Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Research

A pan-cancer analysis of the oncogenic and immunological roles of apolipoprotein F (APOF) in human cancer

verfasst von: Xu Shi, Dechao Feng, Dengxiong Li, Ping Han, Lu Yang, Wuran Wei

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Background

Apolipoprotein F (APOF) has been less studied in cancers. Thus, we aimed to perform a pan-cancer analysis of the oncogenic and immunological effects of APOF on human cancer.

Methods

A standardized TCGA pan-cancer dataset was downloaded. Differential expression, clinical prognosis, genetic mutations, immune infiltration, epigenetic modifications, tumor stemness and heterogeneity were analyzed. We conducted all analyses through software R (version 3.6.3) and its suitable packages.

Results

Overall, we found that the common cancers differentially expressed between tumor and normal samples and prognostic-associated were BRCA, PRAD, KIRP, and LIHC in terms of overall survival (OS), disease-free survival (DFS) and progression-free survival (PFS). The pan-cancer Spearman analysis showed that the mRNA expression of APOF was negatively correlated with four tumor stemness indexes (DMPss, DNAss, ENHss, and EREG-METHss) with statistical significance for PRAD and was positively correlated for LIHC. In terms of BRCA and PRAD patients, we found negative correlation of APOF with TMB, MSI, neo, HRD and LOH. The mutation frequencies of BRCA and LIHC were 0.3%. APOF expression was negatively correlated with immune infiltration and positively correlated with tumor purity for PRAD patients. The mRNA expression of APOF was negatively associated with most TILs for LIHC, B cells, CD4+ T cells, neutrophils, macrophages and dendritic cells, but was positively associated with CD8+ T cells.

Conclusions

Our pan-cancer study offered a relatively comprehensive understanding of the roles of APOF on BRCA, PRAD, KIRP, and LIHC.

Introduction

The apolipoprotein F (APOF) gene is located at 12q13.3, and its product was identified as a minor apolipoprotein in plasma first in 1978, which may be involved in cholesterol (CE) transport and/or esterification [1]. APOF was found to be expressed at a considerable higher level in normal liver than in other parts of human organs, mainly participating in lipoprotein metabolism [2, 3]. APOF inhibits cholesteryl ester transfer protein (CETP) activity, among which it preferentially inhibits transfer events involving low-density lipoprotein (LDL) [4]. In this way, APOF can reduce LDL levels and enhance reverse CE transport in mouse model [5]. Conversely, APOF knockdown enhances the transfer of cholesteryl esters to LDL [6]. Meanwhile, APOF overexpression reduces HDL-CE level in mice by increasing the clearance of HDL-CE [7]. Clinical trials have also revealed that APOF concentrations are lower in patients with hypertriglyceridemia than in healthy controls [8]. APOF is not only associated with lipid metabolism but also potentially with glucose metabolism and has recently been identified as a circulating biomarker associated with the risk of type 2 diabetes [9]. Given the established association between lipid metabolism and disease risk for various types of cancer, for example, prostate cancer and bladder cancer [10, 11], this is not difficult to understand that the expression level of APOF has been found to be helpful in colorectal cancer (CRC) and cholangiocarcinoma (CHOL) screening [12, 13], predicting prognosis in hepatocellular carcinoma (LIHC) [14], and HPV status in oropharyngeal squamous cell carcinoma in previous studies [15].
In this study, we drew the oncological data from the Cancer Genome Atlas (TCGA) to perform a pan-cancer analysis of APOF [16], primarily focusing on four types of cancer in which APOF is both differentially expressed between tumor and normal samples and prognostic-associated, including breast invasive carcinoma (BRCA), kidney chromophobe (KIRP), liver hepatocellular carcinoma (LIHC) and prostate adenocarcinoma (PRAD).

Methods

Differential and prognostic analysis

Similar to our previous studies [17, 18], we downloaded a standardized TCGA pan-cancer dataset from the UCSC database [19] and extracted the expression data of APOF in each sample. We also screened the metastatic samples from primary blood derived cancer-peripheral blood (TCGA-LAML), primary tumor and TCGA-SKCM. In addition, we obtained a high-quality TCGA prognostic datasets from the previous TCGA prognostic study [20]. We filtered the samples with the expression level of 0 and the samples with the follow-up time shorter than 30 days, and further performed log2 (x + 0.001) transformation for each expression value. We eliminated the cancer species with the number of samples less than 10, and finally obtained the expression data of 38 cancers and the data of overall survival (OS), cancer-specific survival (CSS), disease-free survival (DFS) and progression-free survival (PFS). Cox proportional hazards regression model was used to analyze the prognostic value of APOF on cancers, and log rank test was used to obtain prognostic significance. In terms of differential expression between tumor and normal samples, we screened the samples from sloid tissue normal, primary blood derived cancer-peripheral blood, primary tumor and removed the samples with the expression level of 0. Log2 (x + 0.001) transformation for each expression value was performed as well, and cancers with the number of samples less than 3 were removed. Finally, we obtained the expression data of 18 cancers, and unpaired Wilcoxon rank sum and signed rank tests were used to perform differential significance analysis. The clinical correlations of APOF in the pan cancer were evaluated as well. In this study, the abbreviations of each cancer from the TCGA database were shown in the Additional file 1: Fig. S1A.

Tumor stemness, heterogeneity, and mutation landscape

Six tumor stemness indexes, namely differentially methylated probes-based (DMPss), DNA methylation based (DNAss), enhancer elements/DNA methylation-based (ENHss), epigenetically regulated RNA expression-based (EREG.EXPss), epigenetically regulated DNA methylation-based (EREG-METHss), RNA expression-based (RNAss) were used to analyze the correlation between stemness features and APOF expression through the Spearman analysis [21]. In addition, homologous recombination deficiency (HRD) [22], loss of heterozygosity (LOH) [22], neoantigen (NEO) [22], tumor ploidy [22], tumor purity [22], mutant-allele tumor heterogeneity (MATH) and tumor mutation burden (TMB) obtained from the GDC (https://​portal.​gdc.​cancer.​gov/​) and proceeded by MuTect2 software and R package “maftools” [23], and microsatellite instability (MSI) [24] were used to assess the relationship between tumor heterogeneity and APOF expression. We integrated the mutation data and gene expression data, and we filtered the synonymous mutation samples. In each investigated cancer, we assessed the difference in the frequency of gene mutations between high- and low-expression of APOF according to the median expression of APOF through the chi-square test.

RNA modifications and tumor immune microenvironment (TME)

We analyzed the correlations between APOF and 44 marker genes of three types of RNA modification (10 of m1A, 13 of m5C, and 21 of m6A) through the Spearman analysis. The correlations of 24 inhibitory and 36 stimulatory checkpoints [22], and 150 immunoregulatory genes (chemokine, receptor, MHC, immunoinhibitory, immunostimulatory [68]) with the mRNA expression of APOF were conducted as well. Timer [25] and ESTIMATE [26] algorithms were used to assess the TME using the R package “IOBR” [27]. In addition, we also analyzed the relationship between DNA methylation and mRNA expression of APOF, and the correlation of DNA methylation and mRNA expression of APOF with tumor infiltrating lymphocytes (TILs) were performed through the TISIDB database [28].

Statistical analysis

We conducted all analyses through software R (version 3.6.3) and its suitable packages. Unpaired Wilcoxon rank sum and signed rank tests were used to analyze pairwise differences, and Kruskal test was used to test multiple sets of samples. Statistical significance was set as two-sided p < 0.05. Significance was marked as follows: *, p < 0.05; **, p < 0.01; ***, p < 0.001.

Results

Differential and prognostic analysis

Compared to normal samples, we observed that the APOF mRNA expression was significantly upregulated in lung adenocarcinoma (LUAD), low-grade glioma (LGG), PRAD and BRCA while downregulated in kidney renal papillary cell carcinoma (KIRP), pan-kidney cohort (KIPAN), kidney renal clear cell carcinoma (KIRC), LIHC, thyroid carcinoma (THCA), KICH and cholangiocarcinoma (CHOL) patients (Fig. 1A). In terms of OS, we found that high-expression APOF was significantly associated with poor prognosis in glioma (GBMLGG), and low-expression APOF was significantly associated with poor prognosis in LIHC and pancreatic adenocarcinoma (PAAD) (Fig. 1B). For CSS, we observed that overexpression of APOF was significantly related to GBMLGG and downregulation of APOF was significantly related to PAAD (Additional file 1: Fig. S1B). For DFS, high-expression APOF was significantly associated with poor prognosis in KIRP, and low-expression APOF was significantly associated with poor prognosis in BRCA and PAAD (Fig. 1C). In terms of PFS, high-expression APOF was significantly associated with poor prognosis in GBMLGG, and low-expression APOF was significantly associated with poor prognosis for PRAD and LIHC (Fig. 1D). In contrast, APOF expression levels were found to correlate with age and gender for LIHC (Additional file 1: Fig. S1C, D). Differential expression of APOF was significant among clinical grade and stage of LIHC (Additional file 1: Fig. S1E, F) as well as T stage for LIHC, N stage for PRAD (Additional file 1: Fig. S1G–I). Overall, we found that the common cancers differentially expressed between tumor and normal samples and prognostic-associated were BRCA, PRAD, KIRP, and LIHC in terms of DFS and PFS. Moreover, we also found that APOF was differentially expressed at different ages, with positive correlations for BRCA and LIHC, and negative correlations for PRAD.

Tumor stemness, heterogeneity, mutation landscape, RNA modifications and immune regulatory genes and immune checkpoint

The pan-cancer Spearman analysis showed that the mRNA expression of APOF was positively correlated with all six tumor stemness indexes (DNAss, EREG-METHss, DMPss, ENHss, RNAss and EREG.EXPss) with statistical significance for LUAD and with four tumor stemness indexes (DNAss, EREG-METHss, DMPss and ENHss) for LIHC (Fig. 2A–F). While APOF was negatively correlated with two tumor stemness indexes (RNAss and EREG.EXPss) for BRCA (Fig. 2A–F). In terms of tumor heterogeneity, for BRCA, the mRNA expression of APOF was negatively associated with TMB (R = − 0.09), MSI (R = − 0.07), NEO (R = − 0.07), HRD (R = − 0.18) and LOH (R = − 0.18) (Fig. 3A–H). For PRAD, the mRNA expression of APOF was negatively associated with TMB (R = − 0.09), MSI (R = − 0.10), NEO ((R = − 0.10), tumor ploidy (R = − 0.18), HRD (R = − 0.23) and LOH (R = − 0.12) (Fig. 3A–H). For KIRP, the mRNA expression of APOF was positively associated with HRD (R = 0.28) (Fig. 3A–H). For LIHC, the mRNA expression of APOF was positively associated with TMB (R = 0.12) while negatively associated with MATH (R = − 0.12), MSI (R = − 0.13), NEO (R = − 0.13), HRD (R = − 0.21) and LOH (R = − 0.27) (Fig. 3A–H).
The mutation frequencies of BRCA and LIHC were 0.3% (Fig. 4A). We divided tumor patients into two groups according to the median expression of APOF. The mutations of PIK3CA, TP53, KMT2C, MUC17, SRCAP, OBSCN, CENPE, UBR5, GPS2 were significant between high- and low-expression group for BRCA (Fig. 4B). TP53, SPOP, FOXA1, TP53BP1, LRRTM1 and ERF mutations were significant for PRAD (Fig. 4C). In terms of KIRP, the genetic mutations were significant between high- and low-expression group, including TP53, SPOP, FOXA1, TP53BP1, LRRTM1 and ERF (Fig. 4C), and BAP1, RB1, SPEG, IRS4, COL15A1, STK32B, RP1L1, EPG5, TLR8 and CHSY3 mutations were significant for LIHC (Fig. 4E).

RNA modifications and TME

In terms of RNA modifications, for PRAD, the mRNA expression of APOF was positively associated with NSUN6 and FMR1, while for BRCA, the mRNA expression of APOF was positively associated with TRMT61B, TRMT6, ALKBH1, ALKBH3, TRDMT1, NSUN6, METTL3, ALKBH5, FTO and YTHDC2 (Fig. 4F). For KIRP, multiple m1A, m5C and M6A modifications were found to be positively associated with APOF expression, while DNMT3B and KIAA1429 were negatively associated (Fig. 4F). NSUN3, ALKBH5, FTO and FMR1 were positively associated with the mRNA expression of APOF (Fig. 4F). Multiple immunoregulatory genes (Fig. 5A) as well as immune checkpoint genes (Fig. 5B) were found to be associated with APOF expression levels in all four cancer types.
We found that for BRCA, APOF mRNA expression was negatively correlated with immune score (R = − 0.08) (Fig. 6B), while for LIHC, APOF mRNA expression was positively correlated with stromal score (R = 0.1) (Fig. 6C). For PRAD, APOF mRNA expression was negatively correlated with ESTIMATE score (R = − 0.19), immune score (R = − 0.19), and stromal score (R = − 0.15) (Fig. 6A–C). Meanwhile, we observed that the APOF mRNA expression was negatively associated with CD4+ T cells while positively associated with CD8+ T cells for BRCA patients (Fig. 6D). For LIHC, various TILs were negatively associated with APOF mRNA expression [29], including B cells, CD4+ T cells, neutrophils, macrophages and dendritic cells, while CD8+ T cells were positively associated (Fig. 6D).

Discussion

Approximately 75% of circulating apolipoproteins are associated with high-density lipoproteins (HDL), with the remaining 25% present in LDL [30]. Apolipoprotein (APO) has been well studied in relation to cardiovascular illness, and its relationship with cancer has been gradually revealed [31, 32]. Other members of the APO family have been found to be involved in autophagy [33, 34], oxidative stress [35, 36], apoptosis [37], and cancer drug resistance [38, 39]. ApoF, a 162 amino acid C-terminal fragment of pro-APOF, is cleaved by PCSK7 to become mature with an apparent molecular weight of about 30 kDa [40]. ApoF is mainly synthesized and secreted by the liver and was found to be associated with HDL and to a lesser extent with LDL particles in circulation [1, 7, 29, 41]. APOF mainly functions by regulating CETP. Up to 70% of HDL cholesteryl ester clearance to the liver occurs through a CETP-dependent mechanism [42, 43]. Abnormal APOF expression may lead to abnormal lipid metabolism [30, 44]. Reprogramming of lipid metabolism is a known hallmark of cancer [45]. In tumor cells, the rate of lipogenesis is significantly accelerated. The higher rate of lipogenesis in cancer cells is intended to provide substances required for cell proliferation as well as energy generation through β-oxidation of fatty acids [46]. This metabolic reprogramming triggers a series of cascading events in tumor cell physiology and often produces harmful by-products such as ROS. APOF has also been implicated in immune and inflammatory responses in animal studies [47]. For example, transcription of interferon alpha (IFNα)-responsive genes was shown to be impaired in APOF knockout mice [32]. Our study found for the first time a correlation between APOF and the occurrence and prognosis of various cancers, particularly in the four types of cancer: BRCA, PRAD, KIRP, and LIHC. This relationship may be explained from the perspective of tumor metabolism and tumor stemness, heterogeneity, and immune infiltration.
It is known that cancer is closely related to age [4851]. PRAD is one of the most common urinary tumors and its prevalence will be deteriorated as the population ages worldwide [48, 5267]. BRCA and PRAD are two of the most important hormone-related tumors known. For tumorigenesis, since high CE was found to be positively associated with breast cancer, this is consistent with our findings that elevated APOF expression in tumors compared to normal tissue causes high cholesterol [68]. We proposed that APOF increases circulating cholesterol uptake by regulating cholesterol transport and esterification to meet the increased cholesterol demand of proliferating breast cancer cells. In contrast, for prognosis, the low APOF group had larger tumors, higher differentiation and proliferation rates, and more frequently occurring HER2-like phenotypes due to elevated LDL-CE, which further suggesting an important role for APOF in BRCA, by regulating lipid levels [69, 70]. In addition, the effect of hypercholesterolemia on mammary tumor growth and metastasis was also studied in APOE knockout mice [71]. Overall, targeting APOF, i.e., targeting cholesterol transport and esterification, may be one of the targets for BRCA, but might not be as effective as directly targeting downstream cholesterol uptake and its conversion with high specificity. The role of APOF in the carcinogenesis of BRCA patients is controversial, nevertheless. Lower levels of estrogen 2 (E2) can boost ETS-1 production and rapidly induced capillary angiogenesis in BRCA patients [72]. Meanwhile, ETS-1 can activate the APOF promoter [73].
Similar to BRCA, elevated APOF is observed during tumorigenesis, which leads to high cholesterol level and has been found to be positively associated with PRAD and the risk of developing aggressive PRAD [68, 70]. The white adipose tissue around the prostate is a source of lipids used by adjacent prostate cancer cells and a local factor that stimulates the progression of PRAD, where lipids can remodel extracellular matrix and support neovascularization [74]. In addition, hypercholesterolemia is associated with elevated androgen levels as well as the androgen receptor (AR) [7578]. Whereas AR signaling may instead affect cholesterol synthesis. For example, androgen-responsive elements can upregulate the enzyme 3β-hydroxysterol Δ24-reductase (DHCR24) in AR-positive prostate cancer cells, thereby promoting cholesterol accumulation [78, 79]. In addition, our study suggested that APOF expression level was correlated with all prognostic indicators including OS, CSS, DFS and PFS in PAAD patients, although no differences were found between PAAD and normal tissues. Interestingly, we found that PRAD was the only cancer type in which APOF expression levels were negatively correlated with age, combined with our finding that APOF expression levels were much higher in PRAD tumor tissues than in normal tissues, due to the fact that PRAD is the only cancer type in which aging has been identified as a direct risk factor among all cancer types [80]. In addition, PAAD is also a hormone-related cancer and aggressive PAAD was found to be strongly upregulated on LDL-receptors in conjunction with increased cholesterol uptake [81]. Therefore, a prospective combined metabolic therapeutic strategy, in association with  other therapies, is a promising combined metabolic treatment option for PAAD [82].
The liver plays a key role in the metabolism of plasma apolipoproteins, and plasma lipid profiles may be altered in LIHC because plasma levels of apolipoproteins may be a sensitive marker of liver injury [83, 84]. Northern blot analysis showed that APOF mRNA was only found in liver tissue [4, 85]. Our research supported prior research on liver cancer cell lines that APOF expression is down-regulated in LIHC and is associated with low recurrence-free survival [14]. Our study additionally found that APOF was also associated with clinical stage and OS of LIHC patients. Since APOF expression inhibited the proliferation of LIHC cells in vitro and migrated slowly after APOF expression was upregulated [14]. Therefore, we hypothesize that APOF may play a role similar to that of a tumor suppressor gene and the one of the mechanisms of APOF-LIHC association is mediated through the intermediary of nonalcoholic fatty liver disease (NAFLD), which is a precancerous lesion of LIHC and can proceed to cirrhosis through fibrotic phase and can be exacerbated by LIHC [86]. APOF expression levels were reduced in mice on an obesogenic diet, which led to subsequent development of NAFLD and LIHC [87]. In addition, we found a large number of differences in the levels of TILs between the high and low groups of APOF mRNA expression levels, which were negatively correlated with all immune cells except CD8+ T cells. We postulate that the poor prognosis in patients with low APOF expression is associated with remodeling of the hepatic immune cell pool during NAFLD and involvement in the uncontrolled inflammatory environment that promotes liver injury and liver fibrosis [88]. In contrast, CD8+ T cells that can limit tumor load through their ability to initiate anti-tumor immune responses are instead observed to be reduced in the high-risk group [88, 89]. It is also interesting to note that our study found significant differences in APOF expression levels in male and female LIHC patients, with higher expression levels in men. In fact, in normolipidemic plasma, APOF level was 30% higher in men than in women and was positively associated with HDL and TG in normolipidemic men but not in women [8, 90]. This gender difference in APOF expression may be related to the role of HDL and TG in tumorigenesis. Care should be taken when APOF is used as a target of action.
KIRP, the incidence of which is much less studied than for KIRC, is the third most common type of RCC. Renal tumors are known to be characterized by high lipid content [91]. In our study, we found that unlike BRCA, PRAD and LIHC, APOF expression was elevated in normal tissues, and KIRP patients with high APOF expression had a worse prognosis, suggesting that APOF might not be a tumor suppressor gene for KIRP and has a tenuous relationship with immune checkpoints and immunomodulation. It is a question to be explored in the future.
Intuitively, diets rich in cholesterol or fatty acids would reduce liver APOF mRNA levels to less than half of the food-fed value [85]. However, little was previously known about the mechanisms regulating APOF gene expression. Shen et al. found that overexpression of C/EBPα and members of the ETS family increased APOF promoter activity in Huh7 cells, whereas knockdown of C/EBPα resulted in decreased APOF promoter activity in HepG2 and Huh7 cells [73]. FXR binds to and activates the FXR element ER1 in the promoter of the APOF gene [92]. For LIHC, C/EBPα is thought to activate the APOF promoter alone, from which it was hypothesized that mutations in the C/EBP binding site may almost completely eliminate APOF promoter activity [14]. Liu et al. found that APOF is also negatively regulated by agonist-activated LXR or PPARα nuclear receptors binding to a regulatory element ~ 1900 bases 5' to the APOF promoter [93].
Our research has certain limitations. Firstly, despite our observation that APOF was associated with various tumor types and tumor indicators, its specific mechanism cannot be presented through bioinformatics methods. On the other hand, we did not categorize patients of different races, according to ARIC study, there are significant racial disparities in lipid metabolism [10].
APOF is elevated during tumorigenesis in two hormone-dependent tumors, BRCA and PRAD, resulting in elevated circulating cholesterol levels by regulating cholesterol transport and esterification to supply the elevated cholesterol needs of tumor cells. Low APOF expression is then associated with poor prognosis for various tumor types, but the causal relationship might be the opposite of what we thought, and technical levels might in turn regulate a variety of lipid metabolic processes, including cholesterol accumulation. In the case of LIHC, it is most likely that the driving role of genes regulating lipid metabolism, including APOF, in NAFLD as well as in the process of LIHC is a direct result, and various immune cells in the NAFLD microenvironment could confirm this speculation. In contrast, for KIRP, APOF definitely regulates tumors in a different way. In the future, some approaches targeting APOF promoter regulation might be beneficial for the development of future therapeutic targets.

Conclusions

Our pan-cancer study offered a relatively comprehensive understanding of the roles of APOF on BRCA, PRAD, KIRP, and LIHC.

Acknowledgements

The results showed here are in whole or part based upon data generated by the TCGA Research Network: https://​www.​cancer.​gov/​tcga. We appreciated the Figdraw (www.​figdraw.​com) and Chengdu Basebiotech Co.,Ltd for their assistance in drawing and data process.

Declarations

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Not applicable.

Competing interests

The authors declare that there are no conflicts of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Olofsson SO, McConathy WJ, Alaupovic P. Isolation and partial characterization of a new acidic apolipoprotein (apolipoprotein F) from high density lipoproteins of human plasma. Biochemistry. 1978;17(6):1032–6.PubMedCrossRef Olofsson SO, McConathy WJ, Alaupovic P. Isolation and partial characterization of a new acidic apolipoprotein (apolipoprotein F) from high density lipoproteins of human plasma. Biochemistry. 1978;17(6):1032–6.PubMedCrossRef
2.
Zurück zum Zitat Fagerberg L, Hallstrom BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 2014;13(2):397–406.PubMedCrossRef Fagerberg L, Hallstrom BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 2014;13(2):397–406.PubMedCrossRef
3.
Zurück zum Zitat Deprince A, Hennuyer N, Kooijman S, Pronk ACM, Bauge E, Lienard V, et al. Apolipoprotein F is reduced in humans with steatosis and controls plasma triglyceride-rich lipoprotein metabolism. Hepatology. 2023;77(4):1287–302.PubMedCrossRef Deprince A, Hennuyer N, Kooijman S, Pronk ACM, Bauge E, Lienard V, et al. Apolipoprotein F is reduced in humans with steatosis and controls plasma triglyceride-rich lipoprotein metabolism. Hepatology. 2023;77(4):1287–302.PubMedCrossRef
4.
Zurück zum Zitat Wang X, Driscoll DM, Morton RE. Molecular cloning and expression of lipid transfer inhibitor protein reveals its identity with apolipoprotein F. J Biol Chem. 1999;274(3):1814–20.PubMedCrossRef Wang X, Driscoll DM, Morton RE. Molecular cloning and expression of lipid transfer inhibitor protein reveals its identity with apolipoprotein F. J Biol Chem. 1999;274(3):1814–20.PubMedCrossRef
5.
Zurück zum Zitat Liu Y, Morton RE. Apolipoprotein F: a natural inhibitor of cholesteryl ester transfer protein and a key regulator of lipoprotein metabolism. Curr Opin Lipidol. 2020;31(4):194–9.PubMedPubMedCentralCrossRef Liu Y, Morton RE. Apolipoprotein F: a natural inhibitor of cholesteryl ester transfer protein and a key regulator of lipoprotein metabolism. Curr Opin Lipidol. 2020;31(4):194–9.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Morton RE, Liu Y, Izem L. ApoF knockdown increases cholesteryl ester transfer to LDL and impairs cholesterol clearance in fat-fed hamsters. J Lipid Res. 2019;60(11):1868–79.PubMedPubMedCentralCrossRef Morton RE, Liu Y, Izem L. ApoF knockdown increases cholesteryl ester transfer to LDL and impairs cholesterol clearance in fat-fed hamsters. J Lipid Res. 2019;60(11):1868–79.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Lagor WR, Brown RJ, Toh SA, Millar JS, Fuki IV, de la Llera-Moya M, et al. Overexpression of apolipoprotein F reduces HDL cholesterol levels in vivo. Arterioscler Thromb Vasc Biol. 2009;29(1):40–6.PubMedCrossRef Lagor WR, Brown RJ, Toh SA, Millar JS, Fuki IV, de la Llera-Moya M, et al. Overexpression of apolipoprotein F reduces HDL cholesterol levels in vivo. Arterioscler Thromb Vasc Biol. 2009;29(1):40–6.PubMedCrossRef
8.
Zurück zum Zitat Morton RE, Gnizak HM, Greene DJ, Cho KH, Paromov VM. Lipid transfer inhibitor protein (apolipoprotein F) concentration in normolipidemic and hyperlipidemic subjects. J Lipid Res. 2008;49(1):127–35.PubMedCrossRef Morton RE, Gnizak HM, Greene DJ, Cho KH, Paromov VM. Lipid transfer inhibitor protein (apolipoprotein F) concentration in normolipidemic and hyperlipidemic subjects. J Lipid Res. 2008;49(1):127–35.PubMedCrossRef
9.
Zurück zum Zitat Croyal M, Wargny M, Chemello K, Chevalier C, Blanchard V, Bigot-Corbel E, et al. Plasma apolipoprotein concentrations and incident diabetes in subjects with prediabetes. Cardiovasc Diabetol. 2022;21(1):21.PubMedPubMedCentralCrossRef Croyal M, Wargny M, Chemello K, Chevalier C, Blanchard V, Bigot-Corbel E, et al. Plasma apolipoprotein concentrations and incident diabetes in subjects with prediabetes. Cardiovasc Diabetol. 2022;21(1):21.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Du Y, Liu Y, Cao J, Jiang X, Wang Y, Yu J, et al. LDL receptor related protein 1 is an adverse prognostic biomarker that correlates with stromal remodeling and macrophages infiltration in bladder cancer. Front Immunol. 2023;14:1113756.PubMedPubMedCentralCrossRef Du Y, Liu Y, Cao J, Jiang X, Wang Y, Yu J, et al. LDL receptor related protein 1 is an adverse prognostic biomarker that correlates with stromal remodeling and macrophages infiltration in bladder cancer. Front Immunol. 2023;14:1113756.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Chang LC, Hsu YC, Chiu HM, Ueda K, Wu MS, Kao CH, et al. Exploration of the proteomic landscape of small extracellular vesicles in serum as biomarkers for early detection of colorectal neoplasia. Front Oncol. 2021;11: 732743.PubMedPubMedCentralCrossRef Chang LC, Hsu YC, Chiu HM, Ueda K, Wu MS, Kao CH, et al. Exploration of the proteomic landscape of small extracellular vesicles in serum as biomarkers for early detection of colorectal neoplasia. Front Oncol. 2021;11: 732743.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Da Z, Gao L, Su G, Yao J, Fu W, Zhang J, et al. Bioinformatics combined with quantitative proteomics analyses and identification of potential biomarkers in cholangiocarcinoma. Cancer Cell Int. 2020;20:130.PubMedPubMedCentralCrossRef Da Z, Gao L, Su G, Yao J, Fu W, Zhang J, et al. Bioinformatics combined with quantitative proteomics analyses and identification of potential biomarkers in cholangiocarcinoma. Cancer Cell Int. 2020;20:130.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Wang YB, Zhou BX, Ling YB, Xiong ZY, Li RX, Zhong YS, et al. Decreased expression of ApoF associates with poor prognosis in human hepatocellular carcinoma. Gastroenterol Rep (Oxf). 2019;7(5):354–60.PubMedCrossRef Wang YB, Zhou BX, Ling YB, Xiong ZY, Li RX, Zhong YS, et al. Decreased expression of ApoF associates with poor prognosis in human hepatocellular carcinoma. Gastroenterol Rep (Oxf). 2019;7(5):354–60.PubMedCrossRef
15.
Zurück zum Zitat Dickinson A, Saraswat M, Syrjanen S, Tohmola T, Silen R, Randen-Brady R, et al. Comparing serum protein levels can aid in differentiating HPV-negative and -positive oropharyngeal squamous cell carcinoma patients. PLoS ONE. 2020;15(6): e0233974.PubMedPubMedCentralCrossRef Dickinson A, Saraswat M, Syrjanen S, Tohmola T, Silen R, Randen-Brady R, et al. Comparing serum protein levels can aid in differentiating HPV-negative and -positive oropharyngeal squamous cell carcinoma patients. PLoS ONE. 2020;15(6): e0233974.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–27.PubMedCrossRef Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, et al. Cellular senescence: defining a path forward. Cell. 2019;179(4):813–27.PubMedCrossRef
17.
Zurück zum Zitat Zhu W, Feng D, Shi X, Li D, Wei Q, Yang L. A pan-cancer analysis of the oncogenic role of zinc finger protein 419 in human cancer. Front Oncol. 2022;12:1042118. Zhu W, Feng D, Shi X, Li D, Wei Q, Yang L. A pan-cancer analysis of the oncogenic role of zinc finger protein 419 in human cancer. Front Oncol. 2022;12:1042118.
18.
Zurück zum Zitat Feng D, Shi X, Zhu W, Zhang F, Li D, Han P, et al. A pan-cancer analysis of the oncogenic role of leucine zipper protein 2 in human cancer. Exp Hematol Oncol. 2022 Sep 15;11(1):55. Feng D, Shi X, Zhu W, Zhang F, Li D, Han P, et al. A pan-cancer analysis of the oncogenic role of leucine zipper protein 2 in human cancer. Exp Hematol Oncol. 2022 Sep 15;11(1):55.
19.
Zurück zum Zitat Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38(6):675–8.PubMedPubMedCentralCrossRef Goldman MJ, Craft B, Hastie M, Repecka K, McDade F, Kamath A, et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 2020;38(6):675–8.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 2018;173(2):400–16.PubMedPubMedCentralCrossRef Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA pan-cancer clinical data resource to drive high-quality survival outcome analytics. Cell. 2018;173(2):400–16.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173(2):338–54.PubMedPubMedCentralCrossRef Malta TM, Sokolov A, Gentles AJ, Burzykowski T, Poisson L, Weinstein JN, et al. Machine learning identifies stemness features associated with oncogenic dedifferentiation. Cell. 2018;173(2):338–54.PubMedPubMedCentralCrossRef
22.
23.
Zurück zum Zitat Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905.PubMedPubMedCentralCrossRef Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 2017;77(21):e108–10.PubMedPubMedCentralCrossRef Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, et al. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 2017;77(21):e108–10.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.PubMedCrossRef
27.
Zurück zum Zitat Zeng D, Ye Z, Shen R, Yu G, Wu J, Xiong Y, et al. IOBR: multi-omics immuno-oncology biological research to decode tumor microenvironment and signatures. Front Immunol. 2021;12: 687975.PubMedPubMedCentralCrossRef Zeng D, Ye Z, Shen R, Yu G, Wu J, Xiong Y, et al. IOBR: multi-omics immuno-oncology biological research to decode tumor microenvironment and signatures. Front Immunol. 2021;12: 687975.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: an integrated repository portal for tumor-immune system interactions. Bioinformatics. 2019;35(20):4200–2.PubMedCrossRef Ru B, Wong CN, Tong Y, Zhong JY, Zhong SSW, Wu WC, et al. TISIDB: an integrated repository portal for tumor-immune system interactions. Bioinformatics. 2019;35(20):4200–2.PubMedCrossRef
29.
Zurück zum Zitat Morton RE, Greene DJ. Regulation of lipid transfer between lipoproteins by an endogenous plasma protein: selective inhibition among lipoprotein classes. J Lipid Res. 1994;35(5):836–47.PubMedCrossRef Morton RE, Greene DJ. Regulation of lipid transfer between lipoproteins by an endogenous plasma protein: selective inhibition among lipoprotein classes. J Lipid Res. 1994;35(5):836–47.PubMedCrossRef
30.
Zurück zum Zitat He Y, Greene DJ, Kinter M, Morton RE. Control of cholesteryl ester transfer protein activity by sequestration of lipid transfer inhibitor protein in an inactive complex. J Lipid Res. 2008;49(7):1529–37.PubMedPubMedCentralCrossRef He Y, Greene DJ, Kinter M, Morton RE. Control of cholesteryl ester transfer protein activity by sequestration of lipid transfer inhibitor protein in an inactive complex. J Lipid Res. 2008;49(7):1529–37.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Peloso GM, Demissie S, Collins D, Mirel DB, Gabriel SB, Cupples LA, et al. Common genetic variation in multiple metabolic pathways influences susceptibility to low HDL-cholesterol and coronary heart disease. J Lipid Res. 2010;51(12):3524–32.PubMedPubMedCentralCrossRef Peloso GM, Demissie S, Collins D, Mirel DB, Gabriel SB, Cupples LA, et al. Common genetic variation in multiple metabolic pathways influences susceptibility to low HDL-cholesterol and coronary heart disease. J Lipid Res. 2010;51(12):3524–32.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Conlon DM, Thomas T, Fedotova T, Hernandez-Ono A, Di Paolo G, Chan RB, et al. Inhibition of apolipoprotein B synthesis stimulates endoplasmic reticulum autophagy that prevents steatosis. J Clin Invest. 2016;126(10):3852–67.PubMedPubMedCentralCrossRef Conlon DM, Thomas T, Fedotova T, Hernandez-Ono A, Di Paolo G, Chan RB, et al. Inhibition of apolipoprotein B synthesis stimulates endoplasmic reticulum autophagy that prevents steatosis. J Clin Invest. 2016;126(10):3852–67.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Parcon PA, Balasubramaniam M, Ayyadevara S, Jones RA, Liu L, Shmookler Reis RJ, et al. Apolipoprotein E4 inhibits autophagy gene products through direct, specific binding to CLEAR motifs. Alzheimers Dement. 2018;14(2):230–42.PubMedCrossRef Parcon PA, Balasubramaniam M, Ayyadevara S, Jones RA, Liu L, Shmookler Reis RJ, et al. Apolipoprotein E4 inhibits autophagy gene products through direct, specific binding to CLEAR motifs. Alzheimers Dement. 2018;14(2):230–42.PubMedCrossRef
35.
Zurück zum Zitat Trougakos IP, So A, Jansen B, Gleave ME, Gonos ES. Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 2004;64(5):1834–42.PubMedCrossRef Trougakos IP, So A, Jansen B, Gleave ME, Gonos ES. Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 2004;64(5):1834–42.PubMedCrossRef
36.
Zurück zum Zitat Bajo-Graneras R, Crespo-Sanjuan J, Garcia-Centeno RM, Garrote-Adrados JA, Gutierrez G, Garcia-Tejeiro M, et al. Expression and potential role of apolipoprotein D on the death-survival balance of human colorectal cancer cells under oxidative stress conditions. Int J Colorectal Dis. 2013;28(6):751–66.PubMedCrossRef Bajo-Graneras R, Crespo-Sanjuan J, Garcia-Centeno RM, Garrote-Adrados JA, Gutierrez G, Garcia-Tejeiro M, et al. Expression and potential role of apolipoprotein D on the death-survival balance of human colorectal cancer cells under oxidative stress conditions. Int J Colorectal Dis. 2013;28(6):751–66.PubMedCrossRef
37.
Zurück zum Zitat Zhaorigetu S, Yang Z, Toma I, McCaffrey TA, Hu CA. Apolipoprotein L6, induced in atherosclerotic lesions, promotes apoptosis and blocks Beclin 1-dependent autophagy in atherosclerotic cells. J Biol Chem. 2011;286(31):27389–98.PubMedPubMedCentralCrossRef Zhaorigetu S, Yang Z, Toma I, McCaffrey TA, Hu CA. Apolipoprotein L6, induced in atherosclerotic lesions, promotes apoptosis and blocks Beclin 1-dependent autophagy in atherosclerotic cells. J Biol Chem. 2011;286(31):27389–98.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Yiu CC, Sasano H, Ono K, Chow LW. Changes in protein expression after neoadjuvant use of aromatase inhibitors in primary breast cancer: a proteomic approach to search for potential biomarkers to predict response or resistance. Expert Opin Investig Drugs. 2010;19(Suppl 1):S79-89.PubMedCrossRef Yiu CC, Sasano H, Ono K, Chow LW. Changes in protein expression after neoadjuvant use of aromatase inhibitors in primary breast cancer: a proteomic approach to search for potential biomarkers to predict response or resistance. Expert Opin Investig Drugs. 2010;19(Suppl 1):S79-89.PubMedCrossRef
39.
Zurück zum Zitat Cruz IN, Coley HM, Kramer HB, Madhuri TK, Safuwan NA, Angelino AR, et al. Proteomics analysis of ovarian cancer cell lines and tissues reveals drug resistance-associated proteins. Cancer Genomics Proteomics. 2017;14(1):35–51.PubMedCrossRef Cruz IN, Coley HM, Kramer HB, Madhuri TK, Safuwan NA, Angelino AR, et al. Proteomics analysis of ovarian cancer cell lines and tissues reveals drug resistance-associated proteins. Cancer Genomics Proteomics. 2017;14(1):35–51.PubMedCrossRef
40.
Zurück zum Zitat Guillemot J, Essalmani R, Hamelin J, Seidah NG. Is there a link between proprotein convertase PC7 activity and human lipid homeostasis? FEBS Open Bio. 2014;4:741–5.PubMedPubMedCentralCrossRef Guillemot J, Essalmani R, Hamelin J, Seidah NG. Is there a link between proprotein convertase PC7 activity and human lipid homeostasis? FEBS Open Bio. 2014;4:741–5.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Lagor WR, Fields DW, Khetarpal SA, Kumaravel A, Lin W, Weintraub N, et al. The effects of apolipoprotein F deficiency on high density lipoprotein cholesterol metabolism in mice. PLoS ONE. 2012;7(2): e31616.PubMedPubMedCentralCrossRef Lagor WR, Fields DW, Khetarpal SA, Kumaravel A, Lin W, Weintraub N, et al. The effects of apolipoprotein F deficiency on high density lipoprotein cholesterol metabolism in mice. PLoS ONE. 2012;7(2): e31616.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Rader DJ, Alexander ET, Weibel GL, Billheimer J, Rothblat GH. The role of reverse cholesterol transport in animals and humans and relationship to atherosclerosis. J Lipid Res. 2009;50(Suppl):S189–94.PubMedPubMedCentralCrossRef Rader DJ, Alexander ET, Weibel GL, Billheimer J, Rothblat GH. The role of reverse cholesterol transport in animals and humans and relationship to atherosclerosis. J Lipid Res. 2009;50(Suppl):S189–94.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Schwartz CC, VandenBroek JM, Cooper PS. Lipoprotein cholesteryl ester production, transfer, and output in vivo in humans. J Lipid Res. 2004;45(9):1594–607.PubMedCrossRef Schwartz CC, VandenBroek JM, Cooper PS. Lipoprotein cholesteryl ester production, transfer, and output in vivo in humans. J Lipid Res. 2004;45(9):1594–607.PubMedCrossRef
44.
Zurück zum Zitat Morton RE, Greene DJ. Conversion of lipid transfer inhibitor protein (apolipoprotein F) to its active form depends on LDL composition. J Lipid Res. 2011;52(12):2262–71.PubMedPubMedCentralCrossRef Morton RE, Greene DJ. Conversion of lipid transfer inhibitor protein (apolipoprotein F) to its active form depends on LDL composition. J Lipid Res. 2011;52(12):2262–71.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Gupta A, Stokes W, Eguchi M, Hararah M, Amini A, Mueller A, et al. Statin use associated with improved overall and cancer specific survival in patients with head and neck cancer. Oral Oncol. 2019;90:54–66.PubMedPubMedCentralCrossRef Gupta A, Stokes W, Eguchi M, Hararah M, Amini A, Mueller A, et al. Statin use associated with improved overall and cancer specific survival in patients with head and neck cancer. Oral Oncol. 2019;90:54–66.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33.PubMedPubMedCentralCrossRef Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–33.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Yang Y, Fu Q, Zhou T, Li Y, Liu S, Zeng Q, et al. Analysis of apolipoprotein genes and their involvement in disease response of channel catfish after bacterial infection. Dev Comp Immunol. 2017;67:464–70.PubMedCrossRef Yang Y, Fu Q, Zhou T, Li Y, Liu S, Zeng Q, et al. Analysis of apolipoprotein genes and their involvement in disease response of channel catfish after bacterial infection. Dev Comp Immunol. 2017;67:464–70.PubMedCrossRef
48.
Zurück zum Zitat Dechao Feng DL, Ruicheng Wu , Ping Han. Scientific Advancements in Drug Development and Trials for Urothelial Carcinoma: Insights From the 2023 ASCO-GU Cancers Symposium. Aging and disease. 2023. Dechao Feng DL, Ruicheng Wu , Ping Han. Scientific Advancements in Drug Development and Trials for Urothelial Carcinoma: Insights From the 2023 ASCO-GU Cancers Symposium. Aging and disease. 2023.
49.
Zurück zum Zitat Shen W, He J, Hou T, Si J, Chen S. Common Pathogenetic Mechanisms Underlying Aging and Tumor and Means of Interventions. Aging Dis. 2022 Jul 11;13(4):1063–91. Shen W, He J, Hou T, Si J, Chen S. Common Pathogenetic Mechanisms Underlying Aging and Tumor and Means of Interventions. Aging Dis. 2022 Jul 11;13(4):1063–91.
50.
Zurück zum Zitat Schwartz AG. Dehydroepiandrosterone, Cancer, and Aging. Aging Dis. 2022 Apr;13(2):423–32. Schwartz AG. Dehydroepiandrosterone, Cancer, and Aging. Aging Dis. 2022 Apr;13(2):423–32.
51.
Zurück zum Zitat Xu Z, Huang J, Gao M, Guo G, Zeng S, Chen X, et al. Current perspectives on the clinical implications of oxidative RNA damage in aging research: challenges and opportunities. Geroscience. 2021 Apr;43(2):487–505. Xu Z, Huang J, Gao M, Guo G, Zeng S, Chen X, et al. Current perspectives on the clinical implications of oxidative RNA damage in aging research: challenges and opportunities. Geroscience. 2021 Apr;43(2):487–505.
52.
Zurück zum Zitat Zhu WZ, Feng DC, Xiong Q, Shi X, Zhang FC, Wei Q, et al. An autophagy-related gene prognostic index predicting biochemical recurrence, metastasis, and drug resistance for prostate cancer. Asian J Androl. 2023 Mar-Apr;25(2):208–16. Zhu WZ, Feng DC, Xiong Q, Shi X, Zhang FC, Wei Q, et al. An autophagy-related gene prognostic index predicting biochemical recurrence, metastasis, and drug resistance for prostate cancer. Asian J Androl. 2023 Mar-Apr;25(2):208–16.
53.
Zurück zum Zitat Weitao Zheng DF, Xingyu Xiong, Xinyang Liao, Sheng Wang, Hang Xu, Weizhen Le, Qiang Wei, Lu Yang. The Role of cGASSTING in Age-Related Diseases from Mechanisms to Therapies. Aging and Disease. 2023. Weitao Zheng DF, Xingyu Xiong, Xinyang Liao, Sheng Wang, Hang Xu, Weizhen Le, Qiang Wei, Lu Yang. The Role of cGASSTING in Age-Related Diseases from Mechanisms to Therapies. Aging and Disease. 2023.
54.
Zurück zum Zitat Feng DC, Zhu WZ, Shi X, Xiong Q, You J, Wei Q, et al. Identification of senescence-related molecular subtypes and key genes for prostate cancer. Asian J Androl. 2023 Mar-Apr;25(2):223–9. Feng DC, Zhu WZ, Shi X, Xiong Q, You J, Wei Q, et al. Identification of senescence-related molecular subtypes and key genes for prostate cancer. Asian J Androl. 2023 Mar-Apr;25(2):223–9.
55.
Zurück zum Zitat Feng D, Zhu W, Shi X, Wang Z, Wei W, Wei Q, et al. Immune-related gene index predicts metastasis for prostate cancer patients undergoing radical radiotherapy. Exp Hematol Oncol. 2023 Jan 12;12(1):8. Feng D, Zhu W, Shi X, Wang Z, Wei W, Wei Q, et al. Immune-related gene index predicts metastasis for prostate cancer patients undergoing radical radiotherapy. Exp Hematol Oncol. 2023 Jan 12;12(1):8.
56.
Zurück zum Zitat Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology. 2023 Feb;168(2):199–202. Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology. 2023 Feb;168(2):199–202.
57.
Zurück zum Zitat Feng D, Zhu W, Shi X, Xiong Q, Li D, Wei W, et al. Spindle and kinetochore-associated complex subunit 3 could serve as a prognostic biomarker for prostate cancer. Exp Hematol Oncol. 2022 Oct 20;11(1):76. Feng D, Zhu W, Shi X, Xiong Q, Li D, Wei W, et al. Spindle and kinetochore-associated complex subunit 3 could serve as a prognostic biomarker for prostate cancer. Exp Hematol Oncol. 2022 Oct 20;11(1):76.
58.
Zurück zum Zitat Feng D, Zhang F, Li D, Shi X, Xiong Q, Wei Q, et al. Developing an immune-related gene prognostic index associated with progression and providing new insights into the tumor immune microenvironment of prostate cancer. Immunology. 2022 Jun;166(2):197–209. Feng D, Zhang F, Li D, Shi X, Xiong Q, Wei Q, et al. Developing an immune-related gene prognostic index associated with progression and providing new insights into the tumor immune microenvironment of prostate cancer. Immunology. 2022 Jun;166(2):197–209.
59.
Zurück zum Zitat Feng D, Xiong Q, Zhang F, Shi X, Xu H, Wei W, et al. Identification of a Novel Nomogram to Predict Progression Based on the Circadian Clock and Insights Into the Tumor Immune Microenvironment in Prostate Cancer. Front Immunol. 2022;13:777724. Feng D, Xiong Q, Zhang F, Shi X, Xu H, Wei W, et al. Identification of a Novel Nomogram to Predict Progression Based on the Circadian Clock and Insights Into the Tumor Immune Microenvironment in Prostate Cancer. Front Immunol. 2022;13:777724.
60.
Zurück zum Zitat Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology. 2022 Feb 10. Feng D, Xiong Q, Wei Q, Yang L. Cellular landscape of tumour microenvironment in prostate cancer. Immunology. 2022 Feb 10.
61.
Zurück zum Zitat Feng D, Shi X, Zhang F, Xiong Q, Wei Q, Yang L. Energy Metabolism-Related Gene Prognostic Index Predicts Biochemical Recurrence for Patients With Prostate Cancer Undergoing Radical Prostatectomy. Front Immunol. 2022;13:839362. Feng D, Shi X, Zhang F, Xiong Q, Wei Q, Yang L. Energy Metabolism-Related Gene Prognostic Index Predicts Biochemical Recurrence for Patients With Prostate Cancer Undergoing Radical Prostatectomy. Front Immunol. 2022;13:839362.
62.
Zurück zum Zitat Feng D, Shi X, Zhang F, Xiong Q, Wei Q, Yang L. Mitochondria Dysfunction-Mediated Molecular Subtypes and Gene Prognostic Index for Prostate Cancer Patients Undergoing Radical Prostatectomy or Radiotherapy. Front Oncol. 2022;12:858479. Feng D, Shi X, Zhang F, Xiong Q, Wei Q, Yang L. Mitochondria Dysfunction-Mediated Molecular Subtypes and Gene Prognostic Index for Prostate Cancer Patients Undergoing Radical Prostatectomy or Radiotherapy. Front Oncol. 2022;12:858479.
63.
Zurück zum Zitat Feng D, Shi X, You J, Xiong Q, Zhu W, Wei Q, et al. A cellular senescence-related gene prognostic index for biochemical recurrence and drug resistance in patients with prostate cancer. Am J Cancer Res. 2022;12(8):3811–28. Feng D, Shi X, You J, Xiong Q, Zhu W, Wei Q, et al. A cellular senescence-related gene prognostic index for biochemical recurrence and drug resistance in patients with prostate cancer. Am J Cancer Res. 2022;12(8):3811–28.
64.
Zurück zum Zitat Feng D, Shi X, Xiong Q, Zhang F, Li D, Wei W, et al. A Ferroptosis-Related Gene Prognostic Index Associated With Biochemical Recurrence and Radiation Resistance for Patients With Prostate Cancer Undergoing Radical Radiotherapy. Front Cell Dev Biol. 2022;10:803766. Feng D, Shi X, Xiong Q, Zhang F, Li D, Wei W, et al. A Ferroptosis-Related Gene Prognostic Index Associated With Biochemical Recurrence and Radiation Resistance for Patients With Prostate Cancer Undergoing Radical Radiotherapy. Front Cell Dev Biol. 2022;10:803766.
65.
Zurück zum Zitat Feng D, Li D, Shi X, Xiong Q, Zhang F, Wei Q, et al. A gene prognostic index from cellular senescence predicting metastasis and radio resistance for prostate cancer. J Transl Med. 2022 Jun 3;20(1):252. Feng D, Li D, Shi X, Xiong Q, Zhang F, Wei Q, et al. A gene prognostic index from cellular senescence predicting metastasis and radio resistance for prostate cancer. J Transl Med. 2022 Jun 3;20(1):252.
66.
Zurück zum Zitat Feng D, Shi X, Xiong Q, Zhang F, Li D, Yang L. A Gene Prognostic Index Associated With Epithelial-Mesenchymal Transition Predicting Biochemical Recurrence and Tumor Chemoresistance for Prostate Cancer. Front Oncol. 2021;11:805571. Feng D, Shi X, Xiong Q, Zhang F, Li D, Yang L. A Gene Prognostic Index Associated With Epithelial-Mesenchymal Transition Predicting Biochemical Recurrence and Tumor Chemoresistance for Prostate Cancer. Front Oncol. 2021;11:805571.
67.
Zurück zum Zitat Feng D, Wang J, Shi X, Li D, Wei W, Han P. Membrane tension-mediated stiff and soft tumor subtypes closely associated with prognosis for prostate cancer patients. Eur J Med Res. 2023 May 13;28(1):172. Feng D, Wang J, Shi X, Li D, Wei W, Han P. Membrane tension-mediated stiff and soft tumor subtypes closely associated with prognosis for prostate cancer patients. Eur J Med Res. 2023 May 13;28(1):172.
68.
Zurück zum Zitat Kitahara CM, Berrington de Gonzalez A, Freedman ND, Huxley R, Mok Y, Jee SH, et al. Total cholesterol and cancer risk in a large prospective study in Korea. J Clin Oncol. 2011;29(12):1592–8.PubMedPubMedCentralCrossRef Kitahara CM, Berrington de Gonzalez A, Freedman ND, Huxley R, Mok Y, Jee SH, et al. Total cholesterol and cancer risk in a large prospective study in Korea. J Clin Oncol. 2011;29(12):1592–8.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Rodrigues Dos Santos C, Fonseca I, Dias S, Mendes de Almeida JC. Plasma level of LDL-cholesterol at diagnosis is a predictor factor of breast tumor progression. BMC Cancer. 2014;14:132.PubMedPubMedCentralCrossRef Rodrigues Dos Santos C, Fonseca I, Dias S, Mendes de Almeida JC. Plasma level of LDL-cholesterol at diagnosis is a predictor factor of breast tumor progression. BMC Cancer. 2014;14:132.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Alikhani N, Ferguson RD, Novosyadlyy R, Gallagher EJ, Scheinman EJ, Yakar S, et al. Mammary tumor growth and pulmonary metastasis are enhanced in a hyperlipidemic mouse model. Oncogene. 2013;32(8):961–7.PubMedCrossRef Alikhani N, Ferguson RD, Novosyadlyy R, Gallagher EJ, Scheinman EJ, Yakar S, et al. Mammary tumor growth and pulmonary metastasis are enhanced in a hyperlipidemic mouse model. Oncogene. 2013;32(8):961–7.PubMedCrossRef
72.
Zurück zum Zitat Lincoln DW 2nd, Phillips PG, Bove K. Estrogen-induced Ets-1 promotes capillary formation in an in vitro tumor angiogenesis model. Breast Cancer Res Treat. 2003;78(2):167–78.PubMedCrossRef Lincoln DW 2nd, Phillips PG, Bove K. Estrogen-induced Ets-1 promotes capillary formation in an in vitro tumor angiogenesis model. Breast Cancer Res Treat. 2003;78(2):167–78.PubMedCrossRef
73.
Zurück zum Zitat Shen XB, Huang L, Zhang SH, Wang DP, Wu YL, Chen WN, et al. Transcriptional regulation of the apolipoprotein F (ApoF) gene by ETS and C/EBPalpha in hepatoma cells. Biochimie. 2015;112:1–9.PubMedCrossRef Shen XB, Huang L, Zhang SH, Wang DP, Wu YL, Chen WN, et al. Transcriptional regulation of the apolipoprotein F (ApoF) gene by ETS and C/EBPalpha in hepatoma cells. Biochimie. 2015;112:1–9.PubMedCrossRef
76.
Zurück zum Zitat Allott EH, Freedland SJ. Words of wisdom. Re: impact of circulating cholesterol levels on growth and intratumoral androgen concentration of prostate tumors. Eur Urol. 2013;63(1):178–9.PubMedCrossRef Allott EH, Freedland SJ. Words of wisdom. Re: impact of circulating cholesterol levels on growth and intratumoral androgen concentration of prostate tumors. Eur Urol. 2013;63(1):178–9.PubMedCrossRef
77.
Zurück zum Zitat Swinnen JV, Brusselmans K, Verhoeven G. Increased lipogenesis in cancer cells: new players, novel targets. Curr Opin Clin Nutr Metab Care. 2006;9(4):358–65.PubMedCrossRef Swinnen JV, Brusselmans K, Verhoeven G. Increased lipogenesis in cancer cells: new players, novel targets. Curr Opin Clin Nutr Metab Care. 2006;9(4):358–65.PubMedCrossRef
78.
Zurück zum Zitat Heemers HV, Verhoeven G, Swinnen JV. Androgen activation of the sterol regulatory element-binding protein pathway: current insights. Mol Endocrinol. 2006;20(10):2265–77.PubMedCrossRef Heemers HV, Verhoeven G, Swinnen JV. Androgen activation of the sterol regulatory element-binding protein pathway: current insights. Mol Endocrinol. 2006;20(10):2265–77.PubMedCrossRef
79.
Zurück zum Zitat Bonaccorsi L, Luciani P, Nesi G, Mannucci E, Deledda C, Dichiara F, et al. Androgen receptor regulation of the seladin-1/DHCR24 gene: altered expression in prostate cancer. Lab Invest. 2008;88(10):1049–56.PubMedCrossRef Bonaccorsi L, Luciani P, Nesi G, Mannucci E, Deledda C, Dichiara F, et al. Androgen receptor regulation of the seladin-1/DHCR24 gene: altered expression in prostate cancer. Lab Invest. 2008;88(10):1049–56.PubMedCrossRef
80.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.PubMedCrossRef Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.PubMedCrossRef
81.
Zurück zum Zitat Gonias SL, Karimi-Mostowfi N, Murray SS, Mantuano E, Gilder AS. Expression of LDL receptor-related proteins (LRPs) in common solid malignancies correlates with patient survival. PLoS ONE. 2017;12(10): e0186649.PubMedPubMedCentralCrossRef Gonias SL, Karimi-Mostowfi N, Murray SS, Mantuano E, Gilder AS. Expression of LDL receptor-related proteins (LRPs) in common solid malignancies correlates with patient survival. PLoS ONE. 2017;12(10): e0186649.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Guillaumond F, Bidaut G, Ouaissi M, Servais S, Gouirand V, Olivares O, et al. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2015;112(8):2473–8.PubMedPubMedCentralCrossRef Guillaumond F, Bidaut G, Ouaissi M, Servais S, Gouirand V, Olivares O, et al. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2015;112(8):2473–8.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Muir K, Hazim A, He Y, Peyressatre M, Kim DY, Song X, et al. Proteomic and lipidomic signatures of lipid metabolism in NASH-associated hepatocellular carcinoma. Cancer Res. 2013;73(15):4722–31.PubMedCrossRef Muir K, Hazim A, He Y, Peyressatre M, Kim DY, Song X, et al. Proteomic and lipidomic signatures of lipid metabolism in NASH-associated hepatocellular carcinoma. Cancer Res. 2013;73(15):4722–31.PubMedCrossRef
84.
Zurück zum Zitat Ma XL, Gao XH, Gong ZJ, Wu J, Tian L, Zhang CY, et al. Apolipoprotein A1: a novel serum biomarker for predicting the prognosis of hepatocellular carcinoma after curative resection. Oncotarget. 2016;7(43):70654–68.PubMedPubMedCentralCrossRef Ma XL, Gao XH, Gong ZJ, Wu J, Tian L, Zhang CY, et al. Apolipoprotein A1: a novel serum biomarker for predicting the prognosis of hepatocellular carcinoma after curative resection. Oncotarget. 2016;7(43):70654–68.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Izem L, Morton RE. Molecular cloning of hamster lipid transfer inhibitor protein (apolipoprotein F) and regulation of its expression by hyperlipidemia. J Lipid Res. 2009;50(4):676–84.PubMedPubMedCentralCrossRef Izem L, Morton RE. Molecular cloning of hamster lipid transfer inhibitor protein (apolipoprotein F) and regulation of its expression by hyperlipidemia. J Lipid Res. 2009;50(4):676–84.PubMedPubMedCentralCrossRef
86.
87.
Zurück zum Zitat Asgharpour A, Cazanave SC, Pacana T, Seneshaw M, Vincent R, Banini BA, et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J Hepatol. 2016;65(3):579–88.PubMedPubMedCentralCrossRef Asgharpour A, Cazanave SC, Pacana T, Seneshaw M, Vincent R, Banini BA, et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J Hepatol. 2016;65(3):579–88.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22(7):429–43.PubMedCrossRef Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22(7):429–43.PubMedCrossRef
89.
Zurück zum Zitat Tilg H, Adolph TE, Dudek M, Knolle P. Non-alcoholic fatty liver disease: the interplay between metabolism, microbes and immunity. Nat Metab. 2021;3(12):1596–607.PubMedCrossRef Tilg H, Adolph TE, Dudek M, Knolle P. Non-alcoholic fatty liver disease: the interplay between metabolism, microbes and immunity. Nat Metab. 2021;3(12):1596–607.PubMedCrossRef
90.
Zurück zum Zitat Morton RE, Nunes V, Izem L, Quintao E. Markedly elevated lipid transfer inhibitor protein in hypercholesterolemic subjects is mitigated by plasma triglyceride levels. Arterioscler Thromb Vasc Biol. 2001;21(10):1642–9.PubMedCrossRef Morton RE, Nunes V, Izem L, Quintao E. Markedly elevated lipid transfer inhibitor protein in hypercholesterolemic subjects is mitigated by plasma triglyceride levels. Arterioscler Thromb Vasc Biol. 2001;21(10):1642–9.PubMedCrossRef
91.
Zurück zum Zitat Tugnoli V, Trinchero A, Tosi MR. Evaluation of the lipid composition of human healthy and neoplastic renal tissues. Ital J Biochem. 2004;53(4):169–82.PubMed Tugnoli V, Trinchero A, Tosi MR. Evaluation of the lipid composition of human healthy and neoplastic renal tissues. Ital J Biochem. 2004;53(4):169–82.PubMed
92.
Zurück zum Zitat Li L, Liu H, Peng J, Wang Y, Zhang Y, Dong J, et al. Farnesoid X receptor up-regulates expression of lipid transfer inhibitor protein in liver cells and mice. Biochem Biophys Res Commun. 2013;441(4):880–5.PubMedCrossRef Li L, Liu H, Peng J, Wang Y, Zhang Y, Dong J, et al. Farnesoid X receptor up-regulates expression of lipid transfer inhibitor protein in liver cells and mice. Biochem Biophys Res Commun. 2013;441(4):880–5.PubMedCrossRef
93.
Zurück zum Zitat Liu Y, Izem L, Morton RE. Identification of a hormone response element that mediates suppression of APOF by LXR and PPARalpha agonists. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865(3): 158583.PubMedCrossRef Liu Y, Izem L, Morton RE. Identification of a hormone response element that mediates suppression of APOF by LXR and PPARalpha agonists. Biochim Biophys Acta Mol Cell Biol Lipids. 2020;1865(3): 158583.PubMedCrossRef
Metadaten
Titel
A pan-cancer analysis of the oncogenic and immunological roles of apolipoprotein F (APOF) in human cancer
verfasst von
Xu Shi
Dechao Feng
Dengxiong Li
Ping Han
Lu Yang
Wuran Wei
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01156-w

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe