1
|
Corazzari M, Collavin L. Wild-type and mutant p53 in cancer-related ferroptosis. A matter of stress management? Front Genet 2023; 14:1148192. [PMID: 37021009 PMCID: PMC10067580 DOI: 10.3389/fgene.2023.1148192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/09/2023] [Indexed: 03/22/2023] Open
Abstract
Cancer cells within tumor masses are chronically exposed to stress caused by nutrient deprivation, oxygen limitation, and high metabolic demand. They also accumulate hundreds of mutations, potentially generating aberrant proteins that can induce proteotoxic stress. Finally, cancer cells are exposed to various damages during chemotherapy. In a growing tumor, transformed cells eventually adapt to these conditions, eluding the death-inducing outcomes of signaling cascades triggered by chronic stress. One such extreme outcome is ferroptosis, a form of iron-dependent non-apoptotic cell death mediated by lipid peroxidation. Not surprisingly, the tumor suppressor p53 is involved in this process, with evidence suggesting that it acts as a pro-ferroptotic factor and that its ferroptosis-inducing activity may be relevant for tumor suppression. Missense alterations of the TP53 gene are extremely frequent in human cancers and give rise to mutant p53 proteins (mutp53) that lose tumor suppressive function and can acquire powerful oncogenic activities. This suggests that p53 mutation provides a selective advantage during tumor progression, raising interesting questions on the impact of p53 mutant proteins in modulating the ferroptotic process. Here, we explore the role of p53 and its cancer-related mutants in ferroptosis, using a perspective centered on the resistance/sensitivity of cancer cells to exogenous and endogenous stress conditions that can trigger ferroptotic cell death. We speculate that an accurate molecular understanding of this particular axis may improve cancer treatment options.
Collapse
Affiliation(s)
- Marco Corazzari
- Department of Health Sciences and Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Licio Collavin
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
2
|
Zhu P, Li T, Li Q, Gu Y, Shu Y, Hu K, Chen L, Peng X, Peng J, Hao L. Mechanism and Role of Endoplasmic Reticulum Stress in Osteosarcoma. Biomolecules 2022; 12:1882. [PMID: 36551309 PMCID: PMC9775044 DOI: 10.3390/biom12121882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor, often occurring in children and adolescents. The etiology of most patients is unclear, and the current conventional treatment methods are chemotherapy, radiotherapy, and surgical resection. However, the sensitivity of osteosarcoma to radiotherapy and chemotherapy is low, and the prognosis is poor. The development of new and useful treatment strategies for improving patient survival is an urgent need. It has been found that endoplasmic reticulum (ER) stress (ERS) affects tumor angiogenesis, invasion, etc. By summarizing the literature related to osteosarcoma and ERS, we found that the unfolded protein response (UPR) pathway activated by ERS has a regulatory role in osteosarcoma proliferation, apoptosis, and chemoresistance. In osteosarcoma, the UPR pathway plays an important role by crosstalk with autophagy, oxidative stress, and other pathways. Overall, this article focuses on the relationship between ERS and osteosarcoma and reviews the potential of drugs or gene targets associated with ERS for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Peijun Zhu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Ting Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Qingqing Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yawen Gu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yuan Shu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Kaibo Hu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaogang Peng
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jie Peng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
3
|
The Gut-Ex-Vivo System (GEVS) Is a Dynamic and Versatile Tool for the Study of DNBS-Induced IBD in BALB/C and C57BL/6 Mice, Highlighting the Protective Role of Probiotics. BIOLOGY 2022; 11:biology11111574. [PMID: 36358275 PMCID: PMC9687175 DOI: 10.3390/biology11111574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND IBD is a spectrum of pathologies characterized by dysregulated immune activation leading to uncontrolled response against the intestine, thus resulting in chronic gut inflammation and tissue damage. Due to its complexity, the molecular mechanisms responsible for disease onset and progression are still elusive, thus requiring intense research effort. In this context, the development of models replicating the etiopathology of IBD and allowing the testing of new potential therapies is critical. METHODS Colon from C57BL/6 or BALB/c mice was cultivated in a Gut-Ex-Vivo System (GEVS), exposed for 5 h to DNBS 1.5 or 2.5 mg/mL, in presence or absence of two probiotic formulations (P1 = Bifidobacterium breve BR03 (DSM16604) and B632 (DSM24706); P2 = Lacticaseibacillus rhamnosus LR04 (DSM16605), Lactiplantibacillus plantarum LP14 (DSM33401) and Lacticaseibacillus paracasei LPC09), and the main hallmarks of IBD were evaluated. RESULTS Gene expression analysis revealed the following DNBS-induced effects: (i) compromised tight junction organization, responsible for tissue permeability dysregulation; (ii) induction of ER stress, and (iii) tissue inflammation in colon of C57BL/6 mice. Moreover, the concomitant DNBS-induced apoptosis and ferroptosis pathways were evident in colon from both BALB/c and C57BL/6 mice. Finally, the co-administration of probiotics completely prevented the detrimental effects of DNBS. CONCLUSIONS Overall, we have provided results demonstrating that GEVS is a consistent, reliable, and cost-effective system for modeling DNBS-induced IBD, useful for studying the onset and progression of human disease at the molecular level, while also reducing animal suffering. Moreover, we have confirmed the beneficial effect of probiotics administration in promoting the remission of IBD.
Collapse
|
4
|
Integrative Analysis Reveals the Potential Role and Prognostic Value of GOLM1 in Hepatocellular Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8284500. [PMID: 36211823 PMCID: PMC9535134 DOI: 10.1155/2022/8284500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022]
Abstract
Overexpression of Golgi membrane protein 1 (GOLM1) is closely associated with hepatocellular carcinoma (HCC) vascular invasion. How GOLM1 may be involved in angiogenesis in HCC remains unclear. We explored how GOLM1 promotes angiogenesis in HCC and potential prognostic value. Expression levels of GOLM1 in HCC patients and healthy controls were obtained from The Cancer Genome Atlas (TCGA). Differentially expressed genes (DEGs) between HCC patients and controls were compared. GOLM1 was knocked out in the HCC cell line, and RNA sequencing and DEG expression analysis were performed compared with control cells. Based on TCGA data and cell line RNA sequencing data, DEGs affected by a high expression of GOLM1 were identified. Subsequently, enrichment analysis was performed to explore the functions and pathways of the DEGs affected by a high expression of GOLM1. A relevant network analysis was built. Cox regression, genomic variance analysis scores, minimum absolute shrinkage and selection operator regression, and random forest regression models were applied to determine the best prognostic model and validated using the GSE54236 dataset from the Gene Expression Omnibus (GEO). We determined the effect of GOLM1 expression on immune cell infiltration in liver cancer. GOLM1 was overexpressed in HCC tissues compared with controls, and its level correlated with tumor purity and prognosis. 400 DEGs affected by highly expressed GOLM1 were identified in TCGA and cell line RNA sequencing data. Enrichment analysis revealed that these DEGs may be related to biological processes of oxidative stress and angiogenesis and involved in the VEGF signaling pathway and protein processing in endoplasmic reticulum. We predicted a comprehensive regulatory network in which GOLM1 activated VEGF signaling to promote HCC angiogenesis. GOLM1 may interact with E2F1 and IGF2BP3 to promote angiogenesis. GOLM1 overexpression was associated with greater immune cell infiltration. A random forest regression model was the best prognostic model. Our study reveals a potential molecular mechanism of GOLM1 in promoting HCC. We developed two prognostic models based on DEG associated with GOLM1 overexpression to help stratify HCC prognosis and improve individualized treatment.
Collapse
|
5
|
Kuang Y, Ye N, Kyani A, Ljungman M, Paulsen M, Chen H, Zhou M, Wild C, Chen H, Zhou J, Neamati N. Induction of Genes Implicated in Stress Response and Autophagy by a Novel Quinolin-8-yl-nicotinamide QN523 in Pancreatic Cancer. J Med Chem 2022; 65:6133-6156. [PMID: 35439009 PMCID: PMC9195374 DOI: 10.1021/acs.jmedchem.1c02207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Using a cytotoxicity-based phenotypic screen of a highly diverse library of 20,000 small-molecule compounds, we identified a quinolin-8-yl-nicotinamide, QN519, as a promising lead. QN519 represents a novel scaffold with drug-like properties, showing potent in vitro cytotoxicity in a panel of 12 cancer cell lines. Subsequently, lead optimization campaign generated compounds with IC50 values < 1 μM. An optimized compound, QN523, shows significant in vivo efficacy in a pancreatic cancer xenograft model. QN523 treatment significantly increased the expression of HSPA5, DDIT3, TRIB3, and ATF3 genes, suggesting activation of the stress response pathway. We also observed a significant increase in the expression of WIPI1, HERPUD1, GABARAPL1, and MAP1LC3B, implicating autophagy as a major mechanism of action. Due to the lack of effective treatments for pancreatic cancer, discovery of novel agents such as the QN series of compounds with unique mechanism of action has the potential to fulfill a clear unmet medical need.
Collapse
Affiliation(s)
- Yuting Kuang
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Armita Kyani
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mats Ljungman
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michelle Paulsen
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Haijun Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Mingxiang Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Christopher Wild
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, TX 77550, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
Gebert M, Sobolewska A, Bartoszewska S, Cabaj A, Crossman DK, Króliczewski J, Madanecki P, Dąbrowski M, Collawn JF, Bartoszewski R. Genome-wide mRNA profiling identifies X-box-binding protein 1 (XBP1) as an IRE1 and PUMA repressor. Cell Mol Life Sci 2021; 78:7061-7080. [PMID: 34636989 PMCID: PMC8558229 DOI: 10.1007/s00018-021-03952-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/17/2021] [Accepted: 09/28/2021] [Indexed: 02/06/2023]
Abstract
Accumulation of misfolded proteins in ER activates the unfolded protein response (UPR), a multifunctional signaling pathway that is important for cell survival. The UPR is regulated by three ER transmembrane sensors, one of which is inositol-requiring protein 1 (IRE1). IRE1 activates a transcription factor, X-box-binding protein 1 (XBP1), by removing a 26-base intron from XBP1 mRNA that generates spliced XBP1 mRNA (XBP1s). To search for XBP1 transcriptional targets, we utilized an XBP1s-inducible human cell line to limit XBP1 expression in a controlled manner. We also verified the identified XBP1-dependent genes with specific silencing of this transcription factor during pharmacological ER stress induction with both an N-linked glycosylation inhibitor (tunicamycin) and a non-competitive inhibitor of the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) (thapsigargin). We then compared those results to the XBP1s-induced cell line without pharmacological ER stress induction. Using next‐generation sequencing followed by bioinformatic analysis of XBP1-binding motifs, we defined an XBP1 regulatory network and identified XBP1 as a repressor of PUMA (a proapoptotic gene) and IRE1 mRNA expression during the UPR. Our results indicate impairing IRE1 activity during ER stress conditions accelerates cell death in ER-stressed cells, whereas elevating XBP1 expression during ER stress using an inducible cell line correlated with a clear prosurvival effect and reduced PUMA protein expression. Although further studies will be required to test the underlying molecular mechanisms involved in the relationship between these genes with XBP1, these studies identify a novel repressive role of XBP1 during the UPR.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Aleksandra Sobolewska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Aleksandra Cabaj
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - David K Crossman
- Department of Genetics, Heflin Center for Genomic Science, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jarosław Króliczewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Piotr Madanecki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland
| | - Michał Dąbrowski
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107, 80-416, Gdansk, Poland.
| |
Collapse
|
7
|
A Gut-Ex-Vivo System to Study Gut Inflammation Associated to Inflammatory Bowel Disease (IBD). BIOLOGY 2021; 10:biology10070605. [PMID: 34209277 PMCID: PMC8301106 DOI: 10.3390/biology10070605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022]
Abstract
Simple Summary Inflammatory Bowel Disease (IBD) is a complex and multifactorial systemic disease of the gastrointestinal tract, characterized by chronic inflammation, thus resulting in tissue damage and, occasionally, in cancer development. Although the precise origin is still elusive, it is widely considered a disease of modern society, caused by a complex interaction between environment, genetic, immune system, and gut microflora (microbiota). Potentially affected by all the above-mentioned variables, which interplay are highly heterogeneous, the disease appears to be patient-specific. The latter phenomenon, together with the uncertain origin, also contributes to the lack of optimal clinical treatment of these patients. Therefore, the development of appropriate models is crucial to push the research forward and to define new valuable therapeutic approaches. Although tissue biopsies and/or animal models represent the best models to study IBD onset, progression, and clinical interventions, they are both affected by limitations such as invasiveness, cost- and time-consuming, and ethical issues such as animal suffering. Here we propose a novel approach based on the cultivation of mouse tissues (colon) in an ex vivo microfluidic device (Gut-Ex-Vivo System, GEVS) to study IBD. We demonstrate that explanted mouse tissues cultivated in our GEVS can be appropriately stimulated to recapitulate the onset of the disease, in a time- and cost- effective manner. Abstract Inflammatory bowel disease (IBD) is a complex, chronic, and dysregulated inflammatory condition which etiology is still largely unknown. Its prognosis and disease progression are highly variable and unpredictable. IBD comprises several heterogeneous inflammatory conditions ranging from Ulcerative Colitis (UC) to Crohn’s Disease (CD). Importantly, a definite, well-established, and effective clinical treatment for these pathologies is still lacking. The urgent need for treatment is further supported by the notion that patients affected by UC or CD are also at risk of developing cancer. Therefore, a deeper understanding of the molecular mechanisms at the basis of IBD development and progression is strictly required to design new and efficient therapeutic regimens. Although the development of animal models has undoubtedly facilitated the study of IBD, such in vivo approaches are often expensive and time-consuming. Here we propose an organ ex vivo culture (Gut-Ex-Vivo system, GEVS) based on colon from Balb/c mice cultivated in a dynamic condition, able to model the biochemical and morphological features of the mouse models exposed to DNBS (5–12 days), in 5 h. Indeed, upon DNBS exposure, we observed a dose-dependent: (i) up-regulation of the stress-related protein transglutaminase 2 (TG2); (ii) increased intestinal permeability associated with deregulated tight junction protein expression; (iii) increased expression of pro-inflammatory cytokines, such as TNFα, IFNγ, IL1β, IL6, IL17A, and IL15; (iv) down-regulation of the anti-inflammatory IL10; and (v) induction of Endoplasmic Reticulum stress (ER stress), all markers of IBD. Altogether, these data indicate that the proposed model can be efficiently used to study the pathogenesis of IBD, in a time- and cost-effective manner.
Collapse
|
8
|
Reprogramming of microRNA expression via E2F1 downregulation promotes Salmonella infection both in infected and bystander cells. Nat Commun 2021; 12:3392. [PMID: 34099666 PMCID: PMC8184997 DOI: 10.1038/s41467-021-23593-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Cells infected with pathogens can contribute to clearing infections by releasing signals that instruct neighbouring cells to mount a pro-inflammatory cytokine response, or by other mechanisms that reduce bystander cells’ susceptibility to infection. Here, we show the opposite effect: epithelial cells infected with Salmonella Typhimurium secrete host factors that facilitate the infection of bystander cells. We find that the endoplasmic reticulum stress response is activated in both infected and bystander cells, and this leads to activation of JNK pathway, downregulation of transcription factor E2F1, and consequent reprogramming of microRNA expression in a time-dependent manner. These changes are not elicited by infection with other bacterial pathogens, such as Shigella flexneri or Listeria monocytogenes. Remarkably, the protein HMGB1 present in the secretome of Salmonella-infected cells is responsible for the activation of the IRE1 branch of the endoplasmic reticulum stress response in non-infected, neighbouring cells. Furthermore, E2F1 downregulation and the associated microRNA alterations promote Salmonella replication within infected cells and prime bystander cells for more efficient infection. Cells infected with pathogens can release signals that instruct neighbouring cells to mount an immune response or that reduce these cells’ susceptibility to infection. Here, Aguilar et al. show the opposite effect: cells infected with Salmonella Typhimurium secrete host factors that facilitate the infection of bystander cells by activating their ER-stress response.
Collapse
|
9
|
Activation of the Integrated Stress Response and ER Stress Protect from Fluorizoline-Induced Apoptosis in HEK293T and U2OS Cell Lines. Int J Mol Sci 2021; 22:ijms22116117. [PMID: 34204139 PMCID: PMC8201103 DOI: 10.3390/ijms22116117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The prohibitin (PHB)-binding compound fluorizoline as well as PHB-downregulation activate the integrated stress response (ISR) in HEK293T and U2OS human cell lines. This activation is denoted by phosphorylation of eIF2α and increases in ATF4, ATF3, and CHOP protein levels. The blockage of the activation of the ISR by overexpression of GRP78, as well as an increase in IRE1 activity, indicate the presence of ER stress after fluorizoline treatment. The inhibition of the ER stress response in HEK293T and U2OS led to increased sensitivity to fluorizoline-induced apoptosis, indicating a pro-survival role of this pathway after fluorizoline treatment in these cell lines. Fluorizoline induced an increase in calcium concentration in the cytosol and the mitochondria. Finally, two different calcium chelators reduced fluorizoline-induced apoptosis in U2OS cells. Thus, we have found that fluorizoline causes increased ER stress and activation of the integrated stress response, which in HEK293T and U2OS cells are protective against fluorizoline-induced apoptosis.
Collapse
|
10
|
Li M. The role of P53 up-regulated modulator of apoptosis (PUMA) in ovarian development, cardiovascular and neurodegenerative diseases. Apoptosis 2021; 26:235-247. [PMID: 33783663 PMCID: PMC8197724 DOI: 10.1007/s10495-021-01667-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2021] [Indexed: 12/14/2022]
Abstract
P53 up-regulated modulator of apoptosis (PUMA), a pro-apoptotic BCL-2 homology 3 (BH3)-only member of the BCL-2 family, is a direct transcriptional target of P53 that elicits mitochondrial apoptosis under treatment with radiation and chemotherapy. It also induces excessive apoptosis in cardiovascular and/or neurodegenerative diseases. PUMA has been found to play a critical role in ovarian apoptosis. In the present paper, we review the progress of the study in PUMA over the past two decades in terms of its inducement and/or amplification of programmed cell death and describe recent updates to the understanding of both P53-dependent and P53-independent PUMA-mediated apoptotic pathways that are implicated in physiology and pathology, including the development of the ovary and cardiovascular and neurodegenerative diseases. We propose that PUMA may be a key regulator during ovary development, provide a model for PUMA-mediated apoptotic pathways, including intrinsic and extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Mei Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
| |
Collapse
|
11
|
Ciccosanti F, Corazzari M, Casetti R, Amendola A, Collalto D, Refolo G, Vergori A, Taibi C, D’Offizi G, Antinori A, Agrati C, Fimia GM, Ippolito G, Piacentini M, Nardacci R. High Levels of TRIM5α Are Associated with Xenophagy in HIV-1-Infected Long-Term Nonprogressors. Cells 2021; 10:cells10051207. [PMID: 34069225 PMCID: PMC8156091 DOI: 10.3390/cells10051207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/07/2021] [Accepted: 05/13/2021] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a lysosomal-dependent degradative mechanism essential in maintaining cellular homeostasis, but it is also considered an ancient form of innate eukaryotic fighting against invading microorganisms. Mounting evidence has shown that HIV-1 is a critical target of autophagy that plays a role in HIV-1 replication and disease progression. In a special subset of HIV-1-infected patients that spontaneously and durably maintain extremely low viral replication, namely, long-term nonprogressors (LTNP), the resistance to HIV-1-induced pathogenesis is accompanied, in vivo, by a significant increase in the autophagic activity in peripheral blood mononuclear cells. Recently, a new player in the battle of autophagy against HIV-1 has been identified, namely, tripartite motif protein 5α (TRIM5α). In vitro data demonstrated that TRIM5α directly recognizes HIV-1 and targets it for autophagic destruction, thus protecting cells against HIV-1 infection. In this paper, we analyzed the involvement of this factor in the control of HIV-1 infection through autophagy, in vivo, in LTNP. The results obtained showed significantly higher levels of TRIM5α expression in cells from LTNP with respect to HIV-1-infected normal progressor patients. Interestingly, the colocalization of TRIM5α and HIV-1 proteins in autophagic vacuoles in LTNP cells suggested the participation of TRIM5α in the autophagy containment of HIV-1 in LTNP. Altogether, our results point to a protective role of TRIM5α in the successful control of the chronic viral infection in HIV-1-controllers through the autophagy mechanism. In our opinion, these findings could be relevant in fighting against HIV-1 disease, because autophagy inducers might be employed in combination with antiretroviral drugs.
Collapse
Affiliation(s)
- Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Marco Corazzari
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Rita Casetti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Alessandra Amendola
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Diletta Collalto
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Giulia Refolo
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Alessandra Vergori
- Clinical Department, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (A.V.); (C.T.); (G.D.); (A.A.)
| | - Chiara Taibi
- Clinical Department, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (A.V.); (C.T.); (G.D.); (A.A.)
| | - Gianpiero D’Offizi
- Clinical Department, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (A.V.); (C.T.); (G.D.); (A.A.)
| | - Andrea Antinori
- Clinical Department, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (A.V.); (C.T.); (G.D.); (A.A.)
| | - Chiara Agrati
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppe Ippolito
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
- Department of Biology, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Roberta Nardacci
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases, Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; (F.C.); (M.C.); (R.C.); (A.A.); (D.C.); (G.R.); (C.A.); (G.M.F.); (G.I.); (M.P.)
- Correspondence:
| |
Collapse
|
12
|
Ferrari E, Monzani R, Saverio V, Gagliardi M, Pańczyszyn E, Raia V, Villella VR, Bona G, Pane M, Amoruso A, Corazzari M. Probiotics Supplements Reduce ER Stress and Gut Inflammation Associated with Gliadin Intake in a Mouse Model of Gluten Sensitivity. Nutrients 2021; 13:1221. [PMID: 33917155 PMCID: PMC8067866 DOI: 10.3390/nu13041221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/20/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
Exposure to gluten, a protein present in wheat rye and barley, is the major inducer for human Celiac Disease (CD), a chronic autoimmune enteropathy. CD occurs in about 1% worldwide population, in genetically predisposed individuals bearing human leukocyte antigen (HLA) DQ2/DQ8. Gut epithelial cell stress and the innate immune activation are responsible for the breaking oral tolerance to gliadin, a gluten component. To date, the only treatment available for CD is a long-term gluten-free diet. Several studies have shown that an altered composition of the intestinal microbiota (dysbiosis) could play a key role in the pathogenesis of CD through the modulation of intestinal permeability and the regulation of the immune system. Here, we show that gliadin induces a chronic endoplasmic reticulum (ER) stress condition in the small intestine of a gluten-sensitive mouse model and that the coadministration of probiotics efficiently attenuates both the unfolded protein response (UPR) and gut inflammation. Moreover, the composition of probiotics formulations might differ in their activity at molecular level, especially toward the three axes of the UPR. Therefore, probiotics administration might potentially represent a new valuable strategy to treat gluten-sensitive patients, such as those affected by CD.
Collapse
Affiliation(s)
- Eleonora Ferrari
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Romina Monzani
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Valentina Saverio
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Mara Gagliardi
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Elżbieta Pańczyszyn
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Valeria Raia
- Regional Cystic Fibrosis Center, Pediatric Unit, Department of Translational Medical Sciences, Federico II University Naples, 80134 Naples, Italy;
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Valeria Rachela Villella
- European Institute for Research in Cystic Fibrosis (IERFC-Onlus), San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Gianni Bona
- Division of Pediatrics, Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Marco Pane
- Probiotical Research Srl, 28100 Novara, Italy; (M.P.); (A.A.)
| | - Angela Amoruso
- Probiotical Research Srl, 28100 Novara, Italy; (M.P.); (A.A.)
| | - Marco Corazzari
- Department of Health Science, University of Piemonte Orientale, 28100 Novara, Italy; (E.F.); (R.M.); (V.S.); (M.G.); (E.P.)
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
13
|
Guo Y, Fan W, Cao S, Xie Y, Hong J, Zhou H, Wan H, Jin B. 2,3,5,4'-Tetrahydroxystilbene-2-O-β-D-Glucoside modulated human umbilical vein endothelial cells injury under oxidative stress. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:473-479. [PMID: 33093269 PMCID: PMC7585593 DOI: 10.4196/kjpp.2020.24.6.473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022]
Abstract
Endothelial cell injury is a major contributor to cardiovascular diseases. The 2,3,5,4’-Tetrahydroxystilbene-2-O-β-D-Glucoside (TSG) contributes to alleviate human umbilical vein endothelial cells (HUVECs) injury through mechanisms still know a little. This study aims to clarify the TSG effects on gene expression (mRNA and microRNA) related to oxidative stress and endoplasmic reticulum stress induced by H2O2 in HUVECs. We found that TSG significantly reduced the death rate of cells and increased intracellular superoxide dismutase activity. At qRT-PCR, experimental data showed that TSG significantly counteracted the expressions of miR-9-5p, miR-16, miR-21, miR-29b, miR-145-5p, and miR-204-5p. Besides, TSG prevented the expression of ATF6 and CHOP increasing. In contrast, TSG promoted the expression of E2F1. In conclusion, our results point to the obvious protective effect of TSG on HUVECs injury induced by H2O2, and the mechanism may through miR16/ATF6/ E2F1 signaling pathway.
Collapse
Affiliation(s)
- Yan Guo
- College of Basic Medicine & Public Health, Zhejiang 310053, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wenxue Fan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shuyu Cao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yuefeng Xie
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jiancong Hong
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Huifen Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Haitong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Bo Jin
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| |
Collapse
|
14
|
The critical role of PPARα in the binary switch between life and death induced by endoplasmic reticulum stress. Cell Death Dis 2020; 11:691. [PMID: 32826849 PMCID: PMC7443130 DOI: 10.1038/s41419-020-02811-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 11/08/2022]
Abstract
Endoplasmic reticulum stress (ER stress) just like a double-edged sword depending on different conditions in the development of multiple hepatic diseases. But the molecular mechanisms of functional conversion during ER stress have not been fully elucidated. In this study, we aim to illustrate the role of PPARα and the subtle mechanism in the functional conversion of ER stress. Tunicamycin (TM) and thapsigargin (TG), as ER stress inducers, were used to induce ER stress in AML12 cells. During the ER stress, qRT-PCR and immunoblotting was used to measure the expression levels of GRP78 and CHOP which show a gradually increasing trend, while PPARα and autophagy was significantly activated in the early stage but was inhibited in the late stage. Moreover, PPARα inhibition by siRNA promoted cell injury in the mild-ER stress and PPARα activation by WY-14643 reduced cell apoptosis in the serious ER stress. In the mild-ER stress with PPARα knocked down, activation of autophagy by rapamycin significantly improved cell survival, in the serious ER stress with PPARα activation, inhibition of autophagy by 3-MA aggravate cell injury. In addition, in the mild-ER stress with PPARα knocked down, CHOP knocked down by siRNA reduced cell apoptosis, in the serious ER stress activated PPARα, CHOP over-expression mediated by lentiviral vector contributed to serious cell injury. Furthermore, C57BL/6 mice was used to induce ER stress with TM intraperitoneal injection, PPARα and autophagy was upregulated in the mild-ER stress while downregulated in the serious ER stress, measured by qRT-PCR and immunoblotting, further confirmed the finding in vitro. Our results firstly demonstrated that PPARα is a key molecule in the functional conversion of ER stress: protective effects in the mild ER stress was mediated by PPARα-autophagy pathway and destructive effects in the serious ER stress was mediated by PPARα-CHOP pathway.
Collapse
|
15
|
Abstract
Background A growing body of literature suggests the cell–intrinsic activity of Atf6α during ER stress responses has implications for tissue cell number during growth and development, as well as in adult biology and tumorigenesis [1]. This concept is important, linking the cellular processes of secretory protein synthesis and endoplasmic reticulum stress response with functional tissue capacity and organ size. However, the field contains conflicting observations, especially notable in secretory cell types like the pancreatic beta cell. Scope of review Here we summarize current knowledge of the basic biology of Atf6α, along with the pleiotropic roles Atf6α plays in cell life and death decisions and possible explanations for conflicting observations. We include studies investigating the roles of Atf6α in cell survival, death and proliferation using well-controlled methodology and specific validated outcome measures, with a focus on endocrine and metabolic tissues when information was available. Major conclusions The net outcome of Atf6α on cell survival and cell death depends on cell type and growth conditions, the presence and degree of ER stress, and the duration and intensity of Atf6α activation. It is unquestioned that Atf6α activity influences the cell fate decision between survival and death, although opposite directions of this outcome are reported in different contexts. Atf6α can also trigger cell cycle activity to expand tissue cell number through proliferation. Much work remains to be done to clarify the many gaps in understanding in this important emerging field.
Collapse
Affiliation(s)
- Rohit B Sharma
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jarin T Snyder
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laura C Alonso
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
16
|
Antunes F, Pereira GJS, Saito RF, Buri MV, Gagliardi M, Bincoletto C, Chammas R, Fimia GM, Piacentini M, Corazzari M, Smaili SS. Effective Synergy of Sorafenib and Nutrient Shortage in Inducing Melanoma Cell Death through Energy Stress. Cells 2020; 9:E640. [PMID: 32155825 PMCID: PMC7140454 DOI: 10.3390/cells9030640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
Skin melanoma is one of the most aggressive and difficult-to-treat human malignancies, characterized by poor survival rates, thus requiring urgent novel therapeutic approaches. Although metabolic reprogramming has represented so far, a cancer hallmark, accumulating data indicate a high plasticity of cancer cells in modulating cellular metabolism to adapt to a heterogeneous and continuously changing microenvironment, suggesting a novel therapeutic approach for dietary manipulation in cancer therapy. To this aim, we exposed melanoma cells to combined nutrient-restriction/sorafenib. Results indicate that cell death was efficiently induced, with apoptosis representing the prominent feature. In contrast, autophagy was blocked in the final stage by this treatment, similarly to chloroquine, which also enhanced melanoma cell sensitization to combined treatment. Energy stress was evidenced by associated treatment with mitochondrial dysfunction and glycolysis impairment, suggesting metabolic stress determining melanoma cell death. A reduction of tumor growth after cycles of intermittent fasting together with sorafenib treatment was also observed in vivo, reinforcing that the nutrient shortage can potentiate anti-melanoma therapy. Our findings showed that the restriction of nutrients by intermittent fasting potentiates the effects of sorafenib due to the modulation of cellular metabolism, suggesting that it is possible to harness the energy of cancer cells for the treatment of melanoma.
Collapse
Affiliation(s)
- Fernanda Antunes
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Gustavo J. S. Pereira
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Renata F. Saito
- Center for Translational Research in Oncology, Department of Radiology and Oncology, Faculty of Medicine of the University of São Paulo and Cancer Institute of the State of São Paulo, São Paulo 04021-001, Brazil; (R.F.S.); (R.C.)
| | - Marcus V. Buri
- Department of Molecular Biology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil;
| | - Mara Gagliardi
- Department of Health Sciences (DISS), University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), 28100 Novara, Italy
| | - Claudia Bincoletto
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| | - Roger Chammas
- Center for Translational Research in Oncology, Department of Radiology and Oncology, Faculty of Medicine of the University of São Paulo and Cancer Institute of the State of São Paulo, São Paulo 04021-001, Brazil; (R.F.S.); (R.C.)
| | - Gian Maria Fimia
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘Lazzaro Spallanzani’, 00149 Rome, Italy; (G.M.F.); (M.P.)
- Department of Molecular Medicine, University of Rome La Sapienza, 00185 Rome, Italy
| | - Mauro Piacentini
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS ‘Lazzaro Spallanzani’, 00149 Rome, Italy; (G.M.F.); (M.P.)
- Institute of Cytology of the Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Marco Corazzari
- Department of Health Sciences (DISS), University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), 28100 Novara, Italy
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Federal University of São Paulo, Paulista School of Medicine, São Paulo 04021-001, Brazil; (F.A.); (G.J.S.P.); (C.B.); (S.S.S.)
| |
Collapse
|
17
|
Pecoraro A, Carotenuto P, Russo G, Russo A. Ribosomal protein uL3 targets E2F1 and Cyclin D1 in cancer cell response to nucleolar stress. Sci Rep 2019; 9:15431. [PMID: 31659203 PMCID: PMC6817900 DOI: 10.1038/s41598-019-51723-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022] Open
Abstract
Several experimental strategies in the treatment of cancer include drug alteration of cell cycle regulatory pathways as a useful strategy. Extra-ribosomal functions of human ribosomal protein L3 (uL3) may affect DNA repair, cell cycle arrest and apoptosis. In the present study, we demonstrated that uL3 is required for the activation of G1/S transition genes. Luciferase assays established that uL3 negatively regulates the activity of E2F1 promoter. Induced ribosome-free uL3 reduces Cyclin D1 mRNA and protein levels. Using protein/protein immunoprecipitation methods, we demonstrated that uL3 physically interacts with PARP-1 affecting E2F1 transcriptional activity. Our findings led to the identification of a new pathway mediated by uL3 involving E2F1 and Cyclin D1 in the regulation of cell cycle progression.
Collapse
Affiliation(s)
- Annalisa Pecoraro
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131, Naples, Italy
| | - Pietro Carotenuto
- The Institute of Cancer Research, Cancer Therapeutics Unit 15 Cotswold Road, Sutton, London, SM2 5NG, UK
| | - Giulia Russo
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131, Naples, Italy.
| | - Annapina Russo
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131, Naples, Italy.
| |
Collapse
|
18
|
Dual role of Endoplasmic Reticulum Stress-Mediated Unfolded Protein Response Signaling Pathway in Carcinogenesis. Int J Mol Sci 2019; 20:ijms20184354. [PMID: 31491919 PMCID: PMC6770252 DOI: 10.3390/ijms20184354] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer constitutes a grave problem nowadays in view of the fact that it has become one of the main causes of death worldwide. Poor clinical prognosis is presumably due to cancer cells metabolism as tumor microenvironment is affected by oxidative stress. This event triggers adequate cellular response and thereby creates appropriate conditions for further cancer progression. Endoplasmic reticulum (ER) stress occurs when the balance between an ability of the ER to fold and transfer proteins and the degradation of the misfolded ones become distorted. Since ER is an organelle relatively sensitive to oxidative damage, aforementioned conditions swiftly cause the activation of the unfolded protein response (UPR) signaling pathway. The output of the UPR, depending on numerous factors, may vary and switch between the pro-survival and the pro-apoptotic branch, and hence it displays opposing effects in deciding the fate of the cancer cell. The role of UPR-related proteins in tumorigenesis, such as binding the immunoglobulin protein (BiP) and inositol-requiring enzyme-1α (IRE1α), activating transcription factor 6 (ATF6) or the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), has already been specifically described so far. Nevertheless, due to the paradoxical outcomes of the UPR activation as well as gaps in current knowledge, it still needs to be further investigated. Herein we would like to elicit the actual link between neoplastic diseases and the UPR signaling pathway, considering its major branches and discussing its potential use in the development of a novel, anti-cancer, targeted therapy.
Collapse
|
19
|
Role of the Death Receptor and Endoplasmic Reticulum Stress Signaling Pathways in Polyphyllin I-Regulated Apoptosis of Human Hepatocellular Carcinoma HepG2 Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5241941. [PMID: 30671458 PMCID: PMC6323420 DOI: 10.1155/2018/5241941] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/17/2022]
Abstract
Polyphyllin has been reported to exhibit anticancer effects against various types of cancer via the proapoptotic signaling pathway. The aim of the present study was to investigate the role of the endoplasmic reticulum stress and death receptor signaling pathways in PPI-induced apoptosis of human hepatocellular carcinoma HepG2 cells. Analysis demonstrated that PPI could significantly inhibit the proliferation and induce apoptosis of HepG2 cells in a dose- and time-dependent manner. Investigation into the molecular mechanism of PPI indicated that PPI notably mediated ER stress activation via IRE-1 overexpression and activation of the caspase-12 to protect HepG2 cells against apoptosis. In addition, PPI markedly induced the expression of death receptors signaling pathways-associated factors, including tumor necrosis factor (TNF) receptor 1/TNF-α and FAS/FASL. Additionally, suppression of the death receptor signaling pathways with a caspase-8 inhibitor, Z-IETD-FMK, revealed an increase in the death rate and apoptotic rate of HepG2 cells. Collectively, the findings of the present study suggested that the ER stress and death receptor signaling pathways were associated with PPI-induced HepG2 cell apoptosis; however, endoplasmic reticulum stress may serve a protective role in this process. The combination of PPI and Z-IETD-FMK may activate necroptosis, which enhances apoptosis. Therefore, the results of the present study may improve understanding regarding the roles of signaling pathways in PPI regulated apoptosis and contribute to the development of novel therapies for the treatment of HCC.
Collapse
|
20
|
Obiedat A, Seidel E, Mahameed M, Berhani O, Tsukerman P, Voutetakis K, Chatziioannou A, McMahon M, Avril T, Chevet E, Mandelboim O, Tirosh B. Transcription of the NKG2D ligand MICA is suppressed by the IRE1/XBP1 pathway of the unfolded protein response through the regulation of E2F1. FASEB J 2018; 33:3481-3495. [PMID: 30452881 DOI: 10.1096/fj.201801350rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The unfolded protein response (UPR) is an adaptive signaling pathway activated in response to endoplasmic reticulum (ER) stress. The effectors of the UPR are potent transcription activators; however, some genes are suppressed by ER stress at the mRNA level. The mechanisms underlying UPR-mediated gene suppression are less known. Exploration of the effect of UPR on NK cells ligand expression found that the transcription of NK group 2 member D (NKG2D) ligand major histocompatibility complex class I polypeptide-related sequence A/B (MICA/B) is suppressed by the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) pathway of the UPR. Deletion of IRE1 or XBP1 was sufficient to promote mRNA and surface levels of MICA. Accordingly, NKG2D played a greater role in the killing of IRE1/XBP1 knockout target cells. Analysis of effectors downstream to XBP1s identified E2F transcription factor 1 (E2F1) as linking UPR and MICA transcription. The inverse correlation between XBP1 and E2F1 or MICA expression was corroborated in RNA-Seq analysis of 470 primary melanoma tumors. While mechanisms that connect XBP1 to E2F1 are not fully understood, we implicate a few microRNA molecules that are modulated by ER stress and possess dual suppression of E2F1 and MICA. Because of the importance of E2F1 and MICA in cancer progression and recognition, these observations could be exploited for cancer therapy by manipulating the UPR in tumor cells.-Obiedat, A., Seidel, E., Mahameed, M., Berhani, O., Tsukerman, P., Voutetakis, K., Chatziioannou, A., McMahon, M., Avril, T., Chevet, E., Mandelboim, O., Tirosh, B. Transcription of the NKG2D ligand MICA is suppressed by the IRE1/XBP1 pathway of the unfolded protein response through the regulation of E2F1.
Collapse
Affiliation(s)
- Akram Obiedat
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einat Seidel
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Hadassah Medical School, Jerusalem, Israel
| | - Mohamed Mahameed
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Orit Berhani
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Hadassah Medical School, Jerusalem, Israel
| | - Pinchas Tsukerman
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Hadassah Medical School, Jerusalem, Israel
| | - Konstantinos Voutetakis
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation (NHRF), Athens, Greece.,Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Aristotelis Chatziioannou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation (NHRF), Athens, Greece.,e-Noesis Inspired Operational Systems Applications Private Company PC, Kallithea-Athens, Greece
| | - Mari McMahon
- INSERM U1242, University of Rennes, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France; and.,Apoptosis Research Centre (ARC), National University of Ireland, Galway (NUIG), Galway, Ireland
| | - Tony Avril
- INSERM U1242, University of Rennes, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France; and
| | - Eric Chevet
- INSERM U1242, University of Rennes, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Rennes, France; and
| | - Ofer Mandelboim
- The Lautenberg Center for Immunology and Cancer Research, The Biomedical Research Institute Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem Hadassah Medical School, Jerusalem, Israel
| | - Boaz Tirosh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
21
|
Al-Hrout A, Chaiboonchoe A, Khraiwesh B, Murali C, Baig B, El-Awady R, Tarazi H, Alzahmi A, Nelson DR, Greish YE, Ramadan W, Salehi-Ashtiani K, Amin A. Safranal induces DNA double-strand breakage and ER-stress-mediated cell death in hepatocellular carcinoma cells. Sci Rep 2018; 8:16951. [PMID: 30446676 PMCID: PMC6240095 DOI: 10.1038/s41598-018-34855-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Poor prognoses remain the most challenging aspect of hepatocellular carcinoma (HCC) therapy. Consequently, alternative therapeutics are essential to control HCC. This study investigated the anticancer effects of safranal against HCC using in vitro, in silico, and network analyses. Cell cycle and immunoblot analyses of key regulators of cell cycle, DNA damage repair and apoptosis demonstrated unique safranal-mediated cell cycle arrest at G2/M phase at 6 and 12 h, and at S-phase at 24 h, and a pronounced effect on DNA damage machinery. Safranal also showed pro-apoptotic effect through activation of both intrinsic and extrinsic initiator caspases; indicating ER stress-mediated apoptosis. Gene set enrichment analysis provided consistent findings where UPR is among the top terms of up-regulated genes in response to safranal treatment. Thus, proteins involved in ER stress were regulated through safranal treatment to induce UPR in HepG2 cells.
Collapse
Affiliation(s)
- Ala'a Al-Hrout
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Amphun Chaiboonchoe
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Basel Khraiwesh
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
- Center for Genomics and Systems Biology (CGSB), Division of Science, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - Chandraprabha Murali
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Badriya Baig
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE
| | - Raafat El-Awady
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Hamadeh Tarazi
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Amnah Alzahmi
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | - David R Nelson
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE
| | | | - Wafaa Ramadan
- College of Pharmacy and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Kourosh Salehi-Ashtiani
- Laboratory of Algal, Synthetic, and Systems Biology, Division of Science and Math, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE.
- Center for Genomics and Systems Biology (CGSB), Division of Science, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, UAE.
| | - Amr Amin
- Biology Department, College of Science, UAE University, P.O. Box 15551, Al-Ain, UAE.
- Zoology Department, Cairo University, Giza, Egypt.
| |
Collapse
|
22
|
Giglio P, Gagliardi M, Bernardini R, Mattei M, Cotella D, Santoro C, Piacentini M, Corazzari M. Ecto-Calreticulin is essential for an efficient immunogenic cell death stimulation in mouse melanoma. Genes Immun 2018; 20:509-513. [PMID: 30282994 DOI: 10.1038/s41435-018-0047-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
Abstract
Skin melanoma remains one of the most aggressive and difficult to treat human malignancy, with an increasing incidence every year. Although surgical resection represents the best therapeutic approach, this is only feasible in cases of early diagnosis. Furthermore, the established malignancy is resistant to all therapeutic strategies employed so far, resulting in an unacceptable patient survival rate. Although the immune-mediated therapeutic approaches, based on anti-PD1 or anti-CTLA4, are very promising and under clinical trial experimentation, they could conceal not yet fully emerged pitfalls such as the development of autoimmune diseases. Therefore, alternative therapeutic approaches are still under investigation, such as the immunogenic cell death (ICD) process. Here we show that the lack of calreticulin translocation onto mouse melanoma cell membrane prevents the stimulation of an effective ICD response in vivo.
Collapse
Affiliation(s)
- Paola Giglio
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy
| | - Mara Gagliardi
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.,Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Roberta Bernardini
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome 'Tor Vergata', Rome, Italy
| | - Maurizio Mattei
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome 'Tor Vergata', Rome, Italy
| | - Diego Cotella
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Claudio Santoro
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Mauro Piacentini
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy.,Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Marco Corazzari
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy.
| |
Collapse
|
23
|
Li M, Gu MM, Tian X, Xiao BB, Lu S, Zhu W, Yu L, Shang ZF. Hydroxylated-Graphene Quantum Dots Induce DNA Damage and Disrupt Microtubule Structure in Human Esophageal Epithelial Cells. Toxicol Sci 2018; 164:339-352. [PMID: 29669094 PMCID: PMC6016703 DOI: 10.1093/toxsci/kfy090] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Graphene quantum dots (GQDs) have attracted significant interests due to their unique chemical and physical properties. In this study, we investigated the potential effects of hydroxyl-modified GQDs (OH-GQDs) on the human esophageal epithelial cell line HET-1A. Our data revealed significant cytotoxicity of OH-GQDs which decreased the viability of HET-1A in a dose and time-dependent manner. The moderate concentration (25 or 50 µg/ml) of OH-GQDs significantly blocked HET-1A cells in G0/G1 cell cycle phase. An increased percentage of γH2AX-positive and genomically unstable cells were also detected in cells treated with different doses of OH-GQDs (25, 50, and 100 µg/ml). Microarray data revealed that OH-GQDs treatment down-regulated genes related to DNA damage repair, cell cycle regulation and cytoskeleton signal pathways indicating a novel role of OH-GQDs. Consistent with the microarray data, OH-GQDs disrupted microtubule structure and inhibited microtubule regrowth around centrosomes in HET-1A cells. In conclusion, our findings provide important evidence for considering the application of OH-GQDs in biomedical fields.
Collapse
Affiliation(s)
- Ming Li
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Meng-Meng Gu
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Xin Tian
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Bei-Bei Xiao
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Siyuan Lu
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Wei Zhu
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| | - Lan Yu
- Suzhou Digestive Diseases and Nutrition Research Center, Nanjing Medical University Affiliated Suzhou Hospital, North District of Suzhou Municipal Hospital, Suzhou 215000, People’s Republic of China
| | - Zeng-Fu Shang
- State Key Laboratory of Radiation Medicine and Protection, Department of Radiobiology, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, People’s Republic of China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People’s Republic of China
| |
Collapse
|
24
|
Purnell MC, Butawan MBA, Bingol K, Tolley EA, Whitt MA. Modulation of endoplasmic reticulum stress and the unfolded protein response in cancerous and noncancerous cells. SAGE Open Med 2018; 6:2050312118783412. [PMID: 29977552 PMCID: PMC6024343 DOI: 10.1177/2050312118783412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/25/2018] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES The bio-field array is a device that generates a dielectrophoretic electromagnetic field when placed in a hypotonic saline solution and a direct current of approximately 3 A is applied. It is known that cell physiology is guided by bioelectrical properties, and there is a significant growth inhibition in cancerous (MDA-MB-231) cells that are grown in media that has been reconstituted with the saline that has been exposed to the bio-field array direct current dielectrophoretic electromagnetic field, alternatively there is no growth inhibition noted in noncancerous cells (MCF-10A) when grown in the bio-field array direct current dielectrophoretic electromagnetic field treated versus control media. METHODS To examine the basis for selective growth inhibition in human breast carcinoma, we employed cell death assays, cell cycle assays, microarray analysis and reverse transcription-quantitative polymerase chain reaction. RESULTS We found a large transcriptional reprogramming in the cell lines and of the genes affected, those involved in endoplasmic reticulum stress and the unfolded protein response pathways showed some of the most dramatic changes. Cancerous cells grown in media that has been reconstituted with a hypotonic saline solution that has been exposed to the bio-field array direct current dielectrophoretic electromagnetic field show a significant and strong upregulation of the apoptotic arms of the unfolded protein response while the noncancerous cells show a decrease in endoplasmic reticulum stress via microarray analyses and reverse transcription-quantitative polymerase chain reaction. CONCLUSION The bio-field array shows potential to initiate apoptosis in cancerous cells while relieving cell stress in noncancerous cells in vitro. These studies lay a foundation for nurses to conduct future in vivo models for the possible development of future adjunct treatments in chronic disease.
Collapse
Affiliation(s)
- Marcy C Purnell
- Department of Microbiology, Immunology
and Biochemistry, College of Medicine, The University of Tennessee Health Science
Center, Memphis, TN, USA
- The Loewenberg College of Nursing, The
University of Memphis, Memphis, TN, USA
| | | | - Kemal Bingol
- Department of Microbiology, Immunology
and Biochemistry, College of Medicine, The University of Tennessee Health Science
Center, Memphis, TN, USA
| | - Elizabeth A Tolley
- Department of Preventive Medicine, The
University of Tennessee Health Science Center, Memphis, TN, USA
| | - Michael A Whitt
- Department of Microbiology, Immunology
and Biochemistry, College of Medicine, The University of Tennessee Health Science
Center, Memphis, TN, USA
| |
Collapse
|
25
|
Giglio P, Gagliardi M, Tumino N, Antunes F, Smaili S, Cotella D, Santoro C, Bernardini R, Mattei M, Piacentini M, Corazzari M. PKR and GCN2 stress kinases promote an ER stress-independent eIF2α phosphorylation responsible for calreticulin exposure in melanoma cells. Oncoimmunology 2018; 7:e1466765. [PMID: 30221067 DOI: 10.1080/2162402x.2018.1466765] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 04/03/2018] [Accepted: 04/13/2018] [Indexed: 12/21/2022] Open
Abstract
The immunogenic cell death (ICD) process represents a novel therapeutic approach to treat tumours, in which cytotoxic compounds promote both cancer cell death and the emission of damage-associated molecular patterns (DAMPs) from dying cells, to activate the immune system against the malignancy. Therefore, we explored the possibility to stimulate the key molecular players with a pivotal role in the execution of the ICD program in melanoma cells. To this aim, we used the pro-ICD agents mitoxantrone and doxorubicin and found that both agents could induce cell death and stimulate the release/exposure of the strictly required DAMPs in melanoma cells: i) calreticulin (CRT) exposure on the cell membrane; ii) ATP secretion; iii) type I IFNs gene up-regulation and iv) HMGB1 secretion, highlighting no interference by oncogenic BRAF. Importantly, although the ER stress-related PERK activation has been linked to CRT externalization, through the phosphorylation of eIF2α, we found that this stress pathway together with PERK were not involved in melanoma cells. Notably, we identified PKR and GCN2 as key mediators of eIF2α phosphorylation, facilitating the translocation of CTR on melanoma cells surface, under pro-ICD drugs stimulation. Therefore, our data indicate that pro-ICD drugs are able to stimulate the production/release of DAMPs in melanoma cells at least in vitro, indicating in this approach a potential new valuable therapeutic strategy to treat human skin melanoma malignancy.
Collapse
Affiliation(s)
- Paola Giglio
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.,Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy
| | - Mara Gagliardi
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.,Department of Health Science (DISS), University of Piemonte Orientale, Novara, Italy
| | - Nicola Tumino
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy
| | - Fernanda Antunes
- Department of Pharmacology, Federal University of São Paulo, Brazil
| | - Soraya Smaili
- Department of Pharmacology, Federal University of São Paulo, Brazil
| | - Diego Cotella
- Department of Health Science (DISS), University of Piemonte Orientale, Novara, Italy
| | - Claudio Santoro
- Department of Health Science (DISS), University of Piemonte Orientale, Novara, Italy
| | - Roberta Bernardini
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome Tor Vergata, Rome, Italy
| | - Maurizio Mattei
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome Tor Vergata, Rome, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.,Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy
| | - Marco Corazzari
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy.,Department of Health Science (DISS), University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
26
|
Interplay between P-Glycoprotein Expression and Resistance to Endoplasmic Reticulum Stressors. Molecules 2018; 23:molecules23020337. [PMID: 29415493 PMCID: PMC6017601 DOI: 10.3390/molecules23020337] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Multidrug resistance (MDR) is a phenotype of cancer cells with reduced sensitivity to a wide range of unrelated drugs. P-glycoprotein (P-gp)—a drug efflux pump (ABCB1 member of the ABC transporter gene family)—is frequently observed to be a molecular cause of MDR. The drug-efflux activity of P-gp is considered as the underlying mechanism of drug resistance against P-gp substrates and results in failure of cancer chemotherapy. Several pathological impulses such as shortages of oxygen and glucose supply, alterations of calcium storage mechanisms and/or processes of protein N-glycosylation in the endoplasmic reticulum (ER) leads to ER stress (ERS), characterized by elevation of unfolded protein cell content and activation of the unfolded protein response (UPR). UPR is responsible for modification of protein folding pathways, removal of misfolded proteins by ER associated protein degradation (ERAD) and inhibition of proteosynthesis. However, sustained ERS may result in UPR-mediated cell death. Neoplastic cells could escape from the death pathway induced by ERS by switching UPR into pro survival mechanisms instead of apoptosis. Here, we aimed to present state of the art information about consequences of P-gp expression on mechanisms associated with ERS development and regulation of the ERAD system, particularly focused on advances in ERS-associated therapy of drug resistant malignancies.
Collapse
|
27
|
Wang Z, Sun X, Bao Y, Mo J, Du H, Hu J, Zhang X. E2F1 silencing inhibits migration and invasion of osteosarcoma cells via regulating DDR1 expression. Int J Oncol 2017; 51:1639-1650. [PMID: 29039472 PMCID: PMC5673022 DOI: 10.3892/ijo.2017.4165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/04/2017] [Indexed: 12/17/2022] Open
Abstract
In the present study, knockdown of E2F1 impaired the migration and invasion of osteosarcoma cells. Further analysis showed that E2F1 knockdown decreased the expression of discoidin domain receptor 1 (DDR1) which plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Luciferase and ChIP assays confirmed that E2F1 silencing attenuated the expression of DDR1 through disrupting E2F1-mediated transcription of DDR1 in osteosarcoma cells. Similarly with the effect of E2F1 silencing, DDR1 knockdown weakened the migratory and invasive capabilities of osteosarcoma cells; while overexpression of DDR1 resulted in a significant increase of cell motility and invasiveness, even after knocking down E2F1. Interestingly, inactivation of E2F1/DDR1 pathway by shRNA weakened STAT3 signaling and subsequently suppressed the epithelial-mesenchymal transition (EMT) of osteosarcoma cells, as shown with decreased vimentin, MMP2, MMP9, and increased E-cadherin. Consistently, high expressions of E2F1 and DDR1 observed in osteosarcoma tissues were related to TNM stage and metastasis. In addition, high level of E2F1 or DDR1 was associated with poor prognosis in osteosarcoma patients. These results suggest that E2F1/DDR1/STAT3 pathway is critical for malignancy of osteosarcoma, which may provide a novel prognostic indicator or approach for osteosarcoma therapy.
Collapse
Affiliation(s)
- Zhaofeng Wang
- Clinical Laboratory, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Xianjie Sun
- Clinical Laboratory, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Yi Bao
- Central Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Juanfen Mo
- Central Laboratory, The Second Hospital of Jiaxing, Jiaxing, Zhejiang 314000, P.R. China
| | - Hengchao Du
- Clinical Laboratory, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Jichao Hu
- Clinical Laboratory, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Xingen Zhang
- Clinical Laboratory, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
28
|
Ma L, Peng L, Fang S, He B, Liu Z. Celastrol downregulates E2F1 to induce growth inhibitory effects in hepatocellular carcinoma HepG2 cells. Oncol Rep 2017; 38:2951-2958. [PMID: 29048668 DOI: 10.3892/or.2017.5971] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/09/2017] [Indexed: 11/06/2022] Open
Abstract
Celastrol, a natural compound extracted from Tripterygium wilfordii, is known to exhibit potential anticancer activities in various types of tumor cells. E2F1 is reported to be overexpressed in several types of human tumors and its inactivation may be a valuable novel potential therapeutic strategy for cancer treatment. However, the molecular mechanism underlying the pro-apoptotic effects of celastrol on hepatocellular carcinoma (HCC) cells remains unclear, and E2F1-targeted compounds have been rarely identified. In the present study, we demonstrated that celastrol inhibited the proliferation of human HCC cells and triggered apoptosis of HepG2 cells in a caspase-dependent manner. E2F1 was potently downregulated by celastrol in a dose- and time-dependent manner at both the mRNA and protein levels. Moreover, siRNA-mediated E2F1 silencing enhanced celastrol-induced apoptosis and inhibition of proliferation. Our data imply that downregulation of E2F1 may be a key factor in the celastrol-mediated inhibitory effects in HepG2 cells, and celastrol can serve as a leading compound for the development of compounds designed to inactivate E2F1 for HCC therapy.
Collapse
Affiliation(s)
- Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui 243002, P.R. China
| | - Lei Peng
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui 243002, P.R. China
| | - Sheng Fang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui 243002, P.R. China
| | - Bangguo He
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui 243002, P.R. China
| | - Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui 243002, P.R. China
| |
Collapse
|
29
|
Mammalian ECD Protein Is a Novel Negative Regulator of the PERK Arm of the Unfolded Protein Response. Mol Cell Biol 2017; 37:MCB.00030-17. [PMID: 28652267 PMCID: PMC5574048 DOI: 10.1128/mcb.00030-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/17/2017] [Indexed: 01/01/2023] Open
Abstract
Mammalian Ecdysoneless (ECD) is a highly conserved ortholog of the DrosophilaEcd gene product whose mutations impair the synthesis of Ecdysone and produce cell-autonomous survival defects, but the mechanisms by which ECD functions are largely unknown. Here we present evidence that ECD regulates the endoplasmic reticulum (ER) stress response. ER stress induction led to a reduced ECD protein level, but this effect was not seen in PKR-like ER kinase knockout (PERK-KO) or phosphodeficient eukaryotic translation initiation factor 2α (eIF2α) mouse embryonic fibroblasts (MEFs); moreover, ECD mRNA levels were increased, suggesting impaired ECD translation as the mechanism for reduced protein levels. ECD colocalizes and coimmunoprecipitates with PERK and GRP78. ECD depletion increased the levels of both phospho-PERK (p-PERK) and p-eIF2α, and these effects were enhanced upon ER stress induction. Reciprocally, overexpression of ECD led to marked decreases in p-PERK, p-eIF2α, and ATF4 levels but robust increases in GRP78 protein levels. However, GRP78 mRNA levels were unchanged, suggesting a posttranscriptional event. Knockdown of GRP78 reversed the attenuating effect of ECD overexpression on PERK signaling. Significantly, overexpression of ECD provided a survival advantage to cells upon ER stress induction. Taken together, our data demonstrate that ECD promotes survival upon ER stress by increasing GRP78 protein levels to enhance the adaptive folding protein in the ER to attenuate PERK signaling.
Collapse
|
30
|
Corazzari M, Gagliardi M, Fimia GM, Piacentini M. Endoplasmic Reticulum Stress, Unfolded Protein Response, and Cancer Cell Fate. Front Oncol 2017; 7:78. [PMID: 28491820 PMCID: PMC5405076 DOI: 10.3389/fonc.2017.00078] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/10/2017] [Indexed: 12/24/2022] Open
Abstract
Perturbation of endoplasmic reticulum (ER) homeostasis results in a stress condition termed "ER stress" determining the activation of a finely regulated program defined as unfolded protein response (UPR) and whose primary aim is to restore this organelle's physiological activity. Several physiological and pathological stimuli deregulate normal ER activity causing UPR activation, such as hypoxia, glucose shortage, genome instability, and cytotoxic compounds administration. Some of these stimuli are frequently observed during uncontrolled proliferation of transformed cells, resulting in tumor core formation and stage progression. Therefore, it is not surprising that ER stress is usually induced during solid tumor development and stage progression, becoming an hallmark of such malignancies. Several UPR components are in fact deregulated in different tumor types, and accumulating data indicate their active involvement in tumor development/progression. However, although the UPR program is primarily a pro-survival process, sustained and/or prolonged stress may result in cell death induction. Therefore, understanding the mechanism(s) regulating the cell survival/death decision under ER stress condition may be crucial in order to specifically target tumor cells and possibly circumvent or overcome tumor resistance to therapies. In this review, we discuss the role played by the UPR program in tumor initiation, progression and resistance to therapy, highlighting the recent advances that have improved our understanding of the molecular mechanisms that regulate the survival/death switch.
Collapse
Affiliation(s)
- Marco Corazzari
- Department of Health Sciences, University of Piemonte Orientale "A. Avogadro", Novara, Italy.,Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy
| | - Mara Gagliardi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Gian Maria Fimia
- Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy.,Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Mauro Piacentini
- Department Clinical Epidemiology and Translational Research, INMI-IRCCS "L. Spallanzani", Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
31
|
Antunes F, Corazzari M, Pereira G, Fimia GM, Piacentini M, Smaili S. Fasting boosts sensitivity of human skin melanoma to cisplatin-induced cell death. Biochem Biophys Res Commun 2017; 485:16-22. [PMID: 27693581 DOI: 10.1016/j.bbrc.2016.09.149] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 01/16/2023]
Abstract
Melanoma is one of leading cause of tumor death worldwide. Anti-cancer strategy includes combination of different chemo-therapeutic agents as well as radiation; however these treatments have limited efficacy and induce significant toxic effects on healthy cells. One of most promising novel therapeutic approach to cancer therapy is the combination of anti-cancer drugs with calorie restriction. Here we investigated the effect Cisplatin (CDDP), one of the most potent chemotherapeutic agent used to treat tumors, in association with fasting in wild type and mutated BRAFV600E melanoma cell lines. Here we show that nutrient deprivation can consistently enhance the sensitivity of tumor cells to cell death induction by CDDP, also of those malignancies particularly resistant to any treatment, such as oncogenic BRAF melanomas. Mechanistic studies revealed that the combined therapy induced cell death is characterized by ROS accumulation and ATF4 in the absence of ER-stress. In addition, we show that autophagy is not involved in the enhanced sensitivity of melanoma cells to combined CDDP/EBSS-induced apoptosis. While, the exposure to 2-DG further enhanced the apoptotic rate observed in SK Mel 28 cells upon treatment with both CDDP and EBSS.
Collapse
Affiliation(s)
- Fernanda Antunes
- Department of Pharmacology, Federal University of São Paulo, Brazil
| | - Marco Corazzari
- Department of Biology, University of Rome "Tor Vergata", Italy; National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Italy.
| | - Gustavo Pereira
- Department of Pharmacology, Federal University of São Paulo, Brazil
| | - Gian Maria Fimia
- Department of Biology, University of Rome "Tor Vergata", Italy; Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, 73100, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Italy; National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Italy.
| | - Soraya Smaili
- Department of Pharmacology, Federal University of São Paulo, Brazil
| |
Collapse
|
32
|
Bhat TA, Chaudhary AK, Kumar S, O'Malley J, Inigo JR, Kumar R, Yadav N, Chandra D. Endoplasmic reticulum-mediated unfolded protein response and mitochondrial apoptosis in cancer. Biochim Biophys Acta Rev Cancer 2016; 1867:58-66. [PMID: 27988298 DOI: 10.1016/j.bbcan.2016.12.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/11/2016] [Accepted: 12/13/2016] [Indexed: 12/12/2022]
Abstract
Abrogation of endoplasmic reticulum (ER) protein folding triggered by exogenous or endogenous factors, stimulates a cellular stress response, termed ER stress. ER stress re-establishes ER homeostasis through integrated signaling termed the ER-unfolded protein response (UPRER). In the presence of severe toxic or prolonged ER stress, the pro-survival function of UPRER is transformed into a lethal signal transmitted to and executed through mitochondria. Mitochondria are key for both apoptotic and autophagic cell death. Thus ER is vital in sensing and coordinating stress pathways to maintain overall physiological homeostasis. However, this function is deregulated in cancer, resulting in resistance to apoptosis induction in response to various stressors including therapeutic agents. Here we review the connections between ER stress and mitochondrial apoptosis, describing potential cancer therapeutic targets.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Joseph R Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, United States.
| |
Collapse
|
33
|
Elevated E2F7 expression predicts poor prognosis in human patients with gliomas. J Clin Neurosci 2016; 33:187-193. [PMID: 27460513 DOI: 10.1016/j.jocn.2016.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/16/2022]
Abstract
E2F transcription factors have been studied extensively in a broad range of organisms as major regulators of cell cycle, apoptosis, and differentiation. The E2F family includes the atypical member E2F7, which has been rarely studied in gliomas. The aim of this study is to determine the expression status of E2F7 in gliomas, its relationship to clinicopathological features, and patients' outcome. The mRNA levels of E2F7 in the human brain and different grades of gliomas were analysed using datasets from the publically available Oncomine database. One of the most significant co-expression factors, CDK1, together with E2F7, was further validated by immunohistochemistry in 90 different grades of gliomas. Furthermore, univariate and multivariate analyses were performed to identify prognostic variables relative to patient and tumour characteristics and treatment modalities. E2F7 mRNA expression was found to be elevated in gliomas by Oncomine-database analysis. Immunohistochemistry showed an increase in E2F7 labelling index in high- versus low-grade gliomas (62.1±11.8% vs. 18.9±10.2%, p<0.0001). There was a positive correlation between E2F7 and CDK1 immunoreactivity (Spearman r=0.446, p=0.037). Clinicopathological evaluation suggested that E2F7 expression was associated with tumour grade (p<0.0001) and recurrence (p=0.025). In Cox multivariate analysis, pathological classification and recurrence were independent prognostic factors of gliomas, and E2F7 was significantly related to progression-free survival (p=0.011), but not overall survival (p=0.062). Our findings suggested that E2F7 might act as an independent prognostic factor of gliomas and might constitute a potential therapeutic target for this disease.
Collapse
|
34
|
Nolan K, Walter F, Tuffy LP, Poeschel S, Gallagher R, Haunsberger S, Bray I, Stallings RL, Concannon CG, Prehn JHM. Endoplasmic reticulum stress-mediated upregulation of miR-29a enhances sensitivity to neuronal apoptosis. Eur J Neurosci 2016; 43:640-52. [DOI: 10.1111/ejn.13160] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/24/2015] [Accepted: 01/04/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Katie Nolan
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Franziska Walter
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Liam P. Tuffy
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Simone Poeschel
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Ross Gallagher
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Stefan Haunsberger
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Isabella Bray
- Cancer Genetics; Molecular and Cellular Therapeutics; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Raymond L. Stallings
- Cancer Genetics; Molecular and Cellular Therapeutics; Royal College of Surgeons in Ireland; Dublin Ireland
| | - Caoimhín G. Concannon
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| | - Jochen H. M. Prehn
- Centre for the Study of Neurological Disorders and Department of Physiology and Medical Physics; Royal College of Surgeons in Ireland; 123 St. Stephens Green Dublin 2 Ireland
| |
Collapse
|