51
|
Pham TLA, Binh TD, Liu G, Nguyen TQC, Nguyen YDH, Sahashi R, Men TT, Kamei K. Role of Serotonin Transporter in Eye Development of Drosophila melanogaster. Int J Mol Sci 2020; 21:ijms21114086. [PMID: 32521639 PMCID: PMC7312876 DOI: 10.3390/ijms21114086] [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: 04/15/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 11/30/2022] Open
Abstract
Serotonin transporter (SerT) in the brain is an important neurotransmitter transporter involved in mental health. However, its role in peripheral organs is poorly understood. In this study, we investigated the function of SerT in the development of the compound eye in Drosophila melanogaster. We found that SerT knockdown led to excessive cell death and an increased number of cells in S-phase in the posterior eye imaginal disc. Furthermore, the knockdown of SerT in the eye disc suppressed the activation of Akt, and the introduction of PI3K effectively rescued this phenotype. These results suggested that SerT plays a role in the healthy eye development of D.melanogaster by controlling cell death through the regulation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Tuan L. A. Pham
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Tran Duy Binh
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Guanchen Liu
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Thanh Q. C. Nguyen
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Yen D. H. Nguyen
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Ritsuko Sahashi
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
| | - Tran Thanh Men
- Department of Biology, Cantho University, Cantho 900000, Vietnam;
| | - Kaeko Kamei
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto 606-8585, Japan; (T.L.A.P.); (T.D.B.); (G.L.); (T.Q.C.N.); (Y.D.H.N.); (R.S.)
- Correspondence:
| |
Collapse
|
52
|
Zhang Y, Qin L, Xie J, Li J, Wang C. Eupatilin prevents behavioral deficits and dopaminergic neuron degeneration in a Parkinson's disease mouse model. Life Sci 2020; 253:117745. [PMID: 32376269 DOI: 10.1016/j.lfs.2020.117745] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 11/28/2022]
Abstract
AIMS Neuroinflammation and apoptosis play a crucial role in Parkinson's disease (PD) pathogenesis. Eupatilin is a lipophilic flavonoid isolated from Artemisia species and exerts anti-apoptotic and anti-inflammatory activities. In this study, we investigated the effects of Eupatilin on a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MAIN METHODS The rotarod test and traction test were constructed to examine the motor function. Immunofluorescent staining was performed to detect the expression of TH, Iba-1 and GFAP. Apoptosis was examined by the TUNEL assay. Real-time PCR was used to determine the mRNA expression and Western blot and ELISA were used to determine the protein expression. KEY FINDINGS Eupatilin improved behavioral impairment caused by MPTP. A loss of TH positive neurons was observed in the substantia nigra pars compacta of MPTP-lesioned brain, while it was rescued by Eupatilin. Moreover, MPTP administration increased the cell number of microglia and astrocytes and the expression of inflammatory factors TNF-α, IL-1β, and IL-6. Whereas Eupatilin suppressed the activation of neuroinflammation. Eupatilin also decreased cell apoptosis enhanced by MPTP/MPP+ exposure in vivo and in vitro. We further revealed that Eupatilin abolished MPTP-induced downregulation of IκBα expression and accumulation of p65 in the nuclear compartment. Besides, MPTP administration led to dephosphorylation of Akt and GSK-3β, but it was restored by Eupatilin. SIGNIFICANCE We demonstrate that Eupatilin alleviates behavioral impairment and dopaminergic neuron loss induced by MPTP through inhibition of neuroinflammation and apoptosis. Our research provides more evidence for Eupatilin as a potential preventative drug for PD.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Lina Qin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Jieru Xie
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Juan Li
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian 361102, PR China
| | - Chong Wang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, Fujian 361102, PR China.
| |
Collapse
|
53
|
Hansen T, Thant C, White JA, Banerjee R, Thuamsang B, Gunawardena S. Excess active P13K rescues huntingtin-mediated neuronal cell death but has no effect on axonal transport defects. Apoptosis 2020; 24:341-358. [PMID: 30725352 DOI: 10.1007/s10495-019-01520-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
High levels of oxidative stress is detected in neurons affected by many neurodegenerative diseases, including huntington's disease. Many of these diseases also show neuronal cell death and axonal transport defects. While nuclear inclusions/accumulations likely cause cell death, we previously showed that cytoplasmic axonal accumulations can also contribute to neuronal death. However, the cellular mechanisms responsible for activating cell death is unclear. One possibility is that perturbations in normal axonal transport alter the function of the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-pathway, a signal transduction pathway that promotes survival/growth in response to extracellular signals. To test this proposal in vivo, we expressed active PI3K in the context of pathogenic huntingtin (HTT-138Q) in Drosophila larval nerves, which show axonal transport defects and neuronal cell death. We found that excess expression of active P13K significantly suppressed HTT-138Q-mediated neuronal cell death, but had no effect on HTT-138Q-mediated axonal transport defects. Expression of active PI3K also rescued Paraquat-mediated cell death. Further, increased levels of pSer9 (inactive) glycogen synthase kinase 3β was seen in HTT-138Q-mediated larval brains, and in dynein loss of function mutants, indicating the modulation of the pro-survival pathway. Intriguingly, proteins in the PI3K/AKT-pathway showed functional interactions with motor proteins. Taken together our observations suggest that proper axonal transport is likely essential for the normal function of the pro-survival PI3K/AKT-signaling pathway and for neuronal survival in vivo. These results have important implications for targeting therapeutics to early insults during neurodegeneration and death.
Collapse
Affiliation(s)
- Timothy Hansen
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US
| | - Claire Thant
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US
| | - Joseph A White
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US
| | - Rupkatha Banerjee
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US
| | - Bhasirie Thuamsang
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, NY, 14260, US. .,The State University of New York at Buffalo, 109 Cooke Hall, North/Amherst Campus, Buffalo, NY, 14260, US.
| |
Collapse
|
54
|
Xie W, Wulin H, Shao G, Wei L, Qi R, Ma B, Chen N, Shi R. Polygalasaponin F inhibits neuronal apoptosis induced by oxygen-glucose deprivation and reoxygenation through the PI3K/Akt pathway. Basic Clin Pharmacol Toxicol 2020; 127:196-204. [PMID: 32237267 DOI: 10.1111/bcpt.13408] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 11/29/2022]
Abstract
Cerebral ischaemia is a common cerebrovascular disease and often induces neuronal apoptosis, leading to brain damage. Polygalasaponin F (PGSF) is one of the components in Polygala japonica Houtt, and it is a triterpenoid saponin monomer. This research focused on anti-apoptotic effect of PGSF during oxygen-glucose deprivation and reoxygenation (OGD/R) injury in rat adrenal pheochromocytoma cells (PC12) and primary rat cortical neurons. OGD/R treatment reduced viability of PC12 cells and primary neurons. This reduced viability was prevented by PGSF, as shown by MTT assay. OGD/R insult decreased expression of Bcl-2/Bax both in PC12 cells and primary neurons but elevated levels of caspase-3 in primary neurons. However, PGSF may up-regulate expression of Bcl-2/Bax and down-regulate caspase-3 in these particular cells. Furthermore, Bcl-2/Bax and the ratio between phosphorylated Akt and total Akt were decreased in PC12 cells treated with OGD/R, and both were increased by PGSF. Moreover, increase in the ratios of Bcl-2/Bax and phosphorylated Akt/total Akt in PC12 cells was suppressed by phosphatidylinositol 3-kinase (PI3K) inhibitor. Data suggest PGSF might prevent OGD/R-induced injury via activation of PI3K/Akt signalling. The ability of PGSF to block the effects of OGD/R appears to involve regulation of Bcl-2, Bax and caspase-3, which are related to apoptosis.
Collapse
Affiliation(s)
- Wei Xie
- Department of Physiology, Baotou Medical College, Baotou, China.,Institute of Neuroscience, Baotou Medical College, Baotou, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Hade Wulin
- Department of Physiology, Baotou Medical College, Baotou, China.,Department of Pharmacy, Inner Mongolia International Mongolian Hospital, Hohhot, China
| | - Guo Shao
- Institute of Neuroscience, Baotou Medical College, Baotou, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Liqin Wei
- Department of Traditional Chinese Medical Science, Baotou Medical College, Baotou, China
| | - Ruifang Qi
- Department of Physiology, Baotou Medical College, Baotou, China.,Institute of Neuroscience, Baotou Medical College, Baotou, China
| | - Baohui Ma
- Department of Physiology, Baotou Medical College, Baotou, China.,Institute of Neuroscience, Baotou Medical College, Baotou, China
| | - Naihong Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruili Shi
- Department of Physiology, Baotou Medical College, Baotou, China.,Institute of Neuroscience, Baotou Medical College, Baotou, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| |
Collapse
|
55
|
Gugliandolo A, Pollastro F, Bramanti P, Mazzon E. Cannabidiol exerts protective effects in an in vitro model of Parkinson's disease activating AKT/mTOR pathway. Fitoterapia 2020; 143:104553. [PMID: 32184097 DOI: 10.1016/j.fitote.2020.104553] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 01/12/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of the nigrostriatal dopaminergic pathway with loss of substantia nigra pars compacta neurons and dopamine depletion. Various natural compounds showed protective actions against PD. In this work, the protective effects of cannabidiol (CBD), obtained from Cannabis sativa, were evaluated in retinoic acid differentiated SH-SY5Y cells exposed to 1-methyl-4-phenylpyridinium (MPP+), an in vitro PD model. In order to evaluate which receptor is involved in CBD actions CB1, CB2 and TRPV1 receptor antagonists were used. CBD counteracted the loss of cell viability caused by MPP+, reducing apoptosis as demonstrated by the reduction of Bax and caspase 3. Moreover, CBD reduced the nuclear levels of PARP-1. The protective effects of CBD seem to be mediated by the activation of ERK and AKT/mTOR pathways. The treatment with AKT1/2 inhibitor and the mTOR inhibitor rapamycin abolished CBD protective effects. The CBD-induced ERK activation may be mediated by CBD interaction with CB2 and TRPV1. We also investigated the protein levels of the autophagic proteins LC3 and beclin 1. CBD reduced the MPP+-induced increase of LC3 by CB2 and TRPV1 receptors. These data suggested the involvement of ERK in the modulation of autophagy. However, beclin 1 levels were not modified neither by MPP+ nor by CBD. These results indicated that CBD may exert preventive and protective actions in PD.
Collapse
Affiliation(s)
- Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Federica Pollastro
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
56
|
Dai DP, Yu LZ, Ye MF. LincRNA-p21 reverses irinotecan resistance in colon cancer cells via the PI3K/AKT signaling pathway. Shijie Huaren Xiaohua Zazhi 2019; 27:1356-1364. [DOI: 10.11569/wcjd.v27.i22.1356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Irinotecan (camptothecin-11, CPT-11) is a first-line chemotherapy drug for advanced colon cancer, but CPT-11 resistance limits its efficacy. Studying the mechanism of CPT-11 resistance in colon cancer and restoring the sensitivity of colon cancer cells to CPT-11 are of great clinical value in prolonging the life time of colon cancer patients.
AIM To investigate the effect and mechanism of long intergenic non-coding RNA-p21 (lincRNA-p21) on CPT-11 resistance in colon cancer cells.
METHODS HCT-8 and SW480 cells were used to construct irinotecan-resistant HCT-8/CPT-11 and SW480/CPT-11 cell lines by continuously exposing them to increasing concentrations of CPT-11, and the expression of lincRNA-p21 in the cells was detected by real-time quantitative polymerase chain reaction (RT-qPCR). After transfection with pcDNA-lincRNA-p21 or si-lincRNA-p21, the effect of CPT-11 on the viability of HCT-8/CPT-11 cells or SW480/CPT-11 cells was measured by cell counting kit-8 (CCK-8) assay. The regulatory effect of lincRNA-p21 on the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway was preliminarily analyzed by Western blot. After pretreatment with PI3K/AKT pathway inhibitor LY294002 prior to transfection with si-lincRNA-p21, or pretreatment with PI3K/AKT pathway agonist Recilisib prior to transfection with pcDNA-lincRNA-p21, the effect of CPT-11 on cell viability in HCT-8/CPT-11 cells or SW480/CPT-11 cells was measured by CCK-8 assay.
RESULTS LincRNA-p21 expression in CPT-11 resistant cells was significantly lower than that in parental cells. Overexpression of lincRNA-p21 inhibited the resistance of HCT-8/CPT-11 cells and SW480/CPT-11 cells to CPT-11, while knockdown of lincRNA-p21 enhanced the resistance of HCT-8/CPT-11 cells and SW480/CPT-11 cells to CPT-11. Western blot results showed that overexpression of lincRNA-p21 inhibited the activity of the PI3K/AKT pathway, while knockdown of lincRNA-p21 enhanced the activity of the PI3K/AKT pathway. LY294002 inhibited the promotive effect of lincRNA-p21 knockdown on CPT-11 resistance, while Recilisib inhibited the inhibitive effect of lincRNA-p21 overexpression on CPT-11 resistance.
CONCLUSION Up-regulation of lincRNA-p21 can inhibit the CPT-11 resistance of colorectal cancer cells, while down-regulation of lincRNA-p21 can promote their CPT-11 resistance, which may be related to the regulation of the PI3K/AKT signaling activity by lincRNA-p21.
Collapse
Affiliation(s)
- Dan-Ping Dai
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| | - Ling-Zhi Yu
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| | - Meng-Fei Ye
- Department of Pharmacy, Taizhou Hospital, Taizhou Grace Medical Center (Group), Taizhou 317000, Zhejiang Province, China
| |
Collapse
|
57
|
Downregulation of lncRNA UCA1 ameliorates the damage of dopaminergic neurons, reduces oxidative stress and inflammation in Parkinson's disease through the inhibition of the PI3K/Akt signaling pathway. Int Immunopharmacol 2019; 75:105734. [PMID: 31301558 DOI: 10.1016/j.intimp.2019.105734] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/21/2019] [Accepted: 06/29/2019] [Indexed: 01/05/2023]
Abstract
This study is conducted to investigate the role of lncRNA urothelial carcinoma-associated 1 (UCA1) in the protection of dopaminergic neurons in Parkinson's disease (PD) through regulating the PI3K/Akt signaling pathway. PD rat model was induced by injection of 6-hydroxydopamine (6-OHDA) to damage the substantia nigra striatum. The successfully modeled PD rats were introduced with siRNA-negative control (NC) or UCA1-siRNA. The expression of UCA1 in neurobehavioral change, neuroinflammatory response and oxidative stress of PD rats were explored. The effect of UCA1 on the PI3K/Akt signaling pathway and downstream proteins IκBα and ERK was also investigated. The rats with PD exhibited aggregated neurobehavioral change, increased neuroinflammatory response and oxidative stress. Down-regulation of UCA1 up-regulated the expression of TH positive cells and DA content, reduced the apoptosis of substantia nigra neurons, the apoptosis of substantia nigra neurons and oxidative stress and improved the neuroinflammatory response in PD rats. Down-regulation of UCA1 inhibited the activation of the PI3K/AKT signaling pathway in substantia nigra of PD rats. Our study suggests that the downregulated lncRNA UCA1 ameliorates the damage of dopaminergic neurons, reduces oxidative stress and inflammation in PD rats through the inhibition of the PI3K/Akt signaling pathway.
Collapse
|
58
|
Yao S, Li L, Sun X, Hua J, Zhang K, Hao L, Liu L, Shi D, Zhou H. FTY720 Inhibits MPP +-Induced Microglial Activation by Affecting NLRP3 Inflammasome Activation. J Neuroimmune Pharmacol 2019; 14:478-492. [PMID: 31069623 DOI: 10.1007/s11481-019-09843-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons and excessive microglial activation in the substantia nigra pars compacta (SNpc). In the present study, we aimed to demonstrate the therapeutic effectiveness of the potent sphingosine-1-phosphate receptor antagonist fingolimod (FTY720) in an animal model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and to identify the potential mechanisms underlying these therapeutic effects. C57BL/6J mice were orally administered FTY720 before subcutaneous injection of MPTP. Open-field and rotarod tests were performed to determine the therapeutic effect of FTY720. The damage to dopaminergic neurons and the production of monoamine neurotransmitters were assessed using immunohistochemistry, high-performance liquid chromatography, and flow cytometry. Immunofluorescence (CD68- positive) and enzyme-linked immunosorbent assay were used to analyze the activation of microglia, and the levels of activated signaling molecules were measured using Western blotting. Our findings indicated that FTY720 significantly attenuated MPTP-induced behavioral deficits, reduced the loss of dopaminergic neurons, and increased dopamine release. FTY720 directly inhibited MPTP-induced microglial activation in the SNpc, suppressed the production of interleukin (IL)-6, IL-1β, and tumor necrosis factor-α in BV-2 microglial cells treated with 1-methyl-4-phenylpyridinium (MPP+), and subsequently decreased apoptosis in SH-SY5Y neuroblastoma cells. Moreover, in MPP+-treated BV-2 cells and primary microglia, FTY720 treatment significantly attenuated the increases in the phosphorylation of PI3K/AKT/GSK-3β, reduced ROS generation and p65 activation, and also inhibited the activation of NLRP3 inflammasome and caspase-1. In conclusion, FTY720 may reduce PD progression by inhibiting NLRP3 inflammasome activation via its effects on ROS generation and p65 activation in microglia. These findings provide novel insights into the mechanisms underlying the therapeutic effects of FTY720, suggesting its potential as a novel therapeutic strategy against PD. Graphical Abstract FTY720 may reduce ROS production by inhibiting the PI3K/AKT/GSK-3β signaling pathway, while at the same time reducing p65 phosphorylation, thus decreasing NLRP3 inflammasome activation through these two pathways, ultimately reducing microglia activation-induced neuronal damage.
Collapse
Affiliation(s)
- Shu Yao
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Longjun Li
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Xin Sun
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Jun Hua
- Department of Clinical Pharmacy, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China
| | - Keqi Zhang
- Institute of Microscope Science and Technology, Ningbo Yongxin Optics Co. Ltd., 385 Mingzhu Road, Hi-tech Industry Park, Ningbo, 315040, China
| | - Li Hao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China.
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China.
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
59
|
Franceschelli S, Lanuti P, Ferrone A, Gatta DMP, Speranza L, Pesce M, Grilli A, Cacciatore I, Ricciotti E, Di Stefano A, Miscia S, Felaco M, Patruno A. Modulation of Apoptotic Cell Death and Neuroprotective Effects of Glutathione-L-Dopa Codrug Against H 2O 2-Induced Cellular Toxicity. Antioxidants (Basel) 2019; 8:antiox8080319. [PMID: 31430883 PMCID: PMC6720001 DOI: 10.3390/antiox8080319] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 11/16/2022] Open
Abstract
The L-3,4-dihydroxyphenylalanine (LD) is the gold standard drug currently used to manage Parkinson’s disease (PD) and to control its symptoms. However, LD could cause disease neurotoxicity due to the generation of pro-oxidant intermediates deriving from its autoxidation. In order to overcome this limitation, we have conjugated LD to the natural antioxidant glutathione (GSH) to form a codrug (GSH-LD). Here we investigated the effect of GSH-LD on H2O2-induced cellular toxicity in undifferentiated and differentiated lymphoma U-937 and dopaminergic neuroblastoma SH-SY5Y cell lines, used respectively as models to study the involvement of macrophages/microglia and dopaminergic neurons in PD. We analyzed the effect of GSH-LD on apoptosis and cellular oxidative stress, both considered strategic targets for the prevention and treatment of neurodegenerative diseases. Compared to LD and GSH, GSH-LD had a stronger effect in preventing hydrogen peroxide (H2O2) induced apoptosis in both cell lines. Moreover, GSH-LD was able to preserve cell viability, cellular redox status, gluthation metabolism and prevent reactive oxygen species (ROS) formation, in a phosphinositide 3-kinase (PI3K)/kinase B (Akt)-dependent manner, in a neurotoxicity cellular model. Our findings indicate that the GSH-LD codrug offers advantages deriving from the additive effect of LD and GSH and it could represent a promising candidate for PD treatment.
Collapse
Affiliation(s)
- Sara Franceschelli
- Department of Psychological, Health and Territorial Sciences, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Paola Lanuti
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Alessio Ferrone
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Daniela Maria Pia Gatta
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Lorenza Speranza
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Mirko Pesce
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Alfredo Grilli
- Department of Psychological, Health and Territorial Sciences, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University "Gabriele D'Annunzio" of Chieti-Pescara, 66100 Chieti-Pescara, Italy
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonio Di Stefano
- Department of Pharmacy, University "Gabriele D'Annunzio" of Chieti-Pescara, 66100 Chieti-Pescara, Italy
| | - Sebastiano Miscia
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Mario Felaco
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy
| | - Antonia Patruno
- Department of Medicine and Science of Aging, University "G. D'Annunzio", 66100 Chieti-Pescara, Italy.
| |
Collapse
|
60
|
Oza M, Becker W, Gummadidala PM, Dias T, Omebeyinje MH, Chen L, Mitra C, Jesmin R, Chakraborty P, Sajish M, Hofseth LJ, Banerjee K, Wang Q, Moeller PDR, Nagarkatti M, Nagarkatti P, Chanda A. Acute and short-term administrations of delta-9-tetrahydrocannabinol modulate major gut metabolomic regulatory pathways in C57BL/6 mice. Sci Rep 2019; 9:10520. [PMID: 31324830 PMCID: PMC6642200 DOI: 10.1038/s41598-019-46478-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 06/19/2019] [Indexed: 01/07/2023] Open
Abstract
Delta-9-tetrahydrocannabinol (THC) is the primary psychoactive compound in Cannabis, which is studied extensively for its medicinal value. A central gap in the science is the underlying mechanisms surrounding THC's therapeutic effects and the role of gut metabolite profiles. Using a mass-spectrometry based metabolomics, we show here that intraperitoneal injection of THC in C57BL/6 mice modulates metabolic profiles that have previously been identified as integral to health. Specifically, we investigated the effects of acute (single THC injection denoted here as '1X') and short -term (five THC injections on alternate days denoted as '5X') THC administration on fecal and intestinal tissue metabolite profiles. Results are consistent with the hypothesis that THC administration alters host metabolism by targeting two prominent lipid metabolism pathways: glycerophospholipid metabolism and fatty acid biosynthesis.
Collapse
Affiliation(s)
- Megha Oza
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - William Becker
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Phani M Gummadidala
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Travis Dias
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Mayomi H Omebeyinje
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Li Chen
- Creative Proteomics Inc., Shirley, New York, USA
| | - Chandrani Mitra
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Rubaiya Jesmin
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | | | - Mathew Sajish
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Lorne J Hofseth
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | | | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Peter D R Moeller
- National Ocean Service, Hollings Marine Laboratory, Charleston, SC, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Anindya Chanda
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
61
|
Bono F, Mutti V, Savoia P, Barbon A, Bellucci A, Missale C, Fiorentini C. Nicotine prevents alpha-synuclein accumulation in mouse and human iPSC-derived dopaminergic neurons through activation of the dopamine D3- acetylcholine nicotinic receptor heteromer. Neurobiol Dis 2019; 129:1-12. [PMID: 31051233 DOI: 10.1016/j.nbd.2019.04.017] [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: 01/23/2019] [Revised: 04/05/2019] [Accepted: 04/29/2019] [Indexed: 12/25/2022] Open
Abstract
We recently found that in mouse dopaminergic neurons, the heteromer formed by the dopamine D3 receptor (D3R) and the β2 subunit of acetylcholine nicotinic receptor (nAChR) exerts neurotrophic effects when activated by nicotine, leading to neurons with enlarged cell bodies and increased dendrite arborization. Beside this action, we now show that nicotine, by activating the D3R-nAChR heteromer, protects dopaminergic neurons against neuronal injury. In primary cultures of mouse dopaminergic neurons, in fact, the ability of nicotine to inhibit both the pathological accumulation of alpha-synuclein induced by glucose deprivation and the consequent morphological defects were strongly prevented by disrupting the D3R-nAChR heteromer with specific interfering TAT-peptides; the relevance of the phosphoinositide 3-kinase (PI3K) intracellular signaling in mediating nicotine prevention of alpha-synuclein aggregation has been also demonstrated. Moreover, the ability of nicotine in restoring the ubiquitin-proteasome system has been found as a mechanism contributing to the neuroprotective properties of nicotine. By using the proximity ligation assay, we have shown that the D3R-nAChR heteromer is also expressed in human dopaminergic neurons derived from induced pluripotent stem cells. In this human cell model, nicotine exerts neuroprotective effects specifically acting through the D3R-nAChR complex thus indicating that this heteromer is a relevant molecular effector involved in the protection of human dopaminergic neurons.
Collapse
Affiliation(s)
- Federica Bono
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; Laboratory of Personalized and Preventive Medicine, University of Brescia, 25123 Brescia, Italy
| | - Veronica Mutti
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Paola Savoia
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alessandro Barbon
- Unit of Biology and Genetic, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Arianna Bellucci
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; Laboratory of Personalized and Preventive Medicine, University of Brescia, 25123 Brescia, Italy
| | - Cristina Missale
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Chiara Fiorentini
- Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
62
|
New Insights into the Mechanisms of Action of Topical Administration of GLP-1 in an Experimental Model of Diabetic Retinopathy. J Clin Med 2019; 8:jcm8030339. [PMID: 30862093 PMCID: PMC6463072 DOI: 10.3390/jcm8030339] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
The main goals of this work were to assess whether the topical administration of glucagon-like peptide-1 (GLP-1) could revert the impairment of the neurovascular unit induced by long-term diabetes (24 weeks) in diabetic mice and to look into the underlying mechanisms. For that reason, db/db mice were treated with eye drops of GLP-1 or vehicle for 3 weeks. Moreover, db/+ mice were used as control. Studies performed in vivo included electroretinogramand the assessment of vascular leakage by using Evans Blue. NF-κB, GFAP and Ki67 proteins were analyzed by immunofluorescence (IF). Additionally, caspase 9, AMPK, IKBα, NF-κB, AKT, GSK3, β-catenin, Bcl-xl, and VEGF were analyzed by WB. Finally, VEGF, IL-1β, IL-6, TNF-α, IL-18, and NLRP3 were studied by reverse transcription-polymerase chain reaction (RT-PCR) and immunofluorescence. We found that topical administration of GLP-1 reverted reactive gliosis and albumin extravasation, and protected against apoptosis and retinal dysfunction. Regarding the involved mechanisms, GLP-1 exerted an anti-inflammatory action by decreasing NF-κB, inflammosome, and pro-inflammatory factors. In addition, it also decreased VEGF expression. Furthermore, GLP-1 promoted cell survival by increasing the anti-apoptotic protein Bcl-xl and the signaling pathway Akt/GSK3b/β-catenin. Finally, Ki67 results revealed that GLP-1 treatment could induce neurogenesis. In conclusion, the topical administration of GLP-1 reverts the impairment of the neurovascular unit by modulating essential pathways involved in the development of diabetic retinopathy (DR). These beneficial effects on the neurovascular unit could pave the way for clinical trials addressed to confirm the effectiveness of GLP-1 in early stages of DR.
Collapse
|
63
|
Park J, Ha SH, Abekura F, Lim H, Chang YC, Lee MJ, Lee M, Lee YC, Kim CH. 4-O-carboxymethylascochlorin protected against microglial-mediated neurotoxicity in SH-SY5Y and BV2 cocultured cells from LPS-induced neuroinflammation and death by inhibiting MAPK, NF-κB, and Akt pathways. J Cell Biochem 2019; 120:1742-1753. [PMID: 30324762 DOI: 10.1002/jcb.27464] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/19/2018] [Indexed: 01/24/2023]
Abstract
In our previous studies, structurally similar compounds of ascochlorin and ascofuranone exhibited anti-inflammatory activity. Neural inflammation plays a significant role in the commence and advancement of neurodegenerative diseases. It is not known whether 4-O-carboxymethylascochlorin (AS-6) regulates the initial stage of inflammatory responses at the cellular level in BV2 microglia cells. We here investigated the anti-inflammatory effects of AS-6 treatment in microglia cells with the microglial protection in neurons. We found that the lipopolysaccharide (LPS)-stimulated production of nitric oxide, a main regulator of inflammation, is suppressed by AS-6 in BV2 microglial cells. In addition, AS-6 dose-dependently suppressed the increase in COX-2 protein and messenger RNA levels in LPS-stimulated BV2 cells. Moreover, AS-6 inhibited the expression and secretion of proinflammatory cytokines in BV2 microglial cells. At the intracellular level, AS-6 inhibited LPS-activated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in BV2 microglial cells. AS-6 negatively affected mitogen-activated protein kinases (MAPK) and Akt phosphorylation: Phosphorylated forms of ERK, JNK, p38, and Akt decreased. To check whether AS-6 protects against inflammatory inducer-mediated neurotoxicity, neuronal SH-SY5Y cells were coincubated with BV2 cells in conditioned medium. AS-6 exerted a neuroprotective effect by suppressing microglial activation by LPS or amyloid-β peptide. AS-6 is a promising suppressor of inflammatory responses in LPS-induced BV2 cells by attenuating NF-κB and MAPKs signaling. AS-6 protected against microglial-mediated neurotoxicity in SH-SY5Y and BV2 cocultured cells from LPS-induced neuroinflammation and death via inhibiting MAPK, NF-κB, and Akt pathways.
Collapse
Affiliation(s)
- Junyoung Park
- Department of Biological Sciences, Molecular and Cellular Glycobiology Unit, SungKyunKwan University, Suwon, Kyunggi-Do, Republic of Korea
| | - Sun-Hyung Ha
- Department of Biological Sciences, Molecular and Cellular Glycobiology Unit, SungKyunKwan University, Suwon, Kyunggi-Do, Republic of Korea
| | - Fukushi Abekura
- Department of Biological Sciences, Molecular and Cellular Glycobiology Unit, SungKyunKwan University, Suwon, Kyunggi-Do, Republic of Korea
| | - Hakseong Lim
- Department of Biological Sciences, Molecular and Cellular Glycobiology Unit, SungKyunKwan University, Suwon, Kyunggi-Do, Republic of Korea
| | - Young-Chae Chang
- Department of Medicine, Research Institute of Biomedical Engineering, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Moon-Jo Lee
- Department of Herb Science, Dong-Eui Institute of Technology, Busan, Republic of Korea
| | - Miri Lee
- Department of Biological Sciences, Faculty of Medicinal Biotechnology, Dong-A University, Busan, Republic of Korea
| | - Young-Choon Lee
- Department of Biological Sciences, Faculty of Medicinal Biotechnology, Dong-A University, Busan, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Sciences, Molecular and Cellular Glycobiology Unit, SungKyunKwan University, Suwon, Kyunggi-Do, Republic of Korea
| |
Collapse
|
64
|
Li J, Luo J, Liu L, Fu H, Tang L. The association between CD157/BST1 polymorphisms and the susceptibility of Parkinson's disease: a meta-analysis. Neuropsychiatr Dis Treat 2019; 15:1089-1102. [PMID: 31118642 PMCID: PMC6500436 DOI: 10.2147/ndt.s190935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/29/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Different studies have provided some evidence for the association between BST1 polymorphisms and Parkinson's disease (PD). The extent to which these genetic effects are consistent across different populations is unknown. Methods: A meta-analysis of PD case-control studies using a common set of three variants was conducted. Published reports were obtained from electronic databases including Pubmed, Embase, Chinese National Knowledge Infrastructure (CNKI) and Cochrane Library databases between August 2010 and January 2018. Results: A total of 11 individual studies with 8,725 cases and 17,079 controls were included. The results showed statistically significant association between the dominant model of rs11931532 and PD risk in Asian populations (P=0.006, OR [95% CI]=1.22 [1.06-1.41]). Significant association was also detected between the allelic, dominant, and recessive models of rs4698412 and PD risk in Asian populations (allelic model: P<0.00001, OR [95% CI]=1.22 [1.16-1.29]; dominant model: P<0.00001, OR [95%CI]=1.35 [1.20-1.52]; recessive model; P=0.0003, OR [95% CI]=1.30 [1.13-1.50]). Nevertheless, the pooled analyses suggested that no significant association was uncovered between rs11724635 and PD risk (P>0.05). Conclusion: The meta-analysis suggests that the rs11931532 and rs4698412, but not rs11724635 might be risk factors for PD in Asian populations.
Collapse
Affiliation(s)
- Jianming Li
- Department of Neurology, Xiang-ya Hospital, Central South University, Changsha 410219, People's Republic of China.,Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, People's Republic of China.,Department of Human Anatomy, School of Basic Medical Science, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Jia Luo
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Li Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Hui Fu
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, People's Republic of China.,Department of Human Anatomy, School of Basic Medical Science, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Liang Tang
- Department of Human Anatomy, Histology and Embryology, Institute of Neuroscience, Changsha Medical University, Changsha, 410219, People's Republic of China
| |
Collapse
|
65
|
Aqueous Extract of Davallia mariesii Attenuates 6-Hydroxydopamine-Induced Oxidative Damage and Apoptosis in B35 Cells Through Inhibition of Caspase Cascade and Activation of PI3K/AKT/GSK-3β Pathway. Nutrients 2018; 10:nu10101449. [PMID: 30301204 PMCID: PMC6213869 DOI: 10.3390/nu10101449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022] Open
Abstract
The medicinal ferns of Polydiaceae and Davalliaceae species are called "Gusuibu" by Chinese physicians and used as antiaging dietary medicines. Our previous report revealed that Drynaria fortunei (Polydiaceae) protected against 6-hydroxydopamine (6-OHDA)-induced oxidative damage via the PI3K/AKT pathway in B35 neuroblastoma cells. The present study compares the antioxidant phytoconstituent contents and radical scavenging capacities of five Davalliaceae species. The further aim was to clarify the protective mechanism of Davallia mariesii (DM) against 6-OHDA-induced oxidative damage and apoptosis in B35 cells. The results show that Araiostegia perdurans (AP) and DM extracts have better radical scavenging capacities against 1,1-diphenyl-2-picryhydrazyl (DPPH) and reactive oxygen species (ROS) than other Davalliaceae species. However, only DM extract inhibited 6-OHDA autoxidation under cell-free systems and increased cell viability, compared to B35 cells solely exposed to 6-OHDA. DM extract decreased apoptosis and restored mitochondrial expression in 6-OHDA-treated B35 cells. Additional data indicated that DM extract decreased intracellular ROS and nitric oxide levels generated by 6-OHDA exposure. DM extract also restored glutathione (GSH) levels and the activities of glutathione peroxidase and reductase, and then decreased the elevated malondialdehyde (MDA) levels. Finally, DM extract regulated the protein expression of the caspase cascade and PI3K/AKT/GSK-3β pathways. These results suggest that the protective mechanism of DM extract against 6-OHDA-induced oxidative damage and apoptosis might be related to its radical scavenging capacity, maintaining the mitochondrial function to inhibit the Bcl-2/caspase cascade pathway and activating intracellular antioxidant defenses (GSH recycling, HO-1 and NQO-1) by modulating the activation of the PI3K/AKT/GSK-3β pathway.
Collapse
|
66
|
Post-treatment with PT302, a long-acting Exendin-4 sustained release formulation, reduces dopaminergic neurodegeneration in a 6-Hydroxydopamine rat model of Parkinson's disease. Sci Rep 2018; 8:10722. [PMID: 30013201 PMCID: PMC6048117 DOI: 10.1038/s41598-018-28449-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
We previously demonstrated that pretreatment with Exendin-4, a glucagon-like peptide-1 (GLP-1) receptor agonist, reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) -mediated dopaminergic neurodegeneration. The use of GLP-1 or Exendin-4 for Parkinson's disease (PD) patients is limited by their short half-lives. The purpose of this study was to evaluate a new extended release Exendin-4 formulation, PT302, in a rat model of PD. Subcutaneous administration of PT302 resulted in sustained elevations of Exendin-4 in plasma for >20 days in adult rats. To define an efficacious dose within this range, rats were administered PT302 once every 2 weeks either before or following the unilaterally 6-hydroxydopamine lesioning. Pre- and post-treatment with PT302 significantly reduced methamphetamine-induced rotation after lesioning. For animals given PT302 post lesion, blood and brain samples were collected on day 47 for measurements of plasma Exendin-4 levels and brain tyrosine hydroxylase immunoreactivity (TH-IR). PT302 significantly increased TH-IR in the lesioned substantia nigra and striatum. There was a significant correlation between plasma Exendin-4 levels and TH-IR in the substantia nigra and striatum on the lesioned side. Our data suggest that post-treatment with PT302 provides long-lasting Exendin-4 release and reduces neurodegeneration of nigrostriatal dopaminergic neurons in a 6-hydroxydopamine rat model of PD at a clinically relevant dose.
Collapse
|
67
|
Haga S, Ozawa T, Morita N, Asano M, Jin S, Ozaki M. Photo-Activatable Akt Probe: A New Tool to Study the Akt-Dependent Physiopathology of Cancer Cells. Oncol Res 2018; 26:467-472. [PMID: 28933316 PMCID: PMC7844718 DOI: 10.3727/096504017x15040166233313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Akt is commonly overexpressed and activated in cancer cells and plays a pivotal role in cell survival, protection, and chemoresistance. Therefore, Akt is one of the target molecules in understanding characters of cancer cells and developing anticancer drugs. Here we examined whether a newly developed photo-activatable Akt (PA-Akt) probe, based on a light-inducible protein interaction module of plant cryptochrome2 (CRY2) and cryptochrome-interacting basic helix–loop–helix (CIB1), can regulate Akt-associated cell functions. By illuminating blue light to the cells stably transfected with PA-Akt probe, CRY2-Akt (a fusion protein of CRY2 and Akt) underwent a structural change and interacted with Myr-CIBN (myristoylated N-terminal portion of CIB1), anchoring it at the cell membrane. Western blot analysis revealed that S473 and T308 of the Akt of probe-Akt were sequentially phosphorylated by intermittent and continuous light illumination. Endogenous Akt and GSK-3β, one of the main downstream signals of Akt, were also phosphorylated, depending on light intensity. These facts indicate that photo-activation of probe-Akt can activate endogenous Akt and its downstream signals. The photo-activated Akt conferred protection against nutritional deprivation and H2O2 stresses to the cells significantly. Using the newly developed PA-Akt probe, endogenous Akt was activated easily, transiently, and repeatedly. This probe will be a unique tool in studying Akt-associated specific cellular functions in cancer cells and developing anticancer drugs.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of TokyoTokyoJapan
| | - Naoki Morita
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)Sapporo, HokkaidoJapan
| | - Mami Asano
- Laboratory of Molecular and Functional Bioimaging, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Shigeki Jin
- Core Research Laboratory, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido UniversitySapporoJapan
| |
Collapse
|
68
|
Proanthocyanidins against Oxidative Stress: From Molecular Mechanisms to Clinical Applications. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8584136. [PMID: 29750172 PMCID: PMC5884402 DOI: 10.1155/2018/8584136] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/14/2018] [Indexed: 12/15/2022]
Abstract
Proanthocyanidins (PCs) are naturally occurring polyphenolic compounds abundant in many vegetables, plant skins (rind/bark), seeds, flowers, fruits, and nuts. Numerous in vitro and in vivo studies have demonstrated myriad effects potentially beneficial to human health, such as antioxidation, anti-inflammation, immunomodulation, DNA repair, and antitumor activity. Accumulation of prooxidants such as reactive oxygen species (ROS) exceeding cellular antioxidant capacity results in oxidative stress (OS), which can damage macromolecules (DNA, lipids, and proteins), organelles (membranes and mitochondria), and whole tissues. OS is implicated in the pathogenesis and exacerbation of many cardiovascular, neurodegenerative, dermatological, and metabolic diseases, both through direct molecular damage and secondary activation of stress-associated signaling pathways. PCs are promising natural agents to safely prevent acute damage and control chronic diseases at relatively low cost. In this review, we summarize the molecules and signaling pathways involved in OS and the corresponding therapeutic mechanisms of PCs.
Collapse
|
69
|
Roy U, Conklin L, Schiller J, Matysik J, Berry JP, Alia A. Metabolic profiling of zebrafish (Danio rerio) embryos by NMR spectroscopy reveals multifaceted toxicity of β-methylamino-L-alanine (BMAA). Sci Rep 2017; 7:17305. [PMID: 29230019 PMCID: PMC5725574 DOI: 10.1038/s41598-017-17409-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/21/2017] [Indexed: 11/09/2022] Open
Abstract
β-methylamino-L-alanine (BMAA) has been linked to several interrelated neurodegenerative diseases. Despite considerable research, specific contributions of BMAA toxicity to neurodegenerative diseases remain to be fully resolved. In the present study, we utilized state-of-the-art high-resolution magic-angle spinning nuclear magnetic resonance (HRMAS NMR), applied to intact zebrafish (Danio rerio) embryos, as a model of vertebrate development, to elucidate changes in metabolic profiles associated with BMAA exposure. Complemented by several alternative analytical approaches (i.e., in vivo visualization and in vitro assay), HRMAS NMR identified robust and dose-dependent effect of BMAA on several relevant metabolic pathways suggesting a multifaceted toxicity of BMAA including: (1) localized production of reactive oxygen species (ROS), in the developing brain, consistent with excitotoxicity; (2) decreased protective capacity against excitotoxicity and oxidative stress including reduced taurine and glutathione; (3) inhibition of several developmentally stereotypical energetic and metabolic transitions, i.e., metabolic reprogramming; and (4) inhibition of lipid biosynthetic pathways. Matrix-assisted laser desorption time-of-flight (MALDI-ToF) mass spectrometry further identified specific effects on phospholipids linked to both neural development and neurodegeneration. Taken together, a unified model of the neurodevelopmental toxicity of BMAA in the zebrafish embryo is presented in relation to the potential contribution of BMAA to neurodegenerative disease.
Collapse
Affiliation(s)
- Upasana Roy
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany.,Institute of Analytical Chemistry, University of Leipzig, D-04103, Leipzig, Germany
| | - Laura Conklin
- Department of Chemistry and Biochemistry, Florida International University, North Miami, FL, 33181, USA
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany
| | - Jörg Matysik
- Institute of Analytical Chemistry, University of Leipzig, D-04103, Leipzig, Germany
| | - John P Berry
- Department of Chemistry and Biochemistry, Florida International University, North Miami, FL, 33181, USA.
| | - A Alia
- Institute for Medical Physics and Biophysics, University of Leipzig, D-04107, Leipzig, Germany. .,Leiden Institute of Chemistry, 2333, Leiden, The Netherlands.
| |
Collapse
|
70
|
Talarek S, Listos J, Barreca D, Tellone E, Sureda A, Nabavi SF, Braidy N, Nabavi SM. Neuroprotective effects of honokiol: from chemistry to medicine. Biofactors 2017; 43:760-769. [PMID: 28817221 DOI: 10.1002/biof.1385] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/21/2017] [Accepted: 07/27/2017] [Indexed: 01/15/2023]
Abstract
The incidence of neurological disorders is growing in developed countries together with increased lifespan. Nowadays, there are still no effective treatments for neurodegenerative pathologies, which make necessary to search for new therapeutic agents. Natural products, most of them used in traditional medicine, are considered promising alternatives for the treatment of neurodegenerative diseases. Honokiol is a natural bioactive phenylpropanoid compound, belonging to the class of neolignan, found in notable amounts in the bark of Magnolia tree, and has been reported to exert diverse pharmacological properties including neuroprotective activities. Honokiol can permeate the blood brain barrier and the blood-cerebrospinal fluid to increase its bioavailability in neurological tissues. Diverse studies have provided evidence on the neuroprotective effect of honokiol in the central nervous system, due to its potent antioxidant activity, and amelioration of the excitotoxicity mainly related to the blockade of glutamate receptors and reduction in neuroinflammation. In addition, recent studies suggest that honokiol can attenuate neurotoxicity exerted by abnormally aggregated Aβ in Alzheimer's disease. The present work summarizes what is currently known concerning the neuroprotective effects of honokiol and its potential molecular mechanisms of action, which make it considered as a promising agent in the treatment and management of neurodegenerative diseases. © 2017 BioFactors, 43(6):760-769, 2017.
Collapse
Affiliation(s)
- Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin 20-093, Poland
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin 20-093, Poland
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), University of Balearic Islands, Balearic Islands, Spain
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Australia
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
71
|
Han SY, Kim J, Kim E, Kim SH, Seo DB, Kim JH, Shin SS, Cho JY. AKT-targeted anti-inflammatory activity of Panax ginseng calyx ethanolic extract. J Ginseng Res 2017; 42:496-503. [PMID: 30337810 PMCID: PMC6187086 DOI: 10.1016/j.jgr.2017.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
Background Korean ginseng (Panax ginseng) plays an anti-inflammatory role in a variety of inflammatory diseases such as gastritis, hepatitis, and colitis. However, inflammation-regulatory activity of the calyx of the P. ginseng berry has not been thoroughly evaluated. To understand whether the calyx portion of the P. ginseng berry is able to ameliorate inflammatory processes, an ethanolic extract of P. ginseng berry calyx (Pg-C-EE) was prepared, and lipopolysaccharide-activated macrophages and HEK293 cells transfected with inflammation-regulatory proteins were used to test the anti-inflammatory action of Pg-C-EE. Methods The ginsenoside contents of Pg-C-EE were analyzed by HPLC. Suppressive activity of Pg-C-EE on NO production, inflammatory gene expression, transcriptional activation, and inflammation signaling events were examined using the Griess assay, reverse transcription-polymerization chain reaction, luciferase activity reporter gene assay, and immunoblotting analysis. Results Pg-C-EE reduced NO production and diminished mRNA expression of inflammatory genes such as cyclooxygenase-2, inducible NO synthase, and tumor necrosis factor-α in a dose-dependent manner. This extract suppressed luciferase activity induced only by nuclear factor-κB. Interestingly, immunoblotting analysis results demonstrated that Pg-C-EE reduced the activities of protein kinase B (AKT)1 and AKT2. Conclusion These results suggest that Pg-C-EE may have nuclear-factor-κB-targeted anti-inflammatory properties through suppression of AKT. The calyx of the P. ginseng berry is an underused part of the ginseng plant, and development of calyx-derived extracts may be useful for treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Sang Yun Han
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Juewon Kim
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Eunji Kim
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Su Hwan Kim
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Dae Bang Seo
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Jong-Hoon Kim
- Department of Physiology, College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Song Seok Shin
- Vital Beautie Research Division, Amorepacific Research and Development Center, Suwon, Republic of Korea
| | - Jae Youl Cho
- Department of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|