101
|
Marima R, Hull R, Penny C, Dlamini Z. Mitotic syndicates Aurora Kinase B (AURKB) and mitotic arrest deficient 2 like 2 (MAD2L2) in cohorts of DNA damage response (DDR) and tumorigenesis. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2021; 787:108376. [PMID: 34083040 DOI: 10.1016/j.mrrev.2021.108376] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/05/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022]
Abstract
Aurora Kinase B (AURKB) and Mitotic Arrest Deficient 2 Like 2 (MAD2L2) are emerging anticancer therapeutic targets. AURKB and MAD2L2 are the least well studied members of their protein families, compared to AURKA and MAD2L1. Both AURKB and MAD2L2 play a critical role in mitosis, cell cycle checkpoint, DNA damage response (DDR) and normal physiological processes. However, the oncogenic roles of AURKB and MAD2L2 in tumorigenesis and genomic instability have also been reported. DDR acts as an arbitrator for cell fate by either repairing the damage or directing the cell to self-destruction. While there is strong evidence of interphase DDR, evidence of mitotic DDR is just emerging and remains largely unelucidated. To date, inhibitors of the DDR components show effective anti-cancer roles. Contrarily, long-term resistance towards drugs that target only one DDR target is becoming a challenge. Targeting interactions between protein-protein or protein-DNA holds prominent therapeutic potential. Both AURKB and MAD2L2 play critical roles in the success of mitosis and their emerging roles in mitotic DDR cannot be ignored. Small molecule inhibitors for AURKB are in clinical trials. A few lead compounds towards MAD2L2 inhibition have been discovered. Targeting mitotic DDR components and their interaction is emerging as a potent next generation anti-cancer therapeutic target. This can be done by developing small molecule inhibitors for AURKB and MAD2L2, thereby targeting DDR components as anti-cancer therapeutic targets and/or targeting mitotic DDR. This review focuses on AURKB and MAD2L2 prospective synergy to deregulate the p53 DDR pathway and promote favourable conditions for uncontrolled cell proliferation.
Collapse
Affiliation(s)
- Rahaba Marima
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa.
| | - Rodney Hull
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa
| | - Clement Penny
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Parktown, 2193, South Africa
| | - Zodwa Dlamini
- SA-MRC/UP Precision Prevention and Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Pan African Cancer Research Institute, Faculty of Health Sciences, University of Pretoria, Hatfield, 0028, South Africa
| |
Collapse
|
102
|
Kastyro IV, Reshetov IV, Khamidulin GV, Shilin SS, Torshin VI, Kostyaeva MG, Popadyuk VI, Yunusov TY, Shmaevsky PE, Shalamov KP, Kupryakova AD, Doroginskaya ES, Sedelnikova AD. Influence of Surgical Trauma in the Nasal Cavity on the Expression of p53 Protein in the Hippocampus of Rats. DOKL BIOCHEM BIOPHYS 2021; 497:99-103. [PMID: 33895923 DOI: 10.1134/s160767292102006x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 11/23/2022]
Abstract
The aim of the study was to determine the role of the stress effect of septoplasty modeling on p53 protein expression in the hippocampus of rats under conditions of sensory olfactory deprivation. Simulation of septoplasty was carried out on 30 sexually mature male rats. A quantitative assessment of the apoptosis of neurons in the pyramidal layer of the hippocampus in the subfields CA1, CA2, CA3, and dentate gyrus (DG) on days 2, 4, and 6 after surgery was carried out. Histological sections were stained by the immunohistochemical method with antibodies to the p53 protein. An increase in the number of p53-positive neurons was noted in all subfields; the maximum increase in the number of apoptotic neurons was noted on day 4 after surgery. The stress effect of modeling septoplasty in rats, accompanied by sensory deprivation of the peripheral part of the olfactory analyzer, provoked the expression of p53 and the initiation of apoptosis mechanisms in various subfields of the hippocampus.
Collapse
Affiliation(s)
- I V Kastyro
- Peoples' Friendship University of Russia, Moscow, Russia.
| | - I V Reshetov
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - G V Khamidulin
- Peoples' Friendship University of Russia, Moscow, Russia
| | - S S Shilin
- Peoples' Friendship University of Russia, Moscow, Russia
| | - V I Torshin
- Peoples' Friendship University of Russia, Moscow, Russia
| | - M G Kostyaeva
- Peoples' Friendship University of Russia, Moscow, Russia
| | - V I Popadyuk
- Peoples' Friendship University of Russia, Moscow, Russia
| | - T Y Yunusov
- City Clinical Hospital No. 40 of the Moscow Department of Health, Moscow, Russia
| | - P E Shmaevsky
- Peoples' Friendship University of Russia, Moscow, Russia
| | - K P Shalamov
- Peoples' Friendship University of Russia, Moscow, Russia
| | - A D Kupryakova
- Peoples' Friendship University of Russia, Moscow, Russia
| | | | | |
Collapse
|
103
|
Gupta A, Storey KB. Coordinated expression of Jumonji and AHCY under OCT transcription factor control to regulate gene methylation in wood frogs during anoxia. Gene 2021; 788:145671. [PMID: 33887369 DOI: 10.1016/j.gene.2021.145671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022]
Abstract
Wood frogs (Rana sylvatica) can survive extended periods of whole body freezing. Freezing imparts multiple stresses on cells that include anoxia and dehydration, but these can also be experienced as independent stresses. Under anoxia stress, energy metabolism is suppressed, and pro-survival pathways are prioritized to differentially regulate some transcription factors including OCT1 and OCT4. Jumonji C domain proteins (JMJD1A and JMJD2C) are hypoxia responsive demethylases whose expression is accelerated by OCT1 and OCT4 which act to demethylate genes related to the methionine cycle. The responses by these factors to 24 h anoxia exposure and 4 h aerobic recovery was analyzed in liver and skeletal muscle of wood frogs to assess their involvement in metabolic adaptation to oxygen limitation. Immunoblot results showed a decrease in JMJD1A levels under anoxia in liver and muscle, but an increase was observed in JMJD2C demethylase protein in anoxic skeletal muscle. Protein levels of adenosylhomocysteinase (AHCY) and methionine adenosyl transferase (MAT), enzymes of the methionine cycle, also showed an increase in the reoxygenated liver, whereas the levels decreased in muscle. A transcription factor ELISA showed a decrease in DNA binding by OCT1 in the reoxygenated liver and anoxic skeletal muscle, and transcript levels also showed tissue specific gene expression. The present study provides the first analysis of the role of the OCT1 transcription factor, associated proteins, and lysine demethylases in mediating responses to anoxia by wood frog tissues.
Collapse
Affiliation(s)
- Aakriti Gupta
- Department of Biology, Carleton University, Ottawa K1S 5B6, Canada
| | - Kenneth B Storey
- Department of Biology, Carleton University, Ottawa K1S 5B6, Canada.
| |
Collapse
|
104
|
Chatterjee M, Viswanathan P. Long noncoding RNAs in the regulation of p53‐mediated apoptosis in human cancers. Cell Biol Int 2021. [DOI: https://doi.org/10.1002/cbin.11597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Manjima Chatterjee
- School of Bio Sciences and Technology, Vellore Institute of Technology Vellore Tamil Nadu India
| | - Pragasam Viswanathan
- School of Bio Sciences and Technology, Vellore Institute of Technology Vellore Tamil Nadu India
| |
Collapse
|
105
|
Toy HI, Karakülah G, Kontou PI, Alotaibi H, Georgakilas AG, Pavlopoulou A. Investigating Molecular Determinants of Cancer Cell Resistance to Ionizing Radiation Through an Integrative Bioinformatics Approach. Front Cell Dev Biol 2021; 9:620248. [PMID: 33898418 PMCID: PMC8058375 DOI: 10.3389/fcell.2021.620248] [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: 10/22/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Eradication of cancer cells through exposure to high doses of ionizing radiation (IR) is a widely used therapeutic strategy in the clinical setting. However, in many cases, cancer cells can develop remarkable resistance to radiation. Radioresistance represents a prominent obstacle in the effective treatment of cancer. Therefore, elucidation of the molecular mechanisms and pathways related to radioresistance in cancer cells is of paramount importance. In the present study, an integrative bioinformatics approach was applied to three publicly available RNA sequencing and microarray transcriptome datasets of human cancer cells of different tissue origins treated with ionizing radiation. These data were investigated in order to identify genes with a significantly altered expression between radioresistant and corresponding radiosensitive cancer cells. Through rigorous statistical and biological analyses, 36 genes were identified as potential biomarkers of radioresistance. These genes, which are primarily implicated in DNA damage repair, oxidative stress, cell pro-survival, and apoptotic pathways, could serve as potential diagnostic/prognostic markers cancer cell resistance to radiation treatment, as well as for therapy outcome and cancer patient survival. In addition, our findings could be potentially utilized in the laboratory and clinical setting for enhancing cancer cell susceptibility to radiation therapy protocols.
Collapse
Affiliation(s)
- Halil Ibrahim Toy
- Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Panagiota I Kontou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Hani Alotaibi
- Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou, National Technical University of Athens, Athens, Greece
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| |
Collapse
|
106
|
Jaiswara PK, Gupta VK, Sonker P, Rawat SG, Tiwari RK, Pathak C, Kumar S, Kumar A. Nimbolide induces cell death in T lymphoma cells: Implication of altered apoptosis and glucose metabolism. ENVIRONMENTAL TOXICOLOGY 2021; 36:628-641. [PMID: 33274819 DOI: 10.1002/tox.23067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 11/21/2020] [Indexed: 06/12/2023]
Abstract
Nimbolide is a tetranortriterpenoid derived from the leaves and flowers of Azadirachta indica (Neem). It exhibits anticancer activity against a variety of cancers by modulating various crucial features, including cell proliferation, apoptosis, and invasion and metastasis. More importantly, the cytotoxic effect of nimbolide has also been observed against T cell lymphoma, but the underlying mechanisms are still unexplored. So far, no study has been conducted to observe the effect of nimbolide on cancer cell metabolism. Therefore, the present investigation was designed to explore the molecular mechanisms of the antitumor potential of nimbolide against T cell lymphoma, a neoplastic disorder of thymic origin. In addition, we also unraveled the anti-glycolytic activity of nimbolide against T lymphoma cells with possible molecular mechanisms. Our results showed the cytotoxic action of nimbolide against three different cell lines of T cell lymphoma, namely Dalton's lymphoma, HuT-78, and J6. Nimbolide-induced apoptosis in T lymphoma cells by altering the level of reactive oxygen species, p53, Bcl2, Bax, and cytochrome c, with subsequent cleavage of caspase 3. Remarkably, nimbolide inhibited the expression of hypoxia-inducible factor-1α, glucose transporter 3, hexokinase II, and pyruvate dehydrogenase kinase 1, which led to the suppression of glycolysis with concomitant activation of oxidative phosphorylation. Hence, the results of the present investigation demonstrate that nimbolide exerts tumoricidal activity against T lymphoma cells via augmentation of apoptosis and reversal of altered cell metabolism. Thus, the present study provides a new insight for the therapeutic utilization of nimbolide against T cell lymphoma.
Collapse
Affiliation(s)
- Pradip Kumar Jaiswara
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Vishal Kumar Gupta
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Pratishtha Sonker
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Shiv Govind Rawat
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Rajan Kumar Tiwari
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Chandramani Pathak
- Amity Institute of Biotechnology, Amity University, Amity Education Valley, Gurgaon, Haryana, India
| | - Santosh Kumar
- Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Ajay Kumar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
107
|
Hairat S, Zaki M. Half sandwiched RutheniumII complexes: En Route towards the targeted delivery by Human Serum Albumin (HSA). J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
108
|
Molani Gol R, Kheirouri S. The Effects of Quercetin on the Apoptosis of Human Breast Cancer Cell Lines MCF-7 and MDA-MB-231: A Systematic Review. Nutr Cancer 2021; 74:405-422. [PMID: 33682528 DOI: 10.1080/01635581.2021.1897631] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This systematic review was performed with a focus on the effects of quercetin (QT) on the human breast cancer cell lines MCF-7 and MDA-MB-231. PubMed, Scopus, Science Direct, and Google Scholar databases were searched up to May 2020 using relevant keywords. All articles written in English evaluating the effects of QT on the human breast cancer cell lines MCF-7 and/or MDA-MB-231 were eligible for the review. Totally, 31 articles were included in this review. Out of them, 23 studies investigated the effects of QT on MCF-7 cells and indicated that QT induces apoptosis in the cells. Of 15 studies that examined the effects of QT on MDA-MB-231 cells, 14 reports showed successful apoptosis. It is concluded that QT might be beneficial in the eliminating of breast cancer cells. However, further clinical trials are warranted to further verify these outcomes.
Collapse
Affiliation(s)
- Roghayeh Molani Gol
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
109
|
Life as a Vector of Dengue Virus: The Antioxidant Strategy of Mosquito Cells to Survive Viral Infection. Antioxidants (Basel) 2021; 10:antiox10030395. [PMID: 33807863 PMCID: PMC8000470 DOI: 10.3390/antiox10030395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Dengue fever is a mosquito-borne viral disease of increasing global importance. The disease has caused heavy burdens due to frequent outbreaks in tropical and subtropical areas of the world. The dengue virus (DENV) is generally transmitted between human hosts via the bite of a mosquito vector, primarily Aedes aegypti and Ae. albopictus as a minor species. It is known that the virus needs to alternately infect mosquito and human cells. DENV-induced cell death is relevant to the pathogenesis in humans as infected cells undergo apoptosis. In contrast, mosquito cells mostly survive the infection; this allows infected mosquitoes to remain healthy enough to serve as an efficient vector in nature. Overexpression of antioxidant genes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), glutaredoxin (Grx), thioredoxin (Trx), and protein disulfide isomerase (PDI) have been detected in DENV2-infected mosquito cells. Additional antioxidants, including GST, eukaryotic translation initiation factor 5A (eIF5a), and p53 isoform 2 (p53-2), and perhaps some others, are also involved in creating an intracellular environment suitable for cell replication and viral infection. Antiapoptotic effects involving inhibitor of apoptosis (IAP) upregulation and subsequent elevation of caspase-9 and caspase-3 activities also play crucial roles in the ability of mosquito cells to survive DENV infection. This article focused on the effects of intracellular responses in mosquito cells to infection primarily by DENVs. It may provide more information to better understand virus/cell interactions that can possibly elucidate the evolutionary pathway that led to the mosquito becoming a vector.
Collapse
|
110
|
Sena-dos-Santos C, Braga-da-Silva C, Marques D, Azevedo dos Santos Pinheiro J, Ribeiro-dos-Santos Â, Cavalcante GC. Unraveling Cell Death Pathways during Malaria Infection: What Do We Know So Far? Cells 2021; 10:479. [PMID: 33672278 PMCID: PMC7926694 DOI: 10.3390/cells10020479] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Malaria is a parasitic disease (caused by different Plasmodium species) that affects millions of people worldwide. The lack of effective malaria drugs and a vaccine contributes to this disease, continuing to cause major public health and socioeconomic problems, especially in low-income countries. Cell death is implicated in malaria immune responses by eliminating infected cells, but it can also provoke an intense inflammatory response and lead to severe malaria outcomes. The study of the pathophysiological role of cell death in malaria in mammalians is key to understanding the parasite-host interactions and design prophylactic and therapeutic strategies for malaria. In this work, we review malaria-triggered cell death pathways (apoptosis, autophagy, necrosis, pyroptosis, NETosis, and ferroptosis) and we discuss their potential role in the development of new approaches for human malaria therapies.
Collapse
Affiliation(s)
- Camille Sena-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Cíntia Braga-da-Silva
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Diego Marques
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Jhully Azevedo dos Santos Pinheiro
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Ândrea Ribeiro-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
- Programa de Pós-Graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66.075-110, Brazil
| | - Giovanna C. Cavalcante
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| |
Collapse
|
111
|
Huang S, Zhang L, Xu M, Li C, Fu H, Huang J, Jin X, Liang S, Wang H. Co-Delivery of 131 I and Prima-1 by Self-Assembled CD44-Targeted Nanoparticles for Anaplastic Thyroid Carcinoma Theranostics. Adv Healthc Mater 2021; 10:e2001029. [PMID: 33326188 DOI: 10.1002/adhm.202001029] [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: 06/18/2020] [Revised: 08/22/2020] [Indexed: 12/20/2022]
Abstract
New radionuclide-labeled targeting nanocarrier systems have generated new opportunities for tumor treatment and imaging. Nevertheless, such therapeutic strategy is clinically unfeasible on anaplastic thyroid carcinoma (ATC) patients, because of lacking suitable targets and resistance to radiation. In order to figure out a potential treatment, immuno-histochemical staining is performed in human ATC tissue species and high expression of cluster determinant 44 (CD44) is found. Therefore, a CD44-targeted delivery system is designed and constructed by self-assembly of tyrosine (Tyr)-hyaluronic acid (HA)-polyethyleneimine (PEI), which can radiolabel 131/125 I and load a p53 mutant restoring regent, Prima-1. The 125 I-labeled nanocomposites display an impressive tumor imaging as well as a long radiation treatment cycle. The 131 I-labeled nanoparticles show remarkable anti ATC-tumor effects in vitro and in vivo, due to radiosensitization of Prima-1 by reactivation of the p53 mutants.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Linlin Zhang
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Miaomiao Xu
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Chao Li
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Hongliang Fu
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Jie Huang
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Xin Jin
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Sheng Liang
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| | - Hui Wang
- Department of Nuclear Medicine Xin Hua Hospital Affiliated To Shanghai Jiao Tong University School 1665 Kongjiang Road Shanghai 200092 China
| |
Collapse
|
112
|
Abdelaal MR, Soror SH, Elnagar MR, Haffez H. Revealing the Potential Application of EC-Synthetic Retinoid Analogues in Anticancer Therapy. Molecules 2021; 26:506. [PMID: 33477997 PMCID: PMC7835894 DOI: 10.3390/molecules26020506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background and Aim: All-trans retinoic acid (ATRA) induces differentiation and inhibits growth of many cancer cells. However, resistance develops rapidly prompting the urgent need for new synthetic and potent derivatives. EC19 and EC23 are two synthetic retinoids with potent stem cell neuro-differentiation activity. Here, these compounds were screened for their in vitro antiproliferative and cytotoxic activity using an array of different cancer cell lines. (2) Methods: MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, AV/PI (annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI)), cell cycle analysis, immunocytochemistry, gene expression analysis, Western blotting, measurement of glutamate and total antioxidant concentrations were recruited. (3) Results: HepG2, Caco-2, and MCF-7 were the most sensitive cell lines; HepG2 (ATRA; 36.2, EC19; 42.2 and EC23; 0.74 µM), Caco-2 (ATRA; 58.0, EC19; 10.8 and EC23; 14.7 µM) and MCF-7 (ATRA; 99.0, EC19; 9.4 and EC23; 5.56 µM). Caco-2 cells were selected for further biochemical investigations. Isobologram analysis revealed the combined synergistic effects with 5-fluorouracil with substantial reduction in IC50. All retinoids induced apoptosis but EC19 had higher potency, with significant cell cycle arrest at subG0-G1, -S and G2/M phases, than ATRA and EC23. Moreover, EC19 reduced cellular metastasis in a transwell invasion assay due to overexpression of E-cadherin, retinoic acid-induced 2 (RAI2) and Werner (WRN) genes. (4) Conclusion: The present study suggests that EC-synthetic retinoids, particularly EC19, can be effective, alone or in combinations, for potential anticancer activity to colorectal cancer. Further in vivo studies are recommended to pave the way for clinical applications.
Collapse
Affiliation(s)
- Mohamed R. Abdelaal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Sameh H. Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Mohamed R. Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt;
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt; (M.R.A.); (S.H.S.)
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
113
|
Khodavirdipour A, Piri M, Jabbari S, Keshavarzi S, Safaralizadeh R, Alikhani MY. Apoptosis Detection Methods in Diagnosis of Cancer and Their Potential Role in Treatment: Advantages and Disadvantages: a Review. J Gastrointest Cancer 2021; 52:422-430. [PMID: 33392962 DOI: 10.1007/s12029-020-00576-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Interruption of regulation of apoptosis can play a leading role in cancers where elevated apoptosis causes neurodegeneration, autoimmunity, AIDS, and ischemia. One famous example can be p53's downregulation, which is a tumor suppressor gene, which consequently can cause a decrease in apoptosis rate and intense tumor growth and progression and development and inactivation of 53; it can be extended to many cancers in human. Anyhow, apoptosis is a double-edge sword. There are many trials and studies are going on observation and understanding of different steps involved in apoptosis. Apoptosis has a very major role in carcinogenesis and the treatment of cancer. AIM In this updated-cum-comprehensive review, we would like to cover what is apoptosis and cancer and also, will discuss all known methods of apoptosisdetection, their applicability in the treatment of cancer, and their advantages, disadvantages, and limitations. MATERIAL AND METHODS Published articles on indexing sources such as PubMed, Scopus from 2000 to date. RESULT By considering all above information including each methods pros and cons, these routine methods could be great tool with distinctive qualities in treatmentwhich can be great help from patient perspective and as well from government ad health care system point of view. CONCLUSION Accurate diagnosis of cell apoptotic biopathways at different stages assists in evaluating near to exact apoptotic index, which is the perfect sign andindicator for metastasis and also prognosis, thus foreseeing treatment outcome.
Collapse
Affiliation(s)
- Amir Khodavirdipour
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.,Division of Human Genetics, Department of Anatomy, St. John's Hospital, Bangalore, India
| | - Motahareh Piri
- Department of Biology, Faculty of Basic Sciences, University of Zabol, Zabol, Iran
| | - Sarvin Jabbari
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Shiva Keshavarzi
- Department of Biology, Faculty of Basic Sciences, University of Zabol, Zabol, Iran
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
114
|
Abstract
Chemical compounds induce cytotoxicity by various mechanisms, including interference in membrane integrity, metabolism, cellular component degradation or release, and cell division. Between the classic death pathways, namely, autophagy, apoptosis, and necrosis, apoptosis have been in the focus for the last several years as an important pathway for the toxicity of different types of xenobiotics. Because of that, having the tools to evaluate it is key for understanding and explaining the toxicodynamics of different classes of substances. Here, we describe a wide array of classic assays that can be easily implemented to evaluate apoptosis induction.
Collapse
Affiliation(s)
- Lilian Cristina Pereira
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Department of Bioprocesses and Biotechnology, Faculty of Agronomic Sciences of Botucatu, São Paulo State University, Botucatu, SP, Brazil
- Center for Evaluation of Environmental Impact on Human Health (TOXICAM), Botucatu, São Paulo, Brazil
| | - Alecsandra Oliveira de Souza
- Federal Institute of Science and Technology Education of Rondônia-Campus Porto Velho Calama, Porto Velho, RO, Brazil
- FFCLRP-USP, Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Raul Ghiraldelli Miranda
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniel Junqueira Dorta
- FFCLRP-USP, Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
- Instituto Nacional de Tecnologias Alternativas de Detecção, Avaliação Toxicológica e Remoção de Micropututantes e Radioativos (INCT-DATREM), Unesp, Instituto de Química, Caixa Postal 355, CEP: 14800-900, Araraquara, SP, Brazil.
| |
Collapse
|
115
|
Joshi A, Farber K, Scheiber IF. Neurotoxicity of copper and copper nanoparticles. ADVANCES IN NEUROTOXICOLOGY 2021:115-157. [DOI: 10.1016/bs.ant.2020.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
116
|
Duan Y, Bai H, Li X, Wang D, Wang Y, Cao M, Zhang N, Chen H, Wang Y. Oncolytic Adenovirus H101 Synergizes with Radiation in Cervical Cancer Cells. Curr Cancer Drug Targets 2021; 21:619-630. [PMID: 33687882 DOI: 10.2174/1568009621666210308103541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/13/2021] [Accepted: 01/31/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND A major challenge in cervical cancer radiotherapy is tailoring the radiation doses efficiently to eliminate malignant cells and reduce the side effects in normal tissues. Oncolytic adenovirus drug H101 was recently tested and approved as a topical adjuvant treatment for several malignancies. OBJECTIVE This study aimed to evaluate the potential neoadjuvant radiotherapy benefits of H101 by testing the inhibitory function of H101 in combination with radiation in different cervical cancer cells. METHODS Human cervical cancer cell lines C33a, SiHa, CaSki, and HeLa were treated with varying concentrations of H101 alone or in combination with radiation (2 Gy or 4 Gy). Cell viability and apoptosis were measured at the indicated time intervals. HPV16 E6 and cellular p53 mRNA expression alteration was measured by qRT-PCR. In situ RNA scope was used to determine HPV E6 status. P53 protein alterations were detected by Western blot. RESULTS Cell viability and apoptosis assays revealed that the combination of a high dose of H101 (MOI=1000, 10000) with radiation yielded a synergistic anticancer effect in all tested cervical cancer cell lines (P<0.05), with the greatest effect achieved in HPV-negative C33a cells (P<0.05). Low-HPV16-viral-load SiHa cells were more sensitive to the combination therapy than high-HPV16- viral-load CaSki cells (P<0.05). The combined treatment reduced HPV16 E6 expression and increased cellular P53 levels compared to those observed with radiation alone in SiHa and CaSki cells (P<0.05). CONCLUSION Oncolytic adenovirus H101 effectively enhances the antitumor efficacy of radiation in cervical cancer cells and may serve as a novel combination therapy for cervical cancer.
Collapse
Affiliation(s)
- Yixin Duan
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Haixia Bai
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Xiang Li
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Depu Wang
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Ying Wang
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Meng Cao
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Nana Zhang
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Hongwei Chen
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Yili Wang
- Department of Radiation Oncology, Institute of Molecular Radiobiology of Cancer, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| |
Collapse
|
117
|
Jang HJ, Yang JH, Hong E, Jo E, Lee S, Lee S, Choi JS, Yoo HS, Kang H. Chelidonine Induces Apoptosis via GADD45a-p53 Regulation in Human Pancreatic Cancer Cells. Integr Cancer Ther 2021; 20:15347354211006191. [PMID: 33884928 PMCID: PMC8077490 DOI: 10.1177/15347354211006191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chelidonium majus has been used as a traditional medicine in China and western countries for various diseases, including inflammation and cancer. However, the anti-cancer effect of chelidonine, a major compound of C. majus extracts, on pancreatic cancer remains poorly understood. In this study, we found that treatment with chelidonine inhibited proliferation of BxPC-3 and MIA PaCa-2 human pancreatic cancer cells. Annexin-V/propidium iodide staining assay showed that this growth inhibitory effect of chelidonine was induced through apoptosis. We found that chelidonine treatment upregulated mRNA levels and transcription factor activity in both cell lines. Increases in protein expression levels of p53, GADD45A, p21 and cleaved caspase-3 were also observed, with more distinct changes in MIA PaCa-2 cells compared to the BxPC-3 cells. These results suggest that chelidonine induces pancreatic cancer apoptosis through the p53 and GADD45A pathways. Our findings provide new insights into the use of chelidonine for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hyun-Jin Jang
- Korea Basic Science Institute, Daejeon,
Republic of Korea
- Sungkyunkwan University, Suwon,
Republic of Korea
| | - Jae Ho Yang
- Daejeon Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
| | - Eunmi Hong
- Korea Basic Science Institute, Daejeon,
Republic of Korea
| | - Eunbi Jo
- Korea Basic Science Institute, Daejeon,
Republic of Korea
- Hanyang University, Seoul, Republic of
Korea
| | - Soon Lee
- Korea Basic Science Institute, Daejeon,
Republic of Korea
- University of Science and Technology,
Daejeon, Republic of Korea
| | - Sanghun Lee
- Korea Institute of Oriental Medicine,
Daejeon, Republic of Korea
| | - Jong Soon Choi
- Korea Basic Science Institute, Daejeon,
Republic of Korea
| | - Hwa Seung Yoo
- Daejeon Korean Medicine Hospital of
Daejeon University, Seoul, Republic of Korea
- Hwa Seung Yoo, East West Cancer Center,
Seoul Korean Medicine Hospital of Daejeon University, Seoul 05836, Rep. of
Korea.
| | - Hyuno Kang
- Korea Basic Science Institute, Daejeon,
Republic of Korea
- Hyuno Kang, Division of Analytical Science,
Korea Basic Science Institute, 169-148, Gwahak-ro, Yuseong-gu, Daejeon 34133,
Republic of Korea.
| |
Collapse
|
118
|
He H, Lin X, Wu D, Wang J, Guo J, Green DR, Zhang H, Xu B. Enzymatic Noncovalent Synthesis for Mitochondrial Genetic Engineering of Cancer Cells. CELL REPORTS. PHYSICAL SCIENCE 2020; 1:100270. [PMID: 33511360 PMCID: PMC7839975 DOI: 10.1016/j.xcrp.2020.100270] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Since mitochondria contribute to tumorigenesis and drug resistance in cancer, mitochondrial genetic engineering promises a new direction for cancer therapy. Here, we report the use of the perimitochondrial enzymatic noncovalent synthesis (ENS) of peptides for delivering genes selectively into the mitochondria of cancer cells for mitochondrial genetic engineering. Specifically, the micelles of peptides bind to the voltage-dependent anion channel (VDAC) on mitochondria for the proteolysis by enterokinase (ENTK), generating perimitochondrial nanofibers in cancer cells. This process, facilitating selective delivery of nucleic acid or gene vectors into mitochondria of cancer cells, enables the mitochondrial transgene expression of CRISPR/Cas9, FUNDC1, p53, and fluorescent proteins. Mechanistic investigation indicates that the interaction of the peptide assemblies with the VDAC and mitochondrial membrane potential are necessary for mitochondria targeting. This local enzymatic control of intermolecular noncovalent interactions enables selective mitochondrial genetic engineering, thus providing a strategy for targeting cancer cells.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Xinyi Lin
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Difei Wu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Jiaqing Wang
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
| | - Douglas R. Green
- Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| | - Hongwei Zhang
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Avenue, Boston, MA 02115, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St., Waltham, MA 02454, USA
- Lead contact
- Correspondence:
| |
Collapse
|
119
|
Wells N, Quigley J, Pascua J, Pinkowski N, Almaiman L, Brasser SM, Hong MY. Effects of low-to-moderate ethanol consumption on colonic growth and gene expression in young adult and middle-aged male rats. PLoS One 2020; 15:e0243499. [PMID: 33326448 PMCID: PMC7743962 DOI: 10.1371/journal.pone.0243499] [Citation(s) in RCA: 4] [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: 07/14/2020] [Accepted: 11/22/2020] [Indexed: 01/24/2023] Open
Abstract
Excessive alcohol consumption is a risk factor associated with colorectal cancer; however, some epidemiological studies have reported that moderate alcohol consumption may not contribute additional risk or may provide a protective effect reducing colorectal cancer risk. Prior research highlights the importance of proliferation, differentiation, and apoptosis as parameters to consider when evaluating colonic cell growth and tumorigenesis. The present study investigated whether chronic low-to-moderate ethanol consumption altered these parameters of colonic cell growth and expression of related genes. Twenty-four nondeprived young adult (109 days old) and 24 nondeprived middle-aged (420 days old) Wistar rats were randomly assigned to an ethanol-exposed or a water control group (n = 12/group). The ethanol group was provided voluntary access to a 20% v/v ethanol solution on alternate days for 13 weeks. Colon tissues were collected for quantitative immunohistochemical analyses of cell proliferation, differentiation and apoptosis using Ki-67, goblet cell and TUNEL, respectively. Gene expression of cyclin D1 (Ccnd1), Cdk2, Cdk4, p21waf1/cip1 (Cdkn1a), E-cadherin (Cdh1) and p53 were determined by quantitative real-time polymerase chain reaction in colonic scraped mucosa. Ethanol treatment resulted in a lower cell proliferation index and proliferative zone, and lower Cdk2 expression in both age groups, as well as trends toward lower Ccnd1 and higher Cdkn1a expression. Cell differentiation was modestly but significantly reduced by ethanol treatment only in older animals. Overall, older rats showed decreases in apoptosis and gene expression of Cdk4, Cdh1, and p53 compared to younger rats, but there was no observed effect of ethanol exposure on these measures. These findings suggest that low-to-moderate ethanol consumption improves at least one notable parameter in colonic tumorigenesis (cell proliferation) and associated gene expression regardless of age, however, selectively decreased cell differentiation among older subjects.
Collapse
Affiliation(s)
- Nicole Wells
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States of America
| | - Jacqueline Quigley
- Department of Psychology, San Diego State University, San Diego, CA, United States of America
| | - Jeremy Pascua
- Department of Psychology, San Diego State University, San Diego, CA, United States of America
| | - Natalie Pinkowski
- Department of Psychology, San Diego State University, San Diego, CA, United States of America
| | - Lama Almaiman
- Department of Psychology, San Diego State University, San Diego, CA, United States of America
| | - Susan M. Brasser
- Department of Psychology, San Diego State University, San Diego, CA, United States of America
| | - Mee Young Hong
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, CA, United States of America
| |
Collapse
|
120
|
Jung EJ, Lee WS, Paramanantham A, Kim HJ, Shin SC, Kim GS, Jung JM, Ryu CH, Hong SC, Chung KH, Kim CW. p53 Enhances Artemisia annua L. Polyphenols-Induced Cell Death Through Upregulation of p53-Dependent Targets and Cleavage of PARP1 and Lamin A/C in HCT116 Colorectal Cancer Cells. Int J Mol Sci 2020; 21:ijms21239315. [PMID: 33297377 PMCID: PMC7730414 DOI: 10.3390/ijms21239315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022] Open
Abstract
Plant-derived natural polyphenols exhibit anticancer activity without showing any noticeable toxicities to normal cells. The aim of this study was to investigate the role of p53 on the anticancer effect of polyphenols isolated from Korean Artemisia annua L. (pKAL) in HCT116 human colorectal cancer cells. We confirmed that pKAL induced reactive oxygen species (ROS) production, propidium iodide (PI) uptake, nuclear structure change, and acidic vesicles in a p53-independent manner in p53-null HCT116 cells through fluorescence microscopy analysis of DCF/PI-, DAPI-, and AO-stained cells. The pKAL-induced anticancer effects were found to be significantly higher in p53-wild HCT116 cells than in p53-null by hematoxylin staining, CCK-8 assay, Western blot, and flow cytometric analysis of annexin V/PI-stained cells. In addition, expression of ectopic p53 in p53-null cells was upregulated by pKAL in both the nucleus and cytoplasm, increasing pKAL-induced cell death. Moreover, Western bot analysis revealed that pKAL-induced cell death was associated with upregulation of p53-dependent targets such as p21, Bax and DR5 and cleavage of PARP1 and lamin A/C in p53-wild HCT116 cells, but not in p53-null. Taken together, these results indicate that p53 plays an important role in enhancing the anticancer effects of pKAL by upregulating p53 downstream targets and inducing intracellular cell death processes.
Collapse
Affiliation(s)
- Eun Joo Jung
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea; (E.J.J.); (C.W.K.)
| | - Won Sup Lee
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
- Correspondence: ; Tel.: +82-55-750-8733; Fax: +82-55-758-9122
| | - Anjugam Paramanantham
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Hye Jung Kim
- Departments of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Jin-Myung Jung
- Departments of Neurosurgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Chung Ho Ryu
- Department of Food Technology, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Soon Chan Hong
- Departments of Surgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Ky Hyun Chung
- Departments of Urology, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Choong Won Kim
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea; (E.J.J.); (C.W.K.)
| |
Collapse
|
121
|
Fernández-Ortiz M, Sayed RKA, Fernández-Martínez J, Cionfrini A, Aranda-Martínez P, Escames G, de Haro T, Acuña-Castroviejo D. Melatonin/Nrf2/NLRP3 Connection in Mouse Heart Mitochondria during Aging. Antioxidants (Basel) 2020; 9:antiox9121187. [PMID: 33260800 PMCID: PMC7760557 DOI: 10.3390/antiox9121187] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/14/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Aging is a major risk for cardiovascular diseases (CVD). Age-related disorders include oxidative stress, mitochondria dysfunction, and exacerbation of the NF-κB/NLRP3 innate immune response pathways. Some of the molecular mechanisms underlying these processes, however, remain unclear. This study tested the hypothesis that NLRP3 inflammasome plays a role in cardiac aging and melatonin is able to counteract its effects. With the aim of investigating the impact of NLRP3 inflammasome and the actions and target of melatonin in aged myocardium, we analyzed the expression of proteins implied in mitochondria dynamics, autophagy, apoptosis, Nrf2-dependent antioxidant response and mitochondria ultrastructure in heart of wild-type and NLRP3-knockout mice of 3, 12, and 24 months-old, with and without melatonin treatment. Our results showed that the absence of NLRP3 prevented age-related mitochondrial dynamic alterations in cardiac muscle with minimal effects in cardiac autophagy during aging. The deficiency of the inflammasome affected Bax/Bcl2 ratio, but not p53 or caspase 9. The Nrf2-antioxidant pathway was also unaffected by the absence of NLRP3. Furthermore, NLRP3-deficiency prevented the drop in autophagy and mice showed less mitochondrial damage than wild-type animals. Interestingly, melatonin treatment recovered mitochondrial dynamics altered by aging and had few effects on cardiac autophagy. Melatonin supplementation also had an anti-apoptotic action in addition to restoring Nrf2-antioxidant capacity and improving mitochondria ultrastructure altered by aging.
Collapse
Affiliation(s)
- Marisol Fernández-Ortiz
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
| | - Ramy K. A. Sayed
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
| | - José Fernández-Martínez
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
| | - Antonia Cionfrini
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
| | - Paula Aranda-Martínez
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
| | - Germaine Escames
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
- CIBERfes, Ibs. Granada, 18016 Granada, Spain
| | - Tomás de Haro
- UGC de Laboratorios Clínicos, Hospital Universitario San Cecilio, 18016 Granada, Spain;
| | - Darío Acuña-Castroviejo
- Centro de Investigación Biomédica, Departamento de Fisiología, Facultad de Medicina, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, 18016 Granada, Spain; (M.F.-O.); (R.K.A.S.); (J.F.-M.); (A.C.); (P.A.-M.); (G.E.)
- CIBERfes, Ibs. Granada, 18016 Granada, Spain
- UGC de Laboratorios Clínicos, Hospital Universitario San Cecilio, 18016 Granada, Spain;
- Correspondence: ; Tel.: +34-958-241-000 (ext. 20169)
| |
Collapse
|
122
|
Soond SM, Savvateeva LV, Makarov VA, Gorokhovets NV, Townsend PA, Zamyatnin AA. Making Connections: p53 and the Cathepsin Proteases as Co-Regulators of Cancer and Apoptosis. Cancers (Basel) 2020; 12:cancers12113476. [PMID: 33266503 PMCID: PMC7700648 DOI: 10.3390/cancers12113476] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/02/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary This article describes an emerging area of significant interest in cancer and cell death and the relationships shared by these through the p53 and cathepsin proteins. While it has been demonstrated that the p53 protein can directly induce the leakage of cathepsin proteases from the lysosome, directly triggering cell death, little is known about what factors set the threshold at which the lysosome can become permeabilized. It appears that the expression levels of cathepsin proteases may be central to this process, with some of them being transcriptionally regulated by p53. The consequences of such a mechanism have serious implications for lysosomal-mediated apoptosis and have significant input into the design of therapeutics and their strategic use. In this review, we highlight the importance of extending such findings to other cathepsin family members and the need to assess the roles of p53 isoforms and mutants in furthering this mechanism. Abstract While viewed as the “guardian of the genome”, the importance of the tumor suppressor p53 protein has increasingly gained ever more recognition in modulating additional modes of action related to cell death. Slowly but surely, its importance has evolved from a mutated genetic locus heavily implicated in a wide array of cancer types to modulating lysosomal-mediated cell death either directly or indirectly through the transcriptional regulation of the key signal transduction pathway intermediates involved in this. As an important step in determining the fate of cells in response to cytotoxicity or during stress response, lysosomal-mediated cell death has also become strongly interwoven with the key components that give the lysosome functionality in the form of the cathepsin proteases. While a number of articles have been published highlighting the independent input of p53 or cathepsins to cellular homeostasis and disease progression, one key area that warrants further focus is the regulatory relationship that p53 and its isoforms share with such proteases in regulating lysosomal-mediated cell death. Herein, we review recent developments that have shaped this relationship and highlight key areas that need further exploration to aid novel therapeutic design and intervention strategies.
Collapse
Affiliation(s)
- Surinder M. Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia; (L.V.S.); (V.A.M.); (N.V.G.)
- Correspondence: (S.M.S.); (A.A.Z.J.)
| | - Lyudmila V. Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia; (L.V.S.); (V.A.M.); (N.V.G.)
| | - Vladimir A. Makarov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia; (L.V.S.); (V.A.M.); (N.V.G.)
| | - Neonila V. Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia; (L.V.S.); (V.A.M.); (N.V.G.)
| | - Paul A. Townsend
- Division of Cancer Sciences and Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, and the NIHR Manchester Biomedical Research Centre, Manchester M13 9PL, UK;
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia; (L.V.S.); (V.A.M.); (N.V.G.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
- Correspondence: (S.M.S.); (A.A.Z.J.)
| |
Collapse
|
123
|
Apoptosis in the fetal testis eliminates developmentally defective germ cell clones. Nat Cell Biol 2020; 22:1423-1435. [DOI: 10.1038/s41556-020-00603-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 10/12/2020] [Indexed: 01/22/2023]
|
124
|
Mulcahy ME, O'Brien EC, O'Keeffe KM, Vozza EG, Leddy N, McLoughlin RM. Manipulation of Autophagy and Apoptosis Facilitates Intracellular Survival of Staphylococcus aureus in Human Neutrophils. Front Immunol 2020; 11:565545. [PMID: 33262756 PMCID: PMC7686353 DOI: 10.3389/fimmu.2020.565545] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 01/13/2023] Open
Abstract
Polymorphonuclear neutrophils (PMN) are critical for first line innate immune defence against Staphylococcus aureus. Mature circulating PMN maintain a short half-life ending in constitutive apoptotic cell death. This makes them unlikely candidates as a bacterial intracellular niche. However, there is significant evidence to suggest that S. aureus can survive intracellularly within PMN and this contributes to persistence and dissemination during infection. The precise mechanism by which S. aureus parasitizes these cells remains to be established. Herein we propose a novel mechanism by which S. aureus subverts both autophagy and apoptosis in PMN in order to maintain an intracellular survival niche during infection. Intracellular survival of S. aureus within primary human PMN was associated with an accumulation of the autophagic flux markers LC3-II and p62, while inhibition of the autophagy pathway led to a significant reduction in intracellular survival of bacteria. This intracellular survival of S. aureus was coupled with a delay in neutrophil apoptosis as well as increased expression of several anti-apoptotic factors. Importantly, blocking autophagy in infected PMN partially restored levels of apoptosis to that of uninfected PMN, suggesting a connection between the autophagic and apoptotic pathways during intracellular survival. These results provide a novel mechanism for S. aureus intracellular survival and suggest that S. aureus may be subverting crosstalk between the autophagic and apoptosis pathways in order to maintain an intracellular niche within human PMN.
Collapse
Affiliation(s)
- Michelle E Mulcahy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Eóin C O'Brien
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate M O'Keeffe
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Neal Leddy
- bioTEM, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
125
|
Xu Y, Sun Y, Zhou K, Xie C, Li T, Wang Y, Zhang Y, Rodriguez J, Zhang X, Shao R, Wang X, Zhu C. Cranial irradiation alters neuroinflammation and neural proliferation in the pituitary gland and induces late-onset hormone deficiency. J Cell Mol Med 2020; 24:14571-14582. [PMID: 33174363 PMCID: PMC7754041 DOI: 10.1111/jcmm.16086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/20/2022] Open
Abstract
Cranial radiotherapy induces endocrine disorders and reproductive abnormalities, particularly in long-term female cancer survivors, and this might in part be caused by injury to the pituitary gland, but the underlying mechanisms are unknown. The aim of this study was to investigate the influence of cranial irradiation on the pituitary gland and related endocrine function. Female Wistar rat pups on postnatal day 11 were subjected to a single dose of 6 Gy whole-head irradiation, and hormone levels and organ structure in the reproductive system were examined at 20 weeks after irradiation. We found that brain irradiation reduced cell proliferation and induced persistent inflammation in the pituitary gland. The whole transcriptome analysis of the pituitary gland revealed that apoptosis and inflammation-related pathways were up-regulated after irradiation. In addition, irradiation led to significantly decreased levels of the pituitary hormones, growth hormone, adrenocorticotropic hormone, thyroid-stimulating hormone and the reproductive hormones testosterone and progesterone. To conclude, brain radiation induces reduction of pituitary and reproduction-related hormone secretion, this may due to reduced cell proliferation and increased pituitary inflammation after irradiation. Our results thus provide additional insight into the molecular mechanisms underlying complications after head irradiation and contribute to the discovery of preventive and therapeutic strategies related to brain injury following irradiation.
Collapse
Affiliation(s)
- Yiran Xu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Henan, China
| | - Kai Zhou
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.,Department of Neonatology, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Tao Li
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Neonatology, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yafeng Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Neonatology, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaodong Zhang
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Neonatology, Children's Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Xiaoan Zhang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruijin Shao
- Department of Physiology/Endocrinology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
126
|
Dos Santos DC, Rafique J, Saba S, Almeida GM, Siminski T, Pádua C, Filho DW, Zamoner A, Braga AL, Pedrosa RC, Ourique F. Apoptosis oxidative damage-mediated and antiproliferative effect of selenylated imidazo[1,2-a]pyridines on hepatocellular carcinoma HepG2 cells and in vivo. J Biochem Mol Toxicol 2020; 35:e22663. [PMID: 33125183 DOI: 10.1002/jbt.22663] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
Imidazo[1,2-a]pyridines (IP) and organoselenium compounds have been widely exploited in medicinal chemistry due to their pharmacological activities. Hepatocellular carcinoma (HCC) has few treatment options, and unfortunately, the prognosis is poor. Thus, the development of novel therapeutic drugs is urgent. The present study aimed at evaluating the antitumor mechanism of selenylated IP against HepG2 cells and in vivo. The selenylated IP named IP-Se-06 (3-((2-methoxyphenyl)selanyl)-7-methyl-2-phenylimidazol[1,2-a]pyridine) showed high cytotoxicity against HepG2 cells (half-maximal inhibitory concentration [IC50 ] = 0.03 µM) and selectivity for this tumor cell line. At nontoxic concentration, IP-Se-06 decreased the protein levels of Bcl-xL and increased the levels of p53, leading to inhibition of cell proliferation and apoptosis. This compound decreased the level of extracellular signal-regulated kinase 1/2 protein and changed the levels of proteins involved in the drive of the cell cycle, tumor growth, and survival (cyclin B1, cyclin-dependent kinase 2). In addition, IP-Se-06 decreased the number of cells in the S phase. In addition, IP-Se-06 led to increased generation of reactive oxygen species, changed antioxidant defenses, and caused DNA fragmentation. Finally, IP-Se-06 significantly inhibited the growth of Ehrlich ascites tumors in mice, increased survival time, and inhibited angiogenesis. Therefore, IP-Se-06 may be an important compound regarding the development of a therapeutic drug for HCC treatment.
Collapse
Affiliation(s)
- Daniela Coelho Dos Santos
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Jamal Rafique
- Departamento de Química, Instituto de Química (INQUI), Universidade Federal do Mato Grosso do Sul (UFMS), Campo Grande, Mato Grosso do Sul, Brazil
| | - Sumbal Saba
- Departamento de Química Orgânica, Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), Santo André, São Paulo, Brazil
| | - Gabriela M Almeida
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Tâmila Siminski
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Cynthia Pádua
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Danilo W Filho
- Departamento de Ecologia e Zoologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Ariane Zamoner
- Departamento de Bioquímica, Laboratório de Bioquímica e Sinalização Celular (LaBioSignal), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Antonio L Braga
- Departamento de Química, Laboratório de Síntese de Substâncias de Selênio Bioativas (LabSelen), Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Rozangela C Pedrosa
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Fabiana Ourique
- Departamento de Bioquímica, Laboratório de Bioquímica Experimental (LABIOEX), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil.,Departamento de Bioquímica, Laboratório de Bioquímica e Sinalização Celular (LaBioSignal), Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
127
|
Anticancer potential of myricetin bulk and nano forms in vitro in lymphocytes from myeloma patients. Arch Toxicol 2020; 95:337-343. [PMID: 33128380 PMCID: PMC7811500 DOI: 10.1007/s00204-020-02938-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 10/20/2020] [Indexed: 01/01/2023]
Abstract
Evading apoptosis and chemo-resistance are considered as very important factors which help tumour progression and metastasis. Hence, to overcome chemo-resistance, there is an urgent requirement for emergence of more effective treatment options. Myricetin, a naturally occurring flavonoid, is present in various plant-derived foods and shows antitumour potential in different cancers. In the present in vitro study, results from the comet assay demonstrated that myricetin bulk (10 µM) and nano (20 µM) forms exhibited a non-significant level of genotoxicity in lymphocytes from multiple myeloma patients when compared to those from healthy individuals. Western blot results showed a decrease in Bcl-2/Bax ratio and an increase in P53 protein levels in lymphocytes from myeloma patients, but not in lymphocytes from healthy individuals. A significant increase in intracellular reactive oxygen species level was also observed, suggesting that regulation of apoptotic proteins triggered by myricetin exposure in lymphocytes from myeloma patients occurred through P53 and oxidative stress-dependent pathways. The potency of myricetin against lymphocytes from myeloma patients marks it a potential candidate to be considered as an alternative to overcome chemo-resistance in cancer therapies.
Collapse
|
128
|
Poppenberg KE, Tutino VM, Li L, Waqas M, June A, Chaves L, Jiang K, Jarvis JN, Sun Y, Snyder KV, Levy EI, Siddiqui AH, Kolega J, Meng H. Classification models using circulating neutrophil transcripts can detect unruptured intracranial aneurysm. J Transl Med 2020; 18:392. [PMID: 33059716 PMCID: PMC7565814 DOI: 10.1186/s12967-020-02550-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022] Open
Abstract
Background Intracranial aneurysms (IAs) are dangerous because of their potential to rupture. We previously found significant RNA expression differences in circulating neutrophils between patients with and without unruptured IAs and trained machine learning models to predict presence of IA using 40 neutrophil transcriptomes. Here, we aim to develop a predictive model for unruptured IA using neutrophil transcriptomes from a larger population and more robust machine learning methods. Methods Neutrophil RNA extracted from the blood of 134 patients (55 with IA, 79 IA-free controls) was subjected to next-generation RNA sequencing. In a randomly-selected training cohort (n = 94), the Least Absolute Shrinkage and Selection Operator (LASSO) selected transcripts, from which we constructed prediction models via 4 well-established supervised machine-learning algorithms (K-Nearest Neighbors, Random Forest, and Support Vector Machines with Gaussian and cubic kernels). We tested the models in the remaining samples (n = 40) and assessed model performance by receiver-operating-characteristic (ROC) curves. Real-time quantitative polymerase chain reaction (RT-qPCR) of 9 IA-associated genes was used to verify gene expression in a subset of 49 neutrophil RNA samples. We also examined the potential influence of demographics and comorbidities on model prediction. Results Feature selection using LASSO in the training cohort identified 37 IA-associated transcripts. Models trained using these transcripts had a maximum accuracy of 90% in the testing cohort. The testing performance across all methods had an average area under ROC curve (AUC) = 0.97, an improvement over our previous models. The Random Forest model performed best across both training and testing cohorts. RT-qPCR confirmed expression differences in 7 of 9 genes tested. Gene ontology and IPA network analyses performed on the 37 model genes reflected dysregulated inflammation, cell signaling, and apoptosis processes. In our data, demographics and comorbidities did not affect model performance. Conclusions We improved upon our previous IA prediction models based on circulating neutrophil transcriptomes by increasing sample size and by implementing LASSO and more robust machine learning methods. Future studies are needed to validate these models in larger cohorts and further investigate effect of covariates.
Collapse
Affiliation(s)
- Kerry E Poppenberg
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Vincent M Tutino
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Biomedical Engineering, University of Buffalo, Buffalo, USA.,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Lu Li
- Department of Computer Science and Engineering, University of Buffalo, Buffalo, USA
| | - Muhammad Waqas
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Armond June
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Lee Chaves
- Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Kaiyu Jiang
- Genetics, Genomics, and Bioinformatics Program, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - James N Jarvis
- Genetics, Genomics, and Bioinformatics Program, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Yijun Sun
- Genetics, Genomics, and Bioinformatics Program, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Kenneth V Snyder
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Radiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Elad I Levy
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Radiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Adnan H Siddiqui
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA.,Department of Radiology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - John Kolega
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA.,Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Hui Meng
- Canon Stroke and Vascular Research Center, Clinical and Translational Research Center, 875 Ellicott Street, Buffalo, NY, 14214, USA. .,Department of Biomedical Engineering, University of Buffalo, Buffalo, USA. .,Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA. .,Department of Mechanical & Aerospace Engineering, University At Buffalo, Buffalo, NY, USA.
| |
Collapse
|
129
|
Muyskens JB, Winbush A, Foote DM, Turnbull DW, Dreyer HC. Essential amino acid supplementation alters the p53 transcriptional response and cytokine gene expression following total knee arthroplasty. J Appl Physiol (1985) 2020; 129:980-991. [PMID: 32881622 DOI: 10.1152/japplphysiol.00022.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reducing muscle atrophy following orthopedic surgery is critical during the postoperative period. Our previous work in patients who underwent total knee arthroplasty (TKA) showed that the vast majority of atrophy occurs within 2 wk following surgery and that essential amino acid (EAA) supplementation attenuates this atrophy. We used RNA-sequencing (RNA-seq) to identify genes associated with atrophy after TKA with and without EAAs. Analysis of overrepresented gene-ontology terms revealed that p53 signaling and the cytokine-cytokine receptor pathways were highly upregulated after TKA. Relative to the placebo group, the EAA group had altered expression of p53 regulators such as MDM2. This altered expression may account for differences between groups in timing of upregulation of some p53 targets such as apoptosis genes, and may account for the reduction in muscle loss in the subjects receiving EAAs. Furthermore, we observed altered expression of a large number of cytokine-signaling genes including TNFRSF12A, which plays a critical role in muscle atrophy, myogenesis, fibrosis, and the noncanonical NF-κB pathway.NEW & NOTEWORTHY Total knee arthroplasty is the most frequently performed inpatient surgical procedure for those over 45 yr in the United States. Following surgery, patients lose a large amount of muscle, which impacts functional mobility. Previously, our laboratory found that supplementing patients' diets with essential amino acids (EAAs) reduces postsurgical muscle loss. Here, our goal was to characterize the transcriptional changes associated with surgery with and without EAA supplementation to uncover the underlying mechanisms by which EAAs attenuate this muscle loss.
Collapse
Affiliation(s)
| | - Ari Winbush
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon
| | - Douglas M Foote
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Douglas W Turnbull
- Genomics and Cell Characterization Core Facility, University of Oregon, Eugene, Oregon
| | - Hans C Dreyer
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|
130
|
Somade OT, Ajayi BO, Adeyi OE, Adeshina AA, James AS, Ayodele PF. Ethylene glycol monomethyl ether-induced testicular oxidative stress and time-dependent up-regulation of apoptotic, pro-inflammatory, and oncogenic markers in rats. Metabol Open 2020; 7:100051. [PMID: 32924002 PMCID: PMC7451700 DOI: 10.1016/j.metop.2020.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/08/2020] [Accepted: 08/08/2020] [Indexed: 12/19/2022] Open
Abstract
Ethylene glycol monomethyl ether (EGME) is a major component of paints, lacquers, inks, and automobile brake fluids. As a result, exposures to humans are inevitable. We therefore, investigated in this study, its effect on testicular cells in a time-course manner in male Wistar rats. Animals were orally administered 50 mg/kg body weight of EGME for duration of 7, 14, and 21 days. Following 7 days of the administration, levels of NO and GSH were significantly reduced, while levels of c-Myc, K-Ras, caspase-3, IL-6, TNF-α, and IL-1β were significantly increased compared with control. At the end of 14 days exposure, GPx, and SOD activities, as well as IL-10 level were significantly decreased, while levels of c-Myc, K-Ras, p53, Bax, caspase-3, IL-6, TNF-α, IL-1β, and GST activity were significantly elevated compared with control. After 21 days of EGME administration, Bcl-2, IL-10, and NO levels were significantly decreased, while levels of c-Myc, K-Ras, p53, Bax, caspase-3, IL-6, TNF-α, IL-1β, MDA and GST activity were significantly increased compared with control. After 7, 14, and 21 days of EGME administrations, testis histopathology showed severe loss of seminiferous tubules, the seminiferous epithelium revealed very few spermatocytes, spermatids, spermatogonia, spermatozoa, and Sertoli cells, while the interstitial tissue is eroded, with scanty abnormal Leydig cells, compared with the control that appeared normal. We therefore, concluded that EGME-induced testicular toxicity as a result of EGME administration could be via the disorganization of the endogenous antioxidant systems as well as up-regulation of pro-inflammatory, apoptotic and oncogenic mediators in rats.
Collapse
Key Words
- Apoptosis
- Bax, Bcl-2 associated X
- Bcl-2, B-cell lymphoma 2
- CAT, catalase
- Ethylene glycol monomethyl ether
- GPx, glutathione peroxidase
- GSH, reduced glutathione
- GST, glutathione S-transferase
- Histopathology
- IL-1β, interleukin-1 beta
- IL-6, interleukin-6
- Inflammation
- K-Ras, Kirsten rat sarcoma viral oncogene
- MDA, malondialdehyde
- NO, nitric oxide
- Oncogenes
- Oxidative stress
- SOD, superoxide dismutase
- TNF-α, tumor necrosis factor alpha
- Testis
- c-Myc, myelocytomatosis
- p53, tumor suppressor protein
Collapse
Affiliation(s)
- Oluwatobi T Somade
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Babajide O Ajayi
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Olubisi E Adeyi
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | | | - Adewale S James
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Peter F Ayodele
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| |
Collapse
|
131
|
Al-Bahlani S, Burney IA, Al-Dhahli B, Al-Kharusi S, Al-Kharousi F, Al-Kalbani A, Ahmed I. Boswellic acid sensitizes gastric cancer cells to Cisplatin-induced apoptosis via p53-mediated pathway. BMC Pharmacol Toxicol 2020; 21:64. [PMID: 32867831 PMCID: PMC7460741 DOI: 10.1186/s40360-020-00442-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/25/2020] [Indexed: 12/18/2022] Open
Abstract
Background Cisplatin (CDDP) is an effective anticancer drug for Gastric cancer (GC) that induces apoptosis by altering pro- (p53) and anti-apoptotic (Akt and NFkB) proteins; however, chemoresistance remains a big challenge. Additional compounds with promising anticancer effects such as AKBA (Acetyl-keto-beta boswellic acid) may overcome the resistance. However, its role in CDDP-induced apoptosis in GC has not been studied. This study aimed to examine the effectiveness of AKBA on p53-mediated, CDDP-induced apoptosis in GC cells. AGS and NCI-N87 cells were treated with different concentrations (0, 25, 50, 100 μM) of CDDP and/or AKBA. Methods P53, Akt and NFkB proteins and apoptosis were assessed by Western blot and flow cytometry. The role of p53 was determined by inhibiting its function via the siRNA approach. Results The results revealed that CDDP and AKBA significantly increased p53 content in both cells, while Akt and NFkB were significantly decreased. Both compounds significantly induced apoptosis in a dose-dependent manner. AKBA sensitized GC cells to CDDP-induced apoptosis by altering the protein expression. P53 downregulation affected Akt and NFkB proteins with a slight increase in apoptosis induction in the combination treated groups. Conclusions Altogether, our findings suggest that AKBA enhances GC cell sensitivity to CDDP-induced apoptosis via the p53 pathway.
Collapse
Affiliation(s)
- Shadia Al-Bahlani
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35, PC 123 AlKhoud, Muscat, Oman.
| | - Ikram A Burney
- Department of Medicine, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Buthaina Al-Dhahli
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35, PC 123 AlKhoud, Muscat, Oman
| | - Safiya Al-Kharusi
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35, PC 123 AlKhoud, Muscat, Oman
| | - Fakhra Al-Kharousi
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35, PC 123 AlKhoud, Muscat, Oman
| | - Amani Al-Kalbani
- Department of Allied Health Sciences, College of Medicine and Health Sciences, Sultan Qaboos University, P. O. Box 35, PC 123 AlKhoud, Muscat, Oman
| | - Ikhlas Ahmed
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
132
|
Abd-Elhakim YM, Mohamed WAM, El Bohi KM, Ali HA, Mahmoud FA, Saber TM. Prevention of melamine-induced hepatorenal impairment by an ethanolic extract of Moringa oleifera: Changes in KIM-1, TIMP-1, oxidative stress, apoptosis, and inflammation-related genes. Gene 2020; 764:145083. [PMID: 32860902 DOI: 10.1016/j.gene.2020.145083] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS Melamine (ML) is a common food adulterant and contaminant. Moringa oleifera is a well-known medicinal plant with many beneficial biological properties. This study investigated the possible prophylactic and therapeutic activity of an ethanolic extract of M. oleifera (MEE) against ML-induced hepatorenal damage. METHOD Fifty male Sprague Dawley rats were orally administered distilled water, MEE (800 mg/kg bw), ML (700 mg/kg bw), MEE/ML (prophylactically) or MEE+ML (therapeutically). Hepatic aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphate (ALP) in serum were measured. Serum total bilirubin, direct bilirubin, indirect bilirubin, protein, albumin, and globulin contents were also assayed, and urea and creatinine levels were determined. Moreover, antioxidant enzyme activity of glutathione peroxidase (GPx) and catalase (CAT) in serum levels were quantified. Complementary histological and histochemical evaluation of renal and hepatic tissues was conducted, and expression of oxidative stress (GPx and CAT) and apoptosis-related genes, p53 and Bcl-2, in hepatic tissue were assessed. In parallel, transcriptional expression of inflammation and renal injury-related genes, including kidney injury molecule 1 (KIM-1), metallopeptidase inhibitor 1 (TIMP1), and tumor necrosis factor alpha (TNF-α) in the kidney tissue were determined. RESULTS ML caused significant increases in serum levels of ALT, AST, ALP, total bilirubin, direct bilirubin, indirect bilirubin, urea, and creatinine. Further, ML treated rats showed significant reductions in serum levels of protein, albumin, globulin, GPx, and CAT. Distinct histopathological damage and disturbances in glycogen and DNA content in hepatic and renal tissues of ML treated rats were observed. KIM-1, TIMP-1, and TNF-α gene expression was significantly upregulated in kidney tissue. Also, GPx, CAT, and Bcl-2 genes were significantly downregulated, and p53 was significantly upregulated in liver tissue after ML treatment. MEE significantly counteracted the ML-induced hepatorenal damage primarily for co-exposed rats. CONCLUSION MEE could be an effective therapeutic supplement for treatment of ML-induced hepato-renal damage, probably via modulating oxidative stress, apoptosis, and inflammation.
Collapse
Affiliation(s)
- Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| | - Wafaa A M Mohamed
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Khlood M El Bohi
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Haytham A Ali
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt; Department of Biochemistry, Collage of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Fagr A Mahmoud
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Egypt
| | - Taghred M Saber
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
133
|
Kang D, Zuo W, Wu Q, Zhu Q, Liu P. Inhibition of Specificity Protein 1 Is Involved in Phloretin-Induced Suppression of Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1358674. [PMID: 32851058 PMCID: PMC7439178 DOI: 10.1155/2020/1358674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/03/2023]
Abstract
Phloretin is a flavonoid existed in various plants and has been reported to possess anticarcinogenic activity. However, the anticancer mechanism of phloretin in prostate cancer (PCa) remains unclear. Here, our in vitro and in vivo experimental data demonstrate that phloretin inhibits the phosphorylation and the activation of EGFR and then inhibits its downstream PI3K/AKT and MEK/ERK1/2 pathways in PCa cells. Inhibition of these two pathways further decreases expression of Sp1 by inhibiting Sp1 gene transcription, induces degradation of Sp1 protein by inhibiting GSK3β phosphorylation, suppresses nucleolin-enhanced translation of Sp1 mRNA by inhibiting nucleolin phosphorylation, and directly inactivates transcription activity of Sp1. Inhibition of Sp1 subsequently decreases the expression of Sp3/4, VEGF, and Survivin and then upregulates apoptosis-related proteins and downregulates cell cycle-related proteins in PCa cells. Finally, phloretin treatment in PCa cells induces cell growth inhibition and apoptosis, suggesting that phloretin may be an effective therapy compound in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Dan Kang
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Wenren Zuo
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Qingxin Wu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Qingyi Zhu
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Ping Liu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
134
|
Ma WP, Hu SM, Xu YL, Li HH, Ma XQ, Wei BH, Li FY, Guan HS, Yu GL, Liu M, Liu HB. Haimufang decoction, a Chinese medicine formula for lung cancer, arrests cell cycle, stimulates apoptosis in NCI-H1975 cells, and induces M1 polarization in RAW 264.7 macrophage cells. BMC Complement Med Ther 2020; 20:243. [PMID: 32758223 PMCID: PMC7404932 DOI: 10.1186/s12906-020-03031-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lung cancer has the highest morbidity and mortality in the world and novel treatment strategies are still needed. Haimufang decoction (HMF) is a patented clinical prescription of traditional Chinese medicine for lung cancer treatment. HMF is composed of four herbs and has been applied clinically in advanced cancer patients. However, its therapeutic mechanisms are still unclear. This study aims to elucidate the possible mechanisms of HMF for the treatment of lung cancer. METHODS 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay was applied for evaluating the proliferative effect of HMF in lung cancer cells and monocyte macrophage RAW264.7 cells. Flow cytometer was used to detect the effects of HMF on cell cycle and apoptosis, and western blotting was employed to explore the potential apoptotic mechanisms of HMF on lung cancer cells. For immunomodulatory effect, co-culture system was used to detect the activation of macrophage RAW264.7 cells when treated with HMF, and neutral red assay was used to measure the effect of HMF on the phagocytosis of the activated macrophages. Enzyme linked immunosorbent assay, flow cytometer, and immunofluorescence staining method were employed for the investigation on the underlying mechanisms of the immunomodulatory effect on RAW264.7 induced by HMF. RESULTS HMF inhibited the proliferation, induced S phase cell cycle arrest, and stimulated apoptosis in lung cancer NCI-H1975 cells, while had negligible cytotoxicity on macrophage RAW264.7 cells. Moreover, HMF could activate macrophage RAW264.7 cells and promote the inhibition activity of RAW264.7 cells against lung cancer cells. And also, HMF activated macrophages and increased their phagocytic activity in a concentration-dependent manner. HMF increased the expression of macrophage activation marker CD40, the level of nitric oxide, the generation of intracellular reactive oxygen species, as well as M1 macrophages cytokines including tumor necrosis factor-α, interleukin-1β, interleukin 12 p70, and interleukin 6. Further investigation showed that HMF induced M1 but not M2 phenotype polarization in RAW264.7 cells. CONCLUSIONS HMF can mainly exert anticancer activity via (1) cytotoxicity to human lung cancer cells by proliferation inhibition, cell cycle arrest, and apoptosis induction; and also via (2) immunomodulation via macrophage cells activation and M1 phenotype polarization induction.
Collapse
Affiliation(s)
- Wei-Ping Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Shu-Man Hu
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Yan-Lai Xu
- Naval Secret Service Nursing Center of Qingdao, Qingdao, 266071, P. R. China
| | - Hai-Hua Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Xiao-Qing Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Bao-Hong Wei
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Fu-Yu Li
- Naval Secret Service Nursing Center of Qingdao, Qingdao, 266071, P. R. China
| | - Hua-Shi Guan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Guang-Li Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Ming Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| | - Hong-Bing Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
135
|
Zhang L, Gao Y, Zhang R, Sun F, Cheng C, Qian F, Duan X, Wei G, Sun C, Pang X, Chen P, Chai R, Yang T, Wu H, Liu D. THOC1 deficiency leads to late-onset nonsyndromic hearing loss through p53-mediated hair cell apoptosis. PLoS Genet 2020; 16:e1008953. [PMID: 32776944 PMCID: PMC7444544 DOI: 10.1371/journal.pgen.1008953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/20/2020] [Accepted: 06/24/2020] [Indexed: 01/04/2023] Open
Abstract
Apoptosis of cochlear hair cells is a key step towards age-related hearing loss. Although numerous genes have been implicated in the genetic causes of late-onset, progressive hearing loss, few show direct links to the proapoptotic process. By genome-wide linkage analysis and whole exome sequencing, we identified a heterozygous p.L183V variant in THOC1 as the probable cause of the late-onset, progressive, non-syndromic hearing loss in a large family with autosomal dominant inheritance. Thoc1, a member of the conserved multisubunit THO/TREX ribonucleoprotein complex, is highly expressed in mouse and zebrafish hair cells. The thoc1 knockout (thoc1 mutant) zebrafish generated by gRNA-Cas9 system lacks the C-startle response, indicative of the hearing dysfunction. Both Thoc1 mutant and knockdown zebrafish have greatly reduced hair cell numbers, while the latter can be rescued by embryonic microinjection of human wild-type THOC1 mRNA but to significantly lesser degree by the c.547C>G mutant mRNA. The Thoc1 deficiency resulted in marked apoptosis in zebrafish hair cells. Consistently, transcriptome sequencing of the mutants showed significantly increased gene expression in the p53-associated signaling pathway. Depletion of p53 or applying the p53 inhibitor Pifithrin-α significantly rescued the hair cell loss in the Thoc1 knockdown zebrafish. Our results suggested that THOC1 deficiency lead to late-onset, progressive hearing loss through p53-mediated hair cell apoptosis. This is to our knowledge the first human disease associated with THOC1 mutations and may shed light on the molecular mechanism underlying the age-related hearing loss.
Collapse
Affiliation(s)
- Luping Zhang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yu Gao
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ru Zhang
- Shanghai East Hospital, Department of Otorhinolaryngology Shanghai, Shanghai, China
| | - Feifei Sun
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Fuping Qian
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Xuchu Duan
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Guanyun Wei
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Cheng Sun
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiuhong Pang
- Department of Otorhinolaryngology-Head and Neck Surgery, Taizhou People’s Hospital, Fifth Affiliated Hospital, Nantong University, Taizhou, China
| | - Penghui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Renjie Chai
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Tao Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Hao Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Dong Liu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital, School of Life Science, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
136
|
Wang J, Du A, Wang H, Li Y. MiR-599 regulates LPS-mediated apoptosis and inflammatory responses through the JAK2/STAT3 signalling pathway via targeting ROCK1 in human umbilical vein endothelial cells. Clin Exp Pharmacol Physiol 2020; 47:1420-1428. [PMID: 32248560 DOI: 10.1111/1440-1681.13316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
MicroRNA plays an integral role in the development of atherosclerosis. Our study aimed to investigate the roles of miR-599 in lipopolysaccharide (LPS)-induced endothelial damage in human umbilical vein endothelial cells (HUVECs). HUVECs were transfected with a miR-599 mimic and negative control, and then exposed to LPS. The expression of miR-599 was detected by quantitative real time-polymerase chain reaction (RT-qPCR). Cell viability was analyzed by CCK-8 assay and trypan blue exclusion assay; the formation of DNA fragments was tested by Cell Death Detection ELISA Plus kit; the incidence of apoptosis was detected by flow cytometry; the expression of p53 and cleaved-caspase 3 (c-caspase 3) was evaluated by western blot. Moreover, the mRNA levels and concentrations of tumour necrosis factor (TNF)-α, interleukin (IL)-6, ICAM-1 and VCAM-1 were assayed by RT-qPCR and ELISA. The results showed that overexpression of miR-599 increased cell viability, reduced DNA fragments, the incidence of apoptosis, as well as the protein levels of p53 and c-caspase 3 in the presence of LPS. TNF-α, IL-6, ICAM-1 and VCAM-1 mRNA levels and concentrations were also decreased upon miR-599 upregulation. In addition, the dual luciferase reporter assay demonstrated that ROCK1 is a direct target of miR-599. MiR-599 overexpression inhibited ROCK1 expression. Induced expression of ROCK1 reversed the roles of miR-599 in apoptosis and inflammation. The gain function of miR-599 function inhibited activation of the JAK2/STAT3 signalling pathway, which was abrogated by overexpression of ROCK1. Taken together, our results indicate that miR-599 attenuates LPS-caused cell apoptosis and inflammatory responses through the JAK2/STAT3 signalling pathway via targeting ROCK1.
Collapse
Affiliation(s)
- Jia Wang
- Department of Cardiology, Nursing Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hexilin Wang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
137
|
Abbaszadeh F, Fakhri S, Khan H. Targeting apoptosis and autophagy following spinal cord injury: Therapeutic approaches to polyphenols and candidate phytochemicals. Pharmacol Res 2020; 160:105069. [PMID: 32652198 DOI: 10.1016/j.phrs.2020.105069] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a neurological disorder associated with the loss of sensory and motor function. Understanding the precise dysregulated signaling pathways, especially apoptosis and autophagy following SCI, is of vital importance in developing innovative therapeutic targets and treatments. The present study lies in the fact that it reveals the precise dysregulated signaling mediators of apoptotic and autophagic pathways following SCI and also examines the effects of polyphenols and other candidate phytochemicals. It provides new insights to develop new treatments for post-SCI complications. Accordingly, a comprehensive review was conducted using electronic databases including, Scopus, Web of Science, PubMed, and Medline, along with the authors' expertise in apoptosis and autophagy as well as their knowledge about the effects of polyphenols and other phytochemicals on SCI pathogenesis. The primary mechanical injury to spinal cord is followed by a secondary cascade of apoptosis and autophagy that play critical roles during SCI. In terms of pharmacological mechanisms, caspases, Bax/Bcl-2, TNF-α, and JAK/STAT in apoptosis along with LC3 and Beclin-1 in autophagy have shown a close interconnection with the inflammatory pathways mainly glutamatergic, PI3K/Akt/mTOR, ERK/MAPK, and other cross-linked mediators. Besides, apoptotic pathways have been shown to regulate autophagy mediators and vice versa. Prevailing evidence has highlighted the importance of modulating these signaling mediators/pathways by polyphenols and other candidate phytochemicals post-SCI. The present review provides dysregulated signaling mediators and therapeutic targets of apoptotic and autophagic pathways following SCI, focusing on the modulatory effects of polyphenols and other potential phytochemical candidates.
Collapse
Affiliation(s)
- Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
138
|
Lunova M, Smolková B, Uzhytchak M, Janoušková KŽ, Jirsa M, Egorova D, Kulikov A, Kubinová Š, Dejneka A, Lunov O. Light-induced modulation of the mitochondrial respiratory chain activity: possibilities and limitations. Cell Mol Life Sci 2020; 77:2815-2838. [PMID: 31583425 PMCID: PMC11104903 DOI: 10.1007/s00018-019-03321-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/11/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022]
Abstract
Biological effects of high fluence low-power (HFLP) lasers have been reported for some time, yet the molecular mechanisms procuring cellular responses remain obscure. A better understanding of the effects of HFLP lasers on living cells will be instrumental for the development of new experimental and therapeutic strategies. Therefore, we investigated sub-cellular mechanisms involved in the laser interaction with human hepatic cell lines. We show that mitochondria serve as sub-cellular "sensor" and "effector" of laser light non-specific interactions with cells. We demonstrated that despite blue and red laser irradiation results in similar apoptotic death, cellular signaling and kinetic of biochemical responses are distinct. Based on our data, we concluded that blue laser irradiation inhibited cytochrome c oxidase activity in electron transport chain of mitochondria. Contrary, red laser triggered cytochrome c oxidase excessive activation. Moreover, we showed that Bcl-2 protein inhibited laser-induced toxicity by stabilizing mitochondria membrane potential. Thus, cells that either overexpress or have elevated levels of Bcl-2 are protected from laser-induced cytotoxicity. Our findings reveal the mechanism how HFLP laser irradiation interfere with cell homeostasis and underscore that such laser irradiation permits remote control of mitochondrial function in the absence of chemical or biological agents.
Collapse
Affiliation(s)
- Mariia Lunova
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic
- Institute for Clinical and Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Mariia Uzhytchak
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Klára Žofie Janoušková
- Institute for Clinical and Experimental Medicine (IKEM), 14021, Prague, Czech Republic
- First Faculty of Medicine, Charles University, 12108, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical and Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | | | | | - Šárka Kubinová
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic
- Institute of Experimental Medicine, Czech Academy of Sciences, 14220, Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics, Czech Academy of Sciences, 18221, Prague, Czech Republic.
| |
Collapse
|
139
|
Ali AS, Perren A, Lindskog C, Welin S, Sorbye H, Grönberg M, Janson ET. Candidate protein biomarkers in pancreatic neuroendocrine neoplasms grade 3. Sci Rep 2020; 10:10639. [PMID: 32606315 PMCID: PMC7327066 DOI: 10.1038/s41598-020-67670-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are rare tumours that compose 1–2% of all pancreatic tumours.
Patients with metastatic grade 3 neoplasia are usually treated with chemotherapy but have a poor progression-free and overall survival. According to the WHO 2017 classification, they are divided into neuroendocrine tumours (NETs) G3 and neuroendocrine carcinomas (NECs). Despite the new classification, new diagnostic and prognostic biomarkers are needed to sub-categorise the patients and to help guide therapy decisions. Blood from 42 patients and 42 healthy controls were screened for the presence of 92 proteins with the Immuno-Oncology panel using the Proximity Extension Assay provided by Olink Biosciences. Immunohistochemical staining of FAS ligand (FASLG) was performed on 16 patient tumour specimens using a commercial antibody. Fifty-four out of 87 evaluable proteins differed significantly in concentration between blood from patients and blood from healthy controls. FASLG was the only protein for which the concentration in blood was significantly lower in patients compared to controls and the levels correlated negatively to Ki-67 index. Seven of 14 evaluable PanNEN G3 specimens showed FASLG immunoreactivity in the tumour cells while there was scattered immunoreactivity in immune cells. Positive FASLG immunoreactivity correlated to well-differentiated morphology.
FASLG concentration in blood was significantly lower in patients with pancreatic NENs G3 compared to controls, and the expression in tumour tissue was variable. Furthermore, FASLG was negatively correlated to Ki-67 and was more frequently expressed in well-differentiated tumours. Taken together, these results may suggest a role of FASLG in PanNENs.
Collapse
Affiliation(s)
- Abir Salwa Ali
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Rudbecklaboratoriet, hus R3, vån 2, Dag Hammarskjölds väg 20, 752 85, Uppsala, Sweden
| | - Aurel Perren
- Department of Pathology, University of Bern, Bern, Switzerland
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Staffan Welin
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Rudbecklaboratoriet, hus R3, vån 2, Dag Hammarskjölds väg 20, 752 85, Uppsala, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Malin Grönberg
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Rudbecklaboratoriet, hus R3, vån 2, Dag Hammarskjölds väg 20, 752 85, Uppsala, Sweden.
| | - Eva Tiensuu Janson
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Rudbecklaboratoriet, hus R3, vån 2, Dag Hammarskjölds väg 20, 752 85, Uppsala, Sweden
| |
Collapse
|
140
|
(-)-Kusunokinin inhibits breast cancer in N-nitrosomethylurea-induced mammary tumor rats. Eur J Pharmacol 2020; 882:173311. [PMID: 32619673 DOI: 10.1016/j.ejphar.2020.173311] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 01/10/2023]
Abstract
Natural and synthetic (-)-kusunokinin inhibited breast cancer, colon cancer and cholangiocarcinoma cells at the G2/M phase and induced apoptosis. However, there is no report on the action and adverse effects of (-)-kusunokinin in animal models. In this study, we investigated the cytotoxic effect of (-)-kusunokinin from Piper nigrum on cancer cells. NMU-induced rat mammary tumors, an ER positive breast cancer model, were treated with (-)-kusunokinin. Proteins of interest related to cell cycle, angiogenesis, migration and signaling proteins were detected in tumor tissues. Results showed that (-)-kusunokinin exhibited strong cytotoxicity against breast, colon and lung cancer cells and caused low toxicity against normal fibroblast cells. For in vivo study, 7.0 mg/kg and 14.0 mg/kg of (-)-kusunokinin reduced tumor growth without side effects on body weight, internal organs and bone marrow. Combination of (-)-kusunokinin with a low effective dose of doxorubicin significantly inhibited tumor growth and provoked cell death in cancer tissues. Mechanistically, 14.0 mg/kg of (-)-kusunokinin decreased cell proliferation (c-Src, PI3K, Akt, p-Erk1/2 and c-Myc), cell cycle (E2f-1, cyclin B1 and CDK1), and metastasis (E-cadherin, MMP-2 and MMP-9) proteins in tumor tissues, which supports its anticancer effect. We further confirmed the antimigration effect of (-)-kusunokinin; the results show that this compound inhibited breast cancer cell (MCF-7) migration in a dose-dependent manner. In conclusion, the results suggest that 14 mg/kg of (-)-kusunokinin inhibited tumors through the reduction of signaling proteins and their downstream molecules. Therefore, (-)-kusunokinin becomes an intriguing candidate for cancer treatment as it provides a strong potency in cancer inhibition.
Collapse
|
141
|
Najmuddin SUFS, Amin ZM, Tan SW, Yeap SK, Kalyanasundram J, Ani MAC, Veerakumarasivam A, Chan SC, Chia SL, Yusoff K, Alitheen NB. Cytotoxicity study of the interleukin-12-expressing recombinant Newcastle disease virus strain, rAF-IL12, towards CT26 colon cancer cells in vitro and in vivo. Cancer Cell Int 2020; 20:278. [PMID: 32612457 PMCID: PMC7325054 DOI: 10.1186/s12935-020-01372-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/20/2020] [Indexed: 12/14/2022] Open
Abstract
Background Oncolytic viruses have emerged as an alternative therapeutic modality for cancer as they can replicate specifically in tumour cells and induce toxic effects leading to apoptosis. Despite the great potentials and promising results shown in multiple studies, it appears that their efficacy is still moderate and deemed as not sufficient in clinical studies. In addressing this issue, genetic/molecular engineering approach has paved its way to improve the therapeutic efficacy as observed in the case of herpes simplex virus (HSV) expressing granulocyte–macrophage colony-stimulating factor (GM-CSF). This study aimed to explore the cytotoxicity effects of recombinant NDV strain AF2240-i expressing interleukin-12 (rAF-IL12) against CT26 colon cancer cells. Methods The cytotoxicity effect of rAF-IL12 against CT26 colon cancer cell line was determined by MTT assay. Based on the IC50 value from the anti-proliferative assay, further downward assays such as Annexin V FITC and cell cycle progression were carried out and measured by flow cytometry. Then, the in vivo study was conducted where the rAF-IL12 viral injections were given at the intra-tumoral site of the CT26 tumour-burden mice. At the end of the experiment, serum biochemical, T cell immunophenotyping, serum cytokine, histopathology of tumour and organ section, TUNEL assay, and Nanostring gene expression analysis were performed. Results The rAF-IL12 induced apoptosis of CT26 colon cancer cells in vitro as revealed in the Annexin V FITC analysis and also arrested the cancer cells progression at G1 phase of the cell cycle analysis. On the other hand, the rAF-IL12 significantly (p < 0.05) inhibited the growth of CT26 tumour in Balb/c mice and had regulated the immune system by increasing the level of CD4 + , CD8 + , IL-2, IL-12, and IFN-γ. Furthermore, the expression level of apoptosis-related genes (bax and p53) was up-regulated as a result of the rAF-IL12 treatment. Additionally, the rAF-IL12 had also down-regulated the expression level of KRAS, BRAF, MAPK1, Notch1, CCL2, and VEGF oncogenes. Besides, rAF-IL12 intra-tumoral delivery was considered safe and not hazardous to the host as evidenced in pathophysiology of the normal tissues and organs of the mice as well as from the serum biochemistry profile of liver and kidney. Conclusions These results indicated that rAF-IL12 had better anti-tumoral and cytotoxicity effects compared to its parental wild-type, AF2240-i in combatting the CT26 colon cancer model.
Collapse
Affiliation(s)
| | - Zahiah Mohamed Amin
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Sheau Wei Tan
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Swee Keong Yeap
- Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, Sepang, Selangor Darul Ehsan Malaysia
| | - Jeevanathan Kalyanasundram
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Muhamad Alhapis Che Ani
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | | | - Soon Choy Chan
- School of Foundation Studies, Perdana University, Block B and D1, MAEPS Building, MARDI Complex, Jalan MAEPS Perdana, 43400 Serdang, Selangor Darul Ehsan Malaysia
| | - Suet Lin Chia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| | - Khatijah Yusoff
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Malaysian Genome Institute, National Institute of Biotechnology, Kajang, Jalan Bangi, 43000 Selangor Darul Ehsan Malaysia
| | - Noorjahan Banu Alitheen
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia.,Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan 43400 Malaysia
| |
Collapse
|
142
|
Bonisoli-Alquati A, Xu W, Stouffer PC, Taylor SS. Transcriptome analysis indicates a broad range of toxic effects of Deepwater Horizon oil on Seaside Sparrows. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 720:137583. [PMID: 32325582 DOI: 10.1016/j.scitotenv.2020.137583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 06/11/2023]
Abstract
In marine species, the transcriptomic response to Deepwater Horizon (DWH) oil implicated many biochemical pathways, with corresponding adverse outcomes on organ development and physiological performance. Terrestrial organisms differ in their mechanisms of exposure to polycyclic aromatic hydrocarbons (PAHs) and their physiological challenges, and may reveal either distinct effects of oil on biochemical pathways or the generality of the responses to oil shown in marine species. Using a cross-species hybridization microarray approach, we investigated the transcriptomic response in the liver of Seaside Sparrows (Ammospiza maritima) exposed to DWH oil compared with birds from a control site. Our analysis identified 295 genes differentially expressed between birds exposed to oil and controls. Gene ontology (GO) and canonical pathway analysis suggested that the identified genes were involved in a coordinated response that promoted hepatocellular proliferation and liver regeneration while inhibiting apoptosis, necrosis, and liver steatosis. Exposure to oil also altered the expression of genes regulating energy homeostasis, including carbohydrate metabolism and gluconeogenesis, and the biosynthesis, transport and metabolism of lipids. These results provide a molecular mechanism for the long-standing observation of hepatic hypertrophy and altered lipid biosynthesis and transport in birds exposed to crude oil. Several of the activated pathways and pathological outcomes shown here overlap with the ones altered in fish species upon exposure to oil. Overall, our study shows that the path of oil contamination from the marine system into salt marshes can lead to similar responses in terrestrial birds to those described in marine organisms, suggesting similar adverse outcomes and shared machinery for detoxification.
Collapse
Affiliation(s)
- A Bonisoli-Alquati
- Department of Biological Sciences, California State Polytechnic University, Pomona, Pomona, CA, United States of America.
| | - W Xu
- Department of Life Sciences, Texas A&M University - Corpus Christi, Corpus Christi, TX, United States of America
| | - P C Stouffer
- School of Renewable Natural Resources, Louisiana State University AgCenter, Baton Rouge, LA, United States of America; LSU AgCenter, Baton Rouge, LA, United States of America
| | - S S Taylor
- School of Renewable Natural Resources, Louisiana State University AgCenter, Baton Rouge, LA, United States of America; LSU AgCenter, Baton Rouge, LA, United States of America
| |
Collapse
|
143
|
Design, synthesis, and cytotoxic screening of novel azole derivatives on hepatocellular carcinoma (HepG2 Cells). Bioorg Chem 2020; 101:103995. [PMID: 32569897 DOI: 10.1016/j.bioorg.2020.103995] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Novel azole derivatives 3-30 were designed, synthesized, and screened for their antitumor activity on HepG2 cell line. The cytotoxicity screening demonstrated that imidazolone 8 and triazoles 25 and 29 exhibited more potent cytotoxic activities by 1.21-, 4.75-, and 1.8-fold compared to Sorafenib (SOR). Furthermore, vascular endothelial growth factor receptor-2 (VEGFR-2) enzyme inhibition assay declared that compounds 25 and 29 had inhibitory activity at the nanomolar concentration. Moreover, the tested compounds exhibited good β-tubulin (TUB) polymerization inhibition percentages. In addition, DNA flow cytometry analysis over HepG2 cells indicated that triazoles 25 and 29 demonstrated arrest at G1 and G2/M phase of the cell cycle and induced apoptotic activity by increasing sub-G1 phase. Finally, mechanistic studies of the proapoptotic activities of compounds 8, 10, 11, 25, and 29 indicated that they induced upregulation of P53, Fas/Fas-ligand, and BAX/BCL-2 ratio expression that resulted in increasing the active caspase 3/7 percentages and trigger apoptosis.
Collapse
|
144
|
Hussain SS, Faizi S, Rafi K, Simjee SU. Novel Mannich base 3FB3FA8H induces apoptosis by upregulating P53 pathway in neuroblastoma cells. Mol Cell Biochem 2020; 471:29-39. [PMID: 32472321 DOI: 10.1007/s11010-020-03755-1] [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: 01/04/2020] [Accepted: 05/16/2020] [Indexed: 10/24/2022]
Abstract
P53 plays an important role in maintaining genetic stability and development of resistance against tumors. Dysregulation of P53 gene is one of the key factors contributing to the etiology of neuroblastoma which causes cells to evade apoptosis. Activating P53 pathway can be a therapeutic alternative to the currently available medicinal strategies. Mannich bases have been known to possess various biological activities including the anticancer activity. In this study, we have targeted the P53 pathway by novel Mannich base (3FB3FA8H) which can be a future prospect to cure neuroblastoma. 3FB3FA8H has shown modulation of P53 pathway leading to apoptosis of neuroblastoma cells. Mitochondrial membrane permeability is also increased by 3FB3FA8H which may be a consequence of P53 pathway modulation. 3FB3FA8H increases the mRNA levels of P53 leading to activation of BAX. Inclining BAX/BCL2 ratio towards apoptotic BAX leads to cleavage of caspase 3, ultimately, causing apoptosis. Series of experiments provide the evidence that Mannich base 3FB3FA8H leads to P53-mediated apoptosis. Inducing apoptosis by this mechanism could be of central importance in reducing tumor burden which can be a good prospect for neuroblastoma patients.
Collapse
Affiliation(s)
- Syed Saad Hussain
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Shaheen Faizi
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Kinza Rafi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Shabana U Simjee
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan. .,Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
145
|
Wang H, Gong P, Li J, Fu Y, Zhou Z, Liu L. Role of CD133 in human embryonic stem cell proliferation and teratoma formation. Stem Cell Res Ther 2020; 11:208. [PMID: 32460847 PMCID: PMC7251672 DOI: 10.1186/s13287-020-01729-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/29/2020] [Accepted: 05/13/2020] [Indexed: 01/16/2023] Open
Abstract
Background Pluripotent stem cells (PSCs), including human embryonic stem cells (hESCs), hold great potential for regenerative medicine and cell therapy. One of the major hurdles hindering the clinical development of PSC-based therapy is the potential risk of tumorigenesis. CD133 (Prominin 1, PROM1) is a transmembrane protein whose mRNA and glycosylated forms are highly expressed in many human cancer cell types. CD133 also serves as a cancer stem cell (CSC) marker associated with cancer progression and patient outcome. Interestingly, CD133 is highly expressed in hESCs as well as in human preimplantation embryos, but its function in hESCs has remained largely unknown. Methods CD133 knockout hESC WA26 cell line was generated with CRISPR/Cas9. CD133 knockout and wide type hESC lines were subjected to pluripotency, proliferation, telomere biology, and teratoma tests; the related global changes and underlying mechanisms were further systemically analyzed by RNA-seq. Results CD133 deficiency did not affect hESC pluripotency or in vivo differentiation into three germ layers but significantly decreased cell proliferation. RNA-seq revealed that CD133 deficiency dysregulated the p53, PI3K-Akt, AMPK, and Wnt signaling pathways. Alterations in these pathways have been implicated in tumor proliferation and apoptotic escape. Conclusions Our data imply that CD133 could be an additional target and used as a selective marker to sort and eliminate undifferentiated cells in reducing potential teratoma formation risk of hESCs in regenerative medicine.
Collapse
Affiliation(s)
- Hua Wang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Peng Gong
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jie Li
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yudong Fu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhongcheng Zhou
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China. .,Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
146
|
Shakya B, Yadav PN. Thiosemicarbazones as Potent Anticancer Agents and their Modes of Action. Mini Rev Med Chem 2020; 20:638-661. [DOI: 10.2174/1389557519666191029130310] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 11/22/2022]
Abstract
:Thiosemicarbazones (TSCs) are a class of Schiff bases usually obtained by the condensation of thiosemicarbazide with a suitable aldehyde or ketone. TSCs have been the focus of chemists and biologists due to their wide range of pharmacological effects. One of the promising areas in which these excellent metal chelators are being developed is their use against cancer. TSCs have a wide clinical antitumor spectrum with efficacy in various tumor types such as leukemia, pancreatic cancer, breast cancer, non-small cell lung cancer, cervical cancer, prostate cancer and bladder cancer. To obtain better activity, different series of TSCs have been developed by modifying the heteroaromatic system in their molecules. These compounds possessed significant antineoplastic activity when the carbonyl attachment of the side chain was located at a position α to the ring nitrogen atom, whereas attachment of the side chain β or γ to the heterocyclic N atom resulted in inactive antitumor agents. In addition, replacement of the heterocyclic ring N with C also resulted in a biologically inactive compound suggesting that a conjugated N,N,S-tridentate donor set is essential for the biological activities of thiosemicarbazones. Several possible mechanisms have been implemented for the anticancer activity of thiosemicarbazones.
Collapse
Affiliation(s)
- Bhushan Shakya
- Amrit Campus, Tribhuvan University, Thamel, Kathmandu, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
147
|
Huang D, Liu H, Zhu A, Zhou Y, Li Y. Forebrain excitatory neuron-specific SENP2 knockout mouse displays hyperactivity, impaired learning and memory, and anxiolytic-like behavior. Mol Brain 2020; 13:59. [PMID: 32290845 PMCID: PMC7155287 DOI: 10.1186/s13041-020-00591-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Sentrin/SUMO-specific protease 2 (SENP2) is a member of SENPs family involved in maturation of SUMO precursors and deSUMOylation of specific target, and is highly expressed in the central nervous system (CNS). Although SENP2 has been shown to modulate embryonic development, fatty acid metabolism, atherosclerosis and epilepsy, the function of SENP2 in the CNS remains poorly understood. To address the role of SENP2 in the CNS and its potential involvement in neuropathology, we generated SENP2 conditional knockout mice by crossing floxed SENP2 mice with CaMKIIα-Cre transgenic mice. Behavioral tests revealed that SENP2 ablation induced hyper-locomotor activity, anxiolytic-like behaviors, spatial working memory impairment and fear-associated learning defect. In line with these observations, our RNA sequencing (RNA-seq) data identified a variety of differential expression genes that are particularly enriched in locomotion, learning and memory related biologic process. Taken together, our results indicated that SENP2 plays a critical role in emotional and cognitive regulation. This SENP2 conditional knockout mice model may help reveal novel mechanisms that underlie a variety of neuropsychiatric disorders associated with anxiety and cognition.
Collapse
Affiliation(s)
- Dehua Huang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Huiqing Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Aoxue Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Yi Zhou
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
148
|
Kooptiwut S, Samon K, Semprasert N, Suksri K, Yenchitsomanus PT. Prunetin Protects Against Dexamethasone-Induced Pancreatic Β-Cell Apoptosis via Modulation of p53 Signaling Pathway. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20916328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Long-term administration of dexamethasone results in insulin resistance and pancreatic β-cell apoptosis. Prunetin (an O-methylated isoflavone, a type of flavonoid) is demonstrated to protect diabetes, but the molecular mechanism of this protection is still unclear. This study thus aims to investigate how prunetin protects against dexamethasone-induced pancreatic β-cell apoptosis. Rat insulinoma (INS-1) cells were cultured in medium with or without dexamethasone in the presence or absence of prunetin or pifithrin-α, a p53 inhibitor. Cell apoptosis was measured by Annexin V/propidium iodide staining. Dexamethasone significantly induced INS-1 apoptosis but dexamethasone plus prunetin significantly reduced INS-1 apoptosis. Dexamethasone-treated INS-1 upregulated p53 protein expression; the induction of p53 was also reduced in the presence of RU486, a glucocorticoid receptor (GR) inhibitor. This suggested that dexamethasone induced P53 via GR. Dexamethasone-treated INS-1 significantly increased p53, Bax, and Rb protein expressions, whereas treatments of dexamethasone plus prunetin or pifithrin-α significantly decreased these protein expressions. In addition, dexamethasone significantly decreased B-cell lymphoma 2 (Bcl2), while dexamethasone plus prunetin or pifithrin-α significantly increased Bcl2. Dexamethasone significantly increased caspase-3 activity while co-treatment of dexamethasone plus prunetin or pifithrin-α significantly decreased caspase-3 activity to the control level. Taken together, our results revealed that prunetin protected against dexamethasone-induced pancreatic β-cells apoptosis via modulation of the p53 signaling pathway.
Collapse
Affiliation(s)
- Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokwan Samon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Namoiy Semprasert
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanchana Suksri
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
149
|
Wang Y, Wang JN, Chen XZ, Hu QX, Liu QQ, Wu G. Heat stress-induced expression of Px-pdrg and Px-aspp2 in insecticide-resistant and -susceptible Plutella xylostella. BULLETIN OF ENTOMOLOGICAL RESEARCH 2020; 110:177-184. [PMID: 31559929 DOI: 10.1017/s0007485319000543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
p53, DNA damage regulated gene (PDRG) and apoptosis-stimulating p53 protein 2 (ASPP2) are p53-related genes which can promote apoptosis. The full-length cDNA sequence of the Px-pdrg and Px-aspp2 genes were characterized and their mRNA expression dynamics under heat stress were studied in diamondback moth (DBM) Plutella xylostella collected from Fuzhou, China. The full-length cDNA of Px-pdrg and Px-aspp2 spans 721 and 4201 bp, containing 395 and 3216 bp of the open reading frame, which encode a putative protein comprising 130 and 1072 amino acids with a calculated molecular weight of 14.58 and 118.91 kDa, respectively. As compared to 25°C, both Px-pdrg and Px-aspp2 were upregulated in chlorpyrifos-resistant (Rc) and -susceptible (Sm) strains of DBM adults and pupae under heat stress. In addition, Rc DBM showed a significantly higher expression level of Px-pdrg and Px-aspp2 in contrast to Sm DBM. The results indicate that high temperature can significantly promote apoptosis process, especially in Rc-DBM. Significant fitness cost in Rc-DBM might be associated with drastically higher transcript abundance of Px-pdrg and Px-aspp2 under the heat stress.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jing Nan Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xue Zhun Chen
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qi Xing Hu
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qi Qing Liu
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| | - Gang Wu
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
150
|
Valcourt DM, Dang MN, Scully MA, Day ES. Nanoparticle-Mediated Co-Delivery of Notch-1 Antibodies and ABT-737 as a Potent Treatment Strategy for Triple-Negative Breast Cancer. ACS NANO 2020; 14:3378-3388. [PMID: 32083466 PMCID: PMC7098846 DOI: 10.1021/acsnano.9b09263] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for nearly one-quarter of all breast cancer cases, but effective targeted therapies for this disease remain elusive because TNBC cells lack expression of the three most common receptors seen on other subtypes of breast cancer. Here, we exploit TNBC cells' overexpression of Notch-1 receptors and Bcl-2 anti-apoptotic proteins to provide an effective targeted therapy. Prior studies have shown that the small molecule drug ABT-737, which inhibits Bcl-2 to reinstate apoptotic signaling, is a promising candidate for TNBC therapy. However, ABT-737 is poorly soluble in aqueous conditions, and its orally bioavailable derivative causes severe thrombocytopenia. To enable targeted delivery of ABT-737 to TNBC and enhance its therapeutic efficacy, we encapsulated the drug in poly(lactic-co-glycolic acid) nanoparticles (NPs) that were functionalized with Notch-1 antibodies to produce N1-ABT-NPs. The antibodies in this NP platform enable both TNBC cell-specific binding and suppression of Notch signaling within TNBC cells by locking the Notch-1 receptors in a ligand unresponsive state. This Notch inhibition potentiates the effect of ABT-737 by up-regulating Noxa, resulting in effective killing of TNBC cells. We present the results of in vitro studies that demonstrate N1-ABT-NPs can preferentially bind TNBC cells versus noncancerous breast epithelial cells to effectively regulate Bcl-2 and Notch signaling to induce cell death. Further, we show that N1-ABT-NPs can accumulate in subcutaneous TNBC xenograft tumors in mice following systemic administration to reduce tumor burden and extend animal survival. Together, these findings demonstrate that NP-mediated co-delivery of Notch-1 antibodies and ABT-737 is a potent treatment strategy for TNBC that may improve patient outcomes with further development and implementation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Biphenyl Compounds/chemistry
- Biphenyl Compounds/metabolism
- Biphenyl Compounds/pharmacology
- Cell Death/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Female
- Humans
- Mammary Neoplasms, Experimental/diagnostic imaging
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mice
- Mice, Nude
- Nanoparticles/chemistry
- Nanoparticles/metabolism
- Nitrophenols/chemistry
- Nitrophenols/metabolism
- Nitrophenols/pharmacology
- Optical Imaging
- Piperazines/chemistry
- Piperazines/metabolism
- Piperazines/pharmacology
- Receptor, Notch1/chemistry
- Receptor, Notch1/metabolism
- Sulfonamides/chemistry
- Sulfonamides/metabolism
- Sulfonamides/pharmacology
- Triple Negative Breast Neoplasms/diagnostic imaging
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/metabolism
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Mackenzie A Scully
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
- Helen F. Graham Cancer Center and Research Institute, 4701 Ogletown Stanton Road, Newark, Delaware 19713, United States
| |
Collapse
|