1
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
2
|
Digman A, Pajarillo E, Kim S, Ajayi I, Son DS, Aschner M, Lee E. Tamoxifen induces protection against manganese toxicity by REST upregulation via the ER-α/Wnt/β-catenin pathway in neuronal cells. J Biol Chem 2025; 301:108529. [PMID: 40280417 DOI: 10.1016/j.jbc.2025.108529] [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: 09/17/2024] [Revised: 03/17/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic exposure to elevated levels of manganese (Mn) causes a neurological disorder referred to as manganism, with symptoms resembling Parkinson's disease (PD). The repressor element-1 silencing transcription factor (REST) has been shown to be neuroprotective in several neurological disorders, including PD, suggesting that identifying REST upregulation mechanisms is an important avenue for the development of novel therapeutics. 17β-estradiol (E2) activates the Wnt/β-catenin signaling, which is known to increase REST transcription. E2 and tamoxifen (TX), a selective estrogen receptor modulator, exerted protection against Mn toxicity. In this study, we tested if TX upregulates REST potentially via Wnt/β-catenin signaling in Cath.a-differentiated (CAD) neuronal cells using luciferase assay, qPCR, Western blot analysis, immunocytochemistry, mutagenesis, chromatin immunoprecipitation, and electrophoretic mobility shift assay. TX (1 μM) increased REST promoter activities and mRNA/protein levels and attenuated Mn (250 μM)-decreased REST transcription in parallel with TX's protective effects against Mn-induced toxicity, potentially via Wnt. TX activated Wnt/β-catenin signaling by preventing β-catenin degradation via inactivation of glycogen synthase kinase-3 beta, leading to increased β-catenin levels and its nuclear translocation and binding to T-cell factor/lymphoid enhancer binding factor sites on Wnt-responsive elements (WRE) of the REST promoter. Mutation of WRE abolished TX-induced REST promoter activity. TX-induced Wnt signaling activation was primarily via the estrogen receptor (ER)-α, although ER-β and G protein-coupled estrogen receptor 1 also mediated TX's action on REST transcription. These findings underscore the critical role of Wnt/β-catenin signaling in TX-induced REST transcription, affording protection mechanisms against Mn toxicity and neurological disorders associated with REST dysfunction.
Collapse
Affiliation(s)
- Alexis Digman
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Edward Pajarillo
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Sanghoon Kim
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Itunu Ajayi
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, USA.
| |
Collapse
|
3
|
Singh A, Prakash A, Mishra J, Luthra PM. Discovery of novel A 2AR antagonist via 3D-QSAR pharmacophore modeling: neuroprotective effects in 6-OHDA-induced SH-SY5Y cells and haloperidol-induced Parkinsonism in C57 bl/6 mice. Mol Divers 2025:10.1007/s11030-025-11120-x. [PMID: 39899125 DOI: 10.1007/s11030-025-11120-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder which is caused by abrupt degeneration of dopaminergic neuronal cells in the substantia nigra pars compacta (SNPc) area of the midbrain. Adenosine A2A receptors have become promising therapeutic targets for PD; however, many A2A receptor antagonists face challenges, such as limited accessibility or failure in clinical trials due to poor selectivity and bioavailability. To identify novel A2A receptor antagonists, a 3D-QSAR-pharmacophore modeling approach was employed, involving virtual screening of ZINC, NCI, and MayBridge databases. The virtual hits were filtered via ADMET criteria to select compounds with favorable bioavailability and solubility profiles. From the MayBridge database, a potent monocyclic A2A receptor antagonist, AW00032 (N-(furan-2-ylmethyl)-5-methylthiazole-4-yl) thiophene-2-sulfonamide, was identified. AW00032 possessed key pharmacophoric features: two lipophilic hydrogen bond acceptors, one hydrophobic aliphatic/aromatic group, and one aromatic ring. Docking analysis revealed AW00032 had a strong binding affinity for A2A receptors (1.23 nM, ∆G - 10.49 kcal/mol), and its ADMET profile indicated good bioavailability. In 6-OHDA induced SH-SY5Y cells, AW00032 increased dopamine levels and tyrosine hydroxylase (TH) expression, demonstrating its potential as an A2A receptor antagonist. AW00032, discovered through 3D-QSAR pharmacophore modeling, also reduced reactive oxygen species (ROS) levels and showed depletion in mitochondrial dysfunction in 6-OHDA-induced SH-SY5Y cells. It exhibited A2A receptor antagonist activity comparable to the standard antagonist ZM241385, partially restoring dopamine and TH levels. Furthermore, AW00032 improved behavioral symptoms in haloperidol-induced C-57 bl/6 mice.
Collapse
Affiliation(s)
- Ankit Singh
- Neuropharmaceutical Chemistry Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Amresh Prakash
- Neuropharmaceutical Chemistry Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Jyoti Mishra
- Neuropharmaceutical Chemistry Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
4
|
Hijam AC, Tongbram YC, Nongthombam PD, Meitei HN, Koijam AS, Rajashekar Y, Haobam R. Traditionally used edible medicinal plants protect against rotenone induced toxicity in SH-SY5Y cells-a prospect for the development of herbal nutraceuticals. Neurochem Int 2024; 180:105855. [PMID: 39244037 DOI: 10.1016/j.neuint.2024.105855] [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: 06/30/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Plants are good sources of pharmacologically active compounds. The present study aimed to examine the neuroprotective potentials of the methanol extracts of Salix tetrasperma Roxb. leaf (STME) and Plantago asiatica L. (PAME), two edibles medicinal plants of Manipur, India against neurotoxicity induced by rotenone in SH-SY5Y cells. Free radical quenching activities were evaluated by ABTS and DPPH assays. The cytotoxicity of rotenone and the neuronal survival were assessed by MTT assay and MAP2 expression analysis. DCF-DA, Rhodamine 123 (Rh-123), and DAPI measured the intracellular reactive oxygen species (ROS) levels, mitochondrial membrane potential (MMP), and apoptotic nuclei, respectively. Superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) activities were also assessed. LC-QTOF-MS analysis was performed for the identification of the compounds present in STME and PAME. The study showed that both the plant extracts (STME and PAME) showed antioxidant and neuroprotective capabilities in rotenone-induced neurotoxicity by preventing oxidative stress through the reduction of intracellular ROS levels and reversing the activities of GPx, SOD, and CAT caused by rotenone. Further, both plants prevented apoptotic cell death by normalizing the steady state of MMP and protecting nuclear DNA condensation. LC-QTOF-MS analysis shows the presence of known neuroprotective compounds like uridine and gabapentin in STME and PAME respectively. The two plants might be an important source of natural antioxidants and nutraceuticals with neuroprotective abilities. This could be investigated further to formulate herbal nutraceuticals for the treatment of neurodegenerative disease like Parkinson's disease.
Collapse
Affiliation(s)
- Aruna Chanu Hijam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Pooja Devi Nongthombam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Arunkumar Singh Koijam
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Yallapa Rajashekar
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India.
| |
Collapse
|
5
|
Hijam AC, Tongbram YC, Nongthombam PD, Meitei HN, Koijam AS, Rajashekar Y, Haobam R. Neuroprotective potential of traditionally used medicinal plants of Manipur against rotenone-induced neurotoxicity in SH-SY5Y neuroblastoma cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118197. [PMID: 38636579 DOI: 10.1016/j.jep.2024.118197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alternanthera sessilis (L.) R. Br. ex DC., Eryngium foetidum L., and Stephania japonica (Thunb.) Miers plants are traditionally used to treat various central nervous system disorders like paralysis, epilepsy, seizure, convulsion, chronic pain, headache, sleep disturbances, sprain, and mental disorders. However, their possible neuroprotective effects have not been evaluated experimentally so far. AIM OF THE STUDY The study aims to examine the neuroprotective potential of the three plants against cytotoxicity induced by rotenone in SH-SY5Y neuroblastoma cells and assess its plausible mechanisms of neuroprotection. MATERIALS AND METHODS The antioxidant properties of the plant extracts were determined chemically by DPPH and ABTS assay methods. The cytotoxicity of rotenone and the cytoprotective activities of the extracts were evaluated using MTT assays. Microtubule-associated protein 2 (MAP2) expression studies in cells were performed to assess neuronal survival after rotenone and extract treatments. Mitochondrial membrane potential and intracellular levels of reactive oxygen species were evaluated using Rhodamine 123 and DCF-DA dye, respectively. Catalase, glutathione peroxidase, and superoxide dismutase activities were also measured. Apoptotic nuclei were examined using DAPI staining. Liquid chromatography-quadrupole time-of-flight tandem mass spectrometry (LC-QTOF-MS) analysis of the plant extracts was also performed. RESULTS The methanol extracts of A. sessilis, S. japonica, and E. foetidum showed excellent free radical scavenging activities. MAP2 expression studies show that A. sessilis and S. japonica have higher neuroprotective effects against rotenone-induced neurotoxicity in SH-SY5Y cells than E. foetidum. Pre-treating cells with the plant extracts reverses the rotenone-induced increase in intracellular ROS. The plant extracts could also restore the reduced mitochondrial membrane potential induced by rotenone treatment and reinstate rotenone-induced increases in catalase, glutathione peroxidase, and superoxide dismutase activities. All the extracts inhibited rotenone-induced changes in nuclear morphology and DNA condensation, an early event of cellular apoptosis. LC-QTOF-MS analysis of the plant extracts shows the presence of neuroprotective compounds. CONCLUSIONS The plant extracts showed neuroprotective activities against rotenone-treated SH-SY5Y cells through antioxidant and anti-apoptotic mechanisms. These findings support the ethnopharmacological uses of these plants in treating neurological disorders. They probably are a good source of neuroprotective compounds that could be further explored to develop treatment strategies for neurodegenerative diseases like Parkinson's disease.
Collapse
Affiliation(s)
- Aruna Chanu Hijam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Pooja Devi Nongthombam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India
| | | | - Arunkumar Singh Koijam
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Yallapa Rajashekar
- Insect Bioresources Laboratory, Animal Bioresources Programme, Institute of Bioresources & Sustainable Development, Department of Biotechnology, Govt. of India, Takyelpat, Imphal, 795001, Manipur, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, Manipur, India.
| |
Collapse
|
6
|
Garg A, Bandyopadhyay S. Role of an interdependent Wnt, GSK3-β/β-catenin and HB-EGF/EGFR mechanism in arsenic-induced hippocampal neurotoxicity in adult mice. CHEMOSPHERE 2024; 352:141375. [PMID: 38325618 DOI: 10.1016/j.chemosphere.2024.141375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
We previously reported the neurotoxic effects of arsenic in the hippocampus. Here, we explored the involvement of Wnt pathway, which contributes to neuronal functions. Administering environmentally relevant arsenic concentrations to postnatal day-60 (PND60) mice demonstrated a dose-dependent increase in hippocampal Wnt3a and its components, Frizzled, phospho-LRP6, Dishevelled and Axin1 at PND90 and PND120. However, p-GSK3-β(Ser9) and β-catenin levels although elevated at PND90, decreased at PND120. Additionally, treatment with Wnt-inhibitor, rDkk1, reduced p-GSK3-β(Ser9) and β-catenin at PND90, but failed to affect their levels at PND120, indicating a time-dependent link with Wnt. To explore other underlying factors, we assessed epidermal growth factor receptor (EGFR) pathway, which interacts with GSK3-β and appears relevant to neuronal functions. We primarily found that arsenic reduced hippocampal phosphorylated-EGFR and its ligand, Heparin-binding EGF-like growth factor (HB-EGF), at both PND90 and PND120. Moreover, treatment with HB-EGF rescued p-GSK3-β(Ser9) and β-catenin levels at PND120, suggesting their HB-EGF/EGFR-dependent regulation at this time point. Additionally, rDkk1, LiCl (GSK3-β-activity inhibitor), or β-catenin protein treatments induced a time-dependent recovery in HB-EGF, indicating potential inter-dependent mechanism between hippocampal Wnt/β-catenin and HB-EGF/EGFR following arsenic exposure. Fluorescence immunolabeling then validated these findings in hippocampal neurons. Further exploration of hippocampal neuronal survival and apoptosis demonstrated that treatment with rDkk1, LiCl, β-catenin and HB-EGF improved Nissl staining and NeuN levels, and reduced cleaved-caspase-3 levels in arsenic-treated mice. Supportively, we detected improved Y-Maze and Passive Avoidance performances for learning-memory functions in these mice. Overall, our study provides novel insights into Wnt/β-catenin and HB-EGF/EGFR pathway interaction in arsenic-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Asmita Garg
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanghamitra Bandyopadhyay
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Sun HY, Gu AX, Huang BY, Zhang T, Li JP, Shan AS. Dietary Grape Seed Proanthocyanidin Alleviates the Liver Injury Induced by Long-Term High-Fat Diets in Sprague Dawley Rats. Front Vet Sci 2022; 9:959906. [PMID: 35990272 PMCID: PMC9382112 DOI: 10.3389/fvets.2022.959906] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022] Open
Abstract
In mammals, the liver is the most important organ that plays a vital function in lipid metabolism. Grape seed proanthocyanidin (GSPE) is a kind of natural polyphenolic compound primarily obtained from grape skin and seeds. Recent research found it had high bioavailability in defending against obesity, hyperlipidemia, inflammatory, oxidative stress, and targeting liver tissue. However, the mechanism of GSPE in regulating obesity induced by dietary high-fat (HF) was not fully understood, particularly the influences on liver functions. Therefore, this study aimed to investigate the effects of GSPE supplementation on the liver function and lipid metabolic parameters in rats fed HF diets long-term. A total of 40 healthy female Sprague Dawley rats were selected. After 8 weeks of obesity model feeding, the rats were randomly divided into four treatments: NC, standard diet; NC + GSPE, standard diet + 500 mg/kg body weight GSPE; HF, high-fat diet; HG + GSPE, high fat diet + 500 mg/kg body weight GSPE. Results indicated that long-term HF feeding caused severe liver problems including megalohepatia, steatosis, inflammation, and hepatocyte apoptosis. The supplementation of GSPE alleviated these symptoms. The results of the current experiment confirmed that GSPE addition up-regulated the expression of the Wnt3a/β-catenin signaling pathway, thereby restraining the liver cell endoplasmic reticulum stress and hepatocyte apoptosis. Furthermore, the microRNA-103 may play a role in this signal-regulated pathway. In summary, GSPE had a protective effect on the liver and the current experiment provided a reference for the application of GSPE in animal nutrition as a kind of natural feed additive.
Collapse
Affiliation(s)
| | | | | | | | - Jian Ping Li
- College of Animal Science and Technology, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - An Shan Shan
- College of Animal Science and Technology, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
8
|
Fathi M, Vakili K, Yaghoobpoor S, Qadirifard MS, Kosari M, Naghsh N, Asgari taei A, Klegeris A, Dehghani M, Bahrami A, Taheri H, Mohamadkhani A, Hajibeygi R, Rezaei Tavirani M, Sayehmiri F. Pre-clinical Studies Identifying Molecular Pathways of Neuroinflammation in Parkinson's Disease: A Systematic Review. Front Aging Neurosci 2022; 14:855776. [PMID: 35912090 PMCID: PMC9327618 DOI: 10.3389/fnagi.2022.855776] [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: 01/15/2022] [Accepted: 05/23/2022] [Indexed: 12/09/2022] Open
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by neuroinflammation, formation of Lewy bodies, and progressive loss of dopaminergic neurons in the substantia nigra of the brain. In this review, we summarize evidence obtained by animal studies demonstrating neuroinflammation as one of the central pathogenetic mechanisms of PD. We also focus on the protein factors that initiate the development of PD and other neurodegenerative diseases. Our targeted literature search identified 40 pre-clinical in vivo and in vitro studies written in English. Nuclear factor kappa B (NF-kB) pathway is demonstrated as a common mechanism engaged by neurotoxins such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA), as well as the bacterial lipopolysaccharide (LPS). The α-synuclein protein, which plays a prominent role in PD neuropathology, may also contribute to neuroinflammation by activating mast cells. Meanwhile, 6-OHDA models of PD identify microsomal prostaglandin E synthase-1 (mPGES-1) as one of the contributors to neuroinflammatory processes in this model. Immune responses are used by the central nervous system to fight and remove pathogens; however, hyperactivated and prolonged immune responses can lead to a harmful neuroinflammatory state, which is one of the key mechanisms in the pathogenesis of PD.
Collapse
Affiliation(s)
- Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Yaghoobpoor
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Qadirifard
- Department of Nursing and Midwifery, Islamic Azad University, Tehran, Iran
- Department of Nursing, Garmsar Branch, Islamic Azad University, Garmsar, Iran
| | - Mohammadreza Kosari
- The First Clinical College, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Navid Naghsh
- Department of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Afsaneh Asgari taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andis Klegeris
- Department of Biology, Faculty of Science, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| | - Mina Dehghani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ashkan Bahrami
- Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran
| | - Hamed Taheri
- Dental School, Kazan Federal University, Kazan, Russia
| | - Ashraf Mohamadkhani
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramtin Hajibeygi
- Department of Cardiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Mostafa Rezaei Tavirani
| | - Fatemeh Sayehmiri
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Fatemeh Sayehmiri
| |
Collapse
|
9
|
Chen T, Gao F, Luo D, Wang S, Zhao Y, Liu S, Huang J, Lin Y, Zhang Z, Huang H, Wan L. Cistanoside A promotes osteogenesis of primary osteoblasts by alleviating apoptosis and activating autophagy through involvement of the Wnt/β-catenin signal pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:64. [PMID: 35282110 PMCID: PMC8848445 DOI: 10.21037/atm-21-6742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
Background As a phenylethanoid glycoside extracted from Cistanche deserticola, cistanoside A has been shown to have antioxidative effects. In recent years, it has been found to play an important role in osteoporosis. Methods Primary osteoblasts were randomly divided into a cistanoside A (Cis A)-1 group (5 µM), a Cis A-2 group (10 µM), and a Cis A-3 group (20 µM) to screen the optimal dose. Then, cells were treated with Rapamycin (Rapa), 3-MA, Dickkopf-1 (DKK-1), 3MA + Cis A (10 µM), and DKK-1 + Cis A (10 µM). After a certain period of routine culture, Alkaline Phosphatase (ALP) and Alizarin Red S Staining were performed again and the cells were collected for subsequent experiments including immunofluorescence staining, western blotting, transmission electron microscopy, mitochondrial membrane measurement, and Annexin-V-Fluorescein isothiocyanate (Annexin-V-FITC). Results The optimal Cis A dose that preserved osteoblast viability and activated osteogenesis was 10 µM. It appeared that Cis A (10 µM) decreased apoptosis and augmented autophagy via increasing microtubule-associated protein light chain 3 (LC3)-I/II expressions as well as raising Wnt/β-catenin signal pathway activity. The addition of 3-MA further inhibited osteogenic differentiation and suppressed Wnt/β-catenin signal pathway activity to increase apoptosis while reducing autophagy levels. A combination of Cis A and DKK-1 resulted in higher levels of apoptosis but lower levels of autophagy. Conclusions Cis A appears to be a potent inducer of autophagy and inhibitor of apoptosis in primary osteoblasts by working through the Wnt/β-catenin signal pathway, thereby resulting in enhanced osteogenic differentiation.
Collapse
Affiliation(s)
- Tongying Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fenghe Gao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Luo
- Research Laboratory of Spine Degenerative Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shihao Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhua Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiachun Huang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanping Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhihai Zhang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongxing Huang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Wan
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
10
|
Redox Homeostasis and Prospects for Therapeutic Targeting in Neurodegenerative Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9971885. [PMID: 34394839 PMCID: PMC8355971 DOI: 10.1155/2021/9971885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/27/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022]
Abstract
Reactive species, such as those of oxygen, nitrogen, and sulfur, are considered part of normal cellular metabolism and play significant roles that can impact several signaling processes in ways that lead to either cellular sustenance, protection, or damage. Cellular redox processes involve a balance in the production of reactive species (RS) and their removal because redox imbalance may facilitate oxidative damage. Physiologically, redox homeostasis is essential for the maintenance of many cellular processes. RS may serve as signaling molecules or cause oxidative cellular damage depending on the delicate equilibrium between RS production and their efficient removal through the use of enzymatic or nonenzymatic cellular mechanisms. Moreover, accumulating evidence suggests that redox imbalance plays a significant role in the progression of several neurodegenerative diseases. For example, studies have shown that redox imbalance in the brain mediates neurodegeneration and alters normal cytoprotective responses to stress. Therefore, this review describes redox homeostasis in neurodegenerative diseases with a focus on Alzheimer's and Parkinson's disease. A clearer understanding of the redox-regulated processes in neurodegenerative disorders may afford opportunities for newer therapeutic strategies.
Collapse
|
11
|
Liu L, Cao Q, Gao W, Li B, Xia Z, Zhao B. Melatonin protects against focal cerebral ischemia-reperfusion injury in diabetic mice by ameliorating mitochondrial impairments: involvement of the Akt-SIRT3-SOD2 signaling pathway. Aging (Albany NY) 2021; 13:16105-16123. [PMID: 34118791 PMCID: PMC8266371 DOI: 10.18632/aging.203137] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022]
Abstract
Diabetic patients are more vulnerable to cerebral ischemia-reperfusion (CIR) injury and have a worse prognosis and higher mortality after ischemic stroke than non-diabetic counterparts. Melatonin can exert neuroprotective effects against CIR injury in nondiabetic animal models. However, its effects on diabetic CIR injury and the underlying mechanisms remain unclarified. Herein, we found that melatonin administration improved neurological deficit, cerebral infarct volume, brain edema, and cell viability, reduced mitochondrial swelling, reactive oxygen species generation, and cytoplasmic cytochrome C release, and increased mitochondrial antioxidant enzymes activities, adenosine triphosphate production, and mitochondrial membrane potential in both streptozotocin-induced diabetic mice and high glucose-treated HT22 cells. Importantly, melatonin also activated protein kinase B (Akt) and sirtuin 3 (SIRT3)/superoxide dismutase 2 (SOD2) signaling and upregulated mitochondrial biogenesis-related transcription factors. However, these effects were largely attenuated by LY294002 (a specific Akt signaling blocker) administration. Additionally, 3-TYP (a selective SIRT3 inhibitor) and SIRT3 siRNA inhibited the above protective effects of melatonin as well as the upregulation of SIRT3 and the decrease of SOD2 acetylation but did not affect the p-Akt/Akt ratio. Overall, we demonstrate that melatonin can alleviate CIR injury in diabetic mice by activating Akt-SIRT3-SOD2 signaling and subsequently improving mitochondrial damage.
Collapse
Affiliation(s)
- Lian Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Quan Cao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Wenwei Gao
- Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Bingyu Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
12
|
Guo LY, Shi FL, Li M, Sun JH, Li CG, Liu ZX. Astragalus protects PC12 cells from 6-hydroxydopamine-induced neuronal damage: A serum pharmacological study. CHINESE J PHYSIOL 2021; 64:24-31. [PMID: 33642341 DOI: 10.4103/cjp.cjp_50_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Accumulating evidence has already indicated that traditional Chinese medicine (TCM) possesses tremendous potential for treating neurodegenerative diseases. Astragalus, also named Huangqi, is a famous traditional medical herb that can be applied to treat cerebral ischemia and prevent neuronal degeneration. Nevertheless, the underlying mechanisms remain largely unexplored. In the present study, Astragalus-containing serum (ASMES) was prepared and added into the culture medium of PC12 cells to explore its neuroprotective effect on 6-hydroxydopamine (6-OHDA)-caused neuronal toxicity. Our data showed that ASMES significantly ameliorated the cellular viability of cultured PC12 cells against the neurotoxicity induced by 6-OHDA (P < 0.05). Moreover, ASMES significantly decreased the cell apoptosis triggered by 6-OHDA (P < 0.01). Furthermore, 2',7'-dichlorofluorescin diacetate assay was performed to detect the changes in oxidative stress, and we showed that 6-OHDA elevated the production of reactive oxygen species (ROS), whereas ASMES significantly reversed these changes (P < 0.01). Besides, mitochondrial membrane potential (MMP) assay showed that ASMES could restore 6-OHDA-damaged MMP in cultured PC12 cells (P < 0.05). In conclusion, Astragalus could protect PC12 cells from 6-OHDA-caused neuronal toxicity, and possibly, the ROS-mediated apoptotic pathway participated in this process. Collectively, our findings provided valuable insights into the potential in treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Li-Ying Guo
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Shandong, China
| | - Feng-Lei Shi
- Department of Orthopaedics, Cheelee College of Medicine, Qilu Hospital (Qingdao), Shandong University, Shandong, China
| | - Meng Li
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Shandong, China
| | - Jin-Hao Sun
- Department of Anatomy and Neurobiology, Shandong University School of Basic Medicine, Shandong, China
| | - Chuan-Gang Li
- Department of Anesthesiology, Second Hospital of Shandong University, Shandong, China
| | - Zeng-Xun Liu
- Department of Psychiatry, Shandong Mental Health Center, Shandong, China
| |
Collapse
|
13
|
Rotherham M, Nahar T, Goodman T, Telling N, Gates M, El Haj A. Magnetic Mechanoactivation of Wnt Signaling Augments Dopaminergic Differentiation of Neuronal Cells. ACTA ACUST UNITED AC 2020; 3:e1900091. [PMID: 32648650 DOI: 10.1002/adbi.201900091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/07/2019] [Indexed: 01/09/2023]
Abstract
Wnt signaling is a key developmental pathway that regulates dopaminergic progenitor cell proliferation and differentiation during neuronal development. This makes Wnt signaling an important therapeutic target for neurodegenerative conditions such as Parkinson's disease. Wnt signaling can be modulated using peptides such as UM206, which bind to the Wnt receptor Frizzled. Previous work has demonstrated remote activation of the Wnt pathway through Frizzled using peptide-functionalized magnetic nanoparticles (MNPs) with magnetic field stimulation. Using this technology, Wnt signaling is remotely activated in the neuronal cell line SH-SY5Y, and the phenotypic response to stimulation is assessed. Results indicate β-catenin translocalization and activation of TCF/LEF responsive transcription in response to MNP and magnetic fields, which result in dopaminergic marker expression when synergistically combined with differentiation factors retinoic acid and the phorbol ester phorbol 12-myristate 13-acetate. This approach is translated into ex vivo postnatal rat brain slices modeling the developing nigrostriatal pathway. Dopaminergic marker expression is maintained in MNP-labeled SH-SY5Y cells after injection and magnetic stimulation. These results demonstrate the translational value of remote control of signal transduction for controlling neuronal precursor cell behavior and highlight the potential applications for controlled cell differentiation as part of cell therapies for neurodegenerative disease.
Collapse
Affiliation(s)
- Michael Rotherham
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Tasmin Nahar
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Timothy Goodman
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Neil Telling
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Monte Gates
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK
| | - Alicia El Haj
- Institute for Science and Technology in Medicine, Keele University, Guy Hilton Research Centre, Thornburrow Drive, Stoke-on-Trent, ST4 7QB, UK.,Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Edgbaston, Birmingham, B15 2TH, UK
| |
Collapse
|
14
|
Nitroxide Radical-Containing Redox Nanoparticles Protect Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9260748. [PMID: 32377313 PMCID: PMC7196160 DOI: 10.1155/2020/9260748] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/06/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) patients can benefit from antioxidant supplementation, and new efficient antioxidants are needed. The aim of this study was to evaluate the protective effect of selected nitroxide-containing redox nanoparticles (NRNPs) in a cellular model of PD. Antioxidant properties of NRNPs were studied in cell-free systems by protection of dihydrorhodamine 123 against oxidation by 3-morpholino-sydnonimine and protection of fluorescein against bleaching by 2,2-azobis(2-amidinopropane) hydrochloride and sodium hypochlorite. Model blood-brain barrier penetration was studied using hCMEC/D3 cells. Human neuroblastoma SH-SY5Y cells, exposed to 6-hydroxydopamine (6-OHDA), were used as an in vitro model of PD. Cells were preexposed to NRNPs or free nitroxides (TEMPO or 4-amino-TEMPO) for 2 h and treated with 6-OHDA for 1 h and 24 h. The reactive oxygen species (ROS) level was estimated with dihydroethidine 123 and Fluorimetric Mitochondrial Superoxide Activity Assay Kit. Glutathione level (GSH) was measured with ortho-phtalaldehyde, ATP by luminometry, changes in mitochondrial membrane potential with JC-1, and mitochondrial mass with 10-Nonyl-Acridine Orange. NRNP1, TEMPO, and 4-amino-TEMPO (25-150 μM) protected SH-SY5Y cells from 6-OHDA-induced viability loss; the protection was much higher for NRNP1 than for free nitroxides. NRNP1 were better antioxidants in vitro and permeated better the model BBB than free nitroxides. Exposure to 6-OHDA decreased the GSH level after 1 h and increased it considerably after 24 h (apparently a compensatory overresponse); NRNPs and free nitroxides prevented this increase. NRNP1 and free nitroxides prevented the decrease in ATP level after 1 h and increased it after 24 h. 6-OHDA increased the intracellular ROS level and mitochondrial superoxide level. Studied antioxidants mostly decreased ROS and superoxide levels. 6-OHDA decreased the mitochondrial potential and mitochondrial mass; both effects were prevented by NRNP1 and nitroxides. These results suggest that the mitochondria are the main site of 6-OHDA-induced cellular damage and demonstrate a protective effect of NRNP1 in a cellular model of PD.
Collapse
|
15
|
Xia SR, Wen XY, Fan XL, Chen XR, Wei ZW, Li QH, Sun L. Wnt2 overexpression protects against PINK1 mutant‑induced mitochondrial dysfunction and oxidative stress. Mol Med Rep 2020; 21:2633-2641. [PMID: 32323790 DOI: 10.3892/mmr.2020.11066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 03/12/2020] [Indexed: 11/06/2022] Open
Abstract
The PTEN induced putative kinase 1 (PINK1) mutation is the second most common cause of autosomal recessive adolescent Parkinson's disease (PD). Furthermore, mitochondrial disorders and oxidative stress are important mechanisms in the pathogenesis of PD. Numerous members of the Wnt family have been found to be associated with neurodegenerative diseases. Therefore, the present study investigated the role of the Wnt2 gene in PINK1B9 transgenic flies, which is a PD model, and its underlying mechanism. It was identified that overexpression of Wnt2 reduced the abnormality rate of PD transgenic Drosophila and improved their flight ability, while other intervention groups had no significant effect. Furthermore, an increase in ATP concentration normalized mitochondrial morphology, and increased the mRNA expression levels of NADH‑ubiquinone oxidoreductase chain 1 (ND1), ND42, ND75, succinate dehydrogenase complex subunits B, Cytochrome b and Cyclooxygenase 1, which are associated with Wnt2 overexpression. Moreover, overexpression of Wnt2 in PD transgenic Drosophila resulted in the downregulation of reactive oxygen species and malondialdehyde production, and increased manganese superoxide dismutase (MnSOD), while glutathione was not significantly affected. It was found that overexpression of Wnt2 did not alter the protein expression of β‑catenin in PINK1B9 transgenic Drosophila, but did increase the expression levels of PPARG coactivator 1α (PGC‑1α) and forkhead box sub‑group O (FOXO). Collectively, the present results indicated that the Wnt2 gene may have a protective effect on PD PINK1B9 transgenic Drosophila. Thus, it was speculated that the reduction of oxidative stress and the restoration of mitochondrial function via Wnt2 overexpression may be related to the PGC‑1α/FOXO/MnSOD signaling pathway in PINK1 mutant transgenic Drosophila.
Collapse
Affiliation(s)
- Sui-Rui Xia
- Department of Hospital Infection‑Control, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi 541002, P.R. China
| | - Xue-Yi Wen
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Xiao-Li Fan
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Xiao-Rong Chen
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Zai-Wa Wei
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Qing-Hua Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Li Sun
- Faculty of Basic Medical Sciences, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| |
Collapse
|
16
|
Wnt1 Promotes EAAT2 Expression and Mediates the Protective Effects of Astrocytes on Dopaminergic Cells in Parkinson's Disease. Neural Plast 2019; 2019:1247276. [PMID: 31582965 PMCID: PMC6754970 DOI: 10.1155/2019/1247276] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/01/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022] Open
Abstract
Background Wnt/β-catenin signaling has been reported to exert cytoprotective effects in a cellular model of Parkinson's disease (PD). Glutamate excitotoxicity has been suggested to contribute to the pathogenesis of PD, and excitatory amino acid transporters (EAATs) play a predominant role in clearing excessive glutamate. EAAT2 is mainly expressed in astrocytes, which are an important source of Wnt signaling in the brain. Methods Wnt1-overexpressing U251 astrocytes were indirectly cocultured with dopaminergic SH-SY5Y cells treated with 6-hydroxydopamine (6-OHDA). Cell toxicity was determined by cell viability and flow cytometric detection. Glutamate level in the culture medium was determined by enzyme-linked immunosorbent assay (ELISA). Western blot analysis was used to detect the expression of Wnt1, β-catenin, and EAAT2. Immunofluorescence was used to display the expression and translocation of NF-κB p65. Results 6-OHDA treatment significantly decreased cell viability in both U251 cells and SH-SY5Y cells, inhibited the expression of Wnt1, β-catenin, and EAAT2 in U251 cells, and increased the glutamate level in the culture medium. Coculture with Wnt1-overexpressing U251 cells attenuated 6-OHDA-induced apoptosis in SH-SY5Y cells. Overexpression of Wnt1 decreased the glutamate level in the culture media, upregulated β-catenin, EAAT2, and NF-κB levels, and promoted the translocation of NF-κB from the cytoplasm to the nucleus in U251 cells. Conclusion Wnt1 promoted EAAT2 expression and mediated the cytoprotective effects of astrocytes on dopaminergic cells. NF-κB might be involved in the regulation of EAAT2 by Wnt1.
Collapse
|
17
|
Conditional Haploinsufficiency of β-Catenin Aggravates Neuronal Damage in a Paraquat-Based Mouse Model of Parkinson Disease. Mol Neurobiol 2018; 56:5157-5166. [PMID: 30519817 DOI: 10.1007/s12035-018-1431-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022]
Abstract
The canonical Wnt pathway is critical for both the development and adulthood survival and homeostatic maintenance of the midbrain dopaminergic (DA) neurons. Expanding evidence has demonstrated that genetic factors associated with familial Parkinson disease (PD) deregulate this important pathway, suggesting that a disturbed canonical Wnt pathway is likely involved in PD pathogenesis; yet, the specific role of this pathway in sporadic PD remains unclear. In this study, we aimed to determine the effects of specific inhibition of the canonical pathway by hemizygous knockout of β-catenin, the obligatory component of the canonical Wnt pathway, on paraquat (PQ)-induced DA neuronal degeneration in the substantia nigra in vivo. We found that while hemizygous conditional knockout of β-catenin in DA neurons did not cause any significant TH+ neuronal loss in the substantia nigra at basal level, it triggered elevated oxidative stress at basal level and further enhanced PQ-induced oxidative damage and loss of TH+ neurons in the substantia nigra and axonal termini in the striatum that manifested as exacerbated motor deficits. These data support the notion that reduced Wnt/β-catenin signaling in sporadic PD likely contributes to DA neuronal loss through an enhanced oxidative stress-response pathway.
Collapse
|
18
|
Zhao L, Yan M, Wang X, Xiong G, Wu C, Wang Z, Zhou Z, Chang X. Modification of Wnt signaling pathway on paraquat-induced inhibition of neural progenitor cell proliferation. Food Chem Toxicol 2018; 121:311-325. [PMID: 30171970 DOI: 10.1016/j.fct.2018.08.064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/16/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023]
Abstract
Paraquat (PQ) is an agricultural chemical used worldwide. As a potential neurotoxicant, PQ adversely affects neurogenesis and inhibits proliferation of neural progenitor cells (NPCs). However, the molecular mechanistic insights of PQ exposure on NPCs remains to be determined. Herein, we determine the extent to which Wnt/β-catenin signaling involved in the inhibition effect of PQ on mouse NPCs from subventricular zone (SVZ). NPCs were treated with different concentrations of PQ (40, 80, and 120 μM). PQ exposure provoked oxidative stress and apoptosis and PQ inhibited cell viability and proliferation in a concentration-dependent manner. Significantly, PQ exposure altered the expression/protein levels of the Wnt pathway genes in NPCs. In addition, PQ reduced cellular β-catenin, p-GSK-3β, and cyclin-D1 and increased the radio of Bax/Bcl2. Further, Wnt pathway activation by treatment with LiCl and Wnt1 attenuated PQ-induced inhibition of mNPCs proliferation. Antioxidant (NAC) treatment alleviated the inhibition of PQ-induced Wnt signaling pathway. Overall, our results suggest significant inhibitory effects of PQ on NPCs proliferation via the Wnt/β-catenin signaling pathway. Interestingly, our results implied that activation of Wnt/β-catenin signaling pathway attenuated PQ-induced autophagic cell death. Our results therefore bring our understanding of the molecular mechanisms of PQ-induced neurotoxicity.
Collapse
Affiliation(s)
- Lina Zhao
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Mengling Yan
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Xinjin Wang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Guiya Xiong
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Chunhua Wu
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Zhibin Wang
- Department of Environmental Health & Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore, 21205, USA
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Xiuli Chang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
19
|
Qiu F, Shin Y, Chen D, Cheng R, Chen Q, Zhou K, Larrick JW, Mendelson AR, Ma JX. Anti-angiogenic effect of a humanized antibody blocking the Wnt/β-catenin signaling pathway. Microvasc Res 2018; 119:29-37. [PMID: 29630973 DOI: 10.1016/j.mvr.2018.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/14/2018] [Accepted: 03/24/2018] [Indexed: 10/17/2022]
Abstract
PURPOSE Our previous study demonstrated that Mab2F1, a murine monoclonal antibody blocking the Wnt/β-catenin signaling pathway, has beneficial effects on experimental diabetic retinopathy and choroidal neovascularization (NV). The aforementioned antibody has been humanized. This study evaluated effects of the humanized antibody, H1L1, on NV. METHODS H1L1 was evaluated in the alkali burn-induced corneal NV rat model. Rats with corneal NV were injected subconjunctivally with Mab2F1 or H1L1 using non-specific mouse or human IgG as controls. Corneal NV and opacity were evaluated using corneal NV area and inflammatory index. Expression of angiogenic and inflammatory factors and components of the Wnt/β-catenin pathway in both the corneas of the animal model and human corneal epithelial (HCE) cells exposed to Wnt3a conditioned medium (WCM) were determined by Western blotting and a luciferase-based promoter assay. Cytotoxicities of these antibodies were evaluated by MTT assay. RESULTS H1L1 reduced the area of corneal NV and opacity, similar to Mab2F1. Both Mab2F1 and H1L1 down-regulated the overexpression of angiogenic and inflammatory factors including VEGF, TNF-α and ICAM-1, and blocked the aberrant activation of the Wnt/β-catenin pathway as shown by down-regulation of phosphorylated LRP6, total LRP6 and non-phosphorylated β-catenin in the cornea of the NV model and cultured HCE cells exposed to WCM. Both antibodies also inhibited the transcriptional activity of β-catenin induced by WCM in HCE cells. No toxic effects of the antibodies were observed in cultured HCE cells. CONCLUSIONS H1L1 exhibits anti-angiogenic activities through blocking the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Fangfang Qiu
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Younghwa Shin
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | - Rui Cheng
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Qian Chen
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kelu Zhou
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | | - Jian-Xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
20
|
Shi ZY, Deng JX, Fu S, Wang L, Wang Q, Liu B, Li YQ, Deng JB. Protective effect of autophagy in neural ischemia and hypoxia: Negative regulation of the Wnt/β-catenin pathway. Int J Mol Med 2017; 40:1699-1708. [PMID: 29039446 PMCID: PMC5716434 DOI: 10.3892/ijmm.2017.3158] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 09/26/2017] [Indexed: 01/08/2023] Open
Abstract
Autophagy is a highly conserved process of self-digestion to promote cell survival in response to nutrient starvation and other metabolic stresses. However, whether ischemic-hypoxic (IH) injury-induced autophagy acts as a neuroprotective mechanism or leads to neuroinjury is a subject of debate. It is known that autophagy is regulated by signaling pathways, including the mammalian target of rapamycin pathway. However, in neural IH injury, whether other signaling pathways are involved in the regulation of autophagy remains to be fully elucidated. In the present study, using the autophagy agonist (rampycin), autophagy antagonist [3-methyl adenine (3-MA)] and lysosome antagonist (MHY1485), autophagy was intervened with at oxygen-glucose deprivation (OGD) 6 h, in order to elucidate the regulatory mechanisms of autophagy. Using immunocytochemistry and western blot analysis, the expression levels of stress-related proteins, such as hypoxia-inducible factor-1α (HIF-1α) (a key regulator in hypoxia) and cyclooxygenase 2 (COX2; inflammatory indicator), were analyzed. In addition, the upstream proteins (Wnt1 and Wnt3a), downstream proteins (Dvl2, β-catenin) and target proteins (C-myc and cyclin D) in the Wnt/β-catenin signaling pathway were examined by immunocytochemistry and western blot analysis. The present study revealed that autophagy was activated with the upregulation of autophagic flux in IH injury; it was demonstrated that autophagy had a protective role in IH injury. The Wnt/β-catenin pathway was involved in IH injury regulation, and the upstream proteins in the Wnt/β-catenin signaling pathway were upregulated, whereas downstream proteins were downregulated by the activity of autophagy accordingly.
Collapse
Affiliation(s)
- Zhen-Yu Shi
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Jie-Xin Deng
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Su Fu
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Lai Wang
- Institute of Neurobiology, College of Life Science, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Qiang Wang
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Bin Liu
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yong-Qiang Li
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Jin-Bo Deng
- Institute of Neurobiology, Nursing College, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
21
|
Kang X, Qiu J, Li Q, Bell KA, Du Y, Jung DW, Lee JY, Hao J, Jiang J. Cyclooxygenase-2 contributes to oxidopamine-mediated neuronal inflammation and injury via the prostaglandin E2 receptor EP2 subtype. Sci Rep 2017; 7:9459. [PMID: 28842681 PMCID: PMC5573328 DOI: 10.1038/s41598-017-09528-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/17/2017] [Indexed: 01/10/2023] Open
Abstract
Cyclooxygenase-2 (COX-2) triggers pro-inflammatory processes that can aggravate neuronal degeneration and functional impairments in many neurological conditions, mainly via producing prostaglandin E2 (PGE2) that activates four membrane receptors, EP1-EP4. However, which EP receptor is the culprit of COX-2/PGE2-mediated neuronal inflammation and degeneration remains largely unclear and presumably depends on the insult types and responding components. Herein, we demonstrated that COX-2 was induced and showed nuclear translocation in two neuronal cell lines – mouse Neuro-2a and human SH-SY5Y – after treatment with neurotoxin 6-hydroxydopamine (6-OHDA), leading to the biosynthesis of PGE2 and upregulation of pro-inflammatory cytokine interleukin-1β. Inhibiting COX-2 or microsomal prostaglandin E synthase-1 suppressed the 6-OHDA-triggered PGE2 production in these cells. Treatment with PGE2 or EP2 selective agonist butaprost, but not EP4 agonist CAY10598, increased cAMP response in both cell lines. PGE2-initiated cAMP production in these cells was blocked by our recently developed novel selective EP2 antagonists – TG4-155 and TG6-10-1, but not by EP4 selective antagonist GW627368X. The 6-OHDA-promoted cytotoxicity was largely blocked by TG4-155, TG6-10-1 or COX-2 selective inhibitor celecoxib, but not by GW627368X. Our results suggest that PGE2 receptor EP2 is a key mediator of COX-2 activity-initiated cAMP signaling in Neuro-2a and SH-SY5Y cells following 6-OHDA treatment, and contributes to oxidopamine-mediated neurotoxicity.
Collapse
Affiliation(s)
- Xu Kang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA
| | - Jiange Qiu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA.,Department of Cell Biology and Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qianqian Li
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA
| | - Katherine A Bell
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA
| | - Yifeng Du
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA
| | - Da Woon Jung
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jiukuan Hao
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA
| | - Jianxiong Jiang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, Ohio, 45267-0514, USA.
| |
Collapse
|
22
|
Angelov B, Angelova A. Nanoscale clustering of the neurotrophin receptor TrkB revealed by super-resolution STED microscopy. NANOSCALE 2017; 9:9797-9804. [PMID: 28682396 DOI: 10.1039/c7nr03454g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The nanoscale organization of the tropomyosin-related kinase receptor type B (TrkB), a promising therapeutic target for severe neurodegenerative and psychiatric disorders, is examined by stimulated emission depletion (STED) microscopy using the deconvoluted gated STED option. The performed immunofluorescence nanoscopic subdiffraction imaging of the membrane receptor localization reveals that clusters of oligomeric TrkB states and randomly organized nanodomains are formed in the membranes of differentiated human neuroblastoma SH-SY5Y cells, which are studied as an in vitro model of neurodegeneration. Despite that the monomeric (isolated) states of the receptor cannot be distinguished from its dimeric forms in such images, TrkB receptor dimers (or couple of individual monomers) are visualized at super-resolution as single pixels in the magnified Huygens-deconvoluted gated STED images. The clusters of higher-order TrkB oligomers are of dynamic nature rather than of a fixed stoichiometry. The propensity for membrane protein clustering as well as the dissociation of the TrkB receptors nanodomains can be modulated by neurotherapeutic formulations containing ω-3 polyunsaturated docosahexaenoic acid (DHA). Nanomolar concentrations of DHA change the receptor topology and lead to disruption of the cluster phases. This result is of therapeutic importance for TrkB receptor availability upon ligand binding as DHA favours the mobility and the dynamic distribution of the protein populations in the cell membranes.
Collapse
Affiliation(s)
- Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, Na Slovance 2, CZ-18221 Prague, Czech Republic
| | | |
Collapse
|
23
|
Hichor M, Sampathkumar NK, Montanaro J, Borderie D, Petit PX, Gorgievski V, Tzavara ET, Eid AA, Charbonnier F, Grenier J, Massaad C. Paraquat Induces Peripheral Myelin Disruption and Locomotor Defects: Crosstalk with LXR and Wnt Pathways. Antioxid Redox Signal 2017; 27:168-183. [PMID: 27788593 DOI: 10.1089/ars.2016.6711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS Paraquat (PQT), a redox-active herbicide, is a free radical-producing molecule, causing damage particularly to the nervous system; thus, it is employed as an animal model for Parkinson's disease. However, its impact on peripheral nerve demyelination is still unknown. Our aim is to decipher the influence of PQT-induced reactive oxygen species (ROS) production on peripheral myelin. RESULTS We report that PQT provokes severe locomotor and sensory defects in mice. PQT elicited an oxidative stress in the nerve, resulting in an increase of lipid peroxidation and protein carbonylation, despite the induction of nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent antioxidant defenses. We observed a dramatic disorganization of myelin sheaths in the sciatic nerves, dysregulation of myelin gene expression, and aggregation of myelin proteins, a hallmark of demyelination. PQT altered myelin gene expression via liver X receptor (LXR) signaling, a negative regulator of peripheral myelin gene expression through its dialog with the Wnt/β-catenin pathway. PQT prevented β-catenin binding on myelin gene promoters, resulting in the inhibition of Wnt/β-catenin-dependent myelin gene expression. Wnt pathway activation by LiCl dampened the deleterious effects of PQT. LiCl blocked PQT-induced oxidative stress and reduced Schwann cell death. LiCl+PQT-treated mice had normal sensorimotor behaviors and a usual nerve structure. INNOVATION We reveal that PQT damages the sciatic nerve by generating an oxidative stress, dysregulating LXR and Wnt/β-catenin pathways. The activation of Wnt signaling by LiCl reduced the deleterious effects of PQT on the nerve. CONCLUSION We demonstrate that PQT instigates peripheral nerve demyelinating neuropathies by enhancing ROS production and deregulating LXR and Wnt pathways. Stimulating Wnt pathway could be a therapeutic strategy for neuropathy treatment. Antioxid. Redox Signal. 27, 168-183.
Collapse
Affiliation(s)
- Mehdi Hichor
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Nirmal Kumar Sampathkumar
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Julia Montanaro
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Didier Borderie
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Patrice X Petit
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Victor Gorgievski
- 2 INSERM UMRS-S 1130, CNRS UMR824, Pierre and Marie Curie University , Paris, France
| | - Eleni T Tzavara
- 2 INSERM UMRS-S 1130, CNRS UMR824, Pierre and Marie Curie University , Paris, France
| | - Assaad A Eid
- 3 Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Frédéric Charbonnier
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Julien Grenier
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| | - Charbel Massaad
- 1 INSERM UMR-S 1124, Sorbonne Paris Cité, Faculté des Sciences Fondamentales et Biomédicales, Paris Descartes University , Paris, France
| |
Collapse
|
24
|
Lopes FM, Bristot IJ, da Motta LL, Parsons RB, Klamt F. Mimicking Parkinson's Disease in a Dish: Merits and Pitfalls of the Most Commonly used Dopaminergic In Vitro Models. Neuromolecular Med 2017; 19:241-255. [PMID: 28721669 DOI: 10.1007/s12017-017-8454-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/12/2017] [Indexed: 12/27/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and has both unknown etiology and non-curative therapeutic options. Patients begin to present the classic motor symptoms of PD-tremor at rest, bradykinesia and rigidity-once 50-70% of the dopaminergic neurons of the nigrostriatal pathway have degenerated. As a consequence of this, it is difficult to investigate the early-stage events of disease pathogenesis. In vitro experimental models are used extensively in PD research because they present a controlled environment that enables the direct investigation of the early molecular mechanisms that are potentially involved with dopaminergic degeneration, as well as for the screening of potential therapeutic drugs. However, the establishment of PD in vitro models is a controversial issue for neuroscience research not only because it is challenging to mimic, in isolated cell systems, the physiological neuronal environment, but also the pathophysiological conditions experienced by human dopaminergic cells in vivo during the progression of the disease. Since no previous work has attempted to systematically review the literature regarding the establishment of an optimal in vitro model, and/or the features presented by available models used in the PD field, this review aims to summarize the merits and limitations of the most widely used dopaminergic in vitro models in PD research, which may help the PD researcher to choose the most appropriate model for studies directed at the elucidation of the early-stage molecular events underlying PD onset and progression.
Collapse
Affiliation(s)
- Fernanda Martins Lopes
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil. .,Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Ivi Juliana Bristot
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Leonardo Lisbôa da Motta
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil
| | - Richard B Parsons
- Institute of Pharmaceutical Science, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Fabio Klamt
- Laboratório de Bioquímica Celular, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
25
|
Acetyl-L-Carnitine via Upegulating Dopamine D1 Receptor and Attenuating Microglial Activation Prevents Neuronal Loss and Improves Memory Functions in Parkinsonian Rats. Mol Neurobiol 2016; 55:583-602. [PMID: 27975173 DOI: 10.1007/s12035-016-0293-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023]
Abstract
Parkinson's disease is accompanied by nonmotor symptoms including cognitive impairment, which precede the onset of motor symptoms in patients and are regulated by dopamine (DA) receptors and the mesocorticolimbic pathway. The relative contribution of DA receptors and astrocytic glutamate transporter (GLT-1) in cognitive functions is largely unexplored. Similarly, whether microglia-derived increased immune response affects cognitive functions and neuronal survival is not yet understood. We have investigated the effect of acetyl-L-carnitine (ALCAR) on cognitive functions and its possible underlying mechanism of action in 6-hydroxydopamine (6-OHDA)-induced hemiparkinsonian rats. ALCAR treatment in 6-OHDA-lesioned rats improved memory functions as confirmed by decreased latency time and path length in the Morris water maze test. ALCAR further enhanced D1 receptor levels without altering D2 receptor levels in the hippocampus and prefrontal cortex (PFC) regions, suggesting that the D1 receptor is preferentially involved in the regulation of cognitive functions. ALCAR attenuated microglial activation and release of inflammatory mediators through balancing proinflammatory and anti-inflammatory cytokines, which subsequently enhanced the survival of mature neurons in the CA1, CA3, and PFC regions and improved cognitive functions in hemiparkinsonian rats. ALCAR treatment also improved glutathione (GSH) content, while decreasing oxidative stress indices, inducible nitrogen oxide synthase (iNOS) levels, and astrogliosis resulting in the upregulation of GLT-1 levels. Additionally, ALCAR prevented the loss of dopaminergic (DAergic) neurons in ventral tagmental area (VTA)/substantia nigra pars compacta (SNpc) regions of 6-OHDA-lesioned rats, thus maintaining the integrity of the nigrostriatal pathway. Together, these results demonstrate that ALCAR treatment in hemiparkinsonian rats ameliorates neurodegeneration and cognitive deficits, hence suggesting its therapeutic potential in neurodegenerative diseases.
Collapse
|
26
|
Jiang T, Sun Q, Chen S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson's disease and Alzheimer's disease. Prog Neurobiol 2016; 147:1-19. [PMID: 27769868 DOI: 10.1016/j.pneurobio.2016.07.005] [Citation(s) in RCA: 448] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/19/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022]
Abstract
Oxidative stress reflects an imbalance between the overproduction and incorporation of free radicals and the dynamic ability of a biosystem to detoxify reactive intermediates. Free radicals produced by oxidative stress are one of the common features in several experimental models of diseases. Free radicals affect both the structure and function of neural cells, and contribute to a wide range of neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Although the precise mechanisms that result in the degeneration of neurons and the relevant pathological changes remain unclear, the crucial role of oxidative stress in the pathogenesis of neurodegenerative diseases is associated with several proteins (such as α-synuclein, DJ-1, Amyloid β and tau protein) and some signaling pathways (such as extracellular regulated protein kinases, phosphoinositide 3-kinase/Protein Kinase B pathway and extracellular signal-regulated kinases 1/2) that are tightly associated with the neural damage. In this review, we present evidence, gathered over the last decade, concerning a variety of pathogenic proteins, their important signaling pathways and pathogenic mechanisms associated with oxidative stress in Parkinson's disease and Alzheimer's disease. Proper control and regulation of these proteins' functions and the related signaling pathways may be a promising therapeutic approach to the patients. We also emphasizes antioxidative options, including some new neuroprotective agents that eliminate excess reactive oxygen species efficiently and have a certain therapeutic effect; however, controversy surrounds some of them in terms of the dose and length of therapy. These agents require further investigation by clinical application in patients suffering Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Tianfang Jiang
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Sun
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Science & Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
27
|
Li Z, Wang H, Wang Q, Sun J. Buyang Huanwu Decoction Vigorously Rescues PC12 Cells Against 6-OHDA-Induced Neurotoxicity via Akt/GSK3β Pathway Based on Serum Pharmacology Methodology. Rejuvenation Res 2016; 19:467-477. [PMID: 26935342 DOI: 10.1089/rej.2015.1798] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Buyang Huanwu decoction (BYHWD), as a popular traditional Chinese medicine formula, was widely used for treating ischemic diseases. However, in the area of neurodegenerative diseases, the researches focused on BYHWD are rare but promising, and molecular mechanisms underlying are largely elusive. 6-Hydroxydopamine (6-OHDA), a dopaminergic-specific neurotoxin, is extensively used to establish neurotoxic model in vivo and in vitro. In our present study, we prepared drug-containing serum of BYHWD (Buyang Huanwu drug-containing serum [BYHWS]) based on serum pharmacology methodology. Neurotoxic model in vitro was established in PC12 cells, and innovative experimental grouping method was adopted to investigate neuroprotective effects of BYHWS on neurotoxicity induced by 6-OHDA exposure. Remarkably, BYHWS vigorously rescued PC12 cells from 6-OHDA-induced neurotoxicity even to surpass 100% in cell viability. Moreover, Hoechst/propidium iodide (PI) staining revealed that cell apoptotic rate was reduced significantly after incubation of BYHWS. Besides, BYHWS effectively restored the disruption of mitochondrial membrane potential and attenuated the elevation of intracellular reactive oxygen species level caused by 6-OHDA insult. Furthermore, BYHWS remarkably reversed the dephosphorylation of Akt (protein kinase B) and glycogen synthase kinase-3β (GSK3β) evoked by 6-OHDA. The above protective effects were attenuated by coculturing with Akt inhibitor LY294002. In summary, we concluded that the BYHWS vigorously blocked 6-OHDA-induced neurotoxicity via Akt/GSK3β pathway and provided a novel insight into roles of BYHWD in the clinical practices on neurodegenerative diseases.
Collapse
Affiliation(s)
- Zeyan Li
- Department of Anatomy, School of Medicine, Shandong University , Jinan, P.R. China
| | - Hui Wang
- Department of Anatomy, School of Medicine, Shandong University , Jinan, P.R. China
| | - Qian Wang
- Department of Anatomy, School of Medicine, Shandong University , Jinan, P.R. China
| | - Jinhao Sun
- Department of Anatomy, School of Medicine, Shandong University , Jinan, P.R. China
| |
Collapse
|
28
|
Liu L, Chen Y, Qi J, Zhang Y, He Y, Ni W, Li W, Zhang S, Sun S, Taketo MM, Wang L, Chai R, Li H. Wnt activation protects against neomycin-induced hair cell damage in the mouse cochlea. Cell Death Dis 2016; 7:e2136. [PMID: 26962686 PMCID: PMC4823936 DOI: 10.1038/cddis.2016.35] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/11/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022]
Abstract
Recent studies have reported the role of Wnt/β-catenin signaling in hair cell (HC) development, regeneration, and differentiation in the mouse cochlea; however, the role of Wnt/β-catenin signaling in HC protection remains unknown. In this study, we took advantage of transgenic mice to specifically knockout or overactivate the canonical Wnt signaling mediator β-catenin in HCs, which allowed us to investigate the role of Wnt/β-catenin signaling in protecting HCs against neomycin-induced damage. We first showed that loss of β-catenin in HCs made them more vulnerable to neomycin-induced injury, while constitutive activation of β-catenin in HCs reduced HC loss both in vivo and in vitro. We then showed that loss of β-catenin in HCs increased caspase-mediated apoptosis induced by neomycin injury, while β-catenin overexpression inhibited caspase-mediated apoptosis. Finally, we demonstrated that loss of β-catenin in HCs led to increased expression of forkhead box O3 transcription factor (Foxo3) and Bim along with decreased expression of antioxidant enzymes; thus, there were increased levels of reactive oxygen species (ROS) after neomycin treatment that might be responsible for the increased aminoglycoside sensitivity of HCs. In contrast, β-catenin overexpression reduced Foxo3 and Bim expression and ROS levels, suggesting that β-catenin is protective against neomycin-induced HC loss. Our findings demonstrate that Wnt/β-catenin signaling has an important role in protecting HCs against neomycin-induced HC loss and thus might be a new therapeutic target for the prevention of HC death.
Collapse
Affiliation(s)
- L Liu
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Y Chen
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - J Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Y Zhang
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - Y He
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - W Ni
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - W Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - S Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - S Sun
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Laboratory Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| | - M M Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - L Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - R Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - H Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, PR China
| |
Collapse
|
29
|
Abstract
Globally, greater than 30 million individuals are afflicted with disorders of the nervous system accompanied by tens of thousands of new cases annually with limited, if any, treatment options. Erythropoietin (EPO) offers an exciting and novel therapeutic strategy to address both acute and chronic neurodegenerative disorders. EPO governs a number of critical protective and regenerative mechanisms that can impact apoptotic and autophagic programmed cell death pathways through protein kinase B (Akt), sirtuins, mammalian forkhead transcription factors, and wingless signaling. Translation of the cytoprotective pathways of EPO into clinically effective treatments for some neurodegenerative disorders has been promising, but additional work is necessary. In particular, development of new treatments with erythropoiesis-stimulating agents such as EPO brings several important challenges that involve detrimental vascular outcomes and tumorigenesis. Future work that can effectively and safely harness the complexity of the signaling pathways of EPO will be vital for the fruitful treatment of disorders of the nervous system.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101
| |
Collapse
|
30
|
Maiese K. FoxO Transcription Factors and Regenerative Pathways in Diabetes Mellitus. Curr Neurovasc Res 2015; 12:404-13. [PMID: 26256004 PMCID: PMC4567483 DOI: 10.2174/1567202612666150807112524] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 02/07/2023]
Abstract
Mammalian forkhead transcription factors of the O class (FoxO) are exciting targets under consideration for the development of new clinical entities to treat metabolic disorders and diabetes mellitus (DM). DM, a disorder that currently affects greater than 350 million individuals globally, can become a devastating disease that leads to cellular injury through oxidative stress pathways and affects multiple systems of the body. FoxO proteins can regulate insulin signaling, gluconeogenesis, insulin resistance, immune cell migration, and cell senescence. FoxO proteins also control cell fate through oxidative stress and pathways of autophagy and apoptosis that either lead to tissue regeneration or cell demise. Furthermore, FoxO signaling can be dependent upon signal transduction pathways that include silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), Wnt, and Wnt1 inducible signaling pathway protein 1 (WISP1). Cellular metabolic pathways driven by FoxO proteins are complex, can lead to variable clinical outcomes, and require in-depth analysis of the epigenetic and post-translation protein modifications that drive FoxO protein activation and degradation.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|