1
|
Zou M, Chen W, Li J, Qi X, Wang X, Liu F, Hu J, Zhang Q. Apoptosis Signal-Regulated Kinase-1 Promotes Nucleus Pulposus Cell Senescence and Apoptosis to Regulate Intervertebral Disc Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1737-1751. [PMID: 38879082 DOI: 10.1016/j.ajpath.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 06/27/2024]
Abstract
This study investigated the role of apoptosis signal-regulated kinase-1 (ASK1) in intervertebral disc degeneration (IDD). The nucleus pulposus (NP) tissues of non-IDD and IDD patients were subjected to hematoxylin and eosin, Safranin O-fast green, and immunohistochemical staining. Quantitative real-time PCR was used to assess the ASK1 mRNA level within NP tissue samples and cells. The Cell Counting Kit-8 assay, senescence-associated β-galactosidase staining, and flow cytometry were conducted to assess the viability, senescence, and apoptosis of NP cells, respectively. Extracellular matrix-related factors were detected using Western blot analysis. Furthermore, the effect of ASK1 on the IDD rat model was evaluated. Finally, c-Jun N-terminal kinase (JNK) inhibitors were used to verify the effect of the JNK/p38 signaling on IDD. ASK1 mRNA and protein were up-regulated within NP tissue samples from the IDD group, IL-1β-stimulated NP cells, and IDD rats. ASK1 inhibition promoted cell viability and repressed the senescence and apoptosis of NP cells, promoted collagen II and aggrecan, inhibited matrix metalloproteinase 3/9 and a disintegrin and metalloproteinase with thrombospondin motifs 4/5 protein levels, and increased NP cells in rat intervertebral disc tissues. ASK1 overexpression exerted the opposite effects of ASK1 inhibition on NP cells. Additionally, JNK/p38 signaling suppression could reverse the ASK1 up-regulation-induced dysfunction. In conclusion, ASK1 facilitated the senescence and apoptosis of NP cells in promoting IDD progression via the JNK/p38 pathway.
Collapse
Affiliation(s)
- Mingxiang Zou
- Department of Spine Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenkang Chen
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Qi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaobin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fubing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiarui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianshi Zhang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Gu J, Guo C, Ruan J, Li K, Zhou Y, Gong X, Shi H. From ferroptosis to cuproptosis, and calcicoptosis, to find more novel metals-mediated distinct form of regulated cell death. Apoptosis 2024; 29:586-604. [PMID: 38324163 DOI: 10.1007/s10495-023-01927-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 02/08/2024]
Abstract
Regulated cell death (RCD), also known as programmed cell death (PCD), plays a critical role in various biological processes, such as tissue injury/repair, development, and homeostasis. Dysregulation of RCD pathways can lead to the development of many human diseases, such as cancer, neurodegenerative disorders, and cardiovascular diseases. Maintaining proper metal ion homeostasis is critical for human health. However, imbalances in metal levels within cells can result in cytotoxicity and cell death, leading to a variety of diseases and health problems. In recent years, new types of metal overload-induced cell death have been identified, including ferroptosis, cuproptosis, and calcicoptosis. This has prompted us to examine the three defined metal-dependent cell death types, and discuss other metals-induced ferroptosis, cuproptosis, and disrupted Ca2+ homeostasis, as well as the roles of Zn2+ in metals' homeostasis and related RCD. We have reviewed the connection between metals-induced RCD and various diseases, as well as the underlying mechanisms. We believe that further research in this area will lead to the discovery of novel types of metal-dependent RCD, a better understanding of the underlying mechanisms, and the development of new therapeutic strategies for human diseases.
Collapse
Affiliation(s)
- Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Jiacheng Ruan
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Kongdong Li
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Yang Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212013, China.
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
3
|
Maslanka R, Bednarska S, Zadrag-Tecza R. Virtually identical does not mean exactly identical: Discrepancy in energy metabolism between glucose and fructose fermentation influences the reproductive potential of yeast cells. Arch Biochem Biophys 2024; 756:110021. [PMID: 38697344 DOI: 10.1016/j.abb.2024.110021] [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: 12/07/2023] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
The physiological efficiency of cells largely depends on the possibility of metabolic adaptations to changing conditions, especially on the availability of nutrients. Central carbon metabolism has an essential role in cellular function. In most cells is based on glucose, which is the primary energy source, provides the carbon skeleton for the biosynthesis of important cell macromolecules, and acts as a signaling molecule. The metabolic flux between pathways of carbon metabolism such as glycolysis, pentose phosphate pathway, and mitochondrial oxidative phosphorylation is dynamically adjusted by specific cellular economics responding to extracellular conditions and intracellular demands. Using Saccharomyces cerevisiae yeast cells and potentially similar fermentable carbon sources i.e. glucose and fructose we analyzed the parameters concerning the metabolic status of the cells and connected with them alteration in cell reproductive potential. Those parameters were related to the specific metabolic network: the hexose uptake - glycolysis and activity of the cAMP/PKA pathway - pentose phosphate pathway and biosynthetic capacities - the oxidative respiration and energy generation. The results showed that yeast cells growing in a fructose medium slightly increased metabolism redirection toward respiratory activity, which decreased pentose phosphate pathway activity and cellular biosynthetic capabilities. These differences between the fermentative metabolism of glucose and fructose, lead to long-term effects, manifested by changes in the maximum reproductive potential of cells.
Collapse
Affiliation(s)
- Roman Maslanka
- Institute of Biology, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland.
| | - Sabina Bednarska
- Institute of Biology, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland
| | - Renata Zadrag-Tecza
- Institute of Biology, College of Natural Sciences, University of Rzeszow, Rzeszow, Poland
| |
Collapse
|
4
|
Meuten TK, Dean GA, Thamm DH. Review: The PI3K-AKT-mTOR signal transduction pathway in canine cancer. Vet Pathol 2024; 61:339-356. [PMID: 37905509 DOI: 10.1177/03009858231207021] [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] [Indexed: 11/02/2023]
Abstract
Tumors in dogs and humans share many similar molecular and genetic features, incentivizing a better understanding of canine neoplasms not only for the purpose of treating companion animals, but also to facilitate research of spontaneously developing tumors with similar biologic behavior and treatment approaches in an immunologically competent animal model. Multiple tumor types of both species have similar dysregulation of signal transduction through phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB; AKT), and mechanistic target of rapamycin (mTOR), collectively known as the PI3K-AKT-mTOR pathway. This review aims to delineate the pertinent aspects of the PI3K-AKT-mTOR signaling pathway in health and in tumor development. It will then present a synopsis of current understanding of PI3K-AKT-mTOR signaling in important canine cancers and advancements in targeted inhibitors of this pathway.
Collapse
|
5
|
Guo Z, Zhang H, Huang T, Liu C. CCN3/NOV inhibition attenuates oxidative stress-induced apoptosis of mouse neural stem/progenitor cells by blocking the activation of p38 MAPK: An in vitro study. Brain Res 2024; 1827:148756. [PMID: 38199307 DOI: 10.1016/j.brainres.2024.148756] [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: 11/14/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Neural stem/progenitor cells (NSPCs) hold immense promise in clinical applications, yet the harsh conditions resulting from central nervous system (CNS) injuries, particularly oxidative stress, lead to the demise of both native and transplanted NSPCs. Cellular communication network factor 3 (CCN3) exhibits a protective effect against oxidative stress in various cell types. This study investigates the impact of CCN3 on NSPCs apoptosis induced by oxidative stress. To establish models of primary cultured mouse NSPCs under oxidative stress, we exposed them to 50 μM H2O2 for 4 h. Remarkably, pre-exposing CCN3 exacerbated the H2O2-induced decline in cell viability in a concentration-dependent manner. However, employing gene-targeted siRNA to inhibit CCN3 protected NSPCs against H2O2-induced cell death. Conversely, CCN3 replenishment reversed this protective effect, as evidenced by TUNEL staining, the ratio of Cleaved-caspase-3 to Pro-caspase-3, and Bcl-2/Bax. Further investigations revealed that CCN3 pretreatment increased the phosphorylation level of p38 MAPK, while silencing CCN3 diminished p38 MAPK activation. Ultimately, the impact of changes in CCN3 protein expression on H2O2-induced apoptosis was nullified using anisomycin (a p38 activator) and SB 203580 (a p38 inhibitor). Our findings suggest that CCN3 inhibition prevents H2O2-induced cell death in cultured mouse NSPCs via the p38 pathway. These discoveries may contribute to the development of strategies aimed at enhancing the survival of both endogenous and transplanted NSPCs following CNS oxidative stress insults.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanyue Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Tingqin Huang
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chongxiao Liu
- Department of Neurosurgery, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
6
|
Tavakoli Pirzaman A, Sadeghnezhad G, Azmoun Z, Eslami A, Mansoori R, Kazemi S, Hosseini SM. The effect of geraniol on nickel-induced embryotoxicity and cardiotoxicity in rats. Int J Immunopathol Pharmacol 2024; 38:3946320241272693. [PMID: 39393811 PMCID: PMC11483796 DOI: 10.1177/03946320241272693] [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: 03/04/2024] [Revised: 06/29/2024] [Accepted: 07/10/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Nickel (Ni), commonly-used heavy metals in industrial activities, can lead to embryo and organ toxicity, especially cardiovascular damage. Geraniol (GER) has various beneficial effects such as anti-oxidant, anti-inflammatory, anti-tumor, anti-ulcer, anti-microbial, and neuroprotective activities. OBJECTIVE The objective of this study was to investigate the effect of GER on Ni-induced embryotoxicity and cardiotoxicity in rats. METHODS 40 mother Wistar rats were randomly divided into five groups: Control, GER 250, Ni, Ni + GER 100, and Ni + GER 250. On the 20th day of pregnancy, the animals were sacrificed and fetuses along with blood and tissue samples were collocated for morphological, serological, biochemical, and histopathologic analysis. RESULTS Morphological assessments revealed GER's capacity to mitigate the incomplete ossification of fetal skeletons, indicating a potential safeguarding against the impact of Ni-induced embryotoxicity. Serological and biochemical analyses further affirm GER's role, with noteworthy reductions in cardiac injury markers, such as CRP, CKMB, CPK, LDH, and troponin, in response to GER administration, thereby suggesting its cardioprotective potential. Moreover, treatment with GER 250 could significantly reduce the level of MDA and increase the level of TAC compared to the Ni group. Histopathological examinations corroborated these findings, underscoring GER's ability to counteract cardiac injury and diminish structural damage in affected tissue. CONCLUSIONS These multidimensional analyses indicate the protective prowess of GER against Ni-induced embryotoxic and cardiotoxic effects, shedding light on its potential therapeutic significance in combating adverse impacts stemming from Ni exposure.
Collapse
Affiliation(s)
| | - Ghazaleh Sadeghnezhad
- Department of Veterinary Pathology, Babol-Branch, Islamic Azad University, Babol, Iran
| | - Zahra Azmoun
- Department of Veterinary Pathology, Babol-Branch, Islamic Azad University, Babol, Iran
| | - Amirreza Eslami
- Department of Veterinary Pathology, Babol-Branch, Islamic Azad University, Babol, Iran
| | - Razieh Mansoori
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | |
Collapse
|
7
|
Caligiuri A, Becatti M, Porro N, Borghi S, Marra F, Pastore M, Taddei N, Fiorillo C, Gentilini A. Oxidative Stress and Redox-Dependent Pathways in Cholangiocarcinoma. Antioxidants (Basel) 2023; 13:28. [PMID: 38247453 PMCID: PMC10812651 DOI: 10.3390/antiox13010028] [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: 10/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a primary liver tumor that accounts for 2% of all cancer-related deaths worldwide yearly. It can arise from cholangiocytes of biliary tracts, peribiliary glands, and possibly from progenitor cells or even hepatocytes. CCA is characterized by high chemoresistance, aggressiveness, and poor prognosis. Potentially curative surgical therapy is restricted to a small number of patients with early-stage disease (up to 35%). Accumulating evidence indicates that CCA is an oxidative stress-driven carcinoma resulting from chronic inflammation. Oxidative stress, due to enhanced reactive oxygen species (ROS) production and/or decreased antioxidants, has been recently suggested as a key factor in cholangiocyte oncogenesis through gene expression alterations and molecular damage. However, due to different experimental models and conditions, contradictory results regarding oxidative stress in cholangiocarcinoma have been reported. The role of ROS and antioxidants in cancer is controversial due to their context-dependent ability to stimulate tumorigenesis and support cancer cell proliferation or promote cell death. On these bases, the present narrative review is focused on illustrating the role of oxidative stress in cholangiocarcinoma and the main ROS-driven intracellular pathways. Heterogeneous data about antioxidant effects on cancer development are also discussed.
Collapse
Affiliation(s)
- Alessandra Caligiuri
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (A.C.); (F.M.); (M.P.)
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (M.B.); (N.P.); (S.B.); (N.T.)
| | - Nunzia Porro
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (M.B.); (N.P.); (S.B.); (N.T.)
| | - Serena Borghi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (M.B.); (N.P.); (S.B.); (N.T.)
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (A.C.); (F.M.); (M.P.)
| | - Mirella Pastore
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (A.C.); (F.M.); (M.P.)
| | - Niccolò Taddei
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (M.B.); (N.P.); (S.B.); (N.T.)
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy; (M.B.); (N.P.); (S.B.); (N.T.)
| | - Alessandra Gentilini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (A.C.); (F.M.); (M.P.)
| |
Collapse
|
8
|
Liu J, Han X, Zhang T, Tian K, Li Z, Luo F. Reactive oxygen species (ROS) scavenging biomaterials for anti-inflammatory diseases: from mechanism to therapy. J Hematol Oncol 2023; 16:116. [PMID: 38037103 PMCID: PMC10687997 DOI: 10.1186/s13045-023-01512-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Inflammation is a fundamental defensive response to harmful stimuli, but the overactivation of inflammatory responses is associated with most human diseases. Reactive oxygen species (ROS) are a class of chemicals that are generated after the incomplete reduction of molecular oxygen. At moderate levels, ROS function as critical signaling molecules in the modulation of various physiological functions, including inflammatory responses. However, at excessive levels, ROS exert toxic effects and directly oxidize biological macromolecules, such as proteins, nucleic acids and lipids, further exacerbating the development of inflammatory responses and causing various inflammatory diseases. Therefore, designing and manufacturing biomaterials that scavenge ROS has emerged an important approach for restoring ROS homeostasis, limiting inflammatory responses and protecting the host against damage. This review systematically outlines the dynamic balance of ROS production and clearance under physiological conditions. We focus on the mechanisms by which ROS regulate cell signaling proteins and how these cell signaling proteins further affect inflammation. Furthermore, we discuss the use of potential and currently available-biomaterials that scavenge ROS, including agents that were engineered to reduce ROS levels by blocking ROS generation, directly chemically reacting with ROS, or catalytically accelerating ROS clearance, in the treatment of inflammatory diseases. Finally, we evaluate the challenges and prospects for the controlled production and material design of ROS scavenging biomaterials.
Collapse
Affiliation(s)
- Jiatong Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoyue Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tingyue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Keyue Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhaoping Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu, 610041, China.
| |
Collapse
|
9
|
Daley R, Maddipatla V, Ghosh S, Chowdhury O, Hose S, Zigler JS, Sinha D, Liu H. Aberrant Akt2 signaling in the RPE may contribute to retinal fibrosis process in diabetic retinopathy. Cell Death Discov 2023; 9:243. [PMID: 37443129 DOI: 10.1038/s41420-023-01545-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/12/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Diabetic Retinopathy (DR) is a complication of diabetes that causes blindness in adults. Retinal fibrosis is closely associated with developing proliferative diabetic retinopathy (PDR). Clinical studies have shown that fibrotic membranes exhibit uncontrolled growth in PDR and contribute to retinal detachment from RPE cells, ultimately leading to vision loss. While anti-VEGF agents and invasive laser treatments are the primary treatments for PDR, retinal fibrosis has received minimal attention as a potential target for therapeutic intervention. Therefore, to investigate the potential role of Akt2 in the diabetes-induced retinal fibrosis process, we generated RPE-specific Akt2 conditional knockout (cKO) mice and induced diabetes in these mice and Akt2fl/fl control mice by intraperitoneal injection of streptozotocin. After an 8-month duration of diabetes (10 months of age), the mice were euthanized and expression of tight junction proteins, epithelial-mesenchymal transition (EMT), and fibrosis markers were examined in the RPE. Diabetes induction in the floxed control mice decreased levels of the RPE tight junction protein ZO-1 and adherens junction proteins occludin and E-cadherin; these decreases were rescued in Akt2 cKO diabetic mice. Loss of Akt2 also inhibited diabetes-induced elevation of RNA and protein levels of the EMT markers Snail/Slug and Twist1 in the RPE as compared to Akt2fl/fl diabetic mice. We also found that in Akt2 cKO mice diabetes-induced increase of fibrosis markers, including collagen IV, Connective tissue growth factor (CTGF), fibronectin, and alpha-SMA was attenuated. Furthermore, we observed that high glucose-induced alterations in EMT and fibrosis markers in wild-type (WT) RPE explants were rescued in the presence of PI3K and ERK inhibitors, indicating diabetes-induced retinal fibrosis may be mediated via the PI3K/Akt2/ERK signaling, which could provide a novel target for DR therapy.
Collapse
Affiliation(s)
- Rachel Daley
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vishnu Maddipatla
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Olivia Chowdhury
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Samuel Zigler
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Sakakibara O, Shimoda M, Yamamoto G, Higashi Y, Ikeda-Imafuku M, Ishima Y, Kawahara M, Tanaka KI. Effectiveness of Albumin-Fused Thioredoxin against 6-Hydroxydopamine-Induced Neurotoxicity In Vitro. Int J Mol Sci 2023; 24:ijms24119758. [PMID: 37298708 DOI: 10.3390/ijms24119758] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by oxidative stress-dependent loss of dopaminergic neurons in the substantia nigra and elevated microglial inflammatory responses. Recent studies show that cell loss also occurs in the hypothalamus in PD. However, effective treatments for the disorder are lacking. Thioredoxin is the major protein disulfide reductase in vivo. We previously synthesized an albumin-thioredoxin fusion protein (Alb-Trx), which has a longer plasma half-life than thioredoxin, and reported its effectiveness in the treatment of respiratory and renal diseases. Moreover, we reported that the fusion protein inhibits trace metal-dependent cell death in cerebrovascular dementia. Here, we investigated the effectiveness of Alb-Trx against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in vitro. Alb-Trx significantly inhibited 6-OHDA-induced neuronal cell death and the integrated stress response. Alb-Trx also markedly inhibited 6-OHDA-induced reactive oxygen species (ROS) production, at a concentration similar to that inhibiting cell death. Exposure to 6-OHDA perturbed the mitogen-activated protein kinase pathway, with increased phosphorylated Jun N-terminal kinase and decreased phosphorylated extracellular signal-regulated kinase levels. Alb-Trx pretreatment ameliorated these changes. Furthermore, Alb-Trx suppressed 6-OHDA-induced neuroinflammatory responses by inhibiting NF-κB activation. These findings suggest that Alb-Trx reduces neuronal cell death and neuroinflammatory responses by ameliorating ROS-mediated disruptions in intracellular signaling pathways. Thus, Alb-Trx may have potential as a novel therapeutic agent for PD.
Collapse
Affiliation(s)
- Okina Sakakibara
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo 202-8585, Japan
| | - Mikako Shimoda
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo 202-8585, Japan
| | - Gaku Yamamoto
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo 202-8585, Japan
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan
| | - Mayumi Ikeda-Imafuku
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-Cho, Wakayama 640-8156, Japan
| | - Yu Ishima
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan
| | - Masahiro Kawahara
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo 202-8585, Japan
| |
Collapse
|
11
|
Alam M, Hasan GM, Eldin SM, Adnan M, Riaz MB, Islam A, Khan I, Hassan MI. Investigating regulated signaling pathways in therapeutic targeting of non-small cell lung carcinoma. Biomed Pharmacother 2023; 161:114452. [PMID: 36878052 DOI: 10.1016/j.biopha.2023.114452] [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: 01/19/2023] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is the most common malignancy worldwide. The signaling cascades are stimulated via genetic modifications in upstream signaling molecules, which affect apoptotic, proliferative, and differentiation pathways. Dysregulation of these signaling cascades causes cancer-initiating cell proliferation, cancer development, and drug resistance. Numerous efforts in the treatment of NSCLC have been undertaken in the past few decades, enhancing our understanding of the mechanisms of cancer development and moving forward to develop effective therapeutic approaches. Modifications of transcription factors and connected pathways are utilized to develop new treatment options for NSCLC. Developing designed inhibitors targeting specific cellular signaling pathways in tumor progression has been recommended for the therapeutic management of NSCLC. This comprehensive review provided deeper mechanistic insights into the molecular mechanism of action of various signaling molecules and their targeting in the clinical management of NSCLC.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Sayed M Eldin
- Center of Research, Faculty of Engineering, Future University in Egypt, New Cairo 11835, Egypt
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Muhammad Bilal Riaz
- Faculty of Applied Physics and Mathematics, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdnask, Poland; Department of Computer Science and Mathematics, Lebanese American University, Byblos, Lebanon
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
12
|
Huang L, He F, Wu B. Mechanism of effects of nickel or nickel compounds on intestinal mucosal barrier. CHEMOSPHERE 2022; 305:135429. [PMID: 35760131 DOI: 10.1016/j.chemosphere.2022.135429] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 06/15/2023]
Abstract
As an important metal in industry, national defense, and production, nickel widely exists in nature and is also a necessary trace element for human beings and animals. Nickel deficiency will affect the growth and development of animals, the contents of related active substances, enzymes and other essential elements in vivo. However, excessive nickel or longer nickel exposure can induce excessive free radicals (reactive oxygen species and reactive nitrogen) in the body, which can lead to a variety of cell damage, apoptosis and canceration, and ultimately pose negative effects on the health of the body. Among them, the intestinal tract, as the largest interface between the body and the external environment, greatly increases the contact probability between nickel or nickel compounds and the intestinal mucosal barrier, thus, the intestinal structure and function are also more vulnerable to nickel damage, leading to a series of related diseases such as enteritis. Therefore, this paper briefly analyzed the damage mechanism of nickel or its compounds to the intestinal tract from the perspective of four intestinal mucosal barriers: mechanical barrier, immune barrier, microbial barrier and chemical barrier, we hope to make a certain theoretical contribution to the further research and the prevention and treatment of nickel related diseases.
Collapse
Affiliation(s)
- Lijing Huang
- College of Life Sciences, China West Normal University, Nanchong, PR China
| | - Fang He
- College of Life Sciences, China West Normal University, Nanchong, PR China
| | - Bangyuan Wu
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education PR China, Nanchong, PR China; College of Life Sciences, China West Normal University, Nanchong, PR China.
| |
Collapse
|
13
|
Mercury-Induced Oxidative Stress Response in Benthic Foraminifera: An In Vivo Experiment on Amphistegina lessonii. BIOLOGY 2022; 11:biology11070960. [PMID: 36101341 PMCID: PMC9312061 DOI: 10.3390/biology11070960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/22/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
The evaluation of the effects of pollution (e.g., Hg pollution) is a difficult task and relies mostly on biomonitoring based on bioindicators. The application of biomarkers may represent a complementary or alternative approach in environmental biomonitoring. Mercury is known to pose a significant health hazard due to its ability to cross cellular membranes, bioaccumulate, and biomagnify. In the present research, the effects of short-term (i.e., 24 h) Hg exposure in the symbiont-bearing benthic foraminiferal species Amphistegina lessonii are evaluated using several biomarkers (i.e., proteins and enzymes). Mercury leads to significant changes in the biochemistry of cells. Its effects are mainly associated with oxidative stress (i.e., production of reactive oxygen species: ROS), depletion of glutathione (GSH), and alteration of protein synthesis. Specifically, our findings reveal that exposure to Hg leads to the consumption of GSH by GPx and GST for the scavenging of ROS and the activation of antioxidant-related enzymes, including SOD and GSH-enzymes (GST, GSR, GPx, and Se-GPx), that are directly related to a defense mechanism against ROS. The Hg exposure also activates the MAPK (e.g., p-p38) and HSP (e.g., HSP 70) pathways. The observed biochemical alterations associated with Hg exposure may represent effective and reliable proxies (i.e., biomarkers) for the evaluation of stress in A. lessonii and lead to a possible application for the detection of early warning signs of environmental stress in biomonitoring.
Collapse
|
14
|
Volynets GP, Pletnova LV, Sapelkin VM, Savytskyi OV, Yarmoluk SM. A computational analysis of the binding free energies of apoptosis signal-regulating kinase 1 inhibitors from different chemotypes. MOLECULAR SIMULATION 2021. [DOI: 10.1080/08927022.2021.1922686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Galyna P. Volynets
- Department of Medicinal Chemistry, Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv, Ukraine
- Scientific Services Company Otava Ltd., Kyiv, Ukraine
| | - Larysa V. Pletnova
- Department of Medicinal Chemistry, Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv, Ukraine
| | - Vladislav M. Sapelkin
- Department of Medicinal Chemistry, Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv, Ukraine
| | - Oleksandr V. Savytskyi
- Department of Protein Engineering and Bioinformatics, Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv, Ukraine
| | - Sergiy M. Yarmoluk
- Department of Medicinal Chemistry, Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
15
|
Arafa ESA, Refaey MS, Abd El-Ghafar OAM, Hassanein EHM, Sayed AM. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition. Heliyon 2021; 7:e08354. [PMID: 34825082 PMCID: PMC8605069 DOI: 10.1016/j.heliyon.2021.e08354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/06/2021] [Accepted: 11/05/2021] [Indexed: 12/21/2022] Open
Abstract
The p38 mitogen-activated protein kinases (p38 MAPK) is a 38kD polypeptide recognized as the target for many potential anti-inflammatory agents. Accumulating evidence indicates that p38 MAPK could perform many roles in human disease pathophysiology. Therefore, great therapeutic benefits can be attained from p38 MAPK inhibitors. Ginseng is an exceptionally valued medicinal plant of the family Araliaceae (Panax genus). Recently, several studies targeted the therapeutic effects of purified individual ginsenoside, the most significant active ingredient of ginseng, and studied its particular molecular mechanism(s) of action rather than whole-plant extracts. Interestingly, several ginsenosides: ginsenosides compound K, F1, Rb1, Rb3, Rc, Rd, Re, Rf, Rg1, Rg2, Rg3, Rg5, Rh1, Rh2, Ro, notoginsenoside R1, and protopanaxadiol have shown to possess great therapeutic potentials mediated by their ability to downregulate p38 MAPK signaling in different cell lines and experimental animal models. Our review compiles the research findings of various ginsenosides as potent anti-inflammatory agents, highlighting the crucial role of p38 MAPK suppression in their pharmacological actions. In addition, in silico studies were conducted to explore the probable binding of these ginsenosides to p38 MAPK. The results obtained proposed p38 MAPK involvement in the beneficial pharmacological activities of ginsenosides in different ailments. p38 MAPK plays many roles in human disease pathophysiology. Therefore, great therapeutic benefits can be attained from p38 MAPK inhibitors. Several ginsenosides showed to possess great therapeutic potentials mediated by its ability to downregulate p38 MAPK signaling. in silico studies were conducted to explore the binding of these ginsenosides to p38 MAPK and evidenced the promising their inhibitory effect.
Collapse
Affiliation(s)
- El-Shaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates.,Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed S Refaey
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Sadat City, Menoufiya, 32958, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
16
|
Zhang Z, Costa M. p62 functions as a signal hub in metal carcinogenesis. Semin Cancer Biol 2021; 76:267-278. [PMID: 33894381 PMCID: PMC9161642 DOI: 10.1016/j.semcancer.2021.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/06/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022]
Abstract
A number of metals are toxic and carcinogenic to humans. Reactive oxygen species (ROS) play an important role in metal carcinogenesis. Oxidative stress acts as the converging point among various stressors with ROS being the main intracellular signal transducer. In metal-transformed cells, persistent expression of p62 and erythroid 2-related factor 2 (Nrf2) result in apoptosis resistance, angiogenesis, inflammatory microenvironment, and metabolic reprogramming, contributing to overall mechanism of metal carcinogenesis. Autophagy, a conserved intracellular process, maintains cellular homeostasis by facilitating the turnover of protein aggregates, cellular debris, and damaged organelles. In addition to being a substrate of autophagy, p62 is also a crucial molecule in a myriad of cellular functions and in molecular events, which include oxidative stress, inflammation, apoptosis, cell proliferation, metabolic reprogramming, that modulate cell survival and tumor growth. The multiple functions of p62 are appreciated by its ability to interact with several key components involved in various oncogenic pathways. This review summarizes the current knowledge and progress in studies of p62 and metal carcinogenesis with emphasis on oncogenic pathways related to oxidative stress, inflammation, apoptosis, and metabolic reprogramming.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Environmental Medicine, NYU School of Medicine, 341 East 25th Street, New York, NY 10010, USA
| | - Max Costa
- Department of Environmental Medicine, NYU School of Medicine, 341 East 25th Street, New York, NY 10010, USA.
| |
Collapse
|
17
|
Lin CL, Ying TH, Yang SF, Chiou HL, Chen YS, Kao SH, Hsieh YH. MTA2 silencing attenuates the metastatic potential of cervical cancer cells by inhibiting AP1-mediated MMP12 expression via the ASK1/MEK3/p38/YB1 axis. Cell Death Dis 2021; 12:451. [PMID: 33958583 PMCID: PMC8102478 DOI: 10.1038/s41419-021-03729-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022]
Abstract
Metastasis-associated protein 2 (MTA2) is a transcription factor that is highly associated with matrix metalloproteinase 12 (MMP12). Thus, we hypothesized that MTA2 may regulate MMP12 expression and is involved in cervical cancer metastasis. Results showed that MTA2 and MMP12 were highly expressed in cervical cancer cells, and MTA2 knockdown reduced MMP12 expression and inhibited the metastasis of cervical cancer cells in xenograft mice. MMP12 knockdown did not influence the viability of cervical cancer cells but clearly inhibited cell migration and invasion both in vitro and in vivo. MMP12 was highly expressed in cervical tumor tissues and correlated with the poor survival rate of patients with cervical cancer. Further investigations revealed that p38 mitogen-activated protein kinase (p38), mitogen-activated protein kinase kinase 3 (MEK3), and apoptosis signal-regulating kinase 1 (ASK1) were involved in MMP12 downregulation in response to MTA2 knockdown. Results also demonstrated that p38-mediated Y-box binding protein1 (YB1) phosphorylation disrupted the binding of AP1 (c-Fos/c-Jun) to the MMP12 promoter, thereby inhibiting MMP12 expression and the metastatic potential of cervical cancer cells. Collectively, targeting both MTA2 and MMP12 may be a promising strategy for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Chia-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yong-Syuan Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shao-Hsuan Kao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan. .,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan. .,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
18
|
Ramalingam V, Rajaram R. A paradoxical role of reactive oxygen species in cancer signaling pathway: Physiology and pathology. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
19
|
ASK1 Enhances Angiotensin II-Induced Liver Fibrosis In Vitro by Mediating Endoplasmic Reticulum Stress-Dependent Exosomes. Mediators Inflamm 2020; 2020:8183713. [PMID: 33223956 PMCID: PMC7669360 DOI: 10.1155/2020/8183713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 01/16/2023] Open
Abstract
Background Apoptosis signal-regulating kinase 1 (ASK1) has been reported to induce fibrotic signaling in the setting of oxidative stress. However, the role of ASK1 and its mechanism of action in angiotensin II- (Ang II-) induced liver fibrosis remain largely unknown. Methods Human hepatic LX-2 stellate cells were treated with Ang II alone or cotreated with Ang II plus an ASK1 inhibitor (GS-4997) or siRNA-targeting ASK1. Immunofluorescent staining, real-time PCR, and western blotting were used to determine the expressionof α-SMA, Col I, and Col III expression. Cell viability was assessed by the CCK-8 assay. The concentrations of IL-1β, IL-18, and TNF-α in conditioned medium were determined by ELISA. The levels of intracellular ROS in LX-2 cells were analyzed using a ROS assay kit. Exosome size was determined by electron microscopy. Results Ang II markedly increased the expression of extracellular matrix (ECM) proteins (α-SMA, Col I, and Col III) and proinflammatory cytokines (IL-1β, IL-18, and TNF-α). Ang II also increased the expression of endoplasmic reticulum stress (ERS) markers (GRP78, p-PERK, and CHOP) and p-ASK1. Results also showed that pretreatment with GS-4997 or siRNA could abolish all the abovementioned effects on LX-2 cells. Furthermore, we found that exosome release caused by ASK1-mediated ERS was involved in the activation of LX-2 cells by Ang II. The activation of LX-2 cells could be blocked by treating the exosomes with annexin. Conclusions In summary, we found that ASK1 mediates Ang II-activated ERS in HSCs and the subsequent activation of HSCs, suggesting a promising strategy for treating liver fibrosis.
Collapse
|
20
|
Demir E. A review on nanotoxicity and nanogenotoxicity of different shapes of nanomaterials. J Appl Toxicol 2020; 41:118-147. [PMID: 33111384 DOI: 10.1002/jat.4061] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
Nanomaterials (NMs) generally display fascinating physical and chemical properties that are not always present in bulk materials; therefore, any modification to their size, shape, or coating tends to cause significant changes in their chemical/physical and biological characteristics. The dramatic increase in efforts to use NMs renders the risk assessment of their toxicity highly crucial due to the possible health perils of this relatively uncharted territory. The different sizes and shapes of the nanoparticles are known to have an impact on organisms and an important place in clinical applications. The shape of nanoparticles, namely, whether they are rods, wires, or spheres, is a particularly critical parameter to affect cell uptake and site-specific drug delivery, representing a significant factor in determining the potency and magnitude of the effect. This review, therefore, intends to offer a picture of research into the toxicity of different shapes (nanorods, nanowires, and nanospheres) of NMs to in vitro and in vivo models, presenting an in-depth analysis of health risks associated with exposure to such nanostructures and benefits achieved by using certain model organisms in genotoxicity testing. Nanotoxicity experiments use various models and tests, such as cell cultures, cores, shells, and coating materials. This review article also attempts to raise awareness about practical applications of NMs in different shapes in biology, to evaluate their potential genotoxicity, and to suggest approaches to explain underlying mechanisms of their toxicity and genotoxicity depending on nanoparticle shape.
Collapse
Affiliation(s)
- Eşref Demir
- Vocational School of Health Services, Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Antalya Bilim University, Dosemealti, Antalya, Turkey
| |
Collapse
|
21
|
Salimi A, Jamali Z, Atashbar S, Khezri S, Ghorbanpour AM, Etefaghi N. Pathogenic Mechanisms and Therapeutic Implication in Nickel-Induced Cell Damage. Endocr Metab Immune Disord Drug Targets 2020; 20:968-984. [DOI: 10.2174/1871530320666200214123118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/24/2019] [Accepted: 03/07/2019] [Indexed: 11/22/2022]
Abstract
Background:
Nickel (Ni) is mostly applied in a number of industrial areas such as printing
inks, welding, alloys, electronics and electrical professions. Occupational or environmental exposure to
nickel may lead to cancer, allergy reaction, nephrotoxicity, hepatotoxicity, neurotoxicity, as well as
cell damage, apoptosis and oxidative stress.
Methods:
In here, we focused on published studies about cell death, carcinogenicity, allergy reactions
and neurotoxicity, and promising agents for the prevention and treatment of the toxicity by Ni.
Results:
Our review showed that in the last few years, more researches have focused on reactive oxygen
species formation, oxidative stress, DNA damages, apoptosis, interaction with involving receptors
in allergy and mitochondrial damages in neuron induced by Ni.
Conclusion:
The collected data in this paper provide useful information about the main toxicities induced
by Ni, also, their fundamental mechanisms, and how to discover new ameliorative agents for
prevention and treatment by reviewing agents with protective and therapeutic consequences on Ni
induced toxicity.
Collapse
Affiliation(s)
- Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zhaleh Jamali
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Saman Atashbar
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saleh Khezri
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amir M. Ghorbanpour
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nahid Etefaghi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
22
|
Tanaka KI, Shimoda M, Kasai M, Ikeda M, Ishima Y, Kawahara M. Involvement of SAPK/JNK Signaling Pathway in Copper Enhanced Zinc-Induced Neuronal Cell Death. Toxicol Sci 2020; 169:293-302. [PMID: 30768131 DOI: 10.1093/toxsci/kfz043] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Zinc (Zn) plays an important role in many organisms in various physiological functions such as cell division, immune mechanisms and protein synthesis. However, excessive Zn release is induced in pathological situations and causes neuronal cell death. Previously, we reported that Cu ions (Cu2+) markedly exacerbates Zn2+-induced neuronal cell death by potentiating oxidative stress and the endoplasmic reticulum stress response. In contrast, the stress-activated protein kinase/c-Jun amino-terminal kinase (SAPK/JNK) signaling pathway is important in neuronal cell death. Thus, in this study, we focused on the SAPK/JNK signaling pathway and examined its involvement in Cu2+/Zn2+-induced neurotoxicity. Initially, we examined expression of factors involved in the SAPK/JNK signaling pathway. Accordingly, we found that phosphorylated (ie, active) forms of SAPK/JNK (p46 and p54) are increased by CuCl2 and ZnCl2 co-treatment in hypothalamic neuronal mouse cells (GT1-7 cells). Downstream factors of SAPK/JNK, phospho-c-Jun, and phospho-activating transcription factor 2 are also induced by CuCl2 and ZnCl2 co-treatment. Moreover, an inhibitor of the SAPK/JNK signaling pathway, SP600125, significantly suppressed neuronal cell death and activation of the SAPK/JNK signaling pathway induced by CuCl2 and ZnCl2 cotreatment. Finally, we examined involvement of oxidative stress in activation of the SAPK/JNK signaling pathway, and found that human serum albumin-thioredoxin fusion protein, an antioxidative protein, suppresses activation of the SAPK/JNK signaling pathway. On the basis of these results, our findings suggest that activation of ZnCl2-dependent SAPK/JNK signaling pathway is important in neuronal cell death, and CuCl2-induced oxidative stress triggers the activation of this pathway.
Collapse
Affiliation(s)
- Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Mikako Shimoda
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Misato Kasai
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Mayumi Ikeda
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Musashino University, Nishitokyo-shi, Tokyo, Japan
| |
Collapse
|
23
|
Alquezar C, Felix JB, McCandlish E, Buckley BT, Caparros-Lefebvre D, Karch CM, Golbe LI, Kao AW. Heavy metals contaminating the environment of a progressive supranuclear palsy cluster induce tau accumulation and cell death in cultured neurons. Sci Rep 2020; 10:569. [PMID: 31953414 PMCID: PMC6969162 DOI: 10.1038/s41598-019-56930-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder characterized by the presence of intracellular aggregates of tau protein and neuronal loss leading to cognitive and motor impairment. Occurrence is mostly sporadic, but rare family clusters have been described. Although the etiopathology of PSP is unknown, mutations in the MAPT/tau gene and exposure to environmental toxins can increase the risk of PSP. Here, we used cell models to investigate the potential neurotoxic effects of heavy metals enriched in a highly industrialized region in France with a cluster of sporadic PSP cases. We found that iPSC-derived iNeurons from a MAPT mutation carrier tend to be more sensitive to cell death induced by chromium (Cr) and nickel (Ni) exposure than an isogenic control line. We hypothesize that genetic variations may predispose to neurodegeneration induced by those heavy metals. Furthermore, using an SH-SY5Y neuroblastoma cell line, we showed that both heavy metals induce cell death by an apoptotic mechanism. Interestingly, Cr and Ni treatments increased total and phosphorylated tau levels in both cell types, implicating Cr and Ni exposure in tau pathology. Overall, this study suggests that chromium and nickel could contribute to the pathophysiology of tauopathies such as PSP by promoting tau accumulation and neuronal cell death.
Collapse
Affiliation(s)
- Carolina Alquezar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Jessica B Felix
- Graduate Program, Department of Molecular and Cellular Biology Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Lawrence I Golbe
- Division of Movement Disorders. Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Aimee W Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA.
| |
Collapse
|
24
|
Manganese Acts upon Insulin/IGF Receptors to Phosphorylate AKT and Increase Glucose Uptake in Huntington's Disease Cells. Mol Neurobiol 2019; 57:1570-1593. [PMID: 31797328 DOI: 10.1007/s12035-019-01824-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
Perturbations in insulin/IGF signaling and manganese (Mn2+) uptake and signaling have been separately reported in Huntington's disease (HD) models. Insulin/IGF supplementation ameliorates HD phenotypes via upregulation of AKT, a known Mn2+-responsive kinase. Limited evidence both in vivo and in purified biochemical systems suggest Mn2+ enhances insulin/IGF receptor (IR/IGFR), an upstream tyrosine kinase of AKT. Conversely, Mn2+ deficiency impairs insulin release and associated glucose tolerance in vivo. Here, we test the hypothesis that Mn2+-dependent AKT signaling is predominantly mediated by direct Mn2+ activation of the insulin/IGF receptors, and HD-related impairments in insulin/IGF signaling are due to HD genotype-associated deficits in Mn2+ bioavailability. We examined the combined effects of IGF-1 and/or Mn2+ treatments on AKT signaling in multiple HD cellular models. Mn2+ treatment potentiates p-IGFR/IR-dependent AKT phosphorylation under physiological (1 nM) or saturating (10 nM) concentrations of IGF-1 directly at the level of intracellular activation of IGFR/IR. Using a multi-pharmacological approach, we find that > 70-80% of Mn2+-associated AKT signaling across rodent and human neuronal cell models is specifically dependent on IR/IGFR, versus other signaling pathways upstream of AKT activation. Mn2+-induced p-IGFR and p-AKT were diminished in HD cell models, and, consistent with our hypothesis, were rescued by co-treatment of Mn2+ and IGF-1. Lastly, Mn2+-induced IGF signaling can modulate HD-relevant biological processes, as the reduced glucose uptake in HD STHdh cells was partially reversed by Mn2+ supplementation. Our data demonstrate that Mn2+ supplementation increases peak IGFR/IR-induced p-AKT likely via direct effects on IGFR/IR, consistent with its role as a cofactor, and suggests reduced Mn2+ bioavailability contributes to impaired IGF signaling and glucose uptake in HD models.
Collapse
|
25
|
Baig MH, Baker A, Ashraf GM, Dong JJ. ASK1 and its role in cardiovascular and other disorders: available treatments and future prospects. Expert Rev Proteomics 2019; 16:857-870. [DOI: 10.1080/14789450.2019.1676735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Abu Baker
- Nanobiotechnology and nanomedicine lab, Department of Biosciences, Integral University, Lucknow, India
| | - Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jae-June Dong
- Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
26
|
Mincarelli L, Tiano L, Craft J, Marcheggiani F, Vischetti C. Evaluation of gene expression of different molecular biomarkers of stress response as an effect of copper exposure on the earthworm EIsenia Andrei. ECOTOXICOLOGY (LONDON, ENGLAND) 2019; 28:938-948. [PMID: 31402411 DOI: 10.1007/s10646-019-02093-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/29/2019] [Indexed: 06/10/2023]
Abstract
The paper reports the results of a laboratory test on the bioaccumulation and toxicological effects of sub-lethal soil concentration of copper, a widely used fungicide in organic farming, on DNA damage, a critical marker increasingly used in ecotoxicology in the earthworm Eisenia andrei. In the same experimental setting we evaluated gene expression of classical biomarker of stress induced by xenobiotic. [Heat Shock Protein 70 (HSP70) and Metallothionein (MET)], as well as genes coding for enzymes involved in detoxification of reactive oxygen species [Superoxide dismutase (SOD) and catalase (CAT)]. Additionally, expression of genes involved in the immune response were investigated: a Toll-like receptor (TLR), a receptor with cytolytic activity named Cytolytic Factor (CCF) and two antimicrobial peptides, fetidin (FET) and lysenin (LYS). Results showed significant time-dependent bioaccumulation of Cu and DNA damage at concentrations remarkably lower than those found in most agricultural soils worldwide. MET was increased as was FET and TLR. The present work gives new insights into the mechanisms of sub-lethal toxicity of copper as an environmental pollutant and in the identification of novel sub-lethal biomarkers of cellular response to the stressor such as immune response genes.
Collapse
Affiliation(s)
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy.
| | - John Craft
- Biological & Biomedical Science Department, School of Health and Life Science, Glasgow Caledonian University, Glasgow, UK
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Costantino Vischetti
- Environmental, Food and Agricultural Sciences Department, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
27
|
Challa TD, Wueest S, Lucchini FC, Dedual M, Modica S, Borsigova M, Wolfrum C, Blüher M, Konrad D. Liver ASK1 protects from non-alcoholic fatty liver disease and fibrosis. EMBO Mol Med 2019; 11:e10124. [PMID: 31595673 PMCID: PMC6783644 DOI: 10.15252/emmm.201810124] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with obesity and may progress to non-alcoholic steatohepatitis (NASH) and liver fibrosis. The deficit of pharmacological therapies for the latter mainly results from an incomplete understanding of involved pathological mechanisms. Herein, we identify apoptosis signal-regulating kinase 1 (ASK1) as a suppressor of NASH and fibrosis formation. High-fat diet-fed and aged chow-fed liver-specific ASK1-knockout mice develop a higher degree of hepatic steatosis, inflammation, and fibrosis compared to controls. In addition, pharmacological inhibition of ASK1 increased hepatic lipid accumulation in wild-type mice. In line, liver-specific ASK1 overexpression protected mice from the development of high-fat diet-induced hepatic steatosis and carbon tetrachloride-induced fibrosis. Mechanistically, ASK1 depletion blunts autophagy, thereby enhancing lipid droplet accumulation and liver fibrosis. In human livers of lean and obese subjects, ASK1 expression correlated negatively with liver fat content and NASH scores, but positively with markers for autophagy. Taken together, ASK1 may be a novel therapeutic target to tackle NAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Tenagne D Challa
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Fabrizio C Lucchini
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Mara Dedual
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Salvatore Modica
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Marcela Borsigova
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
28
|
Nickel Carcinogenesis Mechanism: DNA Damage. Int J Mol Sci 2019; 20:ijms20194690. [PMID: 31546657 PMCID: PMC6802009 DOI: 10.3390/ijms20194690] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/15/2019] [Accepted: 09/20/2019] [Indexed: 12/14/2022] Open
Abstract
Nickel (Ni) is known to be a major carcinogenic heavy metal. Occupational and environmental exposure to Ni has been implicated in human lung and nasal cancers. Currently, the molecular mechanisms of Ni carcinogenicity remain unclear, but studies have shown that Ni-caused DNA damage is an important carcinogenic mechanism. Therefore, we conducted a literature search of DNA damage associated with Ni exposure and summarized known Ni-caused DNA damage effects. In vitro and vivo studies demonstrated that Ni can induce DNA damage through direct DNA binding and reactive oxygen species (ROS) stimulation. Ni can also repress the DNA damage repair systems, including direct reversal, nucleotide repair (NER), base excision repair (BER), mismatch repair (MMR), homologous-recombination repair (HR), and nonhomologous end-joining (NHEJ) repair pathways. The repression of DNA repair is through direct enzyme inhibition and the downregulation of DNA repair molecule expression. Up to now, the exact mechanisms of DNA damage caused by Ni and Ni compounds remain unclear. Revealing the mechanisms of DNA damage from Ni exposure may contribute to the development of preventive strategies in Ni carcinogenicity.
Collapse
|
29
|
Dey A, Manna S, Adhikary J, Chattopadhyay S, De S, Chattopadhyay D, Roy S. Biodistribution and toxickinetic variances of chemical and green Copper oxide nanoparticles in vitro and in vivo. J Trace Elem Med Biol 2019; 55:154-169. [PMID: 31345354 DOI: 10.1016/j.jtemb.2019.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 06/06/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
In this study, chemical (S1) and green (S2) Copper Oxide nanoparticles (NPs) were synthesized to determine their biodistribution and toxicokinetic variances in vitro and in vivo. Both NPs significantly released Copper ions (Cu) in lymphocytes and were primarily deposited in the mononuclear phagocyte system (MPS) such as the liver and spleen in mice. In particular, S2NPs seemed to be prominently stored in the spleen, whereas the S1NPs were widely stored in more organs including the liver, heart, lungs, kidney and intestine. The circulation in the blood and fecal excretions both showed higher S2NPs contents respectively. Measurements of cell viability, Hemolysis assay, Reactive Oxygen Species (ROS) generation, biochemical estimation and apoptotic or necrotic study in lymphocytes after 24 h and measurements of body and organ weight, serum chemistry evaluation, cytokines level, protein expressions and histopathology of Balb/C mice after 15 days indicated significant toxicity difference between the S1NPs and S2NPs. Our observations proved that the NPs physiochemical properties influence toxicity and Biodistribution profiles in vitro and in vivo.
Collapse
Affiliation(s)
- Aditi Dey
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Subhankar Manna
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Jaydeep Adhikary
- Department of Chemical Sciences, Ariel University, Ariel, 40700, Israel
| | - Sourav Chattopadhyay
- Molecular Genetics and Therapeutics Lab, Indian Institute of Technology, Kanpur, India
| | - Sriparna De
- Department of Polymer Science and Technology, USCTA, University of Calcutta, 92 A.P.C road, Kolkata, 700009, West Bengal, India
| | - Dipankar Chattopadhyay
- Department of Polymer Science and Technology, USCTA, University of Calcutta, 92 A.P.C road, Kolkata, 700009, West Bengal, India.
| | - Somenath Roy
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community health, Vidyasagar University, Midnapore, 721102, West Bengal, India.
| |
Collapse
|
30
|
Chung YP, Yen CC, Tang FC, Lee KI, Liu SH, Wu CC, Hsieh SS, Su CC, Kuo CY, Chen YW. Methylmercury exposure induces ROS/Akt inactivation-triggered endoplasmic reticulum stress-regulated neuronal cell apoptosis. Toxicology 2019; 425:152245. [PMID: 31330229 DOI: 10.1016/j.tox.2019.152245] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/06/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have positively linked mercury exposure and neurodegenerative diseases (ND). Methylmercury (MeHg), an organic form of mercury, is a ubiquitous and potent environmental neurotoxicant that easily crosses the blood-brain barrier and causes irreversible injury to the central nervous system (CNS). However, the molecular mechanisms underlying MeHg-induced neurotoxicity remain unclear. Here, the present study found that Neuro-2a cells underwent apoptosis in response to MeHg (1-5 μM), which was accompanied by increased phosphatidylserine (PS) exposure on the outer cellular membrane leaflets, caspase-3 activity, and the activation of caspase cascades and poly (ADP-ribose) polymerase (PARP). Exposure of Neuro-2a cells to MeHg also triggered endoplasmic reticulum (ER) stress, which was identified via several key molecules (including: glucose-regulated protein (GRP)78, GRP94, C/EBP homologous protein (CHOP) X-box binding protein(XBP)-1, protein kinase R-like ER kinase (PERK), eukaryotic initiation factor 2α (eIF2α), inositol-requiring enzyme(IRE)-1, activation transcription factor(AFT)4, and ATF6. Transfection with GRP78-, GRP94-, CHOP-, and XBP-1-specific small interfering (si)RNA significantly suppressed the expression of these proteins, and attenuated cytotoxicity and caspase-12, -7, and -3 activation in MeHg-exposed cells. Furthermore, MeHg dramatically decreased Akt phosphorylation, and the overexpression of activation of Akt1 (myr-Akt1) could significantly prevent MeHg-induced Akt inactivation, as well as apoptotic and ER stress-related signals. Pretreatment with the antioxidant N-acetylcysteine (NAC) effectively prevented MeHg-induced neuronal cell reactive oxygen species (ROS) generation, apoptotic and ER stress-related signals, and Akt inactivation. Collectively, these results indicate that MeHg exerts its cytotoxicity in neurons by inducing ROS-mediated Akt inactivation up-regulated ER stress, which induces apoptosis and ultimately leads to cell death.
Collapse
Affiliation(s)
- Yao-Pang Chung
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Cheng-Chieh Yen
- Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan; Department of Occupational Medicine, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Feng-Cheng Tang
- Department of Occupational Medicine, Changhua Christian Hospital, Changhua County, 500, Taiwan; Department of Leisure Services Management, Chaoyang University of Technology, Taichung, 413, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, 404, Taiwan
| | - Shang-Shu Hsieh
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Ya-Wen Chen
- Department of Physiology and Graduate Institute of Basic Medical Science, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
31
|
Sur S, Nakanishi H, Steele R, Ray RB. Depletion of PCAT-1 in head and neck cancer cells inhibits tumor growth and induces apoptosis by modulating c-Myc-AKT1-p38 MAPK signalling pathways. BMC Cancer 2019; 19:354. [PMID: 30987615 PMCID: PMC6466688 DOI: 10.1186/s12885-019-5562-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/31/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) represents one of the most common malignancies worldwide with a high mortality rate mainly due to lack of early detection markers, frequent association with metastasis and aggressive phenotype. Recently, long non-coding RNAs (lncRNAs) have been shown to have important regulatory roles in human cancers. The lncRNA prostate cancer-associated transcript 1 (PCAT-1) showed potential oncogenic roles in different cancers, however its role in HNSCC is not known. In this study, we evaluated the role of the PCAT-1 in HNSCC. METHODS The expression of PCAT-1 was measured by quantitative real-time PCR in 23 paired human HNSCC tissues and adjacent non-tumor tissue specimens. Cell proliferation after depleting PCAT-1 was determined. Effect of PCAT-1 depletion in HNSCC cell lines was determined by qRT-PCR and Western blot analyses. Finally, JHU029 HNSCC cells was implanted subcutaneously into athymic nude mice and therapeutic potential of PCAT-1 was investigated. RESULTS Up-regulation of PCAT-1 in TCGA dataset of HNSCC was noted. We also observed increased expression of PCAT-1 in archived HNSCC patient samples as compared to adjacent non-tumor tissues. Knockdown of PCAT-1 significantly reduced cell proliferation in HNSCC cell lines. Mechanistic study revealed significant down regulation of c-Myc and AKT1 gene in both RNA and protein levels upon knockdown of PCAT-1. We observed that c-Myc and AKT1 positively correlate with PCAT-1 expression in HNSCC. Further, we observed activation of p38 MAPK and apoptosis signal-regulating kinase 1 upon knockdown of PCAT-1 which induces Caspase 9 and PARP mediated apoptosis. Targeted inhibition of PCAT-1 regresses tumor growth in nude mice. CONCLUSION Together our data demonstrated an important role of the PCAT-1 in HNSCC and might serve as a target for HNSCC therapy.
Collapse
Affiliation(s)
- Subhayan Sur
- 0000 0004 1936 9342grid.262962.bDepartment of Pathology, Saint Louis University, 1100 South Grand Boulevard, St. Louis, MO 63104 USA
| | - Hiroshi Nakanishi
- 0000 0004 1936 9342grid.262962.bDepartment of Pathology, Saint Louis University, 1100 South Grand Boulevard, St. Louis, MO 63104 USA
| | - Robert Steele
- 0000 0004 1936 9342grid.262962.bDepartment of Pathology, Saint Louis University, 1100 South Grand Boulevard, St. Louis, MO 63104 USA
| | - Ratna B. Ray
- 0000 0004 1936 9342grid.262962.bDepartment of Pathology, Saint Louis University, 1100 South Grand Boulevard, St. Louis, MO 63104 USA
| |
Collapse
|
32
|
Chinnakkannu Vijayakumar S, Venkatakrishnan K, Tan B. 3D quantum theranosomes: a new direction for label-free theranostics. NANOSCALE HORIZONS 2019; 4:495-515. [PMID: 32254103 DOI: 10.1039/c8nh00287h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Quantum-scale materials offer great potential in the field of cancer theranostics. At present, quantum materials are severely limited due to 0D & 1D materials lacking biocompatibility, resulting in coated materials with labelled tags for fluorescence excitation. In addition, the application of magnetic quantum materials has not been reported to date for cancer theranostics. In this current research study, we introduce the concept of applying nickel-based magnetic 3D quantum theranosomes for label-free broadband fluorescence enhancement and cancer therapy. To begin with, we present two (primary and secondary) distinct quantum theranosomes for cancer detection and differentiation (HeLa & MDAMB-231) from mammalian fibroblast cells. The primary theranosomes exhibit a metal enhanced fluorescence (MEF) property through localized surface plasmon resonance to act as cancer detectors, whereas the secondary theranosomes act as cancer differentiators through the fluorescence quenching of HeLa cancer cells. Apart from the above, the synthesized magnetic quantum theranosomes introduced therapeutic functionality wherein the theranosomes mimicked a tumor microenvironment by selectively accelerating the proliferation of mammalian fibroblasts cells while at the same time inducing cancer therapy. These quantum theranosomes were synthesized using femtosecond pulse laser ablation and self-assembled to form an interconnected 3D structure. The 3D architecture and the physicochemical properties of the laser synthesized quantum theranosomes closely resembled a tumor microenvironment. Furthermore, we anticipate that our current recorded findings can shed further light upon these unique magnetic quantum theranosomes as potential contenders towards opening an entirely new direction in the field of cancer theranostics.
Collapse
Affiliation(s)
- Sivaprasad Chinnakkannu Vijayakumar
- Ultrashort Laser Manufacturing Research Facility, Department of Mechanical and Industrial Engineering, Ryerson University, 350 Victoria street, Toronto, Ontario M5B 2K3, Canada.
| | | | | |
Collapse
|
33
|
Chirumbolo S, Bjørklund G, Lysiuk R, Vella A, Lenchyk L, Upyr T. Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways. Int J Mol Sci 2018; 19:ijms19113568. [PMID: 30424557 PMCID: PMC6274856 DOI: 10.3390/ijms19113568] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023] Open
Abstract
The role of phytochemicals as potential prodrugs or therapeutic substances against tumors has come in the spotlight in the very recent years, thanks to the huge mass of encouraging and promising results of the in vitro activity of many phenolic compounds from plant raw extracts against many cancer cell lines. Little but important evidence can be retrieved from the clinical and nutritional scientific literature, where flavonoids are investigated as major pro-apoptotic and anti-metastatic compounds. However, the actual role of these compounds in cancer is still far to be fully elucidated. Many of these phytochemicals act in a pleiotropic and poorly specific manner, but, more importantly, they are able to tune the reactive oxygen species (ROS) signaling to activate a survival or a pro-autophagic and pro-apoptosis mechanism, depending on the oxidative stress-responsive endowment of the targeted cell. This review will try to focus on this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy.
- Scientific Secretary-Council for Nutritional and Environmental Medicine (CONEM), 8610 Mo i Rana, Norway.
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), 8610 Mo i Rana, Norway.
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany, DanyloHalytskyLviv National Medical University, 79007 Lviv, Ukraine.
| | - Antonio Vella
- AOUI Verona, University Hospital, Section of Immunology, 37134 Verona, Italy.
| | - Larysa Lenchyk
- Department of Chemistry of Natural Compounds, National University of Pharmacy, 61168 Kharkiv, Ukraine.
| | - Taras Upyr
- Department of Pharmacognosy, National University of Pharmacy, 61168 Kharkiv, Ukraine.
| |
Collapse
|
34
|
Wan X, Shi L, Ma X, Tang H, Wu G. The Elevated ASK1 Expression Inhibits Proliferation and Invasion in Gastric Cancer HGC-27 Cells. Anat Rec (Hoboken) 2018; 301:1815-1819. [PMID: 30324658 DOI: 10.1002/ar.23906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 12/26/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022]
Abstract
This study aimed to evaluate the effects and mechanism of action of ASK1 gene on the growth and migration of gastric cancer (GC) cells. Total RNA was extracted from the gastric cell lines and GC tissues. The expression level of ASK1, and the association between ASK1 expression and clinicopathological characteristics was assessed by real-time polymerase chain reaction. The effects of ASK1 on the proliferation of HGC-27 cells were assessed by the CCK-8 assay. In addition, the effects of ASK1 on the migration of HGC-27 cells were analyzed by the migration assay using transwell chambers. The expression levels of signaling proteins related to cell migration were detected by Western blotting. Although no significant differences were observed in the expression levels of ASK1 between the GC tissue samples and the normal tissue samples (P = 0.241), ASK1 expression correlated with tumor lymph node metastasis (P = 0.008). Furthermore, ASK1 inhibited proliferation and migration of HGC-27 cells. The increase in the expression of ASK1 in HGC-27 cells induced the activation of the JNK and p38 signaling pathways. The findings demonstrated that increased ASK1 expression level inhibited migration and proliferation of HGC-27 gastric cancer cells, whereas the possible mechanism of action may be attributed to the activation of the JNK and p38 signaling pathways. Anat Rec, 301:1815-1819, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaochen Wan
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Liying Shi
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Xiaohong Ma
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Haimin Tang
- Department of Pathology, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| | - Guoyou Wu
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China
| |
Collapse
|
35
|
Kim KB, Lee S, Kang I, Kim JH. Momordica charantia Ethanol Extract Attenuates H₂O₂-Induced Cell Death by Its Antioxidant and Anti-Apoptotic Properties in Human Neuroblastoma SK-N-MC Cells. Nutrients 2018; 10:nu10101368. [PMID: 30249986 PMCID: PMC6213776 DOI: 10.3390/nu10101368] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/20/2018] [Accepted: 09/22/2018] [Indexed: 01/29/2023] Open
Abstract
Oxidative stress, which is induced by reactive oxygen species (ROS), causes cellular damage which contributes to the pathogenesis of neurodegenerative diseases. Momordica charantia (MC), a traditional medicinal plant, is known to have a variety of health benefits, such as antidiabetic, anti-inflammatory, and antioxidant effects. However, it is unknown whether MC has protective effects against oxidative stress-induced neuronal cell death. The aim of this study was to investigate the potential action of MC on oxidative stress induced by H2O2. First, we tested whether the pretreatment of Momordica charantia ethanol extract (MCEE) attenuates H2O2-induced cell death in human neuroblastoma SK-N-MC cells. MCEE pretreatment significantly improved cell viability and apoptosis that deteriorated by H2O2. Further, MCEE ameliorated the imbalance between intracellular ROS production and removal through the enhancement of the intracellular antioxidant system. Intriguingly, the inhibition of apoptosis was followed by the blockage of mitochondria-dependent cell death cascades and suppression of the phosphorylation of the mitogen-activated protein kinase signaling (MAPKs) pathway by MCEE. Taken together, MCEE was shown to be effective in protecting against H2O2-induced cell death through its antioxidant and anti-apoptotic properties.
Collapse
Affiliation(s)
- Kkot Byeol Kim
- Research Institute, Seoul Medical Center, Seoul 02053, Korea.
| | - SeonAh Lee
- Research Institute, Seoul Medical Center, Seoul 02053, Korea.
| | - Inhae Kang
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| | - Jung-Hee Kim
- Research Institute, Seoul Medical Center, Seoul 02053, Korea.
- Department of Neurosurgery, Seoul Medical Center, Seoul 02053, Korea.
| |
Collapse
|
36
|
Li F, Zhang X, Li Y, Lu K, Yin R, Ming J. Phenolics extracted from tartary (Fagopyrum tartaricum L. Gaerth) buckwheat bran exhibit antioxidant activity, and an antiproliferative effect on human breast cancer MDA-MB-231 cells through the p38/MAP kinase pathway. Food Funct 2018; 8:177-188. [PMID: 27942664 DOI: 10.1039/c6fo01230b] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phenolics extracted from tartary buckwheat (Fagopyrum tartaricum L. Gaerth) bran were analyzed quantitatively and qualitatively. The bioactivity of the phenolic extracts was evaluated, such as the antioxidant activity, and the inhibition capacity on the growth of cancer cells. The molecular mechanism for the inhibitive effect on cancer cells was explored. Results indicated that tartary buckwheat bran phenolics mainly exist in a free form, and free phenolics were twice as abundant as bound phenolics. Free caffeic acid (119.75 μg per 100 mg DW) and bound rutin (51.66 μg per 100 mg DW) represented the main free and bound phenolic compounds, respectively. The free phenolic extract contributed to the major (>90%) antioxidant activities including the oxygen radical antioxidant capacity (ORAC) and cellular antioxidant activity (CAA). The free phenolic extract exhibited anticancer activity for human breast cancer MDA-MB-231 cells in a dose-dependent manner. This significant inhibition effect was achieved through the p38/MAP kinase pathway by inducing cell apoptosis (up-regulating p-p38 and p-ASK1 expressions and down-regulating TRAF2 and p-p53 expressions), and negatively regulating the progression of the cell cycle from the G1 to S phase (increased expression of p21 and suppressed expressions of PCNA, cyclin D1 and CDK4). All these results indicated that tartary buckwheat bran could be a rich resource of natural antioxidants and inhibitors for the growth of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Fuhua Li
- College of Food Science, Southwest University, Chongqing, 400715, PR China. and School of Food Science and Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, 510640, China
| | - Xiaoli Zhang
- College of Food Science, Southwest University, Chongqing, 400715, PR China.
| | - Yao Li
- College of Food Science, Southwest University, Chongqing, 400715, PR China.
| | - Keke Lu
- College of Food Science, Southwest University, Chongqing, 400715, PR China.
| | - Ran Yin
- Department of Food Science, 245 Stocking Hall and Cornell University, Ithaca, New York 14853-7201, USA
| | - Jian Ming
- College of Food Science, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
37
|
Nabinger DD, Altenhofen S, Bitencourt PER, Nery LR, Leite CE, Vianna MRMR, Bonan CD. Nickel exposure alters behavioral parameters in larval and adult zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 624:1623-1633. [PMID: 29102187 DOI: 10.1016/j.scitotenv.2017.10.057] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/06/2017] [Accepted: 10/07/2017] [Indexed: 06/07/2023]
Abstract
Nickel is a heavy metal that, at high concentrations, leads to environmental contamination and causes health problems. We evaluated the effects of NiCl2 exposure on cognition and behavior in larval and adult zebrafish. Larval and adult zebrafish were exposed to NiCl2 concentrations (0.025, 2.0, 5.0, and 15.0mg/L) or water (control) in two treatment regimens: acute and subchronic. Larvae were exposed to NiCl2 for 2h (acute treatment: 5-day-old larvae treated for 2h, tested after treatment) or 11days (subchronic treatment: 11-day-old larvae treated since fertilization, tested at 5, 8 and 11days post-fertilization, dpf). Adults were exposed for 12h (acute treatment) or 96h (subchronic treatment) and were tested after the treatment period. In both regimens, exposed zebrafish showed concentration-dependent increases in body nickel levels compared with controls. For larvae, delayed hatching, decreased heart rate and morphological alterations were observed in subchronically treated zebrafish. Larvae from subchronic treatment tested at 5dpf decrease distance and mean speed at a low concentration (0.025mg/L) and increased at higher concentrations (5.0 and 15.0mg/L). Subchronic treated larvae decrease locomotion at 15.0mg/L at 8 and 11dpf, whereas decreased escape responses to an aversive stimulus was observed at 2.0, 5.0 and 15.0mg/L in all developmental stages. For adults, the exploratory behavior test showed that subchronic nickel exposure induced anxiogenic-like behavior and decrease aggression, whereas impaired memory was observed in both treatments. These results indicate that exposure to nickel in early life stages of zebrafish leads to morphological alterations, avoidance response impairment and locomotor deficits whereas acute and subchronic exposure in adults resulst in anxiogenic effects, impaired memory and decreased aggressive behavior. These effects may be associated to neurotoxic actions of nickel and suggest this metal may influence animals' physiology in doses that do not necessarily impact their survival.
Collapse
Affiliation(s)
- Débora Dreher Nabinger
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil
| | - Stefani Altenhofen
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil
| | - Paula Eliete Rodrigues Bitencourt
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil
| | - Laura Roesler Nery
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Biologia e Desenvolvimento do Sistema Nervoso, Porto Alegre, RS, Brazil
| | | | - Mônica Ryff Moreira Roca Vianna
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Biologia e Desenvolvimento do Sistema Nervoso, Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- PUCRS, Faculdade de Biociências, Programa de Pós-Graduação em Biologia Celular e Molecular, Laboratório de Neuroquímica e Psicofarmacologia, Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Lee CH, Ying TH, Chiou HL, Hsieh SC, Wen SH, Chou RH, Hsieh YH. Alpha-mangostin induces apoptosis through activation of reactive oxygen species and ASK1/p38 signaling pathway in cervical cancer cells. Oncotarget 2018; 8:47425-47439. [PMID: 28537893 PMCID: PMC5564576 DOI: 10.18632/oncotarget.17659] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/20/2017] [Indexed: 01/04/2023] Open
Abstract
Alpha-mangostin, a natural xanthonoid, has been reported to possess the anti-cancer property in various types of human cancer. However, its effects and mechanism of α-mangostin in cervical cancer remain unclear. We found that α-mangostin effectively inhibited cell viability, resulted in loss of mitochondrial membrane potential (MMP), release of cytochrome C, increase of Bax, decrease of Bcl-2, and activation of caspase-9/caspase-3 cascade in cervical cancer cells. Alpha-mangostin elevated the contents of reactive oxygen species (ROS) to activate p38. Disrupting ASK1/p38 signaling pathway by a specific inhibitor of p38, or by the siRNAs against ASK1, MKK3/6, or p38, significantly abolished α-mangostin-induced cell death and apoptotic responses. Moreover, α-mangostin also repressed tumor growth in accordance with increased levels of p-ASK1, p-p38, cleaved-PARP and cleaved-caspase-3 in the tumor mass from the mouse xenograft model of cervical cancer. In the current study, we provided first evidence to demonstrate that dietary antioxidant α-mangostin could inhibit the tumor growth of cervical cancer cells through enhancing ROS amounts to activate ASK1/p38 signaling pathway and damage the integrity of mitochondria and thereby induction of apoptosis in cervical cancer cells.
Collapse
Affiliation(s)
- Chien-Hsing Lee
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Division of Pediatric Surgery, Department of Surgery, China Medical University Children's Hospital, Taichung, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Ching Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Shiua-Hua Wen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
39
|
Xu C, Shi Q, Zhang L, Zhao H. High molecular weight hyaluronan attenuates fine particulate matter-induced acute lung injury through inhibition of ROS-ASK1-p38/JNK-mediated epithelial apoptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 59:190-198. [PMID: 29625389 DOI: 10.1016/j.etap.2018.03.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
Inhalation of fine particulate matter (PM2.5) is asscoiated with lung injury. High molecular weight hyaluronan (HMW-HA) is an essential constituent of extracellular matrix (ECM), exhibiting anti-oxidative and anti-inflammatory properties when administered by injection, inhalation, nebulization or gene delivery of HA synthases. The aim of the present study is to determine whether HMW-HA alleviates PM2.5-induced acute lung injury (ALI) and investigate the underlying mechanisms. We observed that HMW-HA suppressed pathological injury, inflammation, oxidative stress, edema and epithelial damage caused by PM2.5 in the lungs of the rats. The protective mechanism of HMW-HA was further explored in vitro. The results elucidated that reactive oxygen species (ROS) was involved in PM2.5-induced cell apoptosis, and HMW-HA mitigated the oxidative potential of PM2.5, subsequently inhibiting phosphorylation of ASK1 at Thr845, downstream phosphorylation of p38 and JNK, and eventual apoptosis. Our study indicates that HMW-HA is a promising strategy in the prevention of PM2.5-induced pulmonary damage.
Collapse
Affiliation(s)
- Chenming Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, PR China
| | - Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, PR China.
| | - Leifang Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, PR China
| | - Hang Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, PR China
| |
Collapse
|
40
|
Han A, Zou L, Gan X, Li Y, Liu F, Chang X, Zhang X, Tian M, Li S, Su L, Sun Y. ROS generation and MAPKs activation contribute to the Ni-induced testosterone synthesis disturbance in rat Leydig cells. Toxicol Lett 2018; 290:36-45. [PMID: 29567110 DOI: 10.1016/j.toxlet.2018.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/07/2018] [Accepted: 03/13/2018] [Indexed: 01/23/2023]
Abstract
Nickel (Ni) can disorder testosterone synthesis in rat Leydig cells, whereas the mechanisms remain unclear. The aim of this study was to investigate the role of reactive oxygen species (ROS) and mitogen-activated protein kinases (MAPKs) in Ni-induced disturbance of testosterone synthesis in rat Leydig cells. The testosterone production and ROS levels were detected in Leydig cells. The mRNA and protein levels of testosterone synthetase, including StAR, CYP11A1, 3β-HSD, CYP17A1 and 17β-HSD, were determined. Effects of Ni on the ERK1/2, p38 and JNK MAPKs were also investigated. The results showed that Ni triggered ROS generation, consequently resulted in the decrease of testosterone synthetase expression and testosterone production in Leydig cells, which were then attenuated by ROS scavengers of N-acetylcysteine (NAC) and 2,2,6,6-tetramethyl-1-piperidinyloxy (TEMPO), indicating that ROS are involved in the Ni-induced testosterone biosynthesis disturbance. Meanwhile Ni activated the ERK1/2, p38 and JNK MAPKs. Furthermore, Ni-inhibited testosterone synthetase expression levels and testosterone secretion were all alleviated by co-treatment with MAPK specific inhibitors (U0126 and SB203580, respectively), implying that Ni inhibited testosterone synthesis through activating ERK1/2 and p38 MAPK signal pathways in Leydig cells. In conclusion, these findings suggest that Ni causes testosterone synthesis disorder, partly, via ROS and MAPK signal pathways.
Collapse
Affiliation(s)
- Aijie Han
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Lingyue Zou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xiaoqin Gan
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Yu Li
- Department of Infectious Diseases, Shaanxi Provincial People's hospital, Xi'an 710068, China
| | - Fangfang Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xiaotian Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Minmin Tian
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Sheng Li
- Lanzhou Municipal Center for Disease Control, Lanzhou, China
| | - Li Su
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
41
|
Xu J, Wise JTF, Wang L, Schumann K, Zhang Z, Shi X. Dual Roles of Oxidative Stress in Metal Carcinogenesis. J Environ Pathol Toxicol Oncol 2018; 36:345-376. [PMID: 29431065 DOI: 10.1615/jenvironpatholtoxicoloncol.2017025229] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It has been well established that environmental and occupational exposure to heavy metal causes cancer in several organs. Although the exact mechanism of heavy metal carcinogenesis remains elusive, metal-generated reactive oxygen species (ROS) are essential. ROS can play two roles in metal carcinogenesis; two stages in the process of metal carcinogenesis differ in the amounts of ROS activating a dual redox-mediated mechanism. In the early stage of metal carcinogenesis, ROS acts in an oncogenic role. However, in the late stage of metal carcinogenesis, ROS plays an antioncogenic role. Similarly, NF-E2-related factor 2 (Nrf2) also has two different roles, which makes it a key molecule for separating metal carcinogenesis into two different stages. In the early stage, inducible Nrf2 fights against elevated ROS to decrease cell transformation by its antioxidant protection property. In the late stage, constitutively activated Nrf2 manipulates reduced ROS to perform a comfortable environment for apoptosis resistance through an oncogenic role. Interestingly, a cunning carcinogenic mechanism takes advantage of the dual role of Nrf2 to implement the dual role of ROS through a series of redox adaption mechanisms. In this review, we discuss the paradox in the rationales behind the two opposite ROS roles and focus on their potential pharmacological application. The dual role of ROS represents a 'double-edged sword' with many possible novel ROS-mediated strategies in cancer therapy in metal carcinogenesis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Anesthesiology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - James T F Wise
- Division of Nutritional Sciences, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kortney Schumann
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
42
|
Guo H, Cui H, Fang J, Zuo Z, Deng J, Wang X, Zhao L, Chen K, Deng J. Nickel chloride (NiCl2) in hepatic toxicity: apoptosis, G2/M cell cycle arrest and inflammatory response. Aging (Albany NY) 2017; 8:3009-3027. [PMID: 27824316 PMCID: PMC5191883 DOI: 10.18632/aging.101108] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/05/2023]
Abstract
Up to now, the precise mechanism of Ni toxicology is still indistinct. Our aim was to test the apoptosis, cell cycle arrest and inflammatory response mechanism induced by NiCl2 in the liver of broiler chickens. NiCl2 significantly increased hepatic apoptosis. NiCl2 activated mitochondria-mediated apoptotic pathway by decreasing Bcl-2, Bcl-xL, Mcl-1, and increasing Bax, Bak, caspase-3, caspase-9 and PARP mRNA expression. In the Fas-mediated apoptotic pathway, mRNA expression levels of Fas, FasL, caspase-8 were increased. Also, NiCl2 induced ER stress apoptotic pathway by increasing GRP78 and GRP94 mRNA expressions. The ER stress was activated through PERK, IRE1 and ATF6 pathways, which were characterized by increasing eIF2α, ATF4, IRE1, XBP1 and ATF6 mRNA expressions. And, NiCl2 arrested G2/M phase cell cycle by increasing p53, p21 and decreasing cdc2, cyclin B mRNA expressions. Simultaneously, NiCl2 increased TNF-α, IL-1β, IL-6, IL-8 mRNA expressions through NF-κB activation. In conclusion, NiCl2 induces apoptosis through mitochondria, Fas and ER stress-mediated apoptotic pathways and causes cell cycle G2/M phase arrest via p53-dependent pathway and generates inflammatory response by activating NF-κB pathway.
Collapse
Affiliation(s)
- Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University Ya'an 625014, China
| | - Kejie Chen
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China
| | - Jie Deng
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an 625014, China
| |
Collapse
|
43
|
Son YO, Pratheeshkumar P, Wang Y, Kim D, Zhang Z, Shi X. Protection from Cr(VI)-induced malignant cell transformation and tumorigenesis of Cr(VI)-transformed cells by luteolin through Nrf2 signaling. Toxicol Appl Pharmacol 2017; 331:24-32. [PMID: 28416455 PMCID: PMC5568479 DOI: 10.1016/j.taap.2017.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/09/2017] [Accepted: 04/13/2017] [Indexed: 11/19/2022]
Abstract
Cr(VI) is a well known environmental carcinogen, but its mechanism of action and the measures required to mitigate its effects remain to be investigated. Our previous studies showed that exposure of human bronchial epithelial (BEAS-2B) cells to Cr(VI) caused malignant transformation, that these transformed cells progressed through tumorigenesis, and that luteolin, a natural compound, inhibited both of these processes. The present study investigates the underlying mechanisms by which luteolin protects cells against Cr(VI)-induced transformation and tumorigenesis. The present study shows that luteolin activates inducible Nrf2 to inhibit Cr(VI)-generated reactive oxygen species (ROS) in normal BEAS-2B cells. The decreased ROS level is likely responsible for the protective effect of luteolin against Cr(VI)-induced malignant cell transformation in normal cells. By contrast, in cells that have been transformed by Cr(VI), Nrf2 is constitutively activated, and its target proteins, heme oxygenase 1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1), and superoxide dismutase 1/2 (SOD1/SOD2) are all constitutively activated, and ROS levels are low. Bcl-2, an anti-apoptotic protein and target protein of Nrf2 is elevated. Cr(VI)-transformed BEAS-2B cells develop apoptosis resistance, increasing the survival of these transformed cells. Luteolin decreases interactions between Nrf2 and the antioxidant response element sites of its target anti-apoptotic and antioxidant proteins, Bcl-2, Bcl-XL, and HO-1, which results in decreased constitutive Nrf2 activation. The decreased constitutive Nrf2 activation, decrease in Nrf2 target proteins and consequent apoptosis resistance by luteolin are possible mechanisms that mediate the protective effect of luteolin in Cr(VI)-transformed cells.
Collapse
Affiliation(s)
- Young-Ok Son
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yuting Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Donghern Kim
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| |
Collapse
|
44
|
Yu L, Gan X, Liu X, An R. Calcium oxalate crystals induces tight junction disruption in distal renal tubular epithelial cells by activating ROS/Akt/p38 MAPK signaling pathway. Ren Fail 2017; 39:440-451. [PMID: 28335665 PMCID: PMC6014313 DOI: 10.1080/0886022x.2017.1305968] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tight junction plays important roles in regulating paracellular transports and maintaining cell polarity. Calcium oxalate monohydrate (COM) crystals, the major crystalline composition of kidney stones, have been demonstrated to be able to cause tight junction disruption to accelerate renal cell injury. However, the cellular signaling involved in COM crystal-induced tight junction disruption remains largely to be investigated. In the present study, we proved that COM crystals induced tight junction disruption by activating ROS/Akt/p38 MAPK pathway. Treating Madin–Darby canine kidney (MDCK) cells with COM crystals induced a substantial increasing of ROS generation and activation of Akt that triggered subsequential activation of ASK1 and p38 mitogen-activated protein kinase (MAPK). Western blot revealed a significantly decreased expression of ZO-1 and occludin, two important structural proteins of tight junction. Besides, redistribution and dissociation of ZO-1 were observed by COM crystals treatment. Inhibition of ROS by N-acetyl-l-cysteine (NAC) attenuated the activation of Akt, ASK1, p38 MAPK, and down-regulation of ZO-1 and occludin. The redistribution and dissociation of ZO-1 were also alleviated by NAC treatment. These results indicated that ROS were involved in the regulation of tight junction disruption induced by COM crystals. In addition, the down-regulation of ZO-1 and occludin, the phosphorylation of ASK1 and p38 MAPK were also attenuated by MK-2206, an inhibitor of Akt kinase, implying Akt was involved in the disruption of tight junction upstream of p38 MAPK. Thus, these results suggested that ROS-Akt-p38 MAPK signaling pathway was activated in COM crystal-induced disruption of tight junction in MDCK cells.
Collapse
Affiliation(s)
- Lei Yu
- a Department of Urology , the First Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang Province , P.R. China
| | - Xiuguo Gan
- a Department of Urology , the First Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang Province , P.R. China
| | - Xukun Liu
- b Department of General Surgery , the People's Hospital of Jixi , Jixi , Heilongjiang Province , P.R. China
| | - Ruihua An
- a Department of Urology , the First Affiliated Hospital of Harbin Medical University , Harbin , Heilongjiang Province , P.R. China
| |
Collapse
|
45
|
Manuel-Manresa P, Korrodi-Gregório L, Hernando E, Villanueva A, Martínez-García D, Rodilla AM, Ramos R, Fardilha M, Moya J, Quesada R, Soto-Cerrato V, Pérez-Tomás R. Novel Indole-based Tambjamine-Analogues Induce Apoptotic Lung Cancer Cell Death through p38 Mitogen-Activated Protein Kinase Activation. Mol Cancer Ther 2017; 16:1224-1235. [DOI: 10.1158/1535-7163.mct-16-0752] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/16/2016] [Accepted: 03/22/2017] [Indexed: 11/16/2022]
|
46
|
Son YO, Pratheeshkumar P, Divya SP, Zhang Z, Shi X. Nuclear factor erythroid 2-related factor 2 enhances carcinogenesis by suppressing apoptosis and promoting autophagy in nickel-transformed cells. J Biol Chem 2017; 292:8315-8330. [PMID: 28330870 DOI: 10.1074/jbc.m116.773986] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/20/2017] [Indexed: 12/13/2022] Open
Abstract
Nickel-containing compounds are widely used in industry. Nickel is a known human carcinogen that primarily affects the lungs. Proposed mechanisms of nickel-induced carcinogenesis include disruption of cellular iron homeostasis, generation of reactive oxygen species (ROS), and induction of hypoxia signaling. However, the precise molecular mechanisms of nickel-induced malignant transformation and tumor development remain unclear. This study shows that the transcription factor Nrf2 is highly expressed in lung tumor tissue and in nickel-transformed human lung bronchial epithelial BEAS-2B cells (NiT cells). Additionally, constitutively high levels of Nrf2 play a critical role in apoptosis resistance in NiT cells. Basal ROS levels were extremely low in NiT cells and were correlated with elevated expression levels of both antioxidant enzymes (e.g. catalase and superoxide dismutases) and antiapoptotic proteins (e.g. Bcl-2 and Bcl-xL). These processes are tightly controlled by Nrf2. Autophagy inhibition, induced pharmacologically or genetically, enhanced Ni2+-induced apoptosis, indicating that the induction of autophagy is the cause of apoptosis resistance in NiT cells. Using similar approaches, we show that in NiT cells the inhibition of apoptosis decreases autophagy. We have shown that Stat3, which is up-regulated by Nrf2, controls autophagy induction in NiT cells. Colony formation and tumor growth were significantly attenuated by knockdown of Nrf2 or Bcl-2. Taken together, this study demonstrates that in NiT cells constitutively high Nrf2 expression inhibits apoptosis by up-regulating antioxidant enzymes and antiapoptotic proteins to increase autophagy via Stat3 signaling. These findings indicate that the Nrf2-mediated suppression of apoptosis and promotion of autophagy contribute to nickel-induced transformation and tumorigenesis.
Collapse
Affiliation(s)
- Young-Ok Son
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305; National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea.
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Sasidharan Padmaja Divya
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Xianglin Shi
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305.
| |
Collapse
|
47
|
Oral Exposure to Atrazine Induces Oxidative Stress and Calcium Homeostasis Disruption in Spleen of Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7978219. [PMID: 27957240 PMCID: PMC5121465 DOI: 10.1155/2016/7978219] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 08/24/2016] [Accepted: 08/29/2016] [Indexed: 01/02/2023]
Abstract
The widely used herbicide atrazine (ATR) can cause many adverse effects including immunotoxicity, but the underlying mechanisms are not fully understood. The current study investigated the role of oxidative stress and calcium homeostasis in ATR-induced immunotoxicity in mice. ATR at doses of 0, 100, 200, or 400 mg/kg body weight was administered to Balb/c mice daily for 21 days by oral gavage. The studies performed 24 hr after the final exposure showed that ATR could induce the generation of reactive oxygen species in the spleen of the mice, increase the level of advanced oxidation protein product (AOPP) in the host serum, and cause the depletion of reduced glutathione in the serum, each in a dose-related manner. In addition, DNA damage was observed in isolated splenocytes as evidenced by increase in DNA comet tail formation. ATR exposure also caused increases in intracellular Ca2+ within splenocytes. Moreover, ATR treatment led to increased expression of genes for some antioxidant enzymes, such as HO-1 and Gpx1, as well as increased expression of NF-κB and Ref-1 proteins in the spleen. In conclusion, it appears that oxidative stress and disruptions in calcium homeostasis might play an important role in the induction of immunotoxicity in mice by ATR.
Collapse
|
48
|
Gomes SIL, Soares AMVM, Amorim MJB. Effect of Cu and Ni on cellular energy allocation in Enchytraeus albidus. ECOTOXICOLOGY (LONDON, ENGLAND) 2016; 25:1523-1530. [PMID: 27582176 DOI: 10.1007/s10646-016-1706-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/03/2016] [Indexed: 06/06/2023]
Abstract
Effects of nickel and copper on Enchytraeus albidus (Oligochaeta) were investigated using the cellular energy allocation approach. This methodology is used to evaluate the energetic status of an organism and is indicative of its overall condition. Enchytraeids were exposed to the reproduction Effect Concentrations (EC50 and EC90), and the parameters measured were the total energy reserves available (protein, carbohydrate and lipid budgets) and the energy consumption [based on electron transport system activity] which were further integrated to obtain the cellular energy allocation over different periods of exposure (0-2, 2-4 and 4-8 days). Carbohydrates (in comparison to lipids and proteins) were the only energy source mobilized in the case of nickel within 8 days of exposure. For copper exposure, protein budgets were also strongly reduced. Energy consumption increased in a time and dose-dependent way for copper and in the longer exposure period (4-8 days) at the EC90 for Ni exposure, indicating that this is a good biomarker for effects of short-time metal exposure, while cellular energy allocation was only significantly reduced for the EC90 of copper (4-8 days) and EC50 of nickel (2-4 days). The effects of nickel at concentrations causing 50 and 90 % decrease in reproduction were likely not due to the changes in cellular energy allocation within 8 days of exposure.
Collapse
Affiliation(s)
- Susana I L Gomes
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal.
| | - Amadeu M V M Soares
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Mónica J B Amorim
- Department of Biology & CESAM, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
49
|
Talreja J, Talwar H, Ahmad N, Rastogi R, Samavati L. Dual Inhibition of Rip2 and IRAK1/4 Regulates IL-1β and IL-6 in Sarcoidosis Alveolar Macrophages and Peripheral Blood Mononuclear Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:1368-78. [PMID: 27402699 DOI: 10.4049/jimmunol.1600258] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Sarcoidosis is a multisystem granulomatous disease of unknown etiology that primarily affects the lungs. Our previous work indicates that activation of p38 plays a pivotal role in sarcoidosis inflammatory response. Therefore, we investigated the upstream kinase responsible for activation of p38 in sarcoidosis alveolar macrophages (AMs) and PBMCs. We identified that sustained p38 phosphorylation in sarcoidosis AMs and PBMCs is associated with active MAPK kinase 4 but not with MAPK kinase 3/6. Additionally, we found that sarcoidosis AMs exhibit a higher expression of IRAK1, IRAK-M, and receptor interacting protein 2 (Rip2). Surprisingly, ex vivo treatment of sarcoidosis AMs or PBMCs with IRAK1/4 inhibitor led to a significant increase in IL-1β mRNA expression both spontaneously and in response to TLR2 ligand. However, a combination of Rip2 and IRAK-1/4 inhibitors significantly decreased both IL-1β and IL-6 production in sarcoidosis PBMCs and moderately in AMs. Importantly, a combination of Rip2 and IRAK-1/4 inhibitors led to decreased IFN-γ and IL-6 and decreased percentage of activated CD4(+)CD25(+) cells in PBMCs. These data suggest that in sarcoidosis, both pathways, namely IRAK and Rip2, are deregulated. Targeted modulation of Rip2 and IRAK pathways may prove to be a novel treatment for sarcoidosis.
Collapse
Affiliation(s)
- Jaya Talreja
- Division of Pulmonary & Critical Care and Sleep Medicine, Department of Internal Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI 48201; and
| | - Harvinder Talwar
- Division of Pulmonary & Critical Care and Sleep Medicine, Department of Internal Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI 48201; and
| | - Nisar Ahmad
- Division of Pulmonary & Critical Care and Sleep Medicine, Department of Internal Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI 48201; and
| | - Ruchi Rastogi
- Division of Pulmonary & Critical Care and Sleep Medicine, Department of Internal Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI 48201; and
| | - Lobelia Samavati
- Division of Pulmonary & Critical Care and Sleep Medicine, Department of Internal Medicine, Wayne State University School of Medicine and Detroit Medical Center, Detroit, MI 48201; and Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| |
Collapse
|
50
|
Deng J, Guo H, Cui H, Fang J, Zuo Z, Deng J, Wang X, Zhao L. Oxidative stress and inflammatory responses involved in dietary nickel chloride (NiCl 2)-induced pulmonary toxicity in broiler chickens. Toxicol Res (Camb) 2016; 5:1421-1433. [PMID: 30090446 DOI: 10.1039/c6tx00197a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/06/2016] [Indexed: 01/12/2023] Open
Abstract
The respiratory system is the primary target of nickel or nickel compound toxicity after inhalation exposure. There are no reports on the effects of nickel or nickel compounds on the lung via dietary administration at present. This study aimed to investigate pulmonary toxicity induced by dietary NiCl2 in broiler chickens by using histopathology, qRT-PCR, and ELISA. In comparison with the control group, NiCl2 intake induced oxidative damage to DNA (upregulation of 8-OHdG) and lipid peroxidation (upregulation of MDA), which was associated with the upregulation of NO and the downregulation of the expression levels and activities of pulmonary CuZn-SOD, Mn-SOD, CAT, GSH-Px, GR and GST mRNA. Also, the T-AOC activity, GSH content, ability to inhibit the generation of hydroxyl radicals, and ratio of GSH/GSSG were decreased in the groups treated with NiCl2. Concurrently, the mRNA expression levels of iNOS, TNF-α, COX-2, IL-1β, IL-6, IL-8, IL-18 and IFN-γ were increased via the activation of NF-κB, and the mRNA expression levels of anti-inflammatory mediators including IL-2, IL-4 and IL-13 were decreased in the groups treated with NiCl2. The above-mentioned results were the first to demonstrate that NiCl2 intake induced pulmonary oxidative stress and inflammatory responses via the dietary pathway, which subsequently contributed to histopathological lesions and dysfunction.
Collapse
Affiliation(s)
- Jie Deng
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China
| | - Hongrui Guo
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China
| | - Hengmin Cui
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| | - Jing Fang
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| | - Zhicai Zuo
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| | - Junliang Deng
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| | - Xun Wang
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| | - Ling Zhao
- College of Veterinary Medicine , Sichuan Agricultural University , Wenjiang , Chengdu , Sichuan 611130 , China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province , Wenjiang , Chengdu , Sichuan 611130 , China . ; ; Tel: +86-136-0826-4628
| |
Collapse
|