , Gene Expression Regulation, Neoplastic , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 4/genetics , Animals , Antineoplastic Agents/isolation & purification , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Fungal Polysaccharides/isolation & purification , Humans , Male , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Shiitake Mushrooms/chemistry , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Toll-Like Receptor 4/immunology
20.
Oncotarget ; 7(28): 43546-43556, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27286458

ABSTRACT

Autoantibodies against intracellular tumor-associated antigens (TAAs) are commonly found in human cancers. In this study, we characterized the serum autoantibody response to the RalA, Ras-like GTPase, in patients with prostate cancer (PCa). The autoantibodies were detected by immunofluorescence assay in PCa cell lines, ELISA, and immunoblotting in 339 serum samples from patients with PCa and benign prostatic hyperplasia (BPH), and in normal human sera (NHS). The expression of RalA in prostate tumor tissues was evaluated by immunohistochemistry (IHC) in tumor microarrays. The autoantibody level to RalA (median) in NHS was significantly lower than in PCa (0.053 vs 0.138; P < 0.001) and BPH (0.053 vs 0.132; P < 0.005) groups. The circulating anti-RalA autoantibody could distinguish PCa patients from normal individuals with the area under the receiver operating characteristic (ROC) curve (AUC) performing at 0.861, with sensitivity of 52.9% and specificity of 91.0%. Elevation in serum immunoreactivity was observed in PCa patients after radical prostatectomy. The combined use of both anti-RalA autoantibody and PSA showed a significantly higher discriminatory ability compared with either of those markers alone. RalA protein expression was detected by IHC in 85.3% of tumor tissues from PCa patients, but without significant difference compared to BPH or normal control tissues. Together, our study shows the additional benefits of anti-RalA autoantibody as a potential serological biomarker for PCa, particularly in patients with normal PSA, and further demonstrate the utility of biomarker combinations in the immunodiagnosis of PCa.


Subject(s)
Antigens, Neoplasm/immunology , Autoantibodies/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , ral GTP-Binding Proteins/immunology , Aged , Area Under Curve , Autoantigens/immunology , Autoantigens/metabolism , Biomarkers, Tumor/blood , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Microscopy, Fluorescence , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prostate/pathology , Prostate-Specific Antigen/blood , Prostatic Hyperplasia/blood , Prostatic Hyperplasia/immunology , Prostatic Neoplasms/pathology , ROC Curve , Recombinant Proteins/immunology , Retrospective Studies , Tissue Array Analysis , ral GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL