Αρχειοθήκη ιστολογίου

Κυριακή 15 Νοεμβρίου 2020

Epigenetic modification‐dependent androgen receptor occupancy facilitates the ectopic TSPY1 expression in prostate cancer cell

Alexandros G.Sfakianakis shared this article with you from Inoreader
Μέσω Cancer Science

Summary

TSPY1 (testis‐specific protein Y‐encoded 1), a Y chromosome‐linked oncogene, is frequently activated in prostate cancers (PCa) and its expression is correlated with the poor prognosis of PCa. However, the cause leading to the ectopic transcription of TSPY1 in PCa is still unclear. Here, we observed that the methylation status in the CpG islands (CGIs) of the TSPY1 promoter was negatively correlated with its expression level in different human samples. The acetyl‐histone H4 and trimethylated histone H3‐lysine 4, two post translational modifications of histones, occupying on the TSPY1 promoter, faciliated the TSPY1 expression in PCa cells. Additionally, we found that androgen accelerated the TSPY1 trancription on condition of DNA demethylation of TSPY1‐CGIs and promoted PCa cell proliferation. Moreover, the binding of androgen receptor (AR) to the TSPY1 promoter, enhancing TSPY1 transcription, was detected in PCa cells. Taken together, our findings identify the regulation of DNA methylation, acting as a primary mechanism, on TSPY1 expression in PCa,and reveal that TSPY1 is an androgen‐AR axis‐regulated oncogene, suggesting a novel and potential target for PCa therapy

View on the web

Using protein microarray to identify and evaluate autoantibodies to tumor‐associated antigens in ovarian cancer

Alexandros G.Sfakianakis shared this article with you from Inoreader
Μέσω Cancer Science

Abstract

The aim of this study was to develop a non‐invasive serological diagnostic approach in identifying and evaluating a panel of candidate autoantibodies to tumor‐associated antigens (TAAs) based on protein microarray technology for early detection of ovarian cancer (OC). Protein microarray based on 154 proteins encoded by 138 cancer driver genes was used to screen candidate anti‐TAA autoantibodies in discovery cohort containing 17 OC and 27 normal controls. Indirect enzyme‐linked immunosorbent assay (ELISA) was used to detect the content of candidate anti‐TAA autoantibodies in sera from 140 subjects in training cohort. Differential anti‐TAA autoantibodies were further validated in the validation cohort with 328 subjects. Subsequently, 112 sera from the patients with ovarian benign diseases with 104 OC sera and 104 NC sera together were recruited to identify the specificity of representative autoantibodies to OC among ovarian diseases. Five TAAs (GNAS, NPM1, FUBP1, p53, KR AS) were screened out in discovery phase, in which four of them presented higher levels in OC than controls (P<0.05) in training cohort, which was consistent with the result in the subsequent validation cohort. An optimized panel of three anti‐TAA (GNAS, p53 and NPM1) autoantibodies was identified to have relative higher sensitivity (51.2%), specificity (86.0%) and accuracy (68.6%), respectively. This panel can identify 51% of OC patients with CA125 negative. This study supports our assumption that anti‐TAA autoantibodies can be considered as potential diagnostic biomarkers for detection of OC, especially a panel of three anti‐TAA autoantibodies could be a good tool in immunodiagnosis of OC.

View on the web

Tumor mutation burden as a biomarker for lung cancer patients treated with pemetrexed and cisplatin (the JIPANG‐TR)

Alexandros G.Sfakianakis shared this article with you from Inoreader
Μέσω Cancer Science

Abstract

The JIPANG study is a randomized phase III study of pemetrexed/cisplatin (Pem/Cis) versus vinorelbine/cisplatin (Vnr/Cis) for completely resected stage II‐IIIA non‐squamous non‐small cell lung cancer (Ns‐NSCLC). This study did not meet the primary endpoint (recurrence‐free survival, RFS) but Pem/Cis had a similar efficacy to Vnr/Cis with a better tolerability. Tumor mutation burden (TMB) is thought to have a predictive value of immune checkpoint inhibitors. However, the relevance of TMB to cytotoxic chemotherapy remains unknown. This exploratory study investigate relationship between tumor mutation profiles and clinical outcome of Pem/Cis. Formalin‐fixed, paraffin‐embedded tumor tissues (n=389) were obtained from the patients. Mutation status of tissue DNA was analyzed by targeted deep sequencing. EGFR mutations were detected frequently in Ns‐NSCLC (139/374). Patients without any EGFR mutations experience longer RFS in the Pem/Cis arm vs Vnr/Cis arms. Pem/Cis in patients with high TMB (≥12‐16 mut/Mb) tended to improve survival. In patients with wild type EGFR, TMB≥12 mut/Mb was significantly associated with improved RFS with Pem/Cis vs Vnr/Cis (not reached vs 52.5 months; hazard ratio (HR) 0.477). It could be proposed that TMB was predictive of RFS benefit with Pem/Cis vs Vnr/Cis in Ns‐NSCLC. Further investigation is required to determine whether TMB combined with EGFR mutation status could be utilized as a predictive biomarker.

View on the web

Aberrant expression of JAM‐A contributes to the malignancy of cervical adenocarcinoma via interaction with PVR/CD155.

Alexandros G.Sfakianakis shared this article with you from Inoreader
Μέσω Cancer Science

Abstract

Recent studies have shown that aberrant expression of tight junction proteins (TJPs) contributes to malignant potentials of various cancers. In this study, we investigated the expression of junctional adhesion molecule‐A (JAM‐A), one of the transmembrane TJPs, in uterine cervical adenocarcinoma and the significance of its expression for malignancy. Immunohistochemistry on human surgical specimens revealed that JAM‐A was aberrantly expressed in neoplastic regions including adenocarcinoma in situ (AIS). Knockout of JAM‐A significantly suppressed cell proliferation and colony‐forming and migration abilities. We also showed that an antibody specific to an extracellular region of JAM‐A reduced cell proliferation ability and that loss of JAM‐A increased drug sensitivity of cervical adenocarcinoma cells. Based on a comprehensive proteome analysis, we found that poliovirus receptor (PVR/CD155) was regulated by JAM‐A and formed a physical interaction with JAM‐A. In human surgical specimens, PVR/CD155 expression was significantly correlated with some clinicopathological features and prognosis of cervical adenocarcinoma. Interestingly, most of the PVR/CD155‐positive cases expressed a high level of JAM‐A, and patients with the expression pattern of PVR/CD155 positive/JAM‐A high had significantly shorter periods of relapse‐free survival (P = 0.00964) and overall survival (P = 0.0204) than those for the other patients. Our observations suggest that aberrant expression of JAM‐A promotes malignancy of uterine cervical adenocarcinoma via regulation of PVR/CD155, and JAM‐A is therefore a potential therapeutic target for this malignancy.

View on the web

Emerging role of AMPA receptor subunit GluA1 in synaptic plasticity: Implications for Alzheimer's disease

Alexandros G.Sfakianakis shared this article with you from Inoreader
Emerging role of AMPA receptor subunit GluA1 in synaptic plasticity: Implications for Alzheimer's disease

GluA1 mediated synaptic plasticity plays a central role in the development of AD. Amyloid beta/Tau protein has been shown to impair GluA1 and cause synaptic dysfunction and acute neurotoxicity, respectively


Abstract

It is well established that GluA1 mediated synaptic plasticity plays a central role in the early development of AD. The complex cellular and molecular mechanisms that enable GluA1‐related synaptic regulation remain to fully understood. Particularly, understanding the mechanisms that disrupt GluA1 related synaptic plasticity is central to the development of disease‐modifying therapies which are sorely needed as the incidence of AD rises. We surmise that the published evidence establishes deficits in synaptic plasticity as a central factor of AD aetiology. We additionally highlight potential therapeutic strategies for the treatment of AD, and we delve into the roles of GluA1 in learning and memory. Particularly, we review the current understanding of the molecular interactions that confer the actions of this ubiquitous excitatory receptor subunit including post‐translational modification and accessory protein recruitment of the GluA1 subunit. These are proposed to regulate rec eptor trafficking, recycling, channel conductance and synaptic transmission and plasticity.

View on the web

Co‐option of PPARα in the regulation of lipogenesis and fatty acid oxidation in CLA‐induced hepatic steatosis

Alexandros G.Sfakianakis shared this article with you from Inoreader
Co‐option of PPARα in the regulation of lipogenesis and fatty acid oxidation in CLA‐induced hepatic steatosis

1.

Hepatic PPARα is responsible for dietary CLA‐induced NAFLD and insulin resistance in mice.

2.

PPARα activation co‐opts lipogenesis and FA oxidation gene programs in hepatic steatosis. However, PPARα inhibition blocks the enrichment of active histone marks H3K27ac and H3K4me1 and cofactors on the target locus.

3.

GW6471 exploits PPARα as a therapeutic target by reprograming genes key to lipid homeostasis (e.g., Acaca and Acads), and confers protection against NAFLD.


Abstract

Nonalcoholic‐fatty‐liver‐disease (NAFLD) is the result of imbalances in hepatic lipid partitioning and is linked to dietary factors. We demonstrate that conjugated linoleic acid (CLA) when given to mice as a dietary supplement, induced an enlarged liver, hepatic steatosis, and increased plasma levels of fatty acid (FA), alanine transaminase, and triglycerides. The progression of NAFLD and insulin resistance was reversed by GW6471 a small‐molecule antagonist of peroxisome proliferator‐activated receptor α (PPARα). Transcriptional profiling of livers revealed that the genes involved in FA oxidation and lipogenesis as two core gene programs controlled by PPARα in response to CLA and GW6471 including Acaca and Acads. Bioinformatic analysis of PPARα ChIP‐seq data set and ChIP‐qPCR showed that GW6471 blocks PPARα binding to Acaca and Acads and abolishes the PPARα‐mediated local histone modifications of H3K27ac and H3K4me1 in CL A‐treated hepatocytes. Thus, our findings reveal a dual role of PPARα in the regulation of lipid homeostasis and highlight its druggable nature in NAFLD.

View on the web

Reduced neuropathy target esterase in pre‐eclampsia suppresses tube formation of HUVECs via dysregulation of phospholipid metabolism

Alexandros G.Sfakianakis shared this article with you from Inoreader

Abstract

Recently, studies have shown that neuropathy target esterase (NTE) is essential to placental and normal blood vessel development. However, whether it is involved in abnormal placenta angiogenesis of pre‐eclampsia remains unknown. Thus, our aim was to observe the expression of NTE in pre‐eclamptic placentas and its effects and mechanism of NTE on the migration and the tube formation of human umbilical vein endothelial cells (HUVECs). Immunohistochemical staining showed that the NTE protein was intensely located in blood vessels of the normal pregnant placenta. However, western blot revealed that the expression level of NTE protein was significantly reduced in pre‐eclamptic placenta. The results indicated that overexpression of NTE significantly promoted the migration and the tube formation of HUVECs compared with those of the control and scramble short hairpin RNA (shRNA) group. Conversely, NTE shRNA obviously inhibited the migration and the tube formation of HUVECs. Addition ally, chromatography assay evidenced that NTE overexpression significantly reduced the level of phosphatidylcholine (PC) of HUVECs, but NTE shRNA obviously increased the level of PC of HUVECs. Furthermore, exogenous PC and lysophosphatidylcholine (LPC) significantly inhibited the tube formation of HUVECs in a dose‐dependent manner. Collectively, our results suggest that reduced NTE in placenta may contribute to abnormal placenta angiogenesis of pre‐eclampsia via the dysregulation of PC and LPC metabolism.

View on the web

DNA polymerase eta: A potential pharmacological target for cancer therapy

Alexandros G.Sfakianakis shared this article with you from Inoreader
DNA polymerase eta: A potential pharmacological target for cancer therapy

Cancer cells utilize multiple lines of self‐defense mechanisms to bypass chemo and radiotherapy. One such mechanism employed by cancer cells is translesion DNA synthesis (TLS), in which specialized TLS polymerases bypass the DNA lesion with the help of monoubiquitinated proliferating cell nuclear antigen. Among all TLS polymerases (Pol η, Pol ι, Pol κ, REV1, Pol ζ, Pol μ, Pol λ, Pol ν, and Pol θ), DNA polymerase eta (Pol η) is well studied and majorly responsible for the bypass of cisplatin and UV‐induced DNA damage.

Please use Figure 4 for graphical abstract from revised manuscipt or from attachment. We have done minor changes in figure 4 during revision.


Abstract

In the last two decades, intensive research has been carried out to improve the survival rates of cancer patients. However, the development of chemoresistance that ultimately leads to tumor relapse poses a critical challenge for the successful treatment of cancer patients. Many cancer patients experience tumor relapse and ultimately die because of treatment failure associated with acquired drug resistance. Cancer cells utilize multiple lines of self‐defense mechanisms to bypass chemotherapy and radiotherapy. One such mechanism employed by cancer cells is translesion DNA synthesis (TLS), in which specialized TLS polymerases bypass the DNA lesion with the help of monoubiquitinated proliferating cell nuclear antigen. Among all TLS polymerases (Pol η, Pol ι, Pol κ, REV1, Pol ζ, Pol μ, Pol λ, Pol ν, and Pol θ), DNA polymerase eta (Pol η) is well studied and majorly responsible for the bypass of cisplatin and UV‐induced DNA damage. TLS polymerases contribute to chemotherape utic drug‐induced mutations as well as therapy resistance. Therefore, targeting these polymerases presents a novel therapeutic strategy to combat chemoresistance. Mounting evidence suggests that inhibition of Pol η may have multiple impacts on cancer therapy such as sensitizing cancer cells to chemotherapeutics, suppressing drug‐induced mutagenesis, and inhibiting the development of secondary tumors. Herein, we provide a general introduction of Pol η and its clinical implications in blocking acquired drug resistance. In addition; this review addresses the existing gaps and challenges of Pol η mediated TLS mechanisms in human cells. A better understanding of the Pol η mediated TLS mechanism will not merely establish it as a potential pharmacological target but also open possibilities to identify novel drug targets for future therapy.

View on the web

Morphine promotes tumorigenesis and cetuximab resistance via EGFR signaling activation in human colorectal cancer

Alexandros G.Sfakianakis shared this article with you from Inoreader
Morphine promotes tumorigenesis and cetuximab resistance via EGFR signaling activation in human colorectal cancer

Our results suggest that morphine can promote cancer progression and resistance to the EGFR targeted drug cetuximab in colorectal cancer (CRC). Furthermore, the morphine‐induced effect was antagonized by adding nalmefene or mu‐opioid receptor (MOR) knockdown, suggesting a MOR‐mediated mechanism.


Abstract

Morphine, a mu‐opioid receptor (MOR) agonist, has been extensively used to treat advanced cancer pain. In particular, in patients with cancer metastasis, both morphine and anticancer drugs are given simultaneously. However, evidence showed that morphine might be a risk factor in promoting the tumor's malignant potential. In this study, we report that treatment with morphine could activate MOR and lead to the promotion of proliferation, migration, and invasion in HCT116 and DLD1 colorectal cancer (CRC) cells with time‐concentration dependence. Moreover, morphine can also contribute to cetuximab's drug resistance, a targeted drug widely used to treat advanced CRC by inducing the activation of epidermal growth factor receptor (EGFR). The cell phenotype includes proliferation, migration, invasion, and drug resistance, which may be reversed by MOR knockdown or adding nalmefene, the MOR receptor antagonist. Receptor tyrosine kinase array analysis revealed that morphine selectiv ely induced the transactivation of EGFR. EGFR transactivation resulted in the activation of ERK1/2 and AKT. In conclusion, morphine induces the transactivation of EGFR via MOR. It activates the downstream signal pathway AKT‐MTOR and RAS‐MAPK, increases proliferation, migration, and invasion, and promotes resistance to EGFR inhibitors in a CRC cell line. Furthermore, we verified that EGFR inhibition by cetuximab strongly reversed the protumoral effects of morphine in vitro and in vivo. Collectively, we provide evidence that morphine‐EGFR signaling might be a promising therapeutic target for CRC patients, especially for cetuximab‐resistant CRC patients.

View on the web

Interleukin‐33 alleviates diabetic cardiomyopathy through regulation of endoplasmic reticulum stress and autophagy via insulin‐like growth factor‐binding protein 3

Alexandros G.Sfakianakis shared this article with you from Inoreader
Interleukin‐33 alleviates diabetic cardiomyopathy through regulation of endoplasmic reticulum stress and autophagy via insulin‐like growth factor‐binding protein 3

Interleukin‐33 (IL‐33) abates diabetic cardiomyopathy (DCM) by alleviating endoplasmic reticulum (ER) stress and promoting autophagy. Insulin‐like growth factor‐binding protein 3 is essential for IL‐33‐induced ER stress resolution and autophagic enhancement during DCM.


Abstract

Prolonged endoplasmic reticulum (ER) stress is the key driving force behind diabetic cardiomyopathy (DCM). Autophagy is extensively implicated in adaptive mechanisms for cell survival. Interleukin‐33 (IL‐33) is known to be a potent cardiac protector, but its roles in DCM, ER stress, and autophagy are currently unknown. We aimed to explore the effects of IL‐33 on DCM and characterize the roles that ER stress and autophagy play in DCM. The effects of IL‐33 on DCM, ER stress, and autophagy were characterized both in db/db mice and in palmitic acid (PA)‐treated cardiomyocytes. The manipulators of ER stress and autophagy were used to clarify their roles in DCM remittance conferred by IL‐33. Gene expression analysis was used to identify IL‐33‐dependent regulators of ER stress and autophagy. Both db/db mice and PA‐treated cells presented with enhanced levels of ER stress, apoptosis, and lipid deposition, as well as impaired autophagy, all of which could be reversed by I L‐33. Treatment with IL‐33 improved the cardiac diastolic function of diabetic mice. Nonselective autophagy inhibitors, such as 3‐methyladenine (3‐MA) or wortmannin, abolished the protective effects of IL‐33, resulting in an increase in both ER stress and apoptosis. Strikingly, insulin‐like growth factor‐binding protein 3 (IGFBP3) was identified as the gene most significantly differentially expressed between IL‐33 and control groups. Knockdown of IGFBP3 expression, similar to the effect of nonselective autophagy inhibitors, resulted in high levels of ER stress, impaired autophagy, and apoptosis that were not rescued upon treatment with IL‐33. IL‐33 abates DCM by alleviating ER stress and promoting autophagy. IGFBP3 is essential for IL‐33‐induced ER stress resolution and autophagic enhancement during DCM.

View on the web

PBK promotes aggressive phenotypes of cervical cancer through ERK/c‐Myc signaling pathway

Alexandros G.Sfakianakis shared this article with you from Inoreader
PBK promotes aggressive phenotypes of cervical cancer through ERK/c‐Myc signaling pathway

PDZ‐binding kinase (PBK) promoted the malignant progression of cervical cancer through ERK/c‐Myc signal pathway. PBK might be a promising molecular target for cervical cancer treatment.


Abstract

Cervical cancer is the fourth most frequent cancer in women worldwide. PDZ‐binding kinase (PBK) is proven to promote the malignant behaviors of various carcinomas. However, its functional roles and oncogenic mechanisms in cervical cancer are poorly understood. In this study, we reported that PBK was highly expressed in cervical cancer tissues. PBK promoted the proliferation, metastasis, and cisplatin resistance of cervical cancer cells. OTS514, a specific PBK inhibitor, could significantly suppress proliferation and metastasis of cervical cancer cells in vitro and in a xenograft model. Besides, OTS514 could enhance cisplatin‐based chemosensitivity in cervical cancer cells. Mechanistically, PBK promoted the expression and stabilization of c‐Myc through phosphorylating ERK1/2. OTS514 suppressed the phosphorylation of ERK1/2 and the transcriptional activity of c‐Myc. Furthermore, inhibition of the ERK signal pathway by U0126 reversed the increased proliferation and metastasis induced by overexpression of PBK. Exogenous expression of c‐Myc counteracted the decreased proliferation and metastasis evoked by knockdown of PBK. In conclusion, PBK promoted the malignant progression of cervical cancer through ERK/c‐Myc signal pathway. PBK might be a promising molecular target for cervical cancer treatment.

View on the web

Αναζήτηση αυτού του ιστολογίου

! # Ola via Alexandros G.Sfakianakis on Inoreader