Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 2923, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36808166

ABSTRACT

The NLR family CARD domain containing 3 (NLRC3) gene has been reported to have a crucial effect on immunity, inflammation, and tumorigenesis. However, the clinical relevance of NLRC3 in lung adenocarcinoma (LUAD) remains unclear. This study analyzed both RNA sequencing data and corresponding clinical outcomes obtained from public databases to identify (i) NLRC3 as a tumor suppressor in LUAD and (ii) its predictive value for the likelihood of patient responsiveness to immunotherapy. The results showed that NLRC3 expression was reduced in LUAD and was lower in advanced-stage tumors. Additionally, reduced NLRC3 expression was correlated with worse patient prognosis. The protein level of NLRC3 was also observed to have prognostic significance. Moreover, downregulation of NLRC3 was found to suppress the chemotaxis and infiltration of antitumor lymphocyte subpopulations as well as natural killer cells. Mechanistic analysis indicated that NLRC3 may be involved in immune infiltration by regulating chemokines and their receptors in LUAD. Furthermore, NLRC3 functions as a molecular switch in macrophages, whereby it mediates the polarization of M1 macrophages. Patients with high NLRC3 expression were also found to exhibit a more promising response to immunotherapy. In conclusion, NLRC3 could serve as a potential prognostic biomarker for LUAD, help predict the immunotherapeutic response of patients, and guide personalized strategies for the treatment of LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Prognosis , Chemotaxis , Biomarkers , Intercellular Signaling Peptides and Proteins
2.
Life Sci ; 310: 121089, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36270423

ABSTRACT

AIMS: There is emerging evidence that antineoplastic agents and the cytotoxic effects on tumor tissues attenuate the benefits of chemotherapy due to tumor microenvironment changes. Nevertheless, the actual relationship between chemotherapy and recurrent tumors in which the genotypes differ from the original tumor after chemotherapy is unclear. MATERIALS AND METHODS: Bone marrow transplantation, flow cytometer, immune inhibition and immunofluorescence will be utilized to investigate the effect of antineoplastic agents on bone-marrow-derived cells (BMDCs) release and recruitment, and to explore the pathways and mechanisms of antineoplastic agents in promoting tumor growth. KEY FINDINGS: Tumor growth and angiogenesis were significantly enhanced in the mouse model after treatment with antineoplastic agents such as cyclophosphamide, 5-fluorouracil, or cisplatin, along with large increases in proangiogenic vascular endothelial growth factor receptor-2 (VEGFR2+), ß3+, CD11b+Gr-1+, and VEGFR2+ß3+ BMDCs in circulating blood. BMDC recruitment and VEGFR2 and ß3 mRNA transcription in tumor tissues were also enhanced by antineoplastic agents. Antineoplastic-agent-treated BMDCs markedly augmented tumor and endothelial cell proliferation, and ß3 mRNA transcription in endothelial cells (ECs). SIGNIFICANCE: The results suggested that antineoplastic-agent treatment augmented the tumor microenvironment by mobilizing proangiogenic BMDCs, enhancing BMDC recruitment and angiogenesis, and increasing BMDC-mediated tumor and EC functions. These results led to tumor growth and angiogenesis aggravation. It is paramount to elucidate the potential mechanism by which the cellular and molecular effects triggered by the antineoplastic agents attenuate the effects of cancer therapy, and thereafter to explore possible methods for improving tumor treatment efficacy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Mice , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Bone Marrow Cells/metabolism , Endothelial Cells/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , RNA, Messenger/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Mol Med Rep ; 19(4): 3148-3158, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816516

ABSTRACT

Brain ischemia, including cerebral ischemia and cerebrovascular ischemia, leads to poor oxygen supply or cerebral hypoxia, and causes brain tissue death or cerebral infarction/ischemic stroke. The troxerutin and cerebroprotein hydrolysate injection (TCHI), is widely applied in China to improve blood supply in ischemic brain tissues and to enhance neuroprotective effects in clinical practice. However, the benefits and detailed underlying mechanism elaborating the effectiveness of TCHI in cerebrovascular diseases require further investigation. Therefore, in the present study, experimental in vivo and in vitro models were employed to investigate the potential mechanisms of TCHI on cerebral ischemic injury. The results demonstrated that TCHI increased the lactate dehydrogenase levels in the brain homogenate and conversely decreased lactic acid levels. TCHI was further observed to significantly increase superoxide dismutase activity and decrease malondialdehyde levels in ischemic brain tissues. In addition, TCHI significantly induced vascular maturation processes, including proliferation, adhesion, migration and tube formation in cultured human umbilical vein endothelial cells. Additionally, TCHI significantly stimulated microvessel formation in the rat aortic ring and chick chorioallantoic membrane assays. Taken together, these results provided strong evidence that TCHI stimulated angiogenesis at multiple steps, and indicated that TCHI attenuated cerebral ischemic damage through the amelioration of oxidative stress and promotion of angiogenesis.


Subject(s)
Anticoagulants/pharmacology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Hydroxyethylrutoside/analogs & derivatives , Neovascularization, Pathologic/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Biomarkers , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Cell Adhesion , Cell Movement , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Hydroxyethylrutoside/pharmacology , Male , Rats , Reactive Oxygen Species/metabolism
4.
Oncol Rep ; 40(4): 2147-2156, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30106454

ABSTRACT

Estradiol (E2) is a prime culprit for enhancing the progression of female hormone­related cancers. Bone marrow­derived cells (BMDCs) have been found to play a pivotal role in tumor growth. Estrogen receptors (ERs) are also found on certain subtypes of BMDCs, in addition to endothelial cells (ECs) and certain tumor cells. However, the role of BMDCs in E2­induced tumor biology is still unclear. Thus, the effects of E2 on ER­negative 4T1 breast cancer growth, the mobilization and recruitment of BMDCs, and interactions among BMDCs, ECs, and 4T1 cells were investigated. The results showed that E2 potentiated 4T1 tumor growth and angiogenesis in mice subjected to sham operation, ovariectomy (OVX), or OVX and E2 replacement treatment. E2 supplementation in mice with OVX upregulated the transcription of stromal cell­derived factor­1 (SDF­1) mRNA in tumor tissues and enhanced the recruitment of BMDCs into tumor tissues in vivo. E2 deficiency significantly decreased proangiogenic CXCR4+, ß3+, Sca­1+ and CXCR4+ß3+ BMDCs circulating in the peripheral blood. Cell­based system analyses showed that E2 augmented the transcription of ß3 mRNA in ECs, increased the adhesion of BMDCs to ECs. In addition, E2 enhanced the BMDC­induced EC proliferation and migration, the BMDC­induced 4T1 proliferation and the 4T1­stimulated EC proliferation in addition to enhancing the proliferation of tumor cells and the migration of ECs in vitro. Therefore, E2 enhanced the growth of breast tumors by stimulating tumor cells and ECs directly, as well as by increasing proangiogenic BMDC mobilization and recruitment leading to augmentation of the tumor and EC functions indirectly by cell proliferation assay. These findings reveal a separate mechanism via which E2 promotes the growth of female hormone­dependent tumors, which may be useful in explorations of new therapies for related cancers.


Subject(s)
Bone Marrow/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Proliferation , Endothelium, Vascular/pathology , Estrogens/toxicity , Neovascularization, Pathologic/pathology , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Bone Marrow/drug effects , Breast Neoplasms/chemically induced , Cell Adhesion , Cell Movement , Culture Media, Conditioned/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/chemically induced , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...