1
|
Bolat İ, Bolat M, Kiliçlioğlu M, Yıldırım S, Sağlam YS, Çomaklı S, Gözegir B, Özmen M, Warda M. Differential TLR2 and TLR4 mediated inflammatory and apoptotic responses in asymptomatic and symptomatic Leptospira interrogans infections in canine uterine tissue. Microb Pathog 2025; 198:107186. [PMID: 39615709 DOI: 10.1016/j.micpath.2024.107186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/05/2024] [Accepted: 11/28/2024] [Indexed: 12/15/2024]
Abstract
Leptospirosis is major zoonotic disease with global implications, affecting both domestic animals and humans. It is caused by Leptospira interrogans (L. interrogans), which can damage multiple organs, including the kidneys, liver, testes, and uterus. Despite this, L. interrogans can also persist asymptomatically in tissues, akin to nonpathogenic strains. The mechanisms driving asymptomatic infections remain poorly understood. This study investigated the role of L. interrogans in asymptomatic infection within the uterine tissue of canines, focusing on the differential expression of Toll-like receptors (TLRs)2 and 4 and their roles in inflammatory and apoptotic pathways. We hypothesized that TLR2 and TLR4 coexpression is crucial for eliciting inflammation and apoptosis, whereas TLR4 alone might be insufficient. Our findings revealed that in symptomatic infections, both TLR2 and TLR4 are coexpressed, leading to markedly elevated levels of the proinflammatory cytokines IL-10, IL-1β, TNF-α, and IL-6. This enhanced inflammatory response is further evidenced by increased CD4 expression, indicating robust T helper cell activation. In contrast, asymptomatic infections are characterized by exclusive TLR4 expression, with inflammatory markers remaining at baseline levels. Additionally, we observed that L. interrogans induces apoptosis in symptomatic animals through TLR2 and TLR4 mediated activation of Caspase 8 and Caspase 3. These findings illustrate that L. interrogans drives both inflammation and apoptosis via the combination of TLR2 and TLR4 actions. When only TLR4 is activated, the immune response is insufficient, resulting in an asymptomatic disease course. This study provides novel insights into the differential roles of TLR receptors in leptospirosis, offering potential directions for targeted therapeutic strategies.
Collapse
Affiliation(s)
- İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Merve Bolat
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Metin Kiliçlioğlu
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Serkan Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Yavuz Selim Sağlam
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Berrah Gözegir
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Murat Özmen
- Molecular Diagnostics and Research Laboratory, Ministry of Agriculture and Forestry, Erzurum, Turkey
| | - Mohamad Warda
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey; Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Bolat İ, Bolat M, Kiliçlioğlu M, Yıldırım S, Sağlam YS, Çomaklı S, Gözegir B. Cellular pathophysiology of Leptospira interrogans infection in canine testicular tissue: role of the TLR4/NF-kB/JNK pathway. J Comp Pathol 2025; 216:10-19. [PMID: 39657408 DOI: 10.1016/j.jcpa.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/10/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024]
Abstract
Leptospirosis is a prevalent zoonotic disease in dogs. Although it is known that leptospires are primarily harboured in kidney tissues of dogs, it has been reported that they also infect testicular tissue. Leptospira interrogans causes various lesions in canine testicular tissues; however, the pathogenesis has not been clearly explained. In this study, 20 canine testicular tissue samples infected with L. interrogans were investigated for evidence of oxidative DNA damage, inflammation, apoptosis and autophagy. Ten samples of canine testicular tissue that were negative for L. interrogans were used as a positive control group. All tissues were examined by histopathological, immunohistochemistry (IHC) and immunofluorescence (IF) methods. Histopathological examination revealed that testicular tissues infected with L. interrogans had inflammation and oedema, and degeneration and necrosis of spermatocytes. In cases of severe disease, enzyme-linked immunosorbent assay, reverse transcriptase polymer chain reaction and IHC and IF indicated significant increases in levels of TLR4, NF-kB, IL-1β, TNF-α, 8-OHdG, JNK1/3, caspase-8, caspase-3, LC3A and LC3B but lower levels in milder cases. These results indicate that L. interrogans stimulated the immune system through the TLR4/NF-kB/JNK pathway in dog testicular tissues, leading to inflammation and apoptosis. The infection also caused oxidative DNA damage and autophagy.
Collapse
Affiliation(s)
- İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye.
| | - Merve Bolat
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| | - Metin Kiliçlioğlu
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| | - Serkan Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| | - Yavuz S Sağlam
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| | - Berrah Gözegir
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, 25240 Erzurum, Türkiye
| |
Collapse
|
3
|
Jayakodi S, Senthilnathan R, Swaminathan A, Shanmugam VK, Shanmugam RK, Krishnan A, Ponnusamy VK, Tsai PC, Lin YC, Chen YH. Bio-inspired nanoparticles mediated from plant extract biomolecules and their therapeutic application in cardiovascular diseases: A review. Int J Biol Macromol 2023:125025. [PMID: 37245774 DOI: 10.1016/j.ijbiomac.2023.125025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/05/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
Nanoparticles (NPs) have gained recognition for diagnosis, drug delivery, and therapy in fatal diseases. This review focuses on the benefits of green synthesis of bioinspired NPs using various plant extract (containing various biomolecules such as sugars, proteins, and other phytochemical compounds) and their therapeutic application in cardiovascular diseases (CVDs). Multiple factors including inflammation, mitochondrial and cardiomyocyte mutations, endothelial cell apoptosis, and administration of non-cardiac drugs, can trigger the cause of cardiac disorders. Furthermore, the interruption of reactive oxygen species (ROS) synchronization from mitochondria causes oxidative stress in the cardiac system, leading to chronic diseases such as atherosclerosis and myocardial infarction. NPs can decrease the interaction with biomolecules and prevent the incitement of ROS. Understanding this mechanism can pave the way for using green synthesized elemental NPs to reduce the risk of CVD. This review delivers information on the different methods, classifications, mechanisms and benefits of using NPs, as well as the formation and progression of CVDs and their effects on the body.
Collapse
Affiliation(s)
- Santhoshkumar Jayakodi
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Raghul Senthilnathan
- Global Business School for Health, University College London, Gower St, London WC1E 6BT, United Kingdom
| | - Akila Swaminathan
- Clinical Virology, Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Venkat Kumar Shanmugam
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Rajesh Kumar Shanmugam
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Chennai, Tamil Nadu 600077, India
| | - Anbarasu Krishnan
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India.
| | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City 807, Taiwan; Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City 807, Taiwan; Center for Emerging Contaminants Research, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Pei-Chien Tsai
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City 807, Taiwan
| | - Yuan-Chung Lin
- Center for Emerging Contaminants Research, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Institute of Environmental Engineering, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
4
|
Impact of chitosan administration on titanium dioxide nanoparticles induced testicular dysfunction. Sci Rep 2022; 12:19667. [PMID: 36385626 PMCID: PMC9669025 DOI: 10.1038/s41598-022-22044-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The potential reproductive toxic effects of oral TiO2 NPs in adult male rats as well as the possible alleviation of chitosan administration was investigated. Animals were allocated to four groups; the first group received deionized water and was assigned as a control group. In the second group, rats received chitosan at a dose of 5 mg/kg BW/day. The third group was designed for administration of TiO2 NPs at a dose of 150 mg/kg BW/day (1/80 LD50). Rats in the fourth group received both TiO2 NPs and chitosan. After 14 days, TiO2 NPs induced testicular lipid peroxidation as well as oxidative stress. Nano-titanium significantly upregulated genes that encode apoptosis and inflammation in testicular tissue. Moreover, it induced histological alteration in the testicular structure with impairment in spermatogenesis via reduction of PCNA immune-staining. Chitosan administration significantly improved the activities of testicular GPx, SOD, and CAT enzymes. In addition, it significantly down-regulated the relative expressions of pro-apoptotic and pro-inflammatory testicular genes. Chitosan was able to improve the testicular architecture as well as spermatogenesis. The current study revealed the capability of chitosan to ameliorate nano-titanium induced testicular toxicity. Thus, attention should be given to the extensive consumption of nano-titanium particles.
Collapse
|
5
|
Kumarasamy V, Nambiar R, Wang J, Rosenheck H, Witkiewicz AK, Knudsen ES. RB loss determines selective resistance and novel vulnerabilities in ER-positive breast cancer models. Oncogene 2022; 41:3524-3538. [PMID: 35676324 PMCID: PMC10680093 DOI: 10.1038/s41388-022-02362-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 05/11/2022] [Accepted: 05/24/2022] [Indexed: 11/09/2022]
Abstract
The management of metastatic estrogen receptor (ER) positive HER2 negative breast cancer (ER+) has improved; however, therapeutic resistance and disease progression emerges in majority of cases. Using unbiased approaches, as expected PI3K and MTOR inhibitors emerge as potent inhibitors to delay proliferation of ER+ models harboring PIK3CA mutations. However, the cytostatic efficacy of these drugs is hindered due to marginal impact on the expression of cyclin D1. Different combination approaches involving the inhibition of ER pathway or cell cycle result in durable growth arrest via RB activation and subsequent inhibition of CDK2 activity. However, cell cycle alterations due to RB loss or ectopic CDK4/cyclin D1 activation yields resistance to these cytostatic combination treatments. To define means to counter resistance to targeted therapies imparted with RB loss; complementary drug screens were performed with RB-deleted isogenic cell lines. In this setting, RB loss renders ER+ breast cancer models more vulnerable to drugs that target DNA replication and mitosis. Pairwise combinations using these classes of drugs defines greater selectivity for RB deficiency. The combination of AURK and WEE1 inhibitors, yields synergistic cell death selectively in RB-deleted ER+ breast cancer cells via apoptosis and yields profound disease control in vivo. Through unbiased efforts the XIAP/CIAP inhibitor birinapant was identified as a novel RB-selective agent. Birinapant further enhances the cytotoxic effect of chemotherapies and targeted therapies used in the treatment of ER+ breast cancer models selectively in the RB-deficient setting. Using organoid culture and xenograft models, we demonstrate the highly selective use of birinapant based combinations for the treatment of RB-deficient tumors. Together, these data illustrate the critical role of RB-pathway in response to many agents used to treat ER+ breast cancer, whilst informing new therapeutic approaches that could be deployed against resistant disease.
Collapse
Affiliation(s)
- Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Ram Nambiar
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Hanna Rosenheck
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA.
| |
Collapse
|
6
|
Choe K, Park HY, Ikram M, Lee HJ, Park TJ, Ullah R, Kim MO. Systematic Review of the Common Pathophysiological Mechanisms in COVID-19 and Neurodegeneration: The Role of Bioactive Compounds and Natural Antioxidants. Cells 2022; 11:cells11081298. [PMID: 35455977 PMCID: PMC9031507 DOI: 10.3390/cells11081298] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus (2019-nCoVCOVID-19) belongs to the Beta coronavirus family, which contains MERS-CoV (Middle East respiratory syndrome coronavirus) and SARS-CoV (severe acute respiratory syndrome coronavirus). SARS-CoV-2 activates the innate immune system, thereby activating the inflammatory mechanism, causing the release of inflammatory cytokines. Moreover, it has been suggested that COVID-19 may penetrate the central nervous system, and release inflammatory cytokines in the brains, inducing neuroinflammation and neurodegeneration. Several links connect COVID-19 with Alzheimer’s disease (AD), such as elevated oxidative stress, uncontrolled release of the inflammatory cytokines, and mitochondrial apoptosis. There are severe concerns that excessive immune cell activation in COVID-19 may aggravate the neurodegeneration and amyloid-beta pathology of AD. Here, we have collected the evidence, showing the links between the two diseases. The focus has been made to collect the information on the activation of the inflammation, its contributors, and shared therapeutic targets. Furthermore, we have given future perspectives, research gaps, and overlapping pathological bases of the two diseases. Lastly, we have given the short touch to the drugs that have equally shown rescuing effects against both diseases. Although there is limited information available regarding the exact links between COVID-19 and neuroinflammation, we have insight into the pathological contributors of the diseases. Based on the shared pathological features and therapeutic targets, we hypothesize that the activation of the immune system may induce neurological disorders by triggering oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Hyun Young Park
- Department of Pediatrics, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands;
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht Medical Center, 6229 ER Maastricht, The Netherlands
| | - Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Hyeon Jin Lee
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences (MVLS), University of Glasgow, Glasgow G12 0ZD, UK;
| | - Rahat Ullah
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (K.C.); (M.I.); (H.J.L.); (R.U.)
- Alz-Dementia Korea Co., Jinju 52828, Korea
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
7
|
Arsenic Nanoparticles are Effective in Reducing 3-Methylcholanthrene Induced Carcinogenesis in Murine Fibrosarcoma by Promoting Anti-tumorigenic Inflammation. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-021-00920-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
8
|
Dual Role of p73 in Cancer Microenvironment and DNA Damage Response. Cells 2021; 10:cells10123516. [PMID: 34944027 PMCID: PMC8700694 DOI: 10.3390/cells10123516] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms that regulate cancer progression is pivotal for the development of new therapies. Although p53 is mutated in half of human cancers, its family member p73 is not. At the same time, isoforms of p73 are often overexpressed in cancers and p73 can overtake many p53 functions to kill abnormal cells. According to the latest studies, while p73 represses epithelial–mesenchymal transition and metastasis, it can also promote tumour growth by modulating crosstalk between cancer and immune cells in the tumor microenvironment, M2 macrophage polarisation, Th2 T-cell differentiation, and angiogenesis. Thus, p73 likely plays a dual role as a tumor suppressor by regulating apoptosis in response to genotoxic stress or as an oncoprotein by promoting the immunosuppressive environment and immune cell differentiation.
Collapse
|
9
|
Zhou L, He Q, Yang X, Zheng S, Ou X. Research on Mechanism of Superparamagnetic Iron Oxide Nanoparticles in Activating Endoplasmic Reticulum and Prompting Apoptosis of Liver Cells Through Mediation of Tumor Necrosis Factor- α/Tumor Necrosis Factor Receptor 1 (TNF- α/TNFR1) Pathway. J Biomed Nanotechnol 2021; 17:2413-2419. [PMID: 34974864 DOI: 10.1166/jbn.2021.3199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of this study was to assess mechanism of superparamagnetic iron oxide nanoparticles (SPIO-NPs) in activating endoplasmic reticulum (ER) and prompting apoptosis of liver cells through mediating the TNF-α/TNFR1 pathway. The SPIO-NPs were prepared and identified, and HegG2 cells were cultivated in vitro, and their apoptosis was detected. The specific pathogen-free (SPF)-grade rats were divided into several groups; which included blank group, low concentration group, high concentration group and control group. The enzymatic activity of Caspase-3 in liver tissue was tested, and expressions of Caspase-3, Bax, Bcl-2, TNF-α, p-TNFR1, IRE1α, and eIF2α were tested. The size of prepared SPIO-NPs was 7.5 nm and there was no coagulation. There was good dispersity and electric potential, and appearance was stable. The apoptotic rate in the high concentration group was notably higher than in the other groups. There was notable inflammatory cell infiltration in the high concentration group, where quantity of apoptosis was highest. The quantity of apoptosis and fluorocyte in the high concentration group were notably higher than in the other groups. Moreover, there were over expressions of Caspase-3, Bax, Caspase-3, p-TNFR1, IRE1α, and eIF2α in the high concentration group while the expression of TNF-α was lowest. The apoptosis of HegG2 cells was prompted by SPIO-NPs, and quantity of apoptosis was increased with increased adopted concentration. The active expression of p-TNFR1, IRE1α, and eIF2α could be prompted to reduce the expression of TNF-α and increase the expression of Caspase-3 and Bax for prompting the apoptosis.
Collapse
Affiliation(s)
- Liyang Zhou
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Qin He
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xiaoàn Yang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Shuo Zheng
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xueting Ou
- Department of Infectious Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| |
Collapse
|
10
|
Lotfipanah S, Yaghmaei P, Zeinali M, Haeri Rohani SA, Kabodanian Ardestani S. Evaluation of TNF Family Gene Expression under the Influence of Single-Walled and Multi-Walled Carboxylated Carbon Nanotubes in Jurkat Cell Line and Rat. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2717. [PMID: 34435060 PMCID: PMC8358176 DOI: 10.30498/ijb.2021.2717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Nanomaterials, e.g.carbon nanotubes (CNTs), have broad usage in medicine for diagnosis, treatment, and drug delivery. Prior to the widespread use of CNTs, any potential toxicity
issues must be considered. Apoptosis is an important issue in toxicological studies, and tumor necrosis factor (TNF) family members execute crucial roles in apoptosis and inflammation.
We examined the survival of Jurkat cells under the influence of single-walled CNTs (SWCNTs) and multi-walled CNTs (MWCNTs) as well as their impacts on the mRNA levels of
TNF family transcripts in Jurkat cells and rats. Objective: To evaluate the toxicity or safety of a specific concentration and form of CNT on the expression of one of the gene families of the apoptotic pathway. Materials and Methods: Jurkat cells were exposed to SWCNTs and MWCNTs in carboxylated form (SWCNTS-COOH and MWCNTs-COOH). MTT assay assessed the cell survival, and using qRT-PCR, the expression
levels of TNF, CD40LG, TNFSF10, TNFSF8, CD40, TNFRSF10A, TNFRSF10B,
TNFRSF11B, TNFRSF1A, TNFRSF21, TNFRSF25, and TNFRSF9 were examined. The housekeeping genes β-actin and
glyceraldehyde 3-phosphate dehydrogenase was utilized for normalization. We also evaluated the expression levels of TNF and TNFRSF10A in rats in vivo 30 and 60 days after being injected with CNTs. Results: After 72 h of carboxylated CNTs at 100 µg. mL-1, no significant change was observed in the survival rate of treated Jurkat cells. The expression of two genes
(TNF and TNFRSF10A) changed significantly. Examining the expression profiles of these two genes in rats demonstrated an insignificant change in the expression of any
of these genes after 30 and 60 days. The qRT-PCR analysis exhibited the elevated levels of TNF and TNFRSF10A mRNA in the CNT-treated cells, while expression of other TNF family
members did not significantly differ from control (untreated) Jurkat cells. There was also no significant change in the gene expression levels of TNF and TNFRSF10A in CNT-treated rats after 30 and 60 days. Conclusions: Administration of SWCNTs-COOH and MWCNTs-COOH could result in the up-regulation of TNF and TNFRSF10A but did not initiate apoptosis in Jurkat cells.
Carboxylated SWCNTs showed more potent activity than MWCNTs in activating TNF gene expression and probably trigger cell death through external apoptotic pathways.
Collapse
Affiliation(s)
- Shirin Lotfipanah
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Majid Zeinali
- Biotechnology Research Center, Research Institute of Petroleum Industry (RIPI), Tehran, Iran
| | - Seyed Ali Haeri Rohani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
11
|
Lin HH, Hsieh MC, Wang CP, Yu PR, Lee MS, Chen JH. Anti-Atherosclerotic Effect of Gossypetin on Abnormal Vascular Smooth Muscle Cell Proliferation and Migration. Antioxidants (Basel) 2021; 10:antiox10091357. [PMID: 34572989 PMCID: PMC8470489 DOI: 10.3390/antiox10091357] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Gossypetin (GTIN), known as 3,5,7,8,3′,4′-hexahydroxyflavone, has been demonstrated to exert anti-atherosclerotic potential against apoptotic injury in oxidized low-density lipoprotein-incubated endothelial cells, and atherosclerotic lesions of cholesterol-fed rabbits. However, the effect and underlying mechanism of GTIN on abnormal vascular smooth muscle cells (VSMCs) proliferation and migration, a major event in the pathogenesis of atherosclerosis, is still unknown. In this study, non-cytotoxic doses of GTIN abolished the VSMCs A7r5 proliferation and cell-cycle S phase distribution. The GTIN-arrested G0/G1 phase might be performed by increasing the expressions of phosphorylated p53 and its downstream molecules that inhibit the activation of cyclin E/cyclin-dependent kinase (cdk)-2, blocking retinoblastoma protein (Rb) phosphorylation and the subsequent dissociation of Rb/transcription factor E2F1 complex. In addition, the results indicated that GTIN inhibited VSMCs wound-healing and migratory abilities through reducing matrix metalloproteinase (MMP)-9 activity and expression, as well as down-regulating protein kinase B (PKB)/nuclear factor-kappaB (NF-κB) signaling. GTIN also revealed potential in diminishing reactive oxygen species (ROS) generation. These findings suggested the inhibitory effects of GTIN on VSMCs dysfunction could likely lead to the containment of atherosclerosis and other cardiovascular illness.
Collapse
Affiliation(s)
- Hui-Hsuan Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan; (H.-H.L.); (M.-C.H.); (C.-P.W.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Ming-Chang Hsieh
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan; (H.-H.L.); (M.-C.H.); (C.-P.W.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Chi-Ping Wang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan; (H.-H.L.); (M.-C.H.); (C.-P.W.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
| | - Pei-Rong Yu
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan;
| | - Ming-Shih Lee
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City 40201, Taiwan; (H.-H.L.); (M.-C.H.); (C.-P.W.)
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
- Correspondence: (M.-S.L.); (J.-H.C.); Tel.: +886-424-730-022 (ext. 12404) (M.-S.L.); +886-424-730-022 (ext. 12195) (J.-H.C.)
| | - Jing-Hsien Chen
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan
- Department of Nutrition, Chung Shan Medical University, Taichung City 40201, Taiwan;
- Correspondence: (M.-S.L.); (J.-H.C.); Tel.: +886-424-730-022 (ext. 12404) (M.-S.L.); +886-424-730-022 (ext. 12195) (J.-H.C.)
| |
Collapse
|
12
|
Xie X, Liang J, Huang R, Luo C, Yang J, Xing H, Zhou L, Qiao H, Ergu E, Chen H. Molecular pathways underlying tissue injuries in the bladder with ketamine cystitis. FASEB J 2021; 35:e21703. [PMID: 34105799 DOI: 10.1096/fj.202100437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/14/2021] [Indexed: 12/11/2022]
Abstract
Ketamine cystitis (KC) is a chronic bladder inflammation leading to urinary urgency, frequency, and pain. The pathogenesis of KC is complicated and involves multiple tissue injuries in the bladder. Recent studies indicated that urothelium disruption, lamina propria fibrosis and inflammation, microvascular injury, neuropathological alterations, and bladder smooth muscle (BSM) abnormalities all contribute to the pathogenesis of KC. Ketamine has been shown to induce these tissue injuries by regulating different signaling pathways. Ketamine can stimulate antiproliferative factor, adenosine triphosphate, and oxidative stress to disrupt urothelium. Lamina propria fibrosis and inflammation are associated with the activation of cyclooxygenase-2, nitric oxide synthase, immunoglobulin E, and transforming growth factor β1. Ketamine contributes to microvascular injury via the N-methyl-D aspartic receptor (NMDAR), and multiple inflammatory and angiogenic factors such as tumor necrosis factor α and vascular endothelial growth factor. For BSM abnormalities, ketamine can depress the protein kinase B, extracellular signal-regulated kinase, Cav1.2, and muscarinic receptor signaling. Elevated purinergic signaling also plays a role in BSM abnormalities. In addition, ketamine affects neuropathological alterations in the bladder by regulating NMDAR- and brain-derived neurotrophic factor-dependent signaling. Inflammatory cells also contribute to neuropathological changes via the secretion of chemical mediators. Clarifying the role and function of these signaling underlying tissue injuries in the bladder with KC can contribute to a better understanding of the pathophysiology of this disease and to the design of effective treatments for KC.
Collapse
Affiliation(s)
- Xiang Xie
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jiayu Liang
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Run Huang
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Chuang Luo
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jiali Yang
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Hongming Xing
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Le Zhou
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Han Qiao
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Erti Ergu
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Huan Chen
- Public Center of Experimental Technology and The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
Gene 33/Mig6/ERRFI1, an Adapter Protein with Complex Functions in Cell Biology and Human Diseases. Cells 2021; 10:cells10071574. [PMID: 34206547 PMCID: PMC8306081 DOI: 10.3390/cells10071574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Gene 33 (also named Mig6, RALT, and ERRFI1) is an adapter/scaffold protein with a calculated molecular weight of about 50 kD. It contains multiple domains known to mediate protein–protein interaction, suggesting that it has the potential to interact with many cellular partners and have multiple cellular functions. The research over the last two decades has confirmed that it indeed regulates multiple cell signaling pathways and is involved in many pathophysiological processes. Gene 33 has long been viewed as an exclusively cytosolic protein. However, recent evidence suggests that it also has nuclear and chromatin-associated functions. These new findings highlight a significantly broader functional spectrum of this protein. In this review, we will discuss the function and regulation of Gene 33, as well as its association with human pathophysiological conditions in light of the recent research progress on this protein.
Collapse
|
14
|
Nath A, Cosgrove PA, Mirsafian H, Christie EL, Pflieger L, Copeland B, Majumdar S, Cristea MC, Han ES, Lee SJ, Wang EW, Fereday S, Traficante N, Salgia R, Werner T, Cohen AL, Moos P, Chang JT, Bowtell DDL, Bild AH. Evolution of core archetypal phenotypes in progressive high grade serous ovarian cancer. Nat Commun 2021; 12:3039. [PMID: 34031395 PMCID: PMC8144406 DOI: 10.1038/s41467-021-23171-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/08/2021] [Indexed: 12/16/2022] Open
Abstract
The evolution of resistance in high-grade serous ovarian cancer (HGSOC) cells following chemotherapy is only partially understood. To understand the selection of factors driving heterogeneity before and through adaptation to treatment, we profile single-cell RNA-sequencing (scRNA-seq) transcriptomes of HGSOC tumors collected longitudinally during therapy. We analyze scRNA-seq data from two independent patient cohorts to reveal that HGSOC is driven by three archetypal phenotypes, defined as oncogenic states that describe the majority of the transcriptome variation. Using a multi-task learning approach to identify the biological tasks of each archetype, we identify metabolism and proliferation, cellular defense response, and DNA repair signaling as consistent cell states found across patients. Our analysis demonstrates a shift in favor of the metabolism and proliferation archetype versus cellular defense response archetype in cancer cells that received multiple lines of treatment. While archetypes are not consistently associated with specific whole-genome driver mutations, they are closely associated with subclonal populations at the single-cell level, indicating that subclones within a tumor often specialize in unique biological tasks. Our study reveals the core archetypes found in progressive HGSOC and shows consistent enrichment of subclones with the metabolism and proliferation archetype as resistance is acquired to multiple lines of therapy.
Collapse
Affiliation(s)
- Aritro Nath
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Patrick A Cosgrove
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Hoda Mirsafian
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Lance Pflieger
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Benjamin Copeland
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Sumana Majumdar
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Mihaela C Cristea
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Ernest S Han
- Division of Gynecologic Oncology, Department of Surgery, City of Hope, Duarte, CA, USA
| | - Stephen J Lee
- Division of Gynecologic Oncology, Department of Surgery, City of Hope, Duarte, CA, USA
| | - Edward W Wang
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Sian Fereday
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Nadia Traficante
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Theresa Werner
- Division of Oncology, Department of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Adam L Cohen
- Division of Oncology, Department of Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Philip Moos
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
| | - Andrea H Bild
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA.
| |
Collapse
|
15
|
Laha D, Grant R, Mishra P, Nilubol N. The Role of Tumor Necrosis Factor in Manipulating the Immunological Response of Tumor Microenvironment. Front Immunol 2021; 12:656908. [PMID: 33986746 PMCID: PMC8110933 DOI: 10.3389/fimmu.2021.656908] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is an intricate system within solid neoplasms. In this review, we aim to provide an updated insight into the TME with a focus on the effects of tumor necrosis factor-α (TNF-α) on its various components and the use of TNF-α to improve the efficiency of drug delivery. The TME comprises the supporting structure of the tumor, such as its extracellular matrix and vasculature. In addition to cancer cells and cancer stem cells, the TME contains various other cell types, including pericytes, tumor-associated fibroblasts, smooth muscle cells, and immune cells. These cells produce signaling molecules such as growth factors, cytokines, hormones, and extracellular matrix proteins. This review summarizes the intricate balance between pro-oncogenic and tumor-suppressive functions that various non-tumor cells within the TME exert. We focused on the interaction between tumor cells and immune cells in the TME that plays an essential role in regulating the immune response, tumorigenesis, invasion, and metastasis. The multifunctional cytokine, TNF-α, plays essential roles in diverse cellular events within the TME. The uses of TNF-α in cancer treatment and to facilitate cancer drug delivery are discussed. The effects of TNF-α on tumor neovasculature and tumor interstitial fluid pressure that improve treatment efficacy are summarized.
Collapse
Affiliation(s)
| | | | | | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
16
|
Prasher P, Sharma M, Zacconi F, Gupta G, Aljabali AA, Mishra V, Tambuwala MM, Kapoor DN, Negi P, Andreoli Pinto TDJ, Singh I, Chellappan DK, Dua K. Synthesis and Anticancer Properties of ‘Azole’ Based Chemotherapeutics as Emerging Chemical Moieties: A Comprehensive Review. CURR ORG CHEM 2021. [DOI: 10.2174/1385272824999200820152501] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Azole frameworks serve as privileged scaffolds in the contemporary drug design
paradigm owing to their unique physicochemical profile that promotes the development
of highly selective, physiological benevolent chemotherapeutics. Several azole nuclei
function as bioisostere in medicinal chemistry and prompt the development of tailored
therapeutics for targeting the desired biological entities. Besides, the azole scaffold forms
an integral part in the advanced drug designing methodologies, such as target template insitu
drug synthesis, that assists in rapid identification of the hit molecules form a diverse
pool of leads; and direct biomolecule-drug conjugation, along with bioorthogonal strategies
that ensure localization, and superior target specificity of the directed therapeutic.
Lastly, the structural diversity of azole framework and high yielding click synthetic methods
provide a comprehensive Structure-Activity Relationship analysis for design optimization of the potential
drug molecules by fine-tuning the placement of different substituents critical for the activity. This review provides
a comprehensive analysis of the synthesis and anticancer potential of azole based chemotherapeutics.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Arcadia Grant, Dehradun 248007, India
| | - Flavia Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago 7820436, Chile
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302 017, Jaipur, India
| | - Alaa A.A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Deepak N. Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Terezinha de Jesus Andreoli Pinto
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, Professor Lineu Prestes Street, São Paulo 05508-000, Brazil
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
17
|
Distinct p63 and p73 Protein Interactions Predict Specific Functions in mRNA Splicing and Polyploidy Control in Epithelia. Cells 2020; 10:cells10010025. [PMID: 33375680 PMCID: PMC7824480 DOI: 10.3390/cells10010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial organs are the first barrier against microorganisms and genotoxic stress, in which the p53 family members p63 and p73 have both overlapping and distinct functions. Intriguingly, p73 displays a very specific localization to basal epithelial cells in human tissues, while p63 is expressed in both basal and differentiated cells. Here, we analyse systematically the literature describing p63 and p73 protein-protein interactions to reveal distinct functions underlying the aforementioned distribution. We have found that p73 and p63 cooperate in the genome stability surveillance in proliferating cells; p73 specific interactors contribute to the transcriptional repression, anaphase promoting complex and spindle assembly checkpoint, whereas p63 specific interactors play roles in the regulation of mRNA processing and splicing in both proliferating and differentiated cells. Our analysis reveals the diversification of the RNA and DNA specific functions within the p53 family.
Collapse
|
18
|
Aghamajidi A, Raoufi E, Parsamanesh G, Jalili A, Salehi-Shadkami M, Mehrali M, Mohsenzadegan M. The attentive focus on T cell-mediated autoimmune pathogenesis of psoriasis, lichen planus and vitiligo. Scand J Immunol 2020; 93:e13000. [PMID: 33190330 DOI: 10.1111/sji.13000] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/16/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022]
Abstract
T cell-mediated autoimmune skin diseases develop as a result of the aberrant immune response to the skin cells with T cells playing a central role. These chronic inflammatory skin diseases encompass various types including psoriasis, lichen planus and vitiligo. These diseases show similarities in their immune-pathophysiology. In the last decade, immunomodulating agents have been very successful in the management of these diseases thanks to a better understanding of the pathophysiology. In this review, we will discuss the immunopathogenic mechanisms and highlight the role of T lymphocytes in psoriasis, lichen planus and vitiligo. This study could provide new insights into a better understanding of targeted therapeutic pathways and biological therapies.
Collapse
Affiliation(s)
- Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Raoufi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Gilda Parsamanesh
- Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Ahmad Jalili
- Dermatology & Skin Care, Buergenstock Medical Center, Obbuergen, Switzerland
| | - Mohammad Salehi-Shadkami
- Student Research Committee, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Mehrali
- Student Research Committee, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Monireh Mohsenzadegan
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Wang B, Huang T, Fang Q, Zhang X, Yuan J, Li M, Ge H. Bone-protective and anti-tumor effect of baicalin in osteotropic breast cancer via induction of apoptosis. Breast Cancer Res Treat 2020; 184:711-721. [PMID: 32939591 DOI: 10.1007/s10549-020-05904-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/29/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE Research suggested that bone is the specific target organ for breast cancer metastasis. The related tumor causes significant morbidity due to a reduction in quality of life and physical function. Increased osteoclast function is implicated in the bone microenvironment during the outgrowth of breast cancer. In the present experimental study, we examined the potential bone-protective effect of baicalin osteotropic breast Cancer and explored the possible mechanism of action. METHODS In vitro cell viability effect of baicalin was assessed on the breast cancer cell lines (MDA-MB-231 and MCF-7). We also estimated the in vitro osteoclast and bone resorption. Further, baicalin-regulated osteoblastogenesis and osteoclastogenesis were also estimated in vitro. Finally, the role of the baicalin in the expansion of osteolytic bone disease was scrutinized in a breast cancer bone metastases model. RESULTS Baicalin significantly (p < 0.001) downregulated the viability of murine and human cancer cell lines and diminished the osteoclastogenesis of osteoclast progenitors via estimation with the help of qRT-PCR. Baicalin showed the downregulation in the mRNA expression of OCN and ALP. Baicalin reduced the TRAP-positive cells in the presence of RANKL. Baicalin considerably upregulated the cytochrome c secretion into the cytoplasm. Baicalin markedly increased the DNA fragmentation, caspase-3, caspase-8, and caspase-9. Baicalin significantly (p < 0.001) reduced the metastatic growth of MDA-MB-231 cells,preserving the bone mass in a bone metastasis model. CONCLUSION Collectively, we can conclude that these results highlight the bone-protective effect of baicalin, which also highlighted the anti-tumor effect; further research is needed into the likely effects on bone health in the bone metastases and osteoporosis populations, such as post-menopausal women with breast cancer.
Collapse
Affiliation(s)
- Bangmin Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Tao Huang
- Department of Galactophore, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Qigen Fang
- Department of Thyroid, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Xu Zhang
- Department of Thyroid, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Junhui Yuan
- Department of Radiology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Mengjie Li
- Department of Stomatology, Zhengzhou Stomatologic Hospital, Zhengzhou, 450008, Henan, China
| | - Hong Ge
- Department of Thyroid, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
- Department of Radiology, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
20
|
Ni G, Chen S, Chen M, Wu J, Yang B, Yuan J, Walton SF, Li H, Wei MQ, Wang Y, Chen G, Liu X, Wang T. Host-Defense Peptides Caerin 1.1 and 1.9 Stimulate TNF-Alpha-Dependent Apoptotic Signals in Human Cervical Cancer HeLa Cells. Front Cell Dev Biol 2020; 8:676. [PMID: 32850805 PMCID: PMC7412766 DOI: 10.3389/fcell.2020.00676] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/03/2020] [Indexed: 01/07/2023] Open
Abstract
Host defense caerin 1.1 and 1.9 peptides, isolated from the glandular secretion of Australian tree frogs, the genus Litoria, have been previously shown to have multiple biological activities, including the inhibition of human papillomavirus (HPV) 16 early protein E7 transformed murine as well as human cancerous cell proliferation both in vitro and in vivo. However, the mechanism underlying their anti-proliferative activities against HPV18+ cervical cancer HeLa cells remains unknown. This study comparatively investigated the anti-proliferation on HeLa cells by caerin 1.1, 1.9, and their mixture, followed by confocal microscopy examination to assess the cellular intake of the peptides. Tandem mass tag labeling proteomics was employed to reveal the proteins that were significantly regulated by the peptide treatment in cells and cell growth environment, to elucidate the signaling pathways that were modulated. Western blot was performed to confirm the modulation of the pathways. Both caerin 1.1 and 1.9 highly inhibited HeLa cell proliferation with a significant additive effect compared to untreated and control peptide. They entered the cells with different magnitudes. Intensive protein-protein interaction was detected among significantly upregulated proteins. Translation, folding and localization of proteins and RNA processing, apoptosis process was significantly enriched post the treatments. The apoptotic signaling was suggested as a result of tumor necrosis factor-α (TNF-α) pathway activation, indicated by the dose-dependent elevated levels of caspase 3 and caspase 9. The epidermal growth factor receptor and androgen receptor pathways appeared inhibited by the peptides. Moreover, the activation of T-cell receptor derived from the quantitation results further implies the likelihood of recruiting more T cells to the cell growth environment post the treatment and more sensitive to T cell mediated killing of HeLa cells. Our results indicate that caerin 1.1 and 1.9 mediate apoptotic signals of HeLa cells and may subsequently enhances adaptive T cell immune responses.
Collapse
Affiliation(s)
- Guoying Ni
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Shu Chen
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
| | - Mo Chen
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Jialing Wu
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
| | - Binbin Yang
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Southport, QLD, Australia
- Department of Laboratory Medicine, Institute of Nanomedicine Technology, Weifang Medical University, Weifang, China
| | - Jianwei Yuan
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Shelley F. Walton
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Hejie Li
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
- Department of Mechanical and Biofunctional System, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Ming Q. Wei
- Menzies Health Institute Queensland and School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Yuejian Wang
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
| | - Guoqiang Chen
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
| | - Xiaosong Liu
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, China
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Tianfang Wang
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore, QLD, Australia
| |
Collapse
|
21
|
Paramanantham A, Kim MJ, Jung EJ, Nagappan A, Yun JW, Kim HJ, Shin SC, Kim GS, Lee WS. Pretreatment of Anthocyanin from the Fruit of Vitis coignetiae Pulliat Acts as a Potent Inhibitor of TNF-α Effect by Inhibiting NF-κB-Regulated Genes in Human Breast Cancer Cells. Molecules 2020; 25:molecules25102396. [PMID: 32455624 PMCID: PMC7287973 DOI: 10.3390/molecules25102396] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/24/2022] Open
Abstract
Vitis coignetiaePulliat (Meoru in Korea) has been used in Korean folk medicine for the treatment of inflammatory diseases and cancers. Evidence suggests that NF-κB activation is mainly involved in cancer cell proliferation, invasion, angiogenesis, and metastasis. TNF-α also enhances the inflammatory process in tumor development. Recently, flavonoids from plants have been reported to have inhibitory effects on NF-κB activities. We investigated the effects of anthocyanins extracted from the fruits of Vitis coignetiae Pulliat (AIM, anthocyanins isolated from Meoru (AIM)) on TNF-α-induced NF-κB activities in MCF-7 human breast cancer cells and the molecules involved in AIM-induced anti-cancer effects, especially on cancer metastasis. We performed cell viability assay, gelatin zymography, invasion assay, and western blot analysis to unravel the anti-NF-κB activity of AIMs on MCF-7 cells. AIM suppressed the TNF-α effects on the NF-κB-regulated proteins involved in cancer cell proliferation (COX-2, C-myc), invasion, and angiogenesis (MMP-2, MMP9, ICAM-1, and VEGF). AIM also increased the expression of E-cadherin, which is one of the hallmarks of the epithelial-mesenchymal transition (EMT) process. In conclusion, this study demonstrates that the anthocyanins isolated from the fruits of Vitis coignetiae Pulliat acts as an inhibitor of TNF-α induced NF-κB activation, and subsequent downstream molecules involved in cancer proliferation, invasion, adhesion, angiogenesis, and thus have anti-metastatic activities in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Anjugam Paramanantham
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (A.P.); (M.J.K.); (A.N.); (J.W.Y.)
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea
| | - Min Jeong Kim
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (A.P.); (M.J.K.); (A.N.); (J.W.Y.)
| | - Eun Joo Jung
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea;
| | - Arulkumar Nagappan
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (A.P.); (M.J.K.); (A.N.); (J.W.Y.)
| | - Jeong Won Yun
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (A.P.); (M.J.K.); (A.N.); (J.W.Y.)
| | - Hye Jung Kim
- Departments of Pharmacology, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea;
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, Korea;
| | - Gon Sup Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea
- Correspondence: (G.S.K.); (W.S.L.); Tel.: +82-55-772-2356 (G.S.K.); +82-55-750-8733 (W.S.L.)
| | - Won Sup Lee
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 660-702, Korea; (A.P.); (M.J.K.); (A.N.); (J.W.Y.)
- Correspondence: (G.S.K.); (W.S.L.); Tel.: +82-55-772-2356 (G.S.K.); +82-55-750-8733 (W.S.L.)
| |
Collapse
|
22
|
HDAC3-ERα Selectively Regulates TNF-α-Induced Apoptotic Cell Death in MCF-7 Human Breast Cancer Cells via the p53 Signaling Pathway. Cells 2020; 9:cells9051280. [PMID: 32455774 PMCID: PMC7290399 DOI: 10.3390/cells9051280] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/29/2022] Open
Abstract
Tumor necrosis factor-α (TNF-α) plays a significant role in inflammation and cancer-related apoptosis. We identified a TNF-α-mediated epigenetic mechanism of apoptotic cell death regulation in estrogen receptor-α (ERα)-positive human breast cancer cells. To assess the apoptotic effect of TNF-α, annexin V/ propidium iodide (PI) double staining, cell viability assays, and Western blotting were performed. To elucidate this mechanism, histone deacetylase (HDAC) activity assay and immunoprecipitation (IP) were conducted; the mechanism was subsequently confirmed through chromatin IP (ChIP) assays. Finally, we assessed HDAC3-ERα-mediated apoptotic cell death after TNF-α treatment in ERα-positive human breast cancer (MCF-7) cells via the transcriptional activation of p53 target genes using luciferase assay and quantitative reverse transcription PCR. The TNF-α-induced selective apoptosis in MCF-7 cells was negatively regulated by the HDAC3-ERα complex in a caspase-7-dependent manner. HDAC3 possessed a p53-binding element, thus suppressing the transcriptional activity of its target genes. In contrast, MCF-7 cell treatment with TNF-α led to dissociation of the HDAC3-ERα complex and substitution of the occupancy on the promoter by the p53-p300 complex, thus accelerating p53 target gene expression. In this process, p53 stabilization was accompanied by its acetylation. This study showed that p53-mediated apoptosis in ERα-positive human breast cancer cells was negatively regulated by HDAC3-ERα in a caspase-7-dependent manner. Therefore, these proteins have potential application in therapeutic strategies.
Collapse
|
23
|
Parada CA, Osbun J, Kaur S, Yakkioui Y, Shi M, Pan C, Busald T, Karasozen Y, Gonzalez-Cuyar LF, Rostomily R, Zhang J, Ferreira M. Kinome and phosphoproteome of high-grade meningiomas reveal AKAP12 as a central regulator of aggressiveness and its possible role in progression. Sci Rep 2018; 8:2098. [PMID: 29391485 PMCID: PMC5794791 DOI: 10.1038/s41598-018-19308-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/29/2017] [Indexed: 01/05/2023] Open
Abstract
There is a need to better understand meningioma oncogenesis for biomarker discovery and development of targeted therapies. Histological or genetic criteria do not accurately predict aggressiveness. Post-translational studies in meningioma progression are lacking. In the present work, we introduce a combination of mass spectrometry-based phosphoproteomics and peptide array kinomics to profile atypical and anaplastic (high-grade) meningiomas. In the discovery set of fresh-frozen tissue specimens (14), the A-kinase anchor protein 12 (AKAP12) protein was found downregulated across the grades. AKAP12 knockdown in benign meningioma cells SF4433 increases proliferation, cell cycle, migration, invasion, and confers an anaplastic profile. Differentially regulated pathways were characteristic of high-grade meningiomas. Low AKAP12 expression in a larger cohort of patients (75) characterized tumor invasiveness, recurrence, and progression, indicating its potential as a prognostic biomarker. These results demonstrate AKAP12 as a central regulator of meningioma aggressiveness with a possible role in progression.
Collapse
Affiliation(s)
- Carolina Angelica Parada
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Joshua Osbun
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Sumanpreet Kaur
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Youssef Yakkioui
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Min Shi
- Department of Pathology/University of Washington School of Medicine, Harborview Medical Center, Seattle/WA, 98104, USA
| | - Catherine Pan
- Department of Pathology/University of Washington School of Medicine, Harborview Medical Center, Seattle/WA, 98104, USA
| | - Tina Busald
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Yigit Karasozen
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Luis Francisco Gonzalez-Cuyar
- Department of Pathology/University of Washington School of Medicine, Harborview Medical Center, Seattle/WA, 98104, USA
| | - Robert Rostomily
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA
| | - Jing Zhang
- Department of Pathology/University of Washington School of Medicine, Harborview Medical Center, Seattle/WA, 98104, USA
| | - Manuel Ferreira
- Departments of Neurosurgery/University of Washington School of Medicine, University of Washington Medical Center, Seattle/WA, 98195, USA.
| |
Collapse
|
24
|
Antonucci LA, Egger JV, Krucher NA. Phosphorylation of the Retinoblastoma protein (Rb) on serine-807 is required for association with Bax. Cell Cycle 2015; 13:3611-7. [PMID: 25483096 PMCID: PMC4614104 DOI: 10.4161/15384101.2014.964093] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The recent finding that the Retinoblastoma protein (Rb) is able to regulate apoptosis in a non-transcriptional manner directly at the mitochondria by interaction with the pro-apoptotic protein Bax prompted this investigation of the complex formed between Rb and Bax. Because the function of Rb in the cellular processes of proliferation, apoptosis, senescence and differentiation is regulated by phosphorylation we endeavored to elucidate the phosphorylation status of Rb with respect to its association with Bax and its role in apoptosis. In this study we found that Rb phosphorylated on at least 4 C-terminal phosphorylation sites (S608, S795, S807/S811, and T821) is present at the mitochondria under non-stressed cellular conditions. An in vitro binding assay showed that Bax binds to Rb phosphorylated at S807/S811 in 3 cancer cell types. Physiologically relevant association between Bax and Rb phosphorylated on S807/S811 was demonstrated by reciprocal co-immunoprecipitation experiments using antibodies specific for Rb phosphorylated on S807/S811 and Bax. Mutant Rb proteins expressed in Rb-null C33A cells showed that phosphorylation of S807 of Rb promotes association with Bax and that mimicking phosphorylation at S807 of Rb can block the induction of apoptosis due to PNUTS downregulation. Finally using siRNA to activate phosphatase activity in MCF7 cells, Rb is dephosphorylated at several sites including S807/S811, dissociates from Bax and apoptosis is triggered. These studies show that phosphorylation of Rb regulates its association with Bax and its role in apoptosis.
Collapse
Affiliation(s)
- Lisa A Antonucci
- a Department of Biology and Health Science ; Pace University ; Pleasantville , NY USA
| | | | | |
Collapse
|
25
|
Thompson PA, Khatami M, Baglole CJ, Sun J, Harris S, Moon EY, Al-Mulla F, Al-Temaimi R, Brown D, Colacci A, Mondello C, Raju J, Ryan E, Woodrick J, Scovassi I, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Hamid RA, Lowe L, Guarnieri T, Bisson WH. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 2015; 36 Suppl 1:S232-S253. [PMID: 26106141 PMCID: PMC4492068 DOI: 10.1093/carcin/bgv038] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 01/09/2015] [Accepted: 01/14/2015] [Indexed: 12/16/2022] Open
Abstract
An emerging area in environmental toxicology is the role that chemicals and chemical mixtures have on the cells of the human immune system. This is an important area of research that has been most widely pursued in relation to autoimmune diseases and allergy/asthma as opposed to cancer causation. This is despite the well-recognized role that innate and adaptive immunity play as essential factors in tumorigenesis. Here, we review the role that the innate immune cells of inflammatory responses play in tumorigenesis. Focus is placed on the molecules and pathways that have been mechanistically linked with tumor-associated inflammation. Within the context of chemically induced disturbances in immune function as co-factors in carcinogenesis, the evidence linking environmental toxicant exposures with perturbation in the balance between pro- and anti-inflammatory responses is reviewed. Reported effects of bisphenol A, atrazine, phthalates and other common toxicants on molecular and cellular targets involved in tumor-associated inflammation (e.g. cyclooxygenase/prostaglandin E2, nuclear factor kappa B, nitric oxide synthesis, cytokines and chemokines) are presented as example chemically mediated target molecule perturbations relevant to cancer. Commentary on areas of additional research including the need for innovation and integration of systems biology approaches to the study of environmental exposures and cancer causation are presented.
Collapse
Affiliation(s)
- Patricia A. Thompson
- *To whom correspondence should be addressed. Tel: +1 631 444 6818; Fax: +1 631 444 3424;
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA
| | - Carolyn J. Baglole
- Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Shelley Harris
- Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Dustin Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Ivana Scovassi
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy
- Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy,
- National Institute of Biostructures and Biosystems, Viale Medaglie d’ Oro, 305, 00136 Roma, Italy and
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
26
|
Zaharieva MM, Kirilov M, Chai M, Berger SM, Konstantinov S, Berger MR. Reduced expression of the retinoblastoma protein shows that the related signaling pathway is essential for mediating the antineoplastic activity of erufosine. PLoS One 2014; 9:e100950. [PMID: 24987858 PMCID: PMC4079453 DOI: 10.1371/journal.pone.0100950] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/02/2014] [Indexed: 01/13/2023] Open
Abstract
Erufosine is a new antineoplastic agent of the group of alkylphosphocholines, which interferes with signal transduction and induces apoptosis in various leukemic and tumor cell lines. The present study was designed to examine for the first time the mechanism of resistance to erufosine in malignant cells with permanently reduced expression of the retinoblastoma (Rb) protein. Bearing in mind the high number of malignancies with reduced level of this tumor-suppressor, this investigation was deemed important for using erufosine, alone or in combination, in patients with compromised RB1 gene expression. For this purpose, clones of the leukemic T-cell line SKW-3 were used, which had been engineered to constantly express differently low Rb levels. The alkylphosphocholine induced apoptosis, stimulated the expression of the cyclin dependent kinase inhibitor p27Kip1 and inhibited the synthesis of cyclin D3, thereby causing a G2 phase cell cycle arrest and death of cells with wild type Rb expression. In contrast, Rb-deficiency impeded the changes induced by eru-fosine in the expression of these proteins and abrogated the induction of G2 arrest, which was correlated with reduced antiproliferative and anticlonogenic activities of the compound. In conclusion, analysis of our results showed for the first time that the Rb signaling pathway is essential for mediating the antineoplastic activity of erufosine and its efficacy in patients with malignant diseases may be predicted by determining the Rb status.
Collapse
Affiliation(s)
- Maya M. Zaharieva
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
| | - Milen Kirilov
- Department of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany
| | - Minquang Chai
- Department of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany
| | - Stefan M. Berger
- Department of Molecular Biology, Central Institute of Mental Health, Mannheim, Germany
| | - Spiro Konstantinov
- Laboratory for Molecular Pharmacology and Experimental Chemotherapy, Department for Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Martin R. Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
27
|
Sridevi P, Nhiayi MK, Setten RL, Wang JYJ. Persistent inhibition of ABL tyrosine kinase causes enhanced apoptotic response to TRAIL and disrupts the pro-apoptotic effect of chloroquine. PLoS One 2013; 8:e77495. [PMID: 24147007 PMCID: PMC3795698 DOI: 10.1371/journal.pone.0077495] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 09/04/2013] [Indexed: 12/14/2022] Open
Abstract
TNF-Related Apoptosis Inducing Ligand (TRAIL) binds to and activates death receptors to stimulate caspase-8 and apoptosis with higher efficiency in cancer than normal cells but the development of apoptosis resistance has limited its clinical efficacy. We found that stable, but not transient knockdown of the ABL tyrosine kinase enhanced the apoptotic response to TRAIL. Re-expression of Abl, but not its nuclear import- or kinase-defective mutant, in the ABL-knockdown cells re-established apoptosis suppression. TRAIL is known to stimulate caspase-8 ubiquitination (Ub-C8), which can facilitate caspase-8 activation or degradation by the lysosomes. In the ABL-knockdown cells, we found a higher basal level of Ub-C8 that was not further increased by lysosomal inhibition. Re-expression of Abl in the ABL-knockdown cells reduced the basal Ub-C8, correlating with apoptosis suppression. We found that lysosomal inhibition by chloroquine (CQ) could also enhance TRAIL-induced apoptosis. However, this pro-apoptotic effect of CQ was lost in the ABL-knockdown cells but restored by Abl re-expression. Interestingly, kinase inhibition at the time of TRAIL stimulation was not sufficient to enhance apoptosis. Instead, persistent treatment for several days with imatinib, an ABL kinase inhibitor, was required to cause the enhanced and the CQ-insensitive apoptotic response to TRAIL. Together, these results show that persistent loss of nuclear ABL tyrosine kinase function can sensitize cells to TRAIL and suggest that long-term exposure to the FDA-approved ABL kinase inhibitors may potentiate apoptotic response to TRAIL-based cancer therapy.
Collapse
Affiliation(s)
- Priya Sridevi
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - May K. Nhiayi
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ryan L. Setten
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jean Y. J. Wang
- Moores Cancer Center, Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Hopkins S, Linderoth E, Hantschel O, Suarez-Henriques P, Pilia G, Kendrick H, Smalley M, Superti-Furga G, Ferby I. Mig6 is a sensor of EGF receptor inactivation that directly activates c-Abl to induce apoptosis during epithelial homeostasis. Dev Cell 2013; 23:547-59. [PMID: 22975324 PMCID: PMC3657149 DOI: 10.1016/j.devcel.2012.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 05/14/2012] [Accepted: 08/01/2012] [Indexed: 11/21/2022]
Abstract
A fundamental aspect of epithelial homeostasis is the dependence on specific growth factors for cell survival, yet the underlying mechanisms remain obscure. We found an “inverse” mode of receptor tyrosine kinase signaling that directly links ErbB receptor inactivation to the induction of apoptosis. Upon ligand deprivation Mig6 dissociates from the ErbB receptor and binds to and activates the tyrosine kinase c-Abl to trigger p73-dependent apoptosis in mammary epithelial cells. Deletion of Errfi1 (encoding Mig6) and inhibition or RNAi silencing of c-Abl causes impaired apoptosis and luminal filling of mammary ducts. Mig6 activates c-Abl by binding to the kinase domain, which is prevented in the presence of epidermal growth factor (EGF) by Src family kinase-mediated phosphorylation on c-Abl-Tyr488. These results reveal a receptor-proximal switch mechanism by which Mig6 actively senses EGF deprivation to directly activate proapoptotic c-Abl. Our findings challenge the common belief that deprivation of growth factors induces apoptosis passively by lack of mitogenic signaling.
Collapse
Affiliation(s)
- Sarah Hopkins
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Emma Linderoth
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Oliver Hantschel
- CeMM – Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Paula Suarez-Henriques
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Giulia Pilia
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 751 24 Uppsala, Sweden
| | - Howard Kendrick
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Matthew J. Smalley
- Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Giulio Superti-Furga
- CeMM – Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, 1090 Vienna, Austria
| | - Ingvar Ferby
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, 751 24 Uppsala, Sweden
- Corresponding author
| |
Collapse
|
29
|
Lentine B, Antonucci L, Hunce R, Edwards J, Marallano V, Krucher NA. Dephosphorylation of threonine-821 of the retinoblastoma tumor suppressor protein (Rb) is required for apoptosis induced by UV and Cdk inhibition. Cell Cycle 2012; 11:3324-30. [PMID: 22895174 DOI: 10.4161/cc.21693] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Retinoblastoma protein (Rb) is important in the control of cell proliferation and apoptosis. Its activity is controlled by reversible phosphorylation on several serine and threonine residues. When Rb is hypophosphorylated, it inhibits proliferation by preventing passage through the G 1- S phase transition. Hyperphosphorylated Rb promotes cell cycle progression. The role of Rb phosphorylation in the control of apoptosis is largely unknown, although several apoptotic stimuli result in dephosphorylation of Rb. It may be that dephosphorylation of specific amino acids signals apoptosis vs. cell cycle arrest. Using glutamic acid mutagenesis, we have generated 15 single phosphorylation site mutants of Rb to alter serine/threonine to glutamic acid to mimic the phosphorylated state. By calcium phosphate transfection, mutant plasmids were introduced into C33A Rb-null cells, and apoptosis was induced using UV. Apoptosis was measured by ELISA detection of degraded DNA and by immunoblotting to assess proteolytic cleavage of PARP. Our results show that only mutation of threonine-821 to glutamic acid (T821E) blocked apoptosis by 50%, whereas other sites tested had little effect. In Rb-null Saos-2 and SKUT-1 cells, the T821E mutation also blocked apoptosis induced by the cdk inhibitor, Roscovitine, by 50%. In addition, we show that endogenous Rb is dephosphorylated on threonine-821 when cells are undergoing apoptosis. Thus, our data indicates that dephosphorylation of threonine-821 of Rb is required for cells to undergo apoptosis.
Collapse
Affiliation(s)
- Brandon Lentine
- Department of Biology and Health Science, Pace University, 219 Pleasantville, NY, USA
| | | | | | | | | | | |
Collapse
|
30
|
Zhan Q, Korngold R, Lezcano C, McKeon F, Murphy GF. Graft-versus-host disease-related cytokine-driven apoptosis depends on p73 in cytokeratin 15-positive target cells. Biol Blood Marrow Transplant 2012; 18:841-51. [PMID: 22469882 DOI: 10.1016/j.bbmt.2012.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/13/2012] [Indexed: 11/26/2022]
Abstract
Acute graft-versus-host disease (GVHD), a major complication of allogeneic stem cell transplantation, involves cytotoxic soluble and cellular effectors that selectively induce apoptosis in normally apoptosis-resistant, cytokeratin 15 (K15)-expressing epithelial stem cells residing at the tips of rete ridges of human epidermis and in analogous rete-like prominences (RLPs) of murine dorsal lingual epithelium. The mechanisms whereby epithelial stem cells are rendered vulnerable to apoptosis during allostimulation are unknown. We hypothesized that GVHD-induced target cell injury may be related to pathways involving the p53 family that are constitutively expressed by epithelial stem cells and designed to trigger physiological apoptosis as a result of environmental danger signals. Among the p53 family members, we found that p73 protein and mRNA were preferentially expressed in K15(+) RLPs of murine lingual squamous epithelium. On in vitro exposure to recombinant TNF-α and IL-1 in an organ culture model previously shown to replicate early GVHD-like target cell injury, apoptosis was selectively induced in K15(+) stem cell regions and was associated with induction of phosphorylated p73, a marker for p73 activation, and apoptosis was abrogated in target tissue obtained from p73-deficient (p73(-/-)) mice. Evaluation of early in vivo lesions in experimental murine GVHD disclosed identical patterns of phosphorylated p73 expression that coincided with the onset of effector T cell infiltration and target cell apoptosis within K15(+) RLPs. This study is the first to suggest that paradoxical apoptosis in GVHD of physiologically protected K15(+) epithelial stem cells is explainable, at least in part, by cytokine-induced activation of suicide pathways designed to eliminate stem cells after exposure to deleterious factors perceived to be harmful to the host.
Collapse
Affiliation(s)
- Qian Zhan
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
31
|
Huang DY, Chao Y, Tai MH, Yu YH, Lin WW. STI571 reduces TRAIL-induced apoptosis in colon cancer cells: c-Abl activation by the death receptor leads to stress kinase-dependent cell death. J Biomed Sci 2012; 19:35. [PMID: 22462553 PMCID: PMC3348077 DOI: 10.1186/1423-0127-19-35] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 03/30/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND In an effort to achieve better cancer therapies, we elucidated the combination cancer therapy of STI571 (an inhibitor of Bcr-Abl and clinically used for chronic myelogenous leukemia) and TNF-related apoptosis-inducing ligand (TRAIL, a developing antitumor agent) in leukemia, colon, and prostate cancer cells. METHODS Colon cancer (HCT116, SW480), prostate cancer (PC3, LNCaP) and leukemia (K562) cells were treated with STI571 and TRAIL. Cell viability was determined by MTT assay and sub-G1 appearance. Protein expression and kinase phosphorylation were determined by Western blotting. c-Abl and p73 activities were inhibited by target-specific small interfering (si)RNA. In vitro kinase assay of c-Abl was conducted using CRK as a substrate. RESULTS We found that STI571 exerts opposite effects on the antitumor activity of TRAIL. It enhanced cytotoxicity in TRAIL-treated K562 leukemia cells and reduced TRAIL-induced apoptosis in HCT116 and SW480 colon cancer cells, while having no effect on PC3 and LNCaP cells. In colon and prostate cancer cells, TRAIL caused c-Abl cleavage to the active form via a caspase pathway. Interestingly, JNK and p38 MAPK inhibitors effectively blocked TRAIL-induced toxicity in the colon, but not in prostate cancer cells. Next, we found that STI571 could attenuate TRAIL-induced c-Abl, JNK and p38 activation in HCT116 cells. In addition, siRNA targeting knockdown of c-Abl and p73 also reduced TRAIL-induced cytotoxicity, rendering HCT116 cells less responsive to stress kinase activation, and masking the cytoprotective effect of STI571. CONCLUSIONS All together we demonstrate a novel mediator role of p73 in activating the stress kinases p38 and JNK in the classical apoptotic pathway of TRAIL. TRAIL via caspase-dependent action can sequentially activate c-Abl, p73, and stress kinases, which contribute to apoptosis in colon cancer cells. Through the inhibition of c-Abl-mediated apoptotic p73 signaling, STI571 reduces the antitumor activity of TRAIL in colon cancer cells. Our results raise additional concerns when developing combination cancer therapy with TRAIL and STI571 in the future.
Collapse
Affiliation(s)
- Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
32
|
Rastogi S, Rizwani W, Joshi B, Kunigal S, Chellappan SP. TNF-α response of vascular endothelial and vascular smooth muscle cells involve differential utilization of ASK1 kinase and p73. Cell Death Differ 2012; 19:274-83. [PMID: 21738216 PMCID: PMC3191254 DOI: 10.1038/cdd.2011.93] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 04/25/2011] [Accepted: 05/19/2011] [Indexed: 01/02/2023] Open
Abstract
Atherosclerosis involves a specialized inflammatory process regulated by an intricate network of cytokine and chemokine signaling. Atherosclerotic lesions lead to the release of cytokines that can have multiple affects on various vascular cell functions either promoting lesion expansion or alternatively retard progression. Tumor necrosis factor-α (TNF-α) is one such cytokine that can activate both cell survival and cell death mechanisms simultaneously. Here we show that TNF-α induces apoptosis in human aortic endothelial cells (HAECs), while it promotes the proliferation of vascular smooth muscle cells (VSMCs). Both events involved the activation of the Rb-E2F1 transcriptional regulatory pathway. Stimulation of HAECs with TNF-α led to an increased expression of p73 protein and a reduction in the levels of p53. This involved apoptosis signal-regulating kinase 1 (ASK1)- mediated inactivation of Rb and its dissociation from the p73 promoter. In contrast, TNF-α stimulation of VSMCs enhanced the association of E2F1 with proliferative promoters like thymidylate synthase and cdc25A, while Rb was dissociated. ASK1 kinase has a critical role in the apoptotic process, as its depletion or dissociation from Rb reduced TNF-α-induced apoptosis. These results show that the cytokine TNF-α can elicit diametrically opposite responses in vascular endothelial cells and VSMCs, utilizing the Rb-E2F pathway.
Collapse
Affiliation(s)
- S Rastogi
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - W Rizwani
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - B Joshi
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - S Kunigal
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - S P Chellappan
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
33
|
Davis R, Pillai S, Lawrence N, Sebti S, Chellappan SP. TNF-α-mediated proliferation of vascular smooth muscle cells involves Raf-1-mediated inactivation of Rb and transcription of E2F1-regulated genes. Cell Cycle 2012; 11:109-18. [PMID: 22185776 DOI: 10.4161/cc.11.1.18473] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is characterized by hyperplastic neointima and an inflammatory response with cytokines such as TNFα. TNFα is a pleiotropic cytokine that mediates inflammatory, proliferative, cytostatic and cytotoxic effects in a variety of cell types, including endothelial cells and vascular smooth muscle cells (VSMCs). Interestingly, TNFα has been shown to play two very opposing roles in these cell types; it inhibits proliferation and induces apoptosis in endothelial cells, while it enhances the proliferation and migration of VSMCs. Here we show that TNFα is capable of stimulating proliferation of rat VSMCs as well as human VSMCs in a Raf-1/MAP K-dependent manner. TNFα could increase the expression of E2F-regulated proliferative cdc6, Thymidylate synthase (TS) and cdc25A genes in Aortic smooth muscle cells (AoSMC), as seen by real time PCR assays. There is an activation of the stress-induced kinase, JNK1, in VSMCs upon TNFα stimulation. TNFα was capable of inducing binding of the Raf-1 kinase to Rb, and treatment with the Rb-Raf-1 inhibitor, RRD-251, could prevent TNFα-induced S-phase entry in AoSMCs. In addition, inhibition of Raf-1 or Src kinases using pharmacologic inhibitors could also prevent S-phase entry, while inhibition of JNK was not as effective. These results suggest that inhibiting the Rb-Raf-1 interaction is a potential avenue to prevent VSMC proliferation associated with atherosclerosis.
Collapse
Affiliation(s)
- Rebecca Davis
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
34
|
D'agostino L, Giordano A. NSP 5a3a: a potential novel cancer target in head and neck carcinoma. Oncotarget 2011; 1:423-35. [PMID: 21311098 DOI: 10.18632/oncotarget.100913] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
NSP 5a3a along with three other distinct though similar splice variants were initially identified corresponding to locus HCMOGT-1 on chromosome 17p11.2 [1]. Secondary structure analysis of the novel structural protein (NSP) isoforms revealed similarity to Spectrin like proteins containing coiled coil domains [1]. The NSP 5a3a isoform had been found to be highly expressed in-vitro in particular cancer cell lines while very low to un-detectable levels in normal body tissues [1]. Subsequent investigation of this isoform revealed its novel interaction with B23 [2], a multifunctional nucleolar protein involved in ribosome biogenesis, rRNA transcription, mitosis, cell growth control, and apoptosis [3]. Subsequent investigation, elucidated NSP 5a3a's potential involvement in cellular processes such as ribosome biogenesis and rRNA processing by validating NSP 5a3a's novel interaction with B23 and ribonuclear protein hnRNP-L possibly implicating NSP 5a3a's involvement in cellular activities such as RNA metabolism and processing [4]. In this preliminary investigation, we wanted to observe the effect that over-expressing NSP 5a3a may have on cell cycle and its potential application in cancer treatment in aggressive cancers such as head and neck carcinomas. Over-expressed NSP 5a3a in HN30 cells induced a significant degree of apoptosis, an average of a 10.85 fold increase compared to controls 3 days post-transfection. This effect was more significant then the apoptosis observed between Fadu cells over-expressing NSP 5a3a and its controls. Though, the apoptosis induced in the WI38 control cell line showed an average of a 13.2 fold increase between treated and controls comparable to the HN30 cell line 3 days post-transfection. Molecular analysis indentified a novel p73 dependent mechanism independent of p53 and caspase 3 activity through which NSP 5a3a is inducing apoptosis. We propose NSP 5a3a as a potential therapeutic target for site directed cancer treatment in perhaps certain head and neck carcinomas by induction of apoptosis.
Collapse
Affiliation(s)
- Luca D'agostino
- Sbarro Institute for Cancer Research and Molecular Medicine and Department of Biology, College of Science and Technology Temple University, 1900 North 12th street room 431, Philadelphia, PA 19122, USA.
| | | |
Collapse
|
35
|
Allington TM, Schiemann WP. The Cain and Abl of epithelial-mesenchymal transition and transforming growth factor-β in mammary epithelial cells. Cells Tissues Organs 2010; 193:98-113. [PMID: 21051857 DOI: 10.1159/000320163] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transforming growth factor-β (TGF-β) normally inhibits breast cancer development by preventing mammary epithelial cell (MEC) proliferation, by inducing MEC apoptosis, and by creating cell microenvironments that maintain MEC homeostasis and prevent their uncontrolled growth and motility. Mammary tumorigenesis elicits dramatic alterations in MEC architecture and microenvironment integrity, which collectively counteract the tumor-suppressing activities of TGF-β and enable its stimulation of breast cancer invasion and metastasis. How malignant MECs overcome the cytostatic actions imposed by normal microenvironments and TGF-β, and how abnormal microenvironments conspire with TGF-β to stimulate the development and progression of mammary tumors remains largely undefined. These knowledge gaps have prevented science and medicine from implementing treatments effective in simultaneously targeting abnormal cellular microenvironments, and in antagonizing the oncogenic activities of TGF-β in developing and progressing breast cancers. c-Abl is a ubiquitously expressed nonreceptor protein tyrosine kinase that essentially oversees all aspects of cell physiology, including the regulation of cell proliferation, migration and adhesion, as well as that of cell survival. Thus, the biological functions of c-Abl are highly reminiscent of those attributed to TGF-β, including the ability to function as either a suppressor or promoter of tumorigenesis. Interestingly, while dysregulated Abl activity clearly promotes tumorigenesis in hematopoietic cells, an analogous role for c-Abl in regulating solid tumor development, including those of the breast, remains controversial. Here, we review the functions of c-Abl in regulating breast cancer development and progression, and in alleviating the oncogenic activities of TGF-β and its stimulation of epithelial-mesenchymal transition during mammary tumorigenesis.
Collapse
Affiliation(s)
- Tressa M Allington
- Department of Pharmacology, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colo., USA
| | | |
Collapse
|
36
|
D'agostino L, Giordano A. NSP 5a3a: a potential novel cancer target in head and neck carcinoma. Oncotarget 2010; 1:423-435. [PMID: 21311098 PMCID: PMC3248119 DOI: 10.18632/oncotarget.176] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 09/29/2010] [Indexed: 11/25/2022] Open
Abstract
NSP 5a3a along with three other distinct though similar splice variants were initially identified corresponding to locus HCMOGT-1 on chromosome 17p11.2 [1]. Secondary structure analysis of the novel structural protein (NSP) isoforms revealed similarity to Spectrin like proteins containing coiled coil domains [1]. The NSP 5a3a isoform had been found to be highly expressed in-vitro in particular cancer cell lines while very low to un-detectable levels in normal body tissues [1]. Subsequent investigation of this isoform revealed its novel interaction with B23 [2], a multifunctional nucleolar protein involved in ribosome biogenesis, rRNA transcription, mitosis, cell growth control, and apoptosis [3]. Subsequent investigation, elucidated NSP 5a3a's potential involvement in cellular processes such as ribosome biogenesis and rRNA processing by validating NSP 5a3a's novel interaction with B23 and ribonuclear protein hnRNP-L possibly implicating NSP 5a3a's involvement in cellular activities such as RNA metabolism and processing [4]. In this preliminary investigation, we wanted to observe the effect that over-expressing NSP 5a3a may have on cell cycle and its potential application in cancer treatment in aggressive cancers such as head and neck carcinomas. Over-expressed NSP 5a3a in HN30 cells induced a significant degree of apoptosis, an average of a 10.85 fold increase compared to controls 3 days post-transfection. This effect was more significant then the apoptosis observed between Fadu cells over-expressing NSP 5a3a and its controls. Though, the apoptosis induced in the WI38 control cell line showed an average of a 13.2 fold increase between treated and controls comparable to the HN30 cell line 3 days post-transfection. Molecular analysis indentified a novel p73 dependent mechanism independent of p53 and caspase 3 activity through which NSP 5a3a is inducing apoptosis. We propose NSP 5a3a as a potential therapeutic target for site directed cancer treatment in perhaps certain head and neck carcinomas by induction of apoptosis.
Collapse
|
37
|
Abstract
Emerging evidence indicates that suppression of protein kinase C (PKC) renders the susceptibility of cells expressing mutated ras to apoptosis. Although the effort has been made, the underlying molecular mechanisms are not fully understood. In this study, using small hairpin RNAs (shRNAs) or PKC inhibitor, we show that the concurrent suppression of PKC-alpha and beta induces cells ectopically expressing v-ras to undergo apoptosis. In this apoptotic process, PKC-delta is upregulated and translocated from the cytosol to the nucleus. The activated PKC-delta associates with and phosphorylates p73 to initiate apoptosis. In this apoptotic process, Akt seems to be downstream of oncogenic Ras. Moreover, overexpression of PKC-delta, without co-suppression of PKC-alpha and beta, is not apoptotic to the cells, suggesting that PKC-delta and PKC-alpha/beta function oppositely to facilitate cells harboring v-ras to survive. Thus, our study shows that PKC-alpha and beta are necessary for sustaining the homeostasis in cells containing a hyperactive Ras. The abrogation of these two isoforms switches on the p73-regulated apoptotic machinery through the activation of PKC-delta.
Collapse
|
38
|
NF-kappaB activity is constitutively elevated in c-Abl null fibroblasts. Proc Natl Acad Sci U S A 2009; 106:17823-8. [PMID: 19805123 DOI: 10.1073/pnas.0905935106] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The c-abl proto-oncogene encodes a nonreceptor tyrosine kinase involved in many cellular processes, including signaling from growth factor and antigen receptors, remodeling the cytoskeleton, and responding to DNA damage and oxidative stress. Many downstream pathways are affected by c-Abl. Elevated c-Abl kinase activity can inhibit NF-kappaB activity by stabilizing the inhibitory protein IkappaB alpha, raising the possibility that c-Abl-deficient cells might have increased NF-kappaB activity. We examined the levels of NF-kappaB activity in primary mouse embryonic fibroblasts (MEFs) derived from wild-type and c-Abl knockout mice and found that the knockout MEFs indeed exhibited elevated NF-kappaB activity in response to stimulation as well as constitutively elevated NF-kappaB activity. Thus, endogenous c-Abl is a negative regulator of basal and inducible NF-kappaB activity. Examination of various points of NF-kappaB regulation revealed that unstimulated c-Abl knockout MEFs do not exhibit an increase in IkappaB alpha degradation, p65/RelA nuclear translocation, or DNA binding of NF-kappaB subunits. They do, however, show reduced levels of the histone deacetylase HDAC1, a negative regulator of basal NF-kappaB activity. Unstimulated c-Abl knockout MEFs are less responsive to induction of NF-kappaB activity by trichostatin A, an HDAC inhibitor, suggesting that c-Abl might play a role in the HDAC-mediated repression of basal NF-kappaB activity.
Collapse
|
39
|
Huang V, Lu X, Jiang Y, Wang JYJ. Effect of hydroxyurea on the promoter occupancy profiles of tumor suppressor p53 and p73. BMC Biol 2009; 7:35. [PMID: 19558638 PMCID: PMC2711048 DOI: 10.1186/1741-7007-7-35] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 06/26/2009] [Indexed: 12/24/2022] Open
Abstract
Background The p53 tumor suppressor and its related protein, p73, share a homologous DNA binding domain, and mouse genetics studies have suggested that they have overlapping as well as distinct biological functions. Both p53 and p73 are activated by genotoxic stress to regulate an array of cellular responses. Previous studies have suggested that p53 and p73 independently activate the cellular apoptotic program in response to cytotoxic drugs. The goal of this study was to compare the promoter-binding activity of p53 and p73 at steady state and after genotoxic stress induced by hydroxyurea. Results We employed chromatin immunoprecipitation, the NimbleGen promoter arrays and a model-based algorithm for promoter arrays to identify promoter sequences enriched in anti-p53 or anti-p73 immunoprecipitates, either before or after treatment with hydroxyurea, which increased the expression of both p53 and p73 in the human colon cancer cell line HCT116-3(6). We calculated a model-based algorithm for promoter array score for each promoter and found a significant correlation between the promoter occupancy profiles of p53 and p73. We also found that after hydroxyurea treatment, the p53-bound promoters were still bound by p73, but p73 became associated with additional promoters that that did not bind p53. In particular, we showed that hydroxyurea induces the binding of p73 but not p53 to the promoter of MLH3, which encodes a mismatch repair protein, and causes an up-regulation of the MLH3 mRNA. Conclusion These results suggest that hydroxyurea exerts differential effects on the promoter-binding functions of p53 and p73 and illustrate the power of model-based algorithm for promoter array in the analyses of promoter occupancy profiles of highly homologous transcription factors.
Collapse
Affiliation(s)
- Vera Huang
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0820, USA.
| | | | | | | |
Collapse
|
40
|
Karten B, Peake KB, Vance JE. Mechanisms and consequences of impaired lipid trafficking in Niemann-Pick type C1-deficient mammalian cells. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:659-70. [PMID: 19416638 DOI: 10.1016/j.bbalip.2009.01.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Accepted: 01/20/2009] [Indexed: 11/18/2022]
Abstract
Niemann-Pick C disease is a fatal progressive neurodegenerative disorder caused in 95% of cases by mutations in the NPC1 gene; the remaining 5% of cases result from mutations in the NPC2 gene. The major biochemical manifestation of NPC1 deficiency is an abnormal sequestration of lipids, including cholesterol and glycosphingolipids, in late endosomes/lysosomes (LE/L) of all cells. In this review, we summarize the current knowledge of the NPC1 protein in mammalian cells with particular focus on how defects in NPC1 alter lipid trafficking and neuronal functions. NPC1 is a protein of LE/L and is predicted to contain thirteen transmembrane domains, five of which constitute a sterol-sensing domain. The precise function of NPC1, and the mechanism by which NPC1 and NPC2 (both cholesterol binding proteins) act together to promote the movement of cholesterol and other lipids out of the LE/L, have not yet been established. Recent evidence suggests that the sequestration of cholesterol in LE/L of cells of the brain (neurons and glial cells) contributes to the widespread death and dysfunction of neurons in the brain. Potential therapies include treatments that promote the removal of cholesterol and glycosphingolipids from LE/L. Currently, the most promising approach for extending life-span and improving the quality of life for NPC patients is a combination of several treatments each of which individually modestly slows disease progression.
Collapse
Affiliation(s)
- Barbara Karten
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | | | | |
Collapse
|
41
|
Cancino GI, Toledo EM, Leal NR, Hernandez DE, Yévenes LF, Inestrosa NC, Alvarez AR. STI571 prevents apoptosis, tau phosphorylation and behavioural impairments induced by Alzheimer's β-amyloid deposits. Brain 2008; 131:2425-42. [DOI: 10.1093/brain/awn125] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
42
|
Wei J, O’Brien D, Vilgelm A, Piazuelo MB, Correa P, Washington MK, El-Rifai W, Peek RM, Zaika A. Interaction of Helicobacter pylori with gastric epithelial cells is mediated by the p53 protein family. Gastroenterology 2008; 134:1412-23. [PMID: 18343378 PMCID: PMC2430883 DOI: 10.1053/j.gastro.2008.01.072] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Accepted: 01/18/2008] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Although the p53 tumor suppressor has been extensively studied, many critical questions remain unanswered about the biological functions of p53 homologs, p73 and p63. Accumulating evidence suggests that both p73 and p63 play important roles in regulation of apoptosis, cell differentiation, and therapeutic drug sensitivity. METHODS Gastric epithelial cells were cocultured with Helicobacter pylori, and the roles of p63 and p73 proteins were assessed by luciferase reporter, real-time polymerase chain reaction, immunoblotting, and cell survival assays. Short hairpin RNA and dominant-negative mutants were used to inhibit activity of p73 and p63 isoforms. Human and murine gastric tissues were analyzed by immunohistochemistry with p73 and p63 antibodies and modified Steiner's silver method. RESULTS Interaction of H pylori with gastric epithelial cells leads to robust up-regulation of p73 protein in vitro and in vivo in human gastritis specimens and H pylori-infected mice. The p73 increase resulted in up-regulation of pro-apoptotic genes, NOXA, PUMA, and FAS receptor in gastric epithelial cells. Down-regulation of p73 activity suppressed cell death and Fas receptor induced by H pylori. Bacterial virulence factors within the cag pathogenicity island, c-Abl tyrosine kinase, and interaction with p63 isoforms control the activity of p73. CONCLUSION Our findings implicate p73 in H pylori-induced apoptosis and more generally suggest that the p53 family may play a role in the epithelial cell response to H pylori infection.
Collapse
Affiliation(s)
- Jinxiong Wei
- Department of Surgery, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Daniel O’Brien
- Division of Gastroenterology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Anna Vilgelm
- Department of Surgery, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Maria B. Piazuelo
- Division of Gastroenterology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Pelayo Correa
- Division of Gastroenterology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Mary K. Washington
- Department of Pathology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Wael El-Rifai
- Department of Surgery, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
- Department of Cancer Biology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Richard M. Peek
- Department of Surgery, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
- Division of Gastroenterology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
- Department of Cancer Biology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| | - Alexander Zaika
- Department of Surgery, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
- Department of Cancer Biology, Vanderbilt University Medical Center and Vanderbilt-Ingram Cancer Center, Nashville, TN
| |
Collapse
|
43
|
Chuang YT, Fang LW, Lin-Feng MH, Chen RH, Lai MZ. The Tumor Suppressor Death-Associated Protein Kinase Targets to TCR-Stimulated NF-κB Activation. THE JOURNAL OF IMMUNOLOGY 2008; 180:3238-49. [DOI: 10.4049/jimmunol.180.5.3238] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
44
|
Abstract
Tumour necrosis factor-alpha (TNF-alpha) is a cytokine that is involved in many functions, including the inflammatory response, immunity and apoptosis. Some of the responses of TNF-alpha are mediated by caspase-1, which is involved in the production of the pro-inflammatory cytokines interleukin-1beta, interleukin-18 and interleukin-33. The molecular mechanisms involved in TNF-alpha-induced caspase-1 gene expression remain poorly defined, despite the fact that signaling by TNF-alpha has been well studied. The present study was undertaken to investigate the mechanisms involved in the induction of caspase-1 gene expression by TNF-alpha. Treatment of A549 cells with TNF-alpha resulted in an increase in caspase-1 mRNA and protein expression, which was preceded by an increase in interferon regulatory factor-1 and p73 protein levels. Caspase-1 promoter reporter was activated by the treatment of cells with TNF-alpha. Mutation of the interferon regulatory factor-1 binding site resulted in the almost complete loss of basal as well as of TNF-alpha-induced caspase-1 promoter activity. Mutation of the p53/p73 responsive site resulted in reduced TNF-alpha-induced promoter activity. Blocking of p73 function by a dominant negative mutant or by a p73-directed small hairpin RNA reduced basal as well as TNF-alpha-induced caspase-1 promoter activity. TNF-alpha-induced caspase-1 mRNA and protein levels were reduced when p73 mRNA was down-regulated by small hairpin RNA. Caspase-5 gene expression was induced by TNF-alpha, which was inhibited by the small hairpin RNA-mediated down-regulation of p73. Our results show that TNF-alpha induces p73 gene expression, which, together with interferon regulatory factor-1, plays an important role in mediating caspase-1 promoter activation by TNF-alpha.
Collapse
Affiliation(s)
- Nishant Jain
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
45
|
Huang X, Masselli A, Frisch SM, Hunton IC, Jiang Y, Wang JYJ. Blockade of tumor necrosis factor-induced Bid cleavage by caspase-resistant Rb. J Biol Chem 2007; 282:29401-13. [PMID: 17686781 DOI: 10.1074/jbc.m702261200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF) activates caspase-8 to cleave effector caspases or Bid, resulting in type-1 or type-2 apoptosis, respectively. We show here that TNF also induces caspase-8-dependent C-terminal cleavage of the retinoblastoma protein (Rb). Interestingly, fibroblasts from Rb(MI/MI) mice, in which the C-terminal caspase cleavage site is mutated, exhibit a defect in Bid cleavage despite caspase-8 activation. Recent results suggest that TNF receptor endocytosis is required for the activation of caspase-8. Consistent with this notion, inhibition of V-ATPase, which plays an essential role in acidification and degradation of endosomes, specifically restores Bid cleavage in Rb(MI/MI) cells. Inhibition of V-ATPase sensitizes Rb(MI/MI) but not wild-type fibroblasts to TNF-induced apoptosis and stimulates inflammation-associated colonic apoptosis in Rb(MI/MI) but not wild-type mice. These results suggest that Rb cleavage is required for Bid cleavage in TNF-induced type-2 apoptosis, and this requirement can be supplanted by the inhibition of V-ATPase.
Collapse
Affiliation(s)
- XiaoDong Huang
- Division of Biological Sciences, Department of Medicine, University of California, San Diego, La Jolla, California 92093-0820, USA
| | | | | | | | | | | |
Collapse
|
46
|
Chen G, Tai AK, Lin M, Chang F, Terhorst C, Huber BT. Increased proliferation of CD8+ T cells in SAP-deficient mice is associated with impaired activation-induced cell death. Eur J Immunol 2007; 37:663-74. [PMID: 17266174 DOI: 10.1002/eji.200636417] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Defective signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) is responsible for the human X-linked lymphoproliferative syndrome. Defects in T helper 2, natural killer, natural killer T and B cells have been demonstrated in SAP-deficient humans and mice, and increased proliferation of CD8+ T cells has been observed. In the current study, we investigated the properties of CD8+ T cell proliferation and activation-induced cell death (AICD), using OT-I T cell receptor (TCR)-transgenic mice on either wild-type (WT) or SAP-/- background. Interestingly, we found that ovalbumin peptide-activated SAP-/- CD8+ T cells have lower AICD compared to their WT counterparts. Furthermore, the induction of p73, a key mediator of TCR-induced apoptosis through the mitochondrial apoptotic pathway, was significantly reduced at both the mRNA and protein levels in the activated mutant cells. Meanwhile, a reduced level of activated caspase 9 was observed in the mutant cells. We conclude that reduced AICD in activated SAP-/- CD8+ T cells is associated with impaired p73 induction, indicating that the initiation of the mitochondrial apoptotic pathway might be impaired. Our data demonstrate an intrinsic defect in SAP-/- CD8+ T cells and shed light on the increased responsiveness of CD8+ T cells in SAP-/- mice.
Collapse
Affiliation(s)
- Gang Chen
- Department of Pathology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
The retinoblastoma protein RB regulates cell proliferation, differentiation and apoptosis. Homozygous knockout of Rb in mice causes embryonic lethality owing to placental defects that result in excessive apoptosis. RB binds to a number of cellular proteins including the nuclear Abl protein and inhibits its tyrosine kinase activity. Ex vivo experiments have shown that genotoxic or inflammatory stress can activate Abl kinase to stimulate apoptosis. Employing the Rb-null embryos as an in vivo model of apoptosis, we have shown that the genetic ablation of Abl can reduce apoptosis in the developing central nervous system and the embryonic liver. These results are consistent with the inhibitory interaction between RB and Abl, and provide in vivo evidence for the proapoptotic function of Abl.
Collapse
Affiliation(s)
- H L Borges
- Division of Hematology/Oncology, Department of Medicine, Moores Cancer Center, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | |
Collapse
|
48
|
Castro JE, Prada CE, Aguillon RA, Kitada S, Fukuda T, Motta M, Wu C, Dicker F, Sun G, Wang JYJ, Carson DA, Reed JC, Kipps TJ. Thymidine-phosphorothioate oligonucleotides induce activation and apoptosis of CLL cells independently of CpG motifs or BCL-2 gene interference. Leukemia 2006; 20:680-8. [PMID: 16498393 DOI: 10.1038/sj.leu.2404144] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We compared antisense phosphorothioate oligonucleotides (PS-ODN) that target BCL-2 such as Genasense (G3139-PS), with other PS-ODN or phosphodiester-ODN (PO-ODN) in their relative capacity to induce apoptosis of chronic lymphocytic leukemia (CLL) B cells in vitro. Surprisingly, we found that thymidine-containing PS-ODN, but not PO-ODN, induced activation and apoptosis of CLL cells independent of BCL-2 antisense sequence or CpG motifs. All tested thimidine-containing PS-ODN, irrespective of their primary sequences, reduced the expression of Bcl-2 protein and increased the levels of the proapoptotic molecules p53, Bid, Bax in CLL cells. Apoptosis induced by thymidine-containing PS-ODN was preceded by cellular activation, could be blocked by the tyrosine-kinase inhibitor imatinib mesylate (Gleevec), and was dependent on ABL kinase. We conclude that thymidine-containing PS-ODN can activate CLL cells and induce apoptosis via a mechanism that is independent of BCL-2 gene interference or CpG motifs.
Collapse
Affiliation(s)
- J E Castro
- John and Rebecca Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tomasini R, Seux M, Nowak J, Bontemps C, Carrier A, Dagorn JC, Pébusque MJ, Iovanna JL, Dusetti NJ. TP53INP1 is a novel p73 target gene that induces cell cycle arrest and cell death by modulating p73 transcriptional activity. Oncogene 2006; 24:8093-104. [PMID: 16044147 DOI: 10.1038/sj.onc.1208951] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
TP53INP1 is an alternatively spliced gene encoding two nuclear protein isoforms (TP53INP1alpha and TP53INP1beta), whose transcription is activated by p53. When overexpressed, both isoforms induce cell cycle arrest in G1 and enhance p53-mediated apoptosis. TP53INP1s also interact with the p53 gene and regulate p53 transcriptional activity. We report here that TP53INP1 expression is induced during experimental acute pancreatitis in p53-/- mice and in cisplatin-treated p53-/- mouse embryo fibroblasts (MEFs). We demonstrate that ectopic expression of p73, a p53 homologue, leads to TP53INP1 induction in p53-deficient cells. In turn, TP53INP1s alters the transactivation capacity of p73 on several p53-target genes, including TP53INP1 itself, demonstrating a functional association between p73 and TP53INP1s. Also, when overexpressed in p53-deficient cells, TP53INP1s inhibit cell growth and promote cell death as assessed by cell cycle analysis and colony formation assays. Finally, we show that TP53INP1s potentiate the capacity of p73 to inhibit cell growth, that effect being prevented when the p53 mutant R175H is expressed or when p73 expression is blocked by a siRNA. These results suggest that TP53INP1s are functionally associated with p73 to regulate cell cycle progression and apoptosis, independently from p53.
Collapse
Affiliation(s)
- Richard Tomasini
- INSERM U624, Stress Cellulaire, IFR 137-Institut de Cancérologie et Immunologie de Marseille, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Borges HL, Bird J, Wasson K, Cardiff RD, Varki N, Eckmann L, Wang JYJ. Tumor promotion by caspase-resistant retinoblastoma protein. Proc Natl Acad Sci U S A 2005; 102:15587-92. [PMID: 16227443 PMCID: PMC1255734 DOI: 10.1073/pnas.0503925102] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The retinoblastoma (RB) protein regulates cell proliferation and cell death. RB is cleaved by caspase during apoptosis. A mutation of the caspase-cleavage site in the RB C terminus has been made in the mouse Rb-1 locus; the resulting Rb-MI mice are resistant to endotoxin-induced apoptosis in the intestine. The Rb-MI mice do not exhibit increased tumor incidence, because the MI mutation does not disrupt the Rb tumor suppressor function. In this study, we show that Rb-MI can promote the formation of colonic adenomas in the p53-null genetic background. Consistent with this tumor phenotype, Rb-MI reduces colorectal epithelial apoptosis and ulceration caused by dextran sulfate sodium. By contrast, Rb-MI does not affect the lymphoma phenotype of p53-null mice, in keeping with its inability to protect thymocytes and splenocytes from apoptosis. The Rb-MI protein is expressed and phosphorylated in the tumors, thereby inactivating its growth suppression function. These results suggest that RB tumor suppressor function, i.e., inhibition of proliferation, is inactivated by phosphorylation, whereas RB tumor promoting function, i.e., inhibition of apoptosis, is inactivated by caspase cleavage.
Collapse
Affiliation(s)
- Helena L Borges
- Division of Hematology/Oncology, Moores Cancer Center, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|