1
|
Hu J, Yang B, Tao Z, Chen J, Zhang X, Wang S, Xing G, Ngeng NA, Malik A, Appiah-Kubi K, Farina M, Skalny AV, Tinkov AA, Aschner M, Lu R. The role of HIF-1α/BNIP3/mitophagy in acrylonitrile-induced neuronal death in HT22 cells and mice: A potential neuroprotection target. Chem Biol Interact 2025; 406:111327. [PMID: 39615733 DOI: 10.1016/j.cbi.2024.111327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
Acrylonitrile (AN) is a widely utilized organic compound in the production of diverse industrial synthetic materials. While acute exposure to AN can cause neurotoxicity, the precise mechanism remains unclear. Hypoxia-inducible factor 1 alpha (HIF-1α) is a pivotal transcription factor that coordinates and orchestrates multiple physiological processes to adapt to hypoxic conditions, ensuring cellular survival and homeostasis. In this study, we used in vitro (cultured mouse hippocampal neuronal cell line, HT22) and in vivo (AN exposed mice) approaches to investigate the potential modulator effects of HIF-1α in AN-induced neurotoxicity. In vitro, AN exposure caused concentration-dependent toxicity in HT22 cells, which was paralleled by increased Bax levels while decreasing Bcl-2. Exposure to AN resulted in reduced protein levels of HIF-1α, Bcl-2 19-kDa interacting protein 3 (BNIP3), microtubule-associated protein 1 light chain 3 beta (LC3B) and Beclin1, while increased the protein levels of the translocase of outer mitochondrial membrane 20 (TOM20). Furthermore, mitochondrial morphology and function were compromised, suggesting that AN impaired HIF-1α/BNIP3-mediated mitochondrial autophagy and promoted apoptosis. Treatment with a HIF-1α activator, cobalt chloride (CoCl2), reversed these effects, while pretreatment with a HIF-1α inhibitor, 2-methoxyestradiol (2-MeOE2), augmented them. In BNIP3 overexpressing HT22 cells, enhanced cell viability and reduced apoptosis rates were observed. Furthermore, the HIF-1α/BNIP3 pathway was activated by the prolyl hydroxylase (PHD2) inhibitor, deferoxamine (DFO), which increased HT22 cell viability. Similarly, the activation of HIF-1α by administering 20 mg/kg of CoCl2 was found to alleviate neurotoxicity in mice. This treatment enhanced elevations of autophagy protein expression and co-localization of BNIP3 and LC3B. In summary, under normoxia, AN induced neurotoxicity by promoting PHD2-mediated HIF-1α degradation, disrupted the BNIP3-mediated mitophagy pathway, and enhanced apoptosis. Our findings underscore the effect of the HIF-1α/BNIP3-mediated mitochondrial autophagy in AN-induced neurotoxicity and suggest potential therapeutic strategies involving HIF-1α activation or BNIP3 overexpression for AN poisoning treatment.
Collapse
Affiliation(s)
- Jing Hu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zehua Tao
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jian Chen
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xinyu Zhang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Ngwa Adeline Ngeng
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Abdul Malik
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Kwaku Appiah-Kubi
- Department of Applied Biology, C. K. Tedam University of Technology and Applied Sciences, Navrongo, UK-0215-5321, Ghana
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Anatoly V Skalny
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia; Orenburg State University, Pobedy Ave.13, Orenburg, 460018, Russia
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia; Orenburg State University, Pobedy Ave.13, Orenburg, 460018, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150000, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; Experimental Research Center, Affiliated Kunshan Hospital, Jiangsu University, Kunshan, Jiangsu, 215300, China.
| |
Collapse
|
2
|
Chen XL, Xu YM, Lau ATY. Toxic metals in the regulation of epithelial-mesenchymal plasticity: demons or angels? Cancer Cell Int 2022; 22:237. [PMID: 35897065 PMCID: PMC9327425 DOI: 10.1186/s12935-022-02638-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 06/21/2022] [Indexed: 02/08/2023] Open
Abstract
Epithelial cells can trans-differentiate into motile mesenchymal cells through a dynamic process known as epithelial-mesenchymal transition (EMT). EMT is crucial in embryonic development and wound healing but also contributes to human diseases such as organ fibrosis and cancer progression. Heavy metals are environmental pollutants that can affect human health in various ways, including causing cancers. The cytotoxicity and carcinogenicity of heavy metals are complex, and studies have demonstrated that some of these metals can affect the progress of EMT. Here, we focus on reviewing the roles of six environmentally common toxic metals concerning EMT: arsenic (AS), cadmium (Cd), cobalt (Co), chromium (Cr), nickel (Ni), and copper (Cu). Noteworthily, the effects of these elements on EMT may vary according to the form, dose, and exposure time; the dual role of heavy metals (e.g., AS, Cd, and Cu) on EMT is also observed, in which, sometimes they can promote while sometimes inhibit the EMT process. Given the vast number of toxicologically relevant metals that exist in nature, we believe a comprehensive understanding of their effects on EMT is required to dictate in what circumstances these metals act more likely as demons or angels.
Collapse
Affiliation(s)
- Xu-Li Chen
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041 People’s Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041 People’s Republic of China
| | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041 People’s Republic of China
| |
Collapse
|
3
|
Huy TXN, Nguyen TT, Reyes AWB, Kim H, Min W, Lee HJ, Lee JH, Kim S. Cobalt (II) Chloride Regulates the Invasion and Survival of Brucella abortus 544 in RAW 264.7 Cells and B6 Mice. Pathogens 2022; 11:596. [PMID: 35631117 PMCID: PMC9143810 DOI: 10.3390/pathogens11050596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
The effects of Cobalt (II) chloride (CoCl2) in the context of Brucella abortus (B. abortus) infection have not been evaluated so far. Firstly, we found that CoCl2 treatment inhibited the phagocytosis of B. abortus into RAW 264.7 cells. The inhibition of bacterial invasion was regulated by F-actin formation and associated with a reduction in the phosphorylation of ERK1/2 and HIF-1α expression. Secondly, the activation of trafficking regulators LAMP1, LAMP2, and lysosomal enzyme GLA at the transcriptional level activated immune responses, weakening the B. abortus growth at 4 h post-infection (pi). The silencing of HIF-1α increased bacterial survival at 24 h pi. At the same time, CoCl2 treatment showed a significant increase in the transcripts of lysosomal enzyme HEXB and cytokine TNF-α and an attenuation of the bacterial survival. Moreover, the enhancement at the protein level of HIF-1α was induced in the CoCl2 treatment at both 4 and 24 h pi. Finally, our results demonstrated that CoCl2 administration induced the production of serum cytokines IFN-γ and IL-6, which is accompanied by dampened Brucella proliferation in the spleen and liver of treated mice, and reduced the splenomegaly and hepatomegaly. Altogether, CoCl2 treatment contributed to host resistance against B. abortus infection with immunomodulatory effects.
Collapse
Affiliation(s)
- Tran X. N. Huy
- Institute of Applied Sciences, HUTECH University, 475A Dien Bien Phu St., Ward 25, Binh Thanh District, Ho Chi Minh City 72300, Vietnam;
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| | - Trang T. Nguyen
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| | - Alisha W. B. Reyes
- Department of Veterinary Paraclinical Sciences, College of Veterinary Medicine, University of the Philippines Los Baños, College, Laguna 4031, Philippines;
| | - Heejin Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| | - WonGi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| | - Hu J. Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| | - John H. Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Korea;
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (T.T.N.); (H.K.); (W.M.); (H.J.L.)
| |
Collapse
|
4
|
Yang J, Zheng XG, Wu YL, Wang AP, Wang CH, Chen WX, Zhong S, Yang H. Intestinal epithelial cell-derived exosomes package microRNA-23a-3p alleviate gut damage after ischemia/reperfusion via targeting MAP4K4. Biomed Pharmacother 2022; 149:112810. [PMID: 35303564 DOI: 10.1016/j.biopha.2022.112810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/19/2022] [Accepted: 03/07/2022] [Indexed: 11/25/2022] Open
Abstract
Intestinal epithelial cells (IECs) contribute to regulation of gut injury after intestinal ischemia/reperfusion (II/R). Exosomes are well documented to deliver bioactive molecules to recipient cells for the purpose of modulating cell function. However, the role of IEC-derived exosomes in gut damage after II/R and the underlying mechanisms remain unclear. Here, we investigated the effects of exosomal miR-23a-3p on gut damage using primary IECs that underwent oxygen-glucose deprivation (OGD) as well as II/R rats. We observed that exosomes released by IECs attenuated damage in IECs that underwent OGD in vitro (P < 0.05) as well as the degree of gut injury after an II/R assault in vivo (P < 0.05). Injection of miR-23a-3p knockdown exosomes aggravated the II/R injury, whereas PF-6260933, a small-molecule inhibitor of MAP4K4, partly reversed the injury. Underlying mechanistic studies revealed that exosomal miR-23a-3p attenuated gut damage by regulating its downstream target, MAP4K4.
Collapse
Affiliation(s)
- Jin Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military (Army) Medical University, 400046 Chongqing, China; Department of Pediatric Anesthesiology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Xin Guo Zheng
- Department of Pediatric Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Yan Ling Wu
- Department of Pediatric Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Ai Ping Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military (Army) Medical University, 400046 Chongqing, China
| | - Chen Hui Wang
- Department of Pediatric Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Wen Xin Chen
- Department of Pediatric Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China
| | - Shan Zhong
- Department of Pediatric Anesthesiology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China.
| | - Hui Yang
- Department of Pediatric Gastroenterology, Children's Hospital of Nanjing Medical University, 210008 Nanjing, China.
| |
Collapse
|
5
|
Xue F, Du W, Chen S, Ma M, Kuang Y, Chen J, Yi T, Chen H. Hypoxia-Induced Photogenic Radicals by Eosin Y for Efficient Phototherapy of Hypoxic Tumors. ACS APPLIED BIO MATERIALS 2020; 3:8962-8969. [PMID: 35019572 DOI: 10.1021/acsabm.0c01223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The current reported photosensitizers generally show a decreased reactive oxygen species (ROS) generation property under hypoxia conditions, which is the main reason for the clinical failure of photodynamic therapy (PDT) in treatment of solid tumors. Herein, for the first time, hypoxia-induced photogenic radicals by eosin Y (Eos) were reported for efficient phototherapy of hypoxic tumors. More importantly, Eos shows a higher ROS and radical production efficiency under hypoxia conditions than under normoxia conditions. The photogenic radicals were captured by electron paramagnetic resonance and further verified by ROS and radical probe. Introducing CoCl2 as a hypoxia inducer, the photoinduced therapy of the hypoxia cancer cell model and tumor-bearing mice indicated that bovine serum albumin-Eos in hypoxic tumor sites can produce even higher tumor toxicity, thereby crossing the clinical obstacles of hypoxic tumor therapy. This non-oxygen-dependent PDT may open up an avenue for fighting with hypoxia.
Collapse
Affiliation(s)
- Fengfeng Xue
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China
| | - Wenxian Du
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, P. R. China
| | - Shixiong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, P. R. China
| | - Ming Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| | - Yichen Kuang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, P. R. China
| | - Jufeng Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, P. R. China
| | - Tao Yi
- Department of Chemistry and Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Songhu Road 2005, Shanghai 200433, P. R. China
| | - Hangrong Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Dingxi Road 1295, Shanghai 200050, P. R. China.,School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Sub-lane Xiangshan, Hangzhou 310024, China
| |
Collapse
|
6
|
Tang J, Zheng C, Zheng F, Li Y, Wang YL, Aschner M, Guo Z, Yu G, Wu S, Li H. Global N6-methyladenosine profiling of cobalt-exposed cortex and human neuroblastoma H4 cells presents epitranscriptomics alterations in neurodegenerative disease-associated genes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 266:115326. [PMID: 32827984 DOI: 10.1016/j.envpol.2020.115326] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/19/2020] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
Excessive exposure to cobalt (Co) is known to make adverse impact on the nervous system, but its detailed mechanisms of neurotoxicity have yet to be determined. In this study, C57BL/6 mice (0, 4, 8, 16 mg/kg CoCl2, 30 days) and human neuroblastoma H4 cells (0, 100, 400, 600 μM CoCl2) were used as in vivo and in vitro models. Our results revealed that CoCl2 intraperitoneal injection caused significant impairments in learning and memory, as well as pathological damage in the nervous system. We further certificated the alteration of m6A methylation induced by CoCl2 exposure. Our findings demonstrate for the first time, significant differences in the degree of m6A modification, the biological function of m6A-modified transcripts between cortex and H4 cell samples. Specifically, MeRIP-seq and RNA-seq elucidate that CoCl2 exposure results in differentially m6A-modified and expressed genes, which were enriched in pathways involving synaptic transmission, and central nervous system (CNS) development. Mechanistic analyses revealed that CoCl2 remarkably changed m6A modification level by affecting the expression of m6A methyltransferase and demethylase, and decreasing the activity of demethylase. We observed variation of m6A modification in neurodegenerative disease-associated genes upon CoCl2 exposure and identified regulatory strategy between m6A and potential targets mRNA. Our novel findings provide novel insight into the functional roles of m6A modification in neurodegenerative damage caused by environmental neurotoxicants and identify Co-mediated specific RNA regulatory strategy for broadening the epigenetic regulatory mechanism of RNA induced by heavy metals.
Collapse
Affiliation(s)
- Jianping Tang
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China
| | - Chunyan Zheng
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China
| | - Fuli Zheng
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China
| | - Yuqing Li
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China
| | - Yuan-Liang Wang
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Zhenkun Guo
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China
| | - Guangxia Yu
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China
| | - Siying Wu
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China
| | - Huangyuan Li
- Fujian Key Lab of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, China; Key Lab of Environment and Health, School of Public Health, Fujian Medical University, China.
| |
Collapse
|
7
|
Hypoxia and EGF Stimulation Regulate VEGF Expression in Human Glioblastoma Multiforme (GBM) Cells by Differential Regulation of the PI3K/Rho-GTPase and MAPK Pathways. Cells 2019; 8:cells8111397. [PMID: 31698752 PMCID: PMC6912653 DOI: 10.3390/cells8111397] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and deadly cancers of the central nervous system (CNS). It is characterized by the presence of hypoxic regions, especially in the core, leading to an increase in vascularity. This increased vascularization is driven by the expression of the major angiogenic inducer VEGF and the indirect angiogenic inducer Epidermal growth factor (EGF), which stimulates VEGF expression. In this study, we examine the regulation of VEGF by both hypoxia and the EGF signaling pathway. We also examine the involvement of pathways downstream from EGF signaling, including the mitogen-activated protein kinase/extracellular regulated kinase (MAPK/ERK) pathway and the Phosphatidylinositol-3-kinase/RhoA/C (PI3K/RhoA/C) pathway in this regulation. Our results show that VEGF expression and secretion levels increase following either hypoxia or EGF stimulation, with the two stimuli signaling in parallel. We also observed an increase in ERK and protein kinase B (Akt) phosphorylation, in response to EGF stimulation, with kinetics that correlated with the kinetics of the effect on VEGF. Using pharmacological inhibitors against ERK and PI3K and small interfering RNAs (siRNAs) against RhoA and RhoC, we found that both the ERK and the PI3K/RhoA/C pathways have to cooperate in order to lead to an increase in VEGF expression, downstream from EGF. In response to hypoxia, however, only ERK was involved in the regulation of VEGF. Hypoxia also led to a surprising decrease in the activation of PI3K and RhoA/C. Finally, the decrease in the activation of these Rho-GTPases was found to be mediated through a hypoxia-driven overexpression of the Rho-GTPase GTPase activating protein (GAP), StarD13. Therefore, while under normoxic conditions, EGF stimulates the activation of both the PI3K and the MAPK pathways and the induction of VEGF, in glioblastoma cells, hypoxic conditions lead to the suppression of the PI3K/RhoA/C pathway and an exclusive switch to the MAPK pathway.
Collapse
|
8
|
Moringa oleifera extract attenuates the CoCl2 induced hypoxia of rat's brain: Expression pattern of HIF-1α, NF-kB, MAO and EPO. Biomed Pharmacother 2019; 109:1688-1697. [DOI: 10.1016/j.biopha.2018.11.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/04/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022] Open
|
9
|
Oyagbemi AA, Omobowale TO, Awoyomi OV, Ajibade TO, Falayi OO, Ogunpolu BS, Okotie UJ, Asenuga ER, Adejumobi OA, Hassan FO, Ola-Davies OE, Saba AB, Adedapo AA, Yakubu MA. Cobalt chloride toxicity elicited hypertension and cardiac complication via induction of oxidative stress and upregulation of COX-2/Bax signaling pathway. Hum Exp Toxicol 2018; 38:519-532. [PMID: 30596275 DOI: 10.1177/0960327118812158] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cobalt is a ferromagnetic metal with extensive industrial and biological applications. To assess the toxic effects of, and mechanisms involved in cobalt chloride (CoCl2)-induced cardio-renal dysfunctions. Male Wistar rats were exposed orally, daily through drinking water to 0 ppm (control), 150 ppm, 300 ppm, and 600 ppm of CoCl2, respectively. Following exposure, results revealed significant ( p < 0.05) rise in markers of oxidative stress, but decreased activities of catalase, glutathione peroxidase, glutathione-S-transferase, and reduced glutathione content in cardiac and renal tissues. There were significant increases in systolic, diastolic, and mean arterial blood pressure at the 300- and 600-ppm level of CoCl2-exposed rats relative to the control. Prolongation of QT and QTc intervals was observed in CoCl2 alone treated rats. Also, there were significant increases in the heart rates, and reduction in P wave, and PR duration of rats administered CoCl2. Histopathology of the kidney revealed peritubular and periglomerular inflammation, focal glomerular necrosis following CoCl2 exposure. Further, cyclooxygenase-2 and B-cell associated protein X expressions were upregulated in the cardiac and renal tissues of CoCl2-exposed rats relative to the control. Combining all, results from this study implicated oxidative stress, inflammation, and apoptosis as pathologic mechanisms in CoCl2-induced hypertension and cardiovascular complications of rats.
Collapse
Affiliation(s)
- A A Oyagbemi
- 1 Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - T O Omobowale
- 2 Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - O V Awoyomi
- 3 Federal College of Animal Health and Production Technology, Moor Plantation, Ibadan, Nigeria
| | - T O Ajibade
- 1 Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - O O Falayi
- 4 Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - B S Ogunpolu
- 2 Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - U J Okotie
- 3 Federal College of Animal Health and Production Technology, Moor Plantation, Ibadan, Nigeria
| | - E R Asenuga
- 5 Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Benin, Ibadan, Nigeria
| | - O A Adejumobi
- 2 Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - F O Hassan
- 1 Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - O E Ola-Davies
- 1 Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - A B Saba
- 4 Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - A A Adedapo
- 4 Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - M A Yakubu
- 4 Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria.,6 Department of Environmental and Interdisciplinary Sciences, College of Science, Engineering and Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, COPHS, Texas Southern University, Houston, TX, USA
| |
Collapse
|
10
|
Wang W, Huang X, Lin W, Qiu Y, He Y, Yu J, Xi Y, Ye X. Hypoxic preconditioned bone mesenchymal stem cells ameliorate spinal cord injury in rats via improved survival and migration. Int J Mol Med 2018; 42:2538-2550. [PMID: 30106084 PMCID: PMC6192716 DOI: 10.3892/ijmm.2018.3810] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
The unique hypoxic inflammatory microenvironment observed in the spinal cord following spinal cord injury (SCI) limits the survival and efficacy of transplanted bone mesenchymal stem cells (BMSCs). The aim of the present study was to determine whether hypoxic preconditioning (HP) increased the therapeutic effects of BMSC on SCI. BMSCs were pretreated with cobalt chloride (CoCl2) in vitro, and the proliferative apoptotic and migratory abilities of these hypoxic BMSCs (H‑BMSCs) were assessed. BMSCs and H‑BMSCs derived from green fluorescent protein (GFP) rats were transplanted into SCI rats in vivo. The neurological function, histopathology, inflammation, and number and migration of transplanted cells were examined. HP significantly enhanced BMSC migration (increased hypoxia inducible factor 1α and C‑X‑C motif chemokine receptor 4 expression) and tolerance to apoptotic conditions (decreased caspase‑3 and increased B‑cell lymphoma 2 expression) in vitro. In vivo, H‑BMSC transplantation significantly improved neurological function, decreased spinal cord damage and suppressed the inflammatory response associated with microglial activation. The number of GFP‑positive cells in the SCI core and peripheral region of H‑BMSC animals was increased compared with that in those of BMSC animals, suggesting that HP may increase the survival and migratory abilities of BMSCs and highlights their therapeutic potential for SCI.
Collapse
Affiliation(s)
- Weiheng Wang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| | - Xiaodong Huang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| | - Wenbo Lin
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| | - Yuanyuan Qiu
- Department of Respiration, Shanghai Electric Power Hospital, Shanghai 200050
| | - Yunfei He
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
- Department of Orthopedics, Lanzhou General Hospital of Lanzhou Military Command Region, Lanzhou, Gansu 730050, P.R. China
| | - Jiangming Yu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| | - Yanhai Xi
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| | - Xiaojian Ye
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003
| |
Collapse
|
11
|
Li L, Wang Q, Yuan Z, Chen A, Liu Z, Li H, Wang Z. Long non-coding RNA H19 contributes to hypoxia-induced CPC injury by suppressing Sirt1 through miR-200a-3p. Acta Biochim Biophys Sin (Shanghai) 2018; 50:950-959. [PMID: 30137188 DOI: 10.1093/abbs/gmy093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/28/2022] Open
Abstract
Cardiomyocyte death is the chief obstacle that prevents the heart function recovery in myocardial infarction (MI)-induced heart failure (HF). Cardiac progenitor cells (CPCs)-based myocardial regeneration has provided a promising method for heart function recovery after MI. However, CPCs can easily lose their proliferation ability due to oxygen deficiency in infarcted myocardium. Revealing the underlying molecular mechanism for CPC proliferation is critical for effective MI therapy. In the present study, we set up a CoCl2-induced hypoxia model in CPCs. We found that the expression of long non-coding RNA H19 was significantly down-regulated in CPCs after hypoxia stimuli. In addition, H19 suppression attenuated the proliferation and migration of CPCs under hypoxia stress. Furthermore, we discovered that H19 regulated the proliferation and migration of CPCs through mediating the expression of Sirt1 which is a target of miR-200a-3p under hypoxia. In conclusion, our findings demonstrate a novel regulatory mechanism for the proliferation and migration of CPCs under hypoxia condition, which provides useful information for the development of new therapeutic targets for MI therapy.
Collapse
Affiliation(s)
- Linlin Li
- College of Life Sciences, Peking University, Beijing, China
| | - Qiuyun Wang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zhize Yuan
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Anqing Chen
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zuyun Liu
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Haiqing Li
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| | - Zhe Wang
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai, China
| |
Collapse
|
12
|
Lespay-Rebolledo C, Perez-Lobos R, Tapia-Bustos A, Vio V, Morales P, Herrera-Marschitz M. Regionally Impaired Redox Homeostasis in the Brain of Rats Subjected to Global Perinatal Asphyxia: Sustained Effect up to 14 Postnatal Days. Neurotox Res 2018; 34:660-676. [PMID: 29959728 DOI: 10.1007/s12640-018-9928-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022]
Abstract
The present report evaluates the effect of global perinatal asphyxia on several parameters of oxidative stress and cell viability in rat brain tissue sampled at an extended neonatal period up to 14 days, a period characterised by intensive neuritogenesis, synaptogenesis, synaptic consolidation, pruning and delayed cell death. Perinatal asphyxia was induced by immersing foetus-containing uterine horns removed by a caesarean section from on term rat dams into a water bath at 37 °C for 21 min. Asphyxia-exposed and sibling caesarean-delivered foetuses were manually resucitated and nurtured by surrogate dams for 1 to 14 postnatal (P) days. Brain samples (mesencephalon, telencephalon and hippocampus) were assayed for glutathione (reduced and oxidated levels; spectrophotometry), tissue reducing capacity (potassium ferricyanide reducing assay, FRAP), catalase (the key enzyme protecting against oxidative stress and reactive oxygen species, Western blots and ELISA) and cleaved caspase-3 (the key executioner of apoptosis, Western blots) levels. It was found that global PA produced a regionally specific and sustained increase in GSSG/GSH ratio, a regionally specific decrease in tissue reducing capacity and a regionally and time specific decrease of catalase activity and increase of cleaved caspase-3 levels. The present study provides evidence for regionally impaired redox homeostasis in the brain of rats subjected to global PA, an effect observed up to P14, mainly affecting mesencephalon and hippocampus, suggesting a sustained oxidative stress after the posthypoxia period. The oxidative stress observed postnatally can in part be associated to a respiratory apneic-like deficit, since there was a statistically significant decrease in respiration frequency in AS compared to CS neonates, also up to P14, together with the signs of a decreased peripheral blood perfusion (pink-blue skin colour in AS, compared to the pink colour observed in all CS neonates). It is proposed that PA implies a long-term metabolic insult, triggered by the length of hypoxia, the resuscitation/reoxigenation manoevres, but also by the developmental stage of the affected brain regions, and the integrity of cardiovascular and respiratory physiological functions, which are fundamental for warrantying a proper development.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Ronald Perez-Lobos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Andrea Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Valentina Vio
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
| | - Paola Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile
- Department Neuroscience, Medical Faculty, University of Chile, Santiago, Chile
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia 1027, PO Box 8389100, Santiago, Chile.
| |
Collapse
|
13
|
Wang W, Wang Y, Deng G, Ma J, Huang X, Yu J, Xi Y, Ye X. Transplantation of Hypoxic-Preconditioned Bone Mesenchymal Stem Cells Retards Intervertebral Disc Degeneration via Enhancing Implanted Cell Survival and Migration in Rats. Stem Cells Int 2018; 2018:7564159. [PMID: 29535780 PMCID: PMC5832130 DOI: 10.1155/2018/7564159] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/13/2017] [Accepted: 11/22/2017] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Special hypoxic and hypertonic microenvironment in intervertebral discs (IVDs) decreases the treatment effect of cell transplantation. We investigated the hypothesis that hypoxic preconditioning (HP) could improve the therapeutic effect of bone mesenchymal stem cells (BMSCs) to IVD degeneration. METHODS BMSCs from green fluorescent protein-transgenic rats were pretreated with cobalt chloride (CoCl2, 100 μM, 24 h) for hypoxic conditions in vitro. Apoptosis (related pathways of caspase-3 and bcl-2) and migration (related pathways of HIF-1α and CXCR4) were detected in BMSCs. In vivo, BMSCs and HP BMSCs (H-BMSCs) were injected into the rat model of IVD degeneration. The IVD height, survival, migration, and differentiation of transplanted BMSCs and matrix protein expression (collagen II, aggrecan, and MMP-13) were tested. RESULTS H-BMSCs could extensively decrease IVD degeneration by increasing IVD height and collagen II and aggrecan expressions when compared with BMSCs. Significantly, more GFP-positive BMSCs were observed in the nucleus pulposus and annulus fibrosus regions of IVD. HP could significantly decrease BMSC apoptosis (activating bcl-2 and inhibiting caspase-3) and improve BMSC migration (increasing HIF-1α and CXCR4) in vitro. CONCLUSION HP could significantly enhance the capacity of BMSCs to repair DDD by increasing the survival and migration of implanted cells and increasing matrix protein expression.
Collapse
Affiliation(s)
- Weiheng Wang
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yang Wang
- Department of Orthopaedics, Nanjing General Hospital, Nanjing 210000, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Jun Ma
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xiaodong Huang
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jiangming Yu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yanhai Xi
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xiaojian Ye
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
14
|
Chen DW, Wang H, Bao YF, Xie K. Notch signaling molecule is involved in the invasion of MiaPaCa2 cells induced by CoCl2 via regulating epithelial‑mesenchymal transition. Mol Med Rep 2018; 17:4965-4972. [PMID: 29393429 PMCID: PMC5865956 DOI: 10.3892/mmr.2018.8502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
Pancreatic cancer exhibits a high mortality rate resulting from metastasis and there is currently no effective treatment strategy. Hypoxia serves an important role in cancer cells, where cellular metabolic rate is high. The underlying mechanisms that trigger hypoxia and the invasion of pancreatic cancer cells remain unknown. Investigation of the importance of hypoxia in the invasion of pancreatic cancer cells for potential, novel treatment strategies is of primary concern. Cell Counting Kit-8 assay, invasion assay, western blotting and reverse transcription-quantitative polymerase chain reaction were used to investigate invasion and epithelial mesenchymal transition (EMT) and the expression of Notch1 in MiaPaCa2 cells treated with cobalt II chloride (CoCl2). Hypoxia-inducible factor 1α (HIF-1α) small interfering (si)RNA and Notch1 inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) were also selected to investigate these mechanisms. Data indicated that CoCl2 increased the invasion ability and altered EMT in MiaPaCa2 cells. CoCl2 regulated the expression of HIF-1α and Notch1 in MiaPaCa2 cells. In addition, HIF-1α siRNA inhibited the effects of CoCl2 on the expression of Notch1 and decreased Snail, EMT and invasion in MiaPaCa2 cells. DAPT increased the expression of epithelial-cadherin and decreased the content of neural-cadherin, Snail and invasion in MiaPaCa2 cells in the presence or absence of CoCl2. CoCl2 promoted invasion by stimulating the expression of HIF-1α and regulating the expression of Notch1 and EMT in MiaPaCa2 cells. Targeting the Notch1 signaling molecule may be a novel treatment strategy for the prevention and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ding-Wei Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Hong Wang
- Zhejiang Medical College, Hangzhou, Zhejiang 310053, P.R. China
| | - Ya-Fang Bao
- Caihe Street Community Health Service Center, Hangzhou, Zhejiang 310016, P.R. China
| | - Kun Xie
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
15
|
Zeeshan M, Murugadas A, Ghaskadbi S, Ramaswamy BR, Akbarsha MA. Ecotoxicological assessment of cobalt using Hydra model: ROS, oxidative stress, DNA damage, cell cycle arrest, and apoptosis as mechanisms of toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 224:54-69. [PMID: 28222982 DOI: 10.1016/j.envpol.2016.12.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 06/06/2023]
Abstract
The mechanisms underlying cobalt toxicity in aquatic species in general and cnidarians in particular remain poorly understood. Herein we investigated cobalt toxicity in a Hydra model from morphological, histological, developmental, and molecular biological perspectives. Hydra, exposed to cobalt (0-60 mg/L), were altered in morphology, histology, and regeneration. Exposure to standardized sublethal doses of cobalt impaired feeding by affecting nematocytes, which in turn affected reproduction. At the cellular level, excessive ROS generation, as the principal mechanism of action, primarily occurred in the lysosomes, which was accompanied by the upregulation of expression of the antioxidant genes SOD, GST, GPx, and G6PD. The number of Hsp70 and FoxO transcripts also increased. Interestingly, the upregulations were higher in the 24-h than in the 48-h time-point group, indicating that ROS overwhelmed the cellular defense mechanisms at the latter time-point. Comet assay revealed DNA damage. Cell cycle analysis indicated the induction of apoptosis accompanied or not by cell cycle arrest. Immunoblot analyses revealed that cobalt treatment triggered mitochondria-mediated apoptosis as inferred from the modulation of the key proteins Bax, Bcl-2, and caspase-3. From this data, we suggest the use of Hydra as a model organism for the risk assessment of heavy metal pollution in aquatic ecosystems.
Collapse
Affiliation(s)
- Mohammed Zeeshan
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Dept. of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India
| | - Anbazhagan Murugadas
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Dept. of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India
| | - Surendra Ghaskadbi
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune 411004, India
| | | | - Mohammad Abdulkader Akbarsha
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Department of Food Science and Nutrition, College of Food and Agriculture, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
16
|
A Special Extract of Bacopa monnieri (CDRI-08)-Restored Memory in CoCl2-Hypoxia Mimetic Mice Is Associated with Upregulation of Fmr-1 Gene Expression in Hippocampus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:347978. [PMID: 26413121 PMCID: PMC4564622 DOI: 10.1155/2015/347978] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 04/01/2015] [Indexed: 11/18/2022]
Abstract
Fragile X mental retardation protein (FMRP) is a neuronal translational repressor and has been implicated in learning, memory, and cognition. However, the role of Bacopa monnieri extract (CDRI-08) in enhancing cognitive abilities in hypoxia-induced memory impairment via Fmr-1 gene expression is not known. Here, we have studied effects of CDRI-08 on the expression of Fmr-1 gene in the hippocampus of well validated cobalt chloride (CoCl2)-induced hypoxia mimetic mice and analyzed the data with alterations in spatial memory. Results obtained from Morris water maze test suggest that CoCl2 treatment causes severe loss of spatial memory and CDRI-08 is capable of reversing it towards that in the normal control mice. Our semiquantitative RT-PCR, Western blot, and immunofluorescence microscopic data reveal that CoCl2-induced hypoxia significantly upregulates the expression of Hif-1α and downregulates the Fmr-1 expression in the hippocampus, respectively. Further, CDRI-08 administration reverses the memory loss and this is correlated with significant downregulation of Hif-1α and upregulation of Fmr-1 expression. Our data are novel and may provide mechanisms of hypoxia-induced impairments in the spatial memory and action of CDRI-08 in the recovery of hypoxia led memory impairment involving Fmr-1 gene encoded protein called FMRP.
Collapse
|