Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Open Med (Wars) ; 19(1): 20240962, 2024.
Article in English | MEDLINE | ID: mdl-38770178

ABSTRACT

Aims: In cancer biology, the aberrant overexpression of eukaryotic translation initiation factor 5A2 (EIF5A2) has been correlative with an ominous prognosis, thereby underscoring its pivotal role in fostering metastatic progression. Consequently, EIF5A2 has garnered significant attention as a compelling prognostic biomarker for various malignancies. Our research endeavors were thus aimed at elucidating the utility and significance of EIF5A2 as a robust indicator of cancer outcome prediction. Method: An exhaustive search of the PubMed, EMBASE, and Web of Science databases found relevant studies. The link between EIF5A2 and survival prognosis was examined using hazard ratios and 95% confidence intervals. Subsequently, The Cancer Genome Atlas (TCGA) and the Gene Expression Profiling Interactive Analysis (GEPIA) databases were employed to validate EIF5A2 expression across various cancer types. Results: Through pooled analysis, we found that increased EIF5A2 expression was significantly associated with decreased overall survival (OS) and disease-free survival/progression-free survival/relapse-free survival (DFS/PFS/RFS). Moreover, TCGA analysis revealed that EIF5A2 was significantly upregulated in 27 types of cancer, with overexpression being linked to shorter OS in three, worse DFS in two, and worse PFS in six types of cancer. GEPIA showed that patients with EIF5A2 overexpression had reduced OS and DFS. Conclusions: In solid tumors, EIF5A2 emerges as a reliable prognostic marker. Our meta-analysis comprehensively analyzed the prognostic value of EIF5A2 in solid tumors and assessed its efficacy as a predictive marker.

2.
Cancer ; 130(S8): 1378-1391, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-37950749

ABSTRACT

Breast cancer (BC) is the fourth most prevalent cancer in China. Despite conventional treatment strategies, BC patients often have poor therapeutic outcomes, leading to significant global cancer mortality rates. Chimeric antigen receptor (CAR)-based immunotherapy is a promising and innovative approach for cancer treatment that redirects immune cells to attack tumor cells expressing selected tumor antigens (TAs). T cells, natural killer (NK) cells, and macrophages, key components of the immune system, are used in CAR-based immunotherapies. Although remarkable progress has been made with CAR-T cells in hematologic malignancies, the application of CAR-based immunotherapy to BC has lagged. This is partly due to obstacles such as tumor heterogeneity, which is further associated with the TA and BC subtypes, and the immunosuppressive tumor microenvironment (TME). Several combinatorial approaches, including the use of immune checkpoint inhibitors, oncolytic viruses, and antitumor drugs, have been proposed to overcome these obstacles in BC treatment. Furthermore, several CAR-based immunotherapies for BC have been translated into clinical trials. This review provides an overview of the recent progress in CAR-based immunotherapy for BC treatment, including targeting of TAs, consideration of BC subtypes, assessment of the TME, and exploration of combinatorial therapies. The authors focused on preclinical studies and clinical trials of CAR-T cells, CAR-NK cells, and CAR-macrophages especially conducted in China, followed by an internal comparison and discussion of current limits. In conclusion, this review elucidates China's contribution to CAR-based immunotherapies for BC and provides inspiration for further research. PLAIN LANGUAGE SUMMARY: Despite conventional treatment strategies, breast cancer (BC) patients in China often have poor therapeutic outcomes. Chimeric antigen receptor (CAR)-based immunotherapy, a promising approach, can redirect immune cells to kill tumor cells expressing selected tumor antigens (TAs). However, obstacles such as TA selection, BC subtypes, and immunosuppressive tumor microenvironment still exist. Therefore, various combinatorial approaches have been proposed. This article elucidates several Chinese CAR-based preclinical and clinical studies in BC treatment with comparisons of foreign research, and CAR-immune cells are analyzed, providing inspiration for further research.


Subject(s)
Breast Neoplasms , Neoplasms , Receptors, Chimeric Antigen , Humans , Female , Receptors, Chimeric Antigen/therapeutic use , Breast Neoplasms/therapy , Immunotherapy, Adoptive , Neoplasms/therapy , Immunotherapy , Antigens, Neoplasm , Tumor Microenvironment
3.
Cell Death Dis ; 14(9): 586, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37666809

ABSTRACT

The tumor microenvironment (TME) is a highly intricate milieu, comprising a multitude of components, including immune cells and stromal cells, that exert a profound influence on tumor initiation and progression. Within the TME, angiogenesis is predominantly orchestrated by endothelial cells (ECs), which foster the proliferation and metastasis of malignant cells. The interplay between tumor and immune cells with ECs is complex and can either bolster or hinder the immune system. Thus, a comprehensive understanding of the intricate crosstalk between ECs and immune cells is essential to advance the development of immunotherapeutic interventions. Despite recent progress, the underlying molecular mechanisms that govern the interplay between ECs and immune cells remain elusive. Nevertheless, the immunomodulatory function of ECs has emerged as a pivotal determinant of the immune response. In light of this, the study of the relationship between ECs and immune checkpoints has garnered considerable attention in the field of immunotherapy. By targeting specific molecular pathways and signaling molecules associated with ECs in the TME, novel immunotherapeutic strategies may be devised to enhance the efficacy of current treatments. In this vein, we sought to elucidate the relationship between ECs, immune cells, and immune checkpoints in the TME, with the ultimate goal of identifying novel therapeutic targets and charting new avenues for immunotherapy.


Subject(s)
Endothelial Cells , Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Cell Transformation, Neoplastic , Immunomodulation , Tumor Microenvironment
4.
Cancers (Basel) ; 14(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36291773

ABSTRACT

Aging is one of the risk factors for advanced breast cancer. With the increasing trend toward population aging, it is important to study the effects of aging on breast cancer in depth. Cellular senescence and changes in the aging microenvironment in vivo are the basis for body aging and death. In this review, we focus on the influence of the aging microenvironment on breast cancer. Increased breast extracellular matrix stiffness in the aging breast extracellular matrix can promote the invasion of breast cancer cells. The role of senescence-associated secretory phenotypes (SASPs) such as interleukin-6 (IL-6), IL-8, and matrix metalloproteases (MMPs), in breast cancer cell proliferation, invasion, and metastasis is worthy of exploration. Furthermore, the impact of senescent fibroblasts, adipocytes, and endothelial cells on the mammary matrix is discussed in detail. We also list potential targets for senotherapeutics and senescence-inducing agents in the aging microenvironment of breast cancer. In conclusion, this review offers an overview of the influence of the aging microenvironment on breast cancer initiation and progression, with the aim of providing some directions for future research on the aging microenvironment in breast cancer.

5.
J Oncol ; 2021: 6641421, 2021.
Article in English | MEDLINE | ID: mdl-34054955

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive disease with poorer prognosis than other subtypes. We aimed to investigate the prognostic efficacy of multiple tumor markers and constructed a prognostic model for stage I-III TNBC patients. Patients and Methods. We included stage I-III TNBC patients whose serum tumor markers levels were measured prior to the treatment. The optimal cut-off value of each tumor marker was determined by X-tile. Then, we adopted two survival models (lasso Cox model and random survival forest model) to build the prognostic model and AUC values of the time-dependent receiver operating characteristic (ROC) were calculated. The Kaplan-Meier method was used to plot the survival curves and the log-rank test was used to test whether there was a significant difference between the predicted high-risk and low-risk groups. We used univariable and multivariable Cox analysis to identify independent prognostic factors and did subgroup analysis further for the lasso Cox model. RESULTS: We included 258 stage I-III TNBC patients. CEA, CA125, and CA211 showed independent prognostic value for DFS when using the optimal cut-off values; their HRs and 95% CI were as follows: 1.787 (1.056-3.226), 2.684 (1.200-3.931), and 2.513 (1.567-4.877). AUC values of lasso Cox model and random survival forest model were 0.740 and 0.663 for DFS at 60 months, respectively. Both the lasso Cox model and random survival forest model demonstrated excellent prognostic value. According to tumor marker risk scores (TMRS) computed by the lasso Cox model, the high TMRS group had worse DFS (HR = 3.138, 95% CI: 1.711-5.033, p < 0.0001) and OS (3.983, 1.637-7.214, p=0.0011) than low TMRS group. Furthermore, subgroup analysis of N0-N1 patients in the lasso Cox model indicated that TMRS still had a significant prognostic effect on DFS (2.278, 1.189-4.346) and OS (2.982, 1.110-7.519). CONCLUSIONS: Our study indicated that pretreatment levels of serum CEA, CA125, and CA211 had independent prognostic significance for TNBC patients. Both lasso Cox model and random survival forest model that we constructed based on tumor markers could strongly predict the survival risk. Higher TMRS was associated with worse DFS and OS both in stage I-III and N0-N1 TNBC patients.

6.
Aging (Albany NY) ; 12(17): 17079-17098, 2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32902412

ABSTRACT

UNC-5 Homolog B (UNC5B) is a member of the dependence receptor family that regulates cell survival and apoptosis in a ligand-dependent manner. UNC5B plays an important role in the development of multiple cancers, including colorectal, bladder, and thyroid cancer. However, the exact expression pattern and mechanism of UNC5B in breast cancer have not been well elucidated. Here, we showed that UNC5B expression was significantly upregulated in breast cancer using bioinformatics analysis and experimental validation. High UNC5B expression was correlated with poor overall survival in breast cancer patients. UNC5B knockdown inhibited breast cancer cell proliferation and metastasis and compromised PI3K/Akt signaling activation. In summary, UNC5B is a promising diagnostic and prognostic biomarker and targeting UNC5B is a potential strategy for individualized breast cancer treatment.

7.
Article in English | MEDLINE | ID: mdl-32923434

ABSTRACT

Cardiovascular disease (CVD) is currently one of the primary causes of mortality and morbidity worldwide. Nanoparticles (NPs) are playing increasingly important roles in regulating stem cell behavior because of their special features, including shape, size, aspect ratio, surface charge, and surface area. In terms of cardiac disease, NPs can facilitate gene delivery in stem cells, track the stem cells in vivo for long-term monitoring, and enhance retention after their transplantation. The advantages of applying NPs in peripheral vascular disease treatments include facilitating stem cell therapy, mimicking the extracellular matrix environment, and utilizing a safe non-viral gene delivery tool. However, the main limitation of NPs is toxicity, which is related to their size, shape, aspect ratio, and surface charge. Currently, there have been many animal models proving NPs' potential in treating CVD, but no extensive applications of stem-cell therapy using NPs are available in clinical practice. In conclusion, NPs might have significant potential uses in clinical trials of CVD in the future, thereby meeting the changing needs of individual patients worldwide.

8.
Front Oncol ; 10: 301, 2020.
Article in English | MEDLINE | ID: mdl-32185139

ABSTRACT

Partner and localizer of BRCA2 (PALB2) is vital for homologous recombination (HR) repair in response to DNA double-strand breaks (DSBs). PALB2 functions as a tumor suppressor and participates in the maintenance of genome integrity. In this review, we summarize the current knowledge of the biological roles of the multifaceted PALB2 protein and of its regulation. Moreover, we describe the link between PALB2 pathogenic variants (PVs) and breast cancer predisposition, aggressive clinicopathological features, and adverse clinical prognosis. We also refer to both the opportunities and challenges that the identification of PALB2 PVs provides in breast cancer genetic counseling and precision medicine.

9.
J Ophthalmol ; 2020: 4858636, 2020.
Article in English | MEDLINE | ID: mdl-33520295

ABSTRACT

PURPOSE: There has not been a recent population-based study regarding the epidemiological trend and survival of eyelid primary malignant melanoma (PMM). Our study aims to evaluate the updated incidence trends and discuss the factors affecting the survival outcomes of eyelid PMM. METHODS: A total of 1397 eyelid PMM cases diagnosed between 1975 and 2016 were retrospectively identified from the Surveillance, Epidemiology, and End Results (SEER) database. Age-adjusted incidence rates and annual percent changes (APC) were calculated. Kaplan-Meier and Cox proportional hazards regression models were used to calculate survival outcomes and identify potential prognostic factors. RESULTS: The overall age-adjusted incidence of eyelid PMM rose from 0.039 (95% confidence interval [CI], 0.012-0.088) in 1975 to 0.103 (95% CI, 0.070-0.143) per 100 000 population in 2016, with significant APC of 1.313% (p < 0.001). Male subjects showed a higher average age-adjusted incidence rate than female subjects (p < 0.001). Survival analyses showed that 5-year accumulative overall survival (OS) and disease-specific survival (DSS) for patients with eyelid PMM were 70.5% and 90.6%. Additionally, 10-year OS and DSS were 51.8% and 86.1%, respectively. Analyses of Kaplan-Meier survival curves with the log-rank test revealed that older age, White race, nodular melanoma, higher American Joint Committee on Cancer (AJCC) stage (II to IV), advanced stage, distant metastasis, and no-surgery treatment were associated with lower OS and DSS rates. Age, histology, AJCC stage, and stage at diagnosis were found to be independent predictors of OS and DSS in multivariate models. CONCLUSION: The incidence of eyelid PMM increased with significant APC and male predominance. Age, histology, AJCC stage, and stage at diagnosis might be independent predictors of prognosis, emphasizing the importance of improved diagnosis of eyelid PMM.

10.
Hepatology ; 68(6): 2359-2375, 2018 12.
Article in English | MEDLINE | ID: mdl-29742804

ABSTRACT

Sterile inflammation is an essential factor causing hepatic ischemia/reperfusion (I/R) injury. As a critical regulator of inflammation, the role of monocyte chemoattractant protein-induced protein 1 (MCPIP1) in hepatic I/R injury remains undetermined. In this study, we discovered that MCPIP1 downregulation was associated with hepatic I/R injury in liver transplant patients and a mouse model. Hepatocyte-specific Mcpip1 gene knockout and transgenic mice demonstrated that MCPIP1 functions to ameliorate liver damage, reduce inflammation, prevent cell death, and promote regeneration. A mechanistic study revealed that MCPIP1 interacted with and maintained hypoxia-inducible factor 1α (HIF-1α) expression by deubiquitinating HIF-1α. Notably, the HIF-1α inhibitor reversed the protective effect of MCPIP1, whereas the HIF-1α activator compensated for the detrimental effect of MCPIP1 deficiency. Thus, we identified the MCPIP1-HIF-1α axis as a critical pathway that may be a good target for intervention in hepatic I/R injury. (Hepatology 2018; 00:000-000).


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Diseases/metabolism , Reperfusion Injury/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Case-Control Studies , Hepatocytes/physiology , Humans , Liver/pathology , Liver Diseases/pathology , Liver Regeneration , Male , Mice , Primary Cell Culture
11.
Nat Med ; 24(2): 213-223, 2018 02.
Article in English | MEDLINE | ID: mdl-29291351

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a common clinical condition that can lead to advanced liver diseases. Lack of effective pharmacotherapies for NASH is largely attributable to an incomplete understanding of its pathogenesis. The deubiquitinase cylindromatosis (CYLD) plays key roles in inflammation and cancer. Here we identified CYLD as a suppressor of NASH in mice and in monkeys. CYLD is progressively degraded upon interaction with the E3 ligase TRIM47 in proportion to NASH severity. We observed that overexpression of Cyld in hepatocytes concomitantly inhibits lipid accumulation, insulin resistance, inflammation and fibrosis in mice with NASH induced in an experimental setting. Mechanistically, CYLD interacts directly with the kinase TAK1 and removes its K63-linked polyubiquitin chain, which blocks downstream activation of the JNK-p38 cascades. Notably, reconstitution of hepatic CYLD expression effectively reverses disease progression in mice with dietary or genetically induced NASH and in high-fat diet-fed monkeys predisposed to metabolic syndrome. Collectively, our findings demonstrate that CYLD mitigates NASH severity and identify the CYLD-TAK1 axis as a promising therapeutic target for management of the disease.


Subject(s)
Cysteine Endopeptidases/genetics , Inflammation/genetics , MAP Kinase Kinase Kinases/genetics , Non-alcoholic Fatty Liver Disease/genetics , Animals , Carrier Proteins/genetics , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Diet, High-Fat/adverse effects , Disease Models, Animal , Haplorhini , Humans , Inflammation/physiopathology , Liver/metabolism , Liver/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Mice , Neoplasm Proteins/genetics , Non-alcoholic Fatty Liver Disease/physiopathology , Nuclear Proteins/genetics , Protein Binding/genetics , Severity of Illness Index , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/genetics
12.
Nat Med ; 24(1): 84-94, 2018 01.
Article in English | MEDLINE | ID: mdl-29227477

ABSTRACT

Activation of apoptosis signal-regulating kinase 1 (ASK1) in hepatocytes is a key process in the progression of nonalcoholic steatohepatitis (NASH) and a promising target for treatment of the condition. However, the mechanism underlying ASK1 activation is still unclear, and thus the endogenous regulators of this kinase remain open to be exploited as potential therapeutic targets. In screening for proteins that interact with ASK1 in the context of NASH, we identified the deubiquitinase tumor necrosis factor alpha-induced protein 3 (TNFAIP3) as a key endogenous suppressor of ASK1 activation, and we found that TNFAIP3 directly interacts with and deubiquitinates ASK1 in hepatocytes. Hepatocyte-specific ablation of Tnfaip3 exacerbated nonalcoholic fatty liver disease- and NASH-related phenotypes in mice, including glucose metabolism disorders, lipid accumulation and enhanced inflammation, in an ASK1-dependent manner. In contrast, transgenic or adeno-associated virus-mediated TNFAIP3 gene delivery in the liver in both mouse and nonhuman primate models of NASH substantially blocked the onset and progression of the disease. These results implicate TNFAIP3 as a functionally important endogenous suppressor of ASK1 hyperactivation in the pathogenesis of NASH and identify it as a potential new molecular target for NASH therapy.


Subject(s)
DNA-Binding Proteins/metabolism , Liver/enzymology , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Apoptosis , Diet, High-Fat , Fibrosis/prevention & control , Humans , Inflammation/prevention & control , Insulin Resistance , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Signal Transduction , Ubiquitination , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Hepatology ; 66(6): 1866-1884, 2017 12.
Article in English | MEDLINE | ID: mdl-28718215

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, impaired insulin sensitivity, and chronic low-grade inflammation. However, the pathogenic mechanism of NAFLD is poorly understood, which hinders the exploration of possible treatments. Here, we report that ubiquitin-specific protease 18 (USP18), a member of the deubiquitinating enzyme family, plays regulatory roles in NAFLD progression. Expression of USP18 was down-regulated in the livers of nonalcoholic steatohepatitis patients and high-fat diet (HFD)-induced or genetically obese mice. When challenged with HFD, hepatocyte-specific USP18 transgenic mice exhibited improved lipid metabolism and insulin sensitivity, whereas mice knocked out of USP18 expression showed adverse trends regarding hepatic steatosis and glucose metabolic disorders. Furthermore, the concomitant inflammatory response was suppressed in USP18-hepatocyte-specific transgenic mice and promoted in USP18-hepatocyte-specific knockout mice treated with HFD. Mechanistically, hepatocyte USP18 ameliorates hepatic steatosis by interacting with and deubiquitinating transforming growth factorß-activated kinase 1 (TAK1), which inhibits TAK1 activation and subsequently suppresses the downstream c-Jun N-terminal kinase and nuclear factor kappa B signaling pathways. This is further validated by alleviated steatotic phenotypes and highly activated insulin signaling in HFD-fed USP18-hepatocyte-specific knockout mice administered a TAK1 inhibitor. The therapeutic effect of USP18 on NAFLD relies on its deubiquitinating activity because HFD-fed mice injected with active-site mutant USP18 failed to inhibit hepatic steatosis. CONCLUSION: USP18 associates with and deubiquitinates TAK1 to protect against hepatic steatosis, insulin resistance, and the inflammatory response. (Hepatology 2017;66:1866-1884).


Subject(s)
Endopeptidases/metabolism , Fatty Liver/enzymology , Inflammation/enzymology , Insulin Resistance , Ubiquitin Thiolesterase/metabolism , Animals , Deubiquitinating Enzymes/metabolism , Humans , Liver/enzymology , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...