1
|
Noureldeen ME, Shahin NN, Amin HAA, El-Sawalhi MM, Ghaiad HR. Parthenolide ameliorates 3-nitropropionic acid-induced Huntington's disease-like aberrations via modulating NLRP3 inflammasome, reducing microglial activation and inducing astrocyte shifting. Mol Med 2024; 30:158. [PMID: 39327568 PMCID: PMC11425901 DOI: 10.1186/s10020-024-00917-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is a progressive neurodegenerative disease that causes motor, cognitive, and psychiatric abnormalities, with no satisfying disease-modifying therapy so far. 3-nitropropionic acid (3NP) induces behavioural deficits, together with biochemical and histological alterations in animals' striata that mimic HD. The role of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome in HD pathogenesis remains largely uncharacterized. Parthenolide (PTL), a naturally occurring nuclear factor kappa B (NF-κB) inhibitor, is also known to inhibit NLRP3 inflammasome. Whether PTL is beneficial in HD has not been established yet. AIM This study evaluated the possible neuroprotective effects of PTL against 3NP-induced behavioural abnormalities, striatal biochemical derangements, and histological aberrations. METHODS Male Wistar rats received PTL (0.5 mg/kg/day, i.p) for 3 weeks and 3NP (10 mg/kg/day, i.p) was administered alongside for the latter 2 weeks to induce HD. Finally, animals were subjected to open-field, Morris water maze and rotarod tests. Rat striata were examined histologically, striatal protein expression levels of glial fibrillary acidic protein (GFAP), cluster of differentiation 45 (CD45) and neuron-specific enolase (NSE) were evaluated immunohistochemically, while those of interleukin (IL)-1β, IL-18, ionized calcium-binding adapter molecule-1 (Iba1) and glutamate were determined by ELISA. Striatal nuclear factor erythroid 2-related factor 2 (Nrf2), Kelch-like ECH-associated protein (Keap1), NF-κB, NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, S100 calcium-binding protein A10 (S100A10) and complement-3 (C3) were assessed by gene expression analysis. RESULTS PTL improved motor, locomotor, cognitive and anxiety-like behaviours, restored neuronal integrity, upregulated Nrf2, and inhibited NLRP3 inflammasome, NF-κB and microglial activation. Additionally, PTL induced astrocyte shifting towards the neuroprotective A2 phenotype. CONCLUSION PTL exhibits neuroprotection against 3NP-induced HD, that might be ascribed, at least in part, to its modulatory effects on Keap1/Nrf2 and NF-κB/NLRP3 inflammasome signaling.
Collapse
Affiliation(s)
- Mona E Noureldeen
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy St., Cairo, 11562, Egypt
| | - Nancy N Shahin
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy St., Cairo, 11562, Egypt
| | - Hebat Allah A Amin
- Pathology Department, Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Maha M El-Sawalhi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy St., Cairo, 11562, Egypt
| | - Heba R Ghaiad
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy St., Cairo, 11562, Egypt.
| |
Collapse
|
2
|
Purohit S, Oswal P, Bahuguna A, Tyagi A, Bhatt N, Kumar A. Catalytic system having an organotellurium ligand on graphene oxide: immobilization of Pd(0) nanoparticles and application in heterogeneous catalysis of cross-coupling reactions. RSC Adv 2024; 14:27092-27109. [PMID: 39193294 PMCID: PMC11348857 DOI: 10.1039/d4ra03401e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
First heterogeneous catalytic system, having a covalently linked hybrid bidentate organotellurium ligand [i.e., PhTe-CH2-CH2-NH2] on the surface of graphene oxide, has been synthesized with immobilized and stabilized Pd(0) nanoparticles. To the best of our knowledge, it is the first such catalytic system in which a heterogenized organotellurium ligand has been used. It has been well-characterized using different physicochemical characterization techniques viz. P-XRD, XPS, HR-TEM, EELS, FE-SEM, EDX, TGA, BET surface area analysis, FT-IR spectroscopy, and Raman spectroscopy. The Pd content of the final system has been quantified using ICP-OES. Its applications have been explored in Suzuki-Miyaura C-C cross coupling and C-O cross coupling reactions. Hot filtration experiments corroborate the heterogeneous nature of the catalysis. It is recyclable for up to five reaction cycles in Suzuki-Miyaura and C-O cross coupling with marginal loss in performance. It also catalyzes the reactions of chloroarenes such as chlorobenzene, 4-chloroaniline, 1-chloro-4-nitrobenzene, 4-chloroacetophenone, 4-chlorobenzophenone for Suzuki coupling, and 1-chloro-4-nitrobenzene, 4-chlorobenzonitrile, chlorobenzene, and 4-chlorotoluene for C-O coupling. P-XRD, FE-SEM, and EDX study reveals that the catalytic system retains its structural originality and functionality after recycling.
Collapse
Affiliation(s)
- Suraj Purohit
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| | - Preeti Oswal
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| | - Anurag Bahuguna
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| | - Anupma Tyagi
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| | - Neeraj Bhatt
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| | - Arun Kumar
- Department of Chemistry, School of Physical Sciences, Doon University Dehradun-248001 India
| |
Collapse
|
3
|
Rabie MA, Ghoneim AT, Fahmy MI, El-Yamany MF, Sayed RH. Activation of alpha-7 nicotinic acetylcholine receptor by tropisetron mitigates 3-nitropropionic acid-induced Huntington's disease in rats: Role of PI3K/Akt and JAK2/NF-κB signaling pathways. Chem Biol Interact 2024; 393:110957. [PMID: 38513929 DOI: 10.1016/j.cbi.2024.110957] [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: 01/30/2024] [Revised: 03/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Huntington's disease (HD) is an inheritable autosomal-dominant disorder that targets mainly the striatum. 3-Nitropropionic acid (3-NP) induces obvious deleterious behavioral, neurochemical, and histological effects similar to the symptoms of HD. Our study aimed to examine the neuroprotective activity of tropisetron, an alpha-7 neuronal nicotinic acetylcholine receptor (α-7nAChR) agonist, against neurotoxic events associated with 3-NP-induced HD in rats. Forty-eight rats were randomly allocated into four groups. Group I received normal saline, while Groups II, III and IV received 3-NP for 2 weeks. In addition, Group III and IV were treated with tropisetron 1 h after 3-NP administration. Meanwhile, Group IV received methyllycaconitine (MLA), an α-7nAChR antagonist, 30 min before tropisetron administration. Treatment with tropisetron improved motor deficits as confirmed by the behavioral tests and restored normal histopathological features of the striatum. Moreover, tropisetron showed an anti-oxidant activity via increasing the activities of SDH and HO-1 as well as Nrf2 expression along with reducing MDA level. Tropisetron also markedly upregulated the protein expression of p-PI3K and p-Akt which in turn hampered JAK2/NF-κB inflammatory cascade. In addition, tropisetron showed an anti-apoptotic activity through boosting the expression of Bcl-2 and reducing Bax expression and caspase-3 level. Interestingly, all the aforementioned effects of tropisetron were blocked by pre-administration of MLA, which confirms that such neuroprotective effects are mediated via activating of α-7nAChR. In conclusion, tropisetron showed a neuroprotective activity against 3-NP-induced HD via activating PI3K/Akt signaling and suppressing JAK2/NF-κB inflammatory axis. Thus, repositioning of tropisetron could represent a promising therapeutic strategy in management of HD.
Collapse
Affiliation(s)
- Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Ahmed T Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| | - Mohamed I Fahmy
- Department of Pharmacology and Toxicology, College of Pharmaceutical sciences and drug manufacturing, Misr University for Science and Technology (MUST), 12585, Giza, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt; School of Pharmacy, Newgiza University, Giza, Egypt
| |
Collapse
|
4
|
Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Bornhorst J, Cubadda F, Dopter A, FitzGerald R, de Sesmaisons Lecarré A, das Neves Ferreira P, Fabiani L, Horvath Z, Matijević L, Naska A. Scientific opinion on the tolerable upper intake level for manganese. EFSA J 2023; 21:e8413. [PMID: 38075631 PMCID: PMC10704406 DOI: 10.2903/j.efsa.2023.8413] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Following a request from the European Commission (EC), the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for manganese. Systematic reviews of the literature of human and animal data were conducted to assess evidence regarding excess manganese intake (including authorised manganese salts) and the priority adverse health effect, i.e. manganese-induced neurotoxicity. Available human and animal studies support neurotoxicity as a critical effect, however, data are not sufficient and suitable to characterise a dose-response relationship and identify a reference point for manganese-induced neurotoxicity. In the absence of adequate data to establish an UL, estimated background dietary intakes (i.e. manganese intakes from natural dietary sources only) observed among high consumers (95th percentile) were used to provide an indication of the highest level of intake where there is reasonable confidence on the absence of adverse effects. A safe level of intake of 8 mg/day was established for adults ≥ 18 years (including pregnant and lactating women) and ranged between 2 and 7 mg/day for other population groups. The application of the safe level of intake is more limited than an UL because the intake level at which the risk of adverse effects starts to increase is not defined.
Collapse
|
5
|
Funguetto-Ribeiro AC, Nakama KA, Pinz MP, Oliveira RLD, Sacramento MD, Pereira FSO, Pinton S, Wilhelm EA, Luchese C, Alves D, Ávila DS, Haas SE. Development and In Vivo Assessment of 4-Phenyltellanyl-7-chloroquinoline-loaded Polymeric Nanocapsules in Alzheimer's Disease Models. Brain Sci 2023; 13:999. [PMID: 37508931 PMCID: PMC10377448 DOI: 10.3390/brainsci13070999] [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: 05/26/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in older people, and available treatments are palliative and produce undesirable side effects. The 4-phenyltellanyl-7-chloroquinoline (TQ) is an organochalcogen compound studied due to its pharmacological properties, particularly its antioxidant potential. However, TQ possesses some drawbacks such as low aqueous solubility and high toxicity, thus warranting the search for tools that improve the safety and effectiveness of new compounds. Here, we developed and investigated the biological effects of TQ-loaded polymeric nanocapsules (NCTQ) in an AD model in transgenic Caenorhabditis elegans expressing human Aβ1-42 in their body-wall muscles and Swiss mice injected with Aβ25-35. The NCTQ displayed good physicochemical properties, including nanometer size and maximum encapsulation capacity. The treatment showed low toxicity, reduced Aβ peptide-induced paralysis, and activated an endoplasmic reticulum chaperone in the C. elegans model. The Aβ injection in mice caused memory impairment, which NCTQ mitigated by improving working, long-term, and aversive memory. Additionally, no changes in biochemical markers were evidenced in mice, demonstrating that there was no hepatotoxicity in the tested doses. Altogether, these findings provide insights into the neuroprotective effects of TQ and indicate that NCTQ is a promising candidate for AD treatment.
Collapse
Affiliation(s)
| | - Kelly Ayumi Nakama
- Pharmaceutical Science Graduate Program, Federal University of Pampa-UNIPAMPA, Uruguaiana 97501-970, Brazil
| | - Mikaela Peglow Pinz
- Biochemistry and Bioprospecting Graduate Program, Biochemical Pharmacology Research Laboratory (LaFarBio), Neurobiotechnology Research Group (GPN), Chemical, Pharmaceutical and Food Science Center (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | - Renata Leivas de Oliveira
- Biochemistry and Bioprospecting Graduate Program, Biochemical Pharmacology Research Laboratory (LaFarBio), Neurobiotechnology Research Group (GPN), Chemical, Pharmaceutical and Food Science Center (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | - Manoela do Sacramento
- Clean Organic Synthesis Laboratory (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | | | - Simone Pinton
- Biochemistry Graduate Program, Federal University of Pampa-UNIPAMPA, Uruguaiana 97501-970, Brazil
| | - Ethel Antunes Wilhelm
- Biochemistry and Bioprospecting Graduate Program, Biochemical Pharmacology Research Laboratory (LaFarBio), Neurobiotechnology Research Group (GPN), Chemical, Pharmaceutical and Food Science Center (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | - Cristiane Luchese
- Biochemistry and Bioprospecting Graduate Program, Biochemical Pharmacology Research Laboratory (LaFarBio), Neurobiotechnology Research Group (GPN), Chemical, Pharmaceutical and Food Science Center (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | - Diego Alves
- Clean Organic Synthesis Laboratory (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas-UFPel, Pelotas 96010-900, Brazil
| | - Daiana Silva Ávila
- Biochemistry Graduate Program, Federal University of Pampa-UNIPAMPA, Uruguaiana 97501-970, Brazil
| | - Sandra Elisa Haas
- Biochemistry Graduate Program, Federal University of Pampa-UNIPAMPA, Uruguaiana 97501-970, Brazil
- Pharmaceutical Science Graduate Program, Federal University of Pampa-UNIPAMPA, Uruguaiana 97501-970, Brazil
| |
Collapse
|
6
|
Pajarillo E, Nyarko-Danquah I, Digman A, Multani HK, Kim S, Gaspard P, Aschner M, Lee E. Mechanisms of manganese-induced neurotoxicity and the pursuit of neurotherapeutic strategies. Front Pharmacol 2022; 13:1011947. [PMID: 36605395 PMCID: PMC9808094 DOI: 10.3389/fphar.2022.1011947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/01/2022] [Indexed: 01/07/2023] Open
Abstract
Chronic exposure to elevated levels of manganese via occupational or environmental settings causes a neurological disorder known as manganism, resembling the symptoms of Parkinson's disease, such as motor deficits and cognitive impairment. Numerous studies have been conducted to characterize manganese's neurotoxicity mechanisms in search of effective therapeutics, including natural and synthetic compounds to treat manganese toxicity. Several potential molecular targets of manganese toxicity at the epigenetic and transcriptional levels have been identified recently, which may contribute to develop more precise and effective gene therapies. This review updates findings on manganese-induced neurotoxicity mechanisms on intracellular insults such as oxidative stress, inflammation, excitotoxicity, and mitophagy, as well as transcriptional dysregulations involving Yin Yang 1, RE1-silencing transcription factor, transcription factor EB, and nuclear factor erythroid 2-related factor 2 that could be targets of manganese neurotoxicity therapies. This review also features intracellular proteins such as PTEN-inducible kinase 1, parkin, sirtuins, leucine-rich repeat kinase 2, and α-synuclein, which are associated with manganese-induced dysregulation of autophagy/mitophagy. In addition, newer therapeutic approaches to treat manganese's neurotoxicity including natural and synthetic compounds modulating excitotoxicity, autophagy, and mitophagy, were reviewed. Taken together, in-depth mechanistic knowledge accompanied by advances in gene and drug delivery strategies will make significant progress in the development of reliable therapeutic interventions against manganese-induced neurotoxicity.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Ivan Nyarko-Danquah
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Alexis Digman
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Harpreet Kaur Multani
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL, United States
| | - Sanghoon Kim
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Patric Gaspard
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, NY, United States
| | - Eunsook Lee
- Department of Pharmaceutical Science, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, United States
| |
Collapse
|
7
|
Akingbade GT, Ijomone OM, Imam A, Aschner M, Ajao MS. D-Ribose-LCysteine attenuates manganese-induced cognitive and motor deficit, oxidative damage, and reactive microglia activation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 93:103872. [PMID: 35513219 DOI: 10.1016/j.etap.2022.103872] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 05/21/2023]
Abstract
Due to overexposure, manganese (Mn) accumulation in the brain can trigger the inhibition of glutathione synthesis and lead to increased generation of reactive oxygen species (ROS) and oxidative stress. D-Ribose-L-Cysteine (RibCys) has been demonstrated to effectively support glutathione synthesis to scavenge ROS and protect cells from oxidative damage. In the present study, we examined the effects of RibCys on weight changes, cognitive and motor associated activities, oxidative stress markers, striatal and cortical histology, and microglia activation following Mn exposure. Rats were exposed to either saline, Mn or/and RibCys for two weeks. The Mn exposed rats received RibCys either as pre-, co-, or post-treatments. Mn caused a significant decrease in weight, memory and motor activities, increased lactate dehydrogenase level, overexpression of IBA1 reflecting microglia activation, and distortion of the neuronal cytoarchitecture of the striatum and motor cortex, respectively. Interventions with RibCys mitigated Mn-induced neurotoxic events. Our novel study demonstrates that RibCys effectively ameliorates the neurotoxicity following Mn treatment and maybe a therapeutic strategy against the neurological consequences of Mn overexposurec.
Collapse
Affiliation(s)
- Grace T Akingbade
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria; The Neuro- Lab, Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Nigeria; Department of Molecular Pharmacology, Albert Einstein College of Medicine, NY, USA.
| | - Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Basic Medical Sciences, Federal University of Technology Akure, Nigeria; Department of Molecular Pharmacology, Albert Einstein College of Medicine, NY, USA
| | - Aminu Imam
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, NY, USA
| | - Moyosore S Ajao
- Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Nigeria.
| |
Collapse
|
8
|
Ibrahim WW, Abdel Rasheed NO. Diapocynin neuroprotective effects in 3-nitropropionic acid Huntington's disease model in rats: emphasis on Sirt1/Nrf2 signaling pathway. Inflammopharmacology 2022; 30:1745-1758. [PMID: 35639233 PMCID: PMC9499906 DOI: 10.1007/s10787-022-01004-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/30/2022] [Indexed: 04/12/2023]
Abstract
Background and Aim Huntington's disease (HD) is a rare inherited disease portrayed with marked cognitive and motor decline owing to extensive neurodegeneration. NADPH oxidase is considered as an important contributor to the oxidative injury in several neurodegenerative disorders including HD. Thus, the present study explored the possible neuroprotective effects of diapocynin, a specific NADPH oxidase inhibitor, against 3-nitropropionic acid (3-NP) model of HD in rats. Methods Animals received diapocynin (10 mg/kg/day, p.o), 30 min before 3-NP (10 mg/kg/day, i.p) over a period of 14 days. Results Diapocynin administration attenuated 3-NP-induced oxidative stress with significant increase in reduced glutathione, glutathione-S-transferase, nuclear factor erythroid 2-related factor 2, and brain-derived neurotrophic factor striatal contents contrary to NADPH oxidase (NOX2; gp91phox subunit) diminished expression. Moreover, diapocynin mitigated 3-NP-associated neuroinflammation
and glial activation with prominent downregulation of nuclear factor-Кβ p65 and marked decrement of inducible nitric oxide synthase content in addition to decreased immunoreactivity of ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein; markers of microglial and astroglial activation, respectively. Treatment with diapocynin hindered 3-NP-induced apoptosis with prominent decrease in tumor suppressor protein and Bcl-2-associated X protein contents whereas the anti-apoptotic marker; B-cell lymphoma-2 content was noticeably increased. Diapocynin neuroprotective effects could be attributed to silent information regulator 1 upregulation which curbed 3-NP-associated hazards resulting in improved motor functions witnessed during open field, rotarod, and grip strength tests as well as attenuated 3-NP-associated histopathological derangements. Conclusion The present findings indicated that diapocynin could serve as an auspicious nominee for HD management. Graphical abstract ![]()
Collapse
Affiliation(s)
- Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
| | - Nora O Abdel Rasheed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
9
|
Telmisartan neuroprotective effects in 3-nitropropionic acid Huntington's disease model in rats: Cross talk between PPAR-γ and PI3K/Akt/GSK-3β pathway. Life Sci 2022; 297:120480. [DOI: 10.1016/j.lfs.2022.120480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 11/18/2022]
|
10
|
Mustafa AM, Rabie MA, Zaki HF, Shaheen AM. Inhibition of Brain GTP Cyclohydrolase I Attenuates 3-Nitropropionic Acid-Induced Striatal Toxicity: Involvement of Mas Receptor/PI3k/Akt/CREB/ BDNF Axis. Front Pharmacol 2022; 12:740966. [PMID: 35002694 PMCID: PMC8727546 DOI: 10.3389/fphar.2021.740966] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
GTP cyclohydrolase I (GTPCH I) is the rate-limiting enzyme for tetrahydrobiopterin (BH4) biosynthesis; the latter is an essential factor for iNOS activation that contributes neuronal loss in Huntington’s disease (HD). The aim of the study was to investigate the neuroprotective effect of 2,4-diamino-6-hydroxypyrimidine (DAHP), GTPCH I enzyme inhibitor, against neuronal loss in 3-nitropropinic acid (3-NP)-induced HD in rats and to reveal the possible involved mechanisms mediated through PI3K/Akt axis and its correlation to Mas receptor (MasR). Rats received 3-NP (10 mg/kg/day; i.p.) with or without administration of DAHP (0.5 g/kg/day; i.p.) or wortmannin (WM), a PI3K inhibitor, (15 μg/kg/day; i.v.) for 14 days. DAHP improved cognitive, memory, and motor abnormalities induced by 3-NP, as confirmed by striatal histopathological specimens and immunohistochemical examination of GFAP. Moreover, DAHP treatment inhibited GTPCH I activity, resulting in decreased BH4 levels and iNOS activation. Also, DAHP upregulated the protein expression of survival protein; p85/p55 (pY458/199)-PI3K and pS473-Akt that, in turn, boosted the activation of striatal neurotrophic factors and receptor, pS133-CREB, BDNF and pY515-TrKB, which positively affect MasR protein expression and improve mitochondrial dysfunction, as indicated by enhancing both SDH and PGC-1α levels. Indeed, DAHP attenuates oxidative stress by increasing SOD activity and Nrf2 expression in addition to reducing neuro-inflammatory status by inhibiting NF-κB p65 and TNF-α expression. Interestingly, all the previous effects were blocked by co-administration of WM with DAHP. In conclusion, DAHP exerts neuroprotective effect against neuronal loss induced by 3-NP administration via inhibition of GTPCH I and iNOS activity and activation of MasR/PI3K/Akt/CREB/BDNF/TrKB axis besides its antioxidant and anti-inflammatory effect.
Collapse
Affiliation(s)
- Aya M Mustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aya M Shaheen
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
11
|
Rai SN, Singh P, Varshney R, Chaturvedi VK, Vamanu E, Singh MP, Singh BK. Promising drug targets and associated therapeutic interventions in Parkinson's disease. Neural Regen Res 2021; 16:1730-1739. [PMID: 33510062 PMCID: PMC8328771 DOI: 10.4103/1673-5374.306066] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is one of the most debilitating brain diseases. Despite the availability of symptomatic treatments, response towards the health of PD patients remains scarce. To fulfil the medical needs of the PD patients, an efficacious and etiological treatment is required. In this review, we have compiled the information covering limitations of current therapeutic options in PD, novel drug targets for PD, and finally, the role of some critical beneficial natural products to control the progression of PD.
Collapse
Affiliation(s)
| | - Payal Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ritu Varshney
- Department of Bioengineering and Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gujarat, India
| | | | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, Bucharest, Romania
| | - M. P. Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj, India
| | - Brijesh Kumar Singh
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
12
|
Capperucci A, Coronnello M, Salvini F, Tanini D, Dei S, Teodori E, Giovannelli L. Synthesis of functionalised organochalcogenides and in vitro evaluation of their antioxidant activity. Bioorg Chem 2021; 110:104812. [PMID: 33744808 DOI: 10.1016/j.bioorg.2021.104812] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/29/2021] [Accepted: 03/05/2021] [Indexed: 11/26/2022]
Abstract
Differently substituted β-hydroxy- and β-amino dialkyl and alkyl-aryl tellurides and selenides have been prepared through ring-opening reactions of epoxides and aziridines with selenium- or tellurium-centered nucleophiles. The antioxidant properties and the cytotoxicity of such compounds have been investigated on normal human dermal fibroblasts. Most of the studied compounds exhibited a low cytotoxicity and a number of them proved to be non-toxic, not showing any effect on cell viability even at the highest concentration used (100 μM). The obtained results showed a significant antioxidant potential of the selected organotellurium compounds, particularly evident under conditions of exogenously induced oxidative stress. The antioxidant activity of selenium-containing analogues of active tellurides has also been evaluated on cells, highlighting that the replacement of Se with Te brought about a significant increase in the peroxidase activity.
Collapse
Affiliation(s)
- Antonella Capperucci
- University of Florence, Department of Chemistry "Ugo Schiff", Via della Lastruccia 3-13, I-50019 Sesto Fiorentino (Florence), Italy
| | - Marcella Coronnello
- University of Florence, Department of Health Sciences - Section of Clinical Pharmacology and Oncology, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Francesca Salvini
- University of Florence, Department of Health Sciences - Section of Clinical Pharmacology and Oncology, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Damiano Tanini
- University of Florence, Department of Chemistry "Ugo Schiff", Via della Lastruccia 3-13, I-50019 Sesto Fiorentino (Florence), Italy.
| | - Silvia Dei
- University of Florence, Department of Neurosciences, Psychology, Drug Research and Child's Health - Section of Pharmaceutical and Nutraceutical Sciences, via Ugo Schiff 6, 50019 Sesto Fiorentino (FI), Italy.
| | - Elisabetta Teodori
- University of Florence, Department of Neurosciences, Psychology, Drug Research and Child's Health - Section of Pharmaceutical and Nutraceutical Sciences, via Ugo Schiff 6, 50019 Sesto Fiorentino (FI), Italy
| | - Lisa Giovannelli
- University of Florence, Department of Neurosciences, Psychology, Drug Research and Child's Health - Section of Pharmacology, Viale Pieraccini 6, 50139 Firenze, Italy
| |
Collapse
|
13
|
Amaral LDO, Lima VS, Soares SM, Bornhorst J, Lemos SS, Gatto CC, Burrow RA, Gubert P. Synthesis, structural characterization and evaluation of the chelating potential in C. elegans involving complexes of mercury (II) with Schiff bases derived from amino acids. J Organomet Chem 2020. [DOI: 10.1016/j.jorganchem.2020.121500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
Freeman DM, O'Neal R, Zhang Q, Bouwer EJ, Wang Z. Manganese-induced Parkinsonism in mice is reduced using a novel contaminated water sediment exposure model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103399. [PMID: 32380377 DOI: 10.1016/j.etap.2020.103399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 06/11/2023]
Abstract
Heavy metals enter the aquatic environment and accumulate within water sediments, but these metal-sediment interactions remain to be explored within toxicity studies. We developed an exposure model in mice that encapsulates the aquatic microenvironment of metals before exposure. Male and female C57/BL6 mice were exposed via their drinking water to manganese contaminated sediment (Sed_Mn) or to manganese without sediment interaction (Mn) for six weeks. Sediment interaction did not alter weekly manganese ingestion from water in males or females. We analyzed motor impairment, a common feature in manganese-induced Parkinsonism, using the beam traversal, cylinder, and accelerating rotarod tests. Sed_Mn mice performed better overall compared to Mn mice and males were more sensitive to manganese than females in both Sed_Mn and Mn treatment groups. Our study indicates that metal-sediment interactions may alter metal toxicity in mammals and introduces a new exposure model to test the toxicity of metal contaminants of drinking water.
Collapse
Affiliation(s)
- Dana M Freeman
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel O'Neal
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Qiang Zhang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Edward J Bouwer
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhibin Wang
- Department of Environmental Health & Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Sayed NH, Fathy N, Kortam MA, Rabie MA, Mohamed AF, Kamel AS. Vildagliptin Attenuates Huntington's Disease through Activation of GLP-1 Receptor/PI3K/Akt/BDNF Pathway in 3-Nitropropionic Acid Rat Model. Neurotherapeutics 2020; 17:252-268. [PMID: 31728850 PMCID: PMC7007456 DOI: 10.1007/s13311-019-00805-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vildagliptin (Vilda), a dipeptidyl peptidase-4 (DPP-4) inhibitor, has been highlighted as a promising therapeutic agent for neurodegenerative diseases as Alzheimer's and Parkinson's diseases. Vilda's effect is mostly linked to PI3K/Akt signaling in CNS. Moreover, PI3K/Akt activation reportedly enhanced survival and dampened progression of Huntington's disease (HD). However, Vilda's role in HD is yet to be elucidated. Thus, the aim of the study is to uncover the potentiality of Vilda in HD and unfold its link with PI3K/Akt pathway in 3-nitropropionic acid (3NP) rat model. Rats were randomly assigned into 4 groups; group 1 received saline, whereas, groups 2, 3 and 4 received 3NP (10 mg/kg/day; i.p.) for 14 days, concomitantly with Vilda (5 mg/kg/day; p.o.) in groups 3 and 4, and wortmannin (WM), a PI3K inhibitor, (15 μg/kg/day; i.v.) in group 4. Vilda improved cognitive and motor perturbations induced by 3NP, as confirmed by striatal histopathological specimens and immunohistochemical examination of GFAP. The molecular signaling of Vilda was estimated by elevation of GLP-1 level and protein expressions of survival proteins; p85/p55 (pY458/199)-PI3K, pS473-Akt. Together, it boosted striatal neurotrophic factors and receptor; pS133-CREB, BDNF, pY515-TrKB, which subsequently maintained mitochondrial integrity, as indicated by enhancing both SDH and COX activities, and the redox modulators; Sirt1, Nrf2. Such neuroprotection restored imbalance of neurotransmitters through increasing GABA and suppressing glutamate as well PDE10A. These effects were reversed by WM pre-administration. In conclusion, Vilda purveyed significant anti-Huntington effect which may be mediated, at least in part, via activation of GLP-1/PI3K/Akt pathway in 3NP rat model.
Collapse
Affiliation(s)
- Noha H Sayed
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt
| | - Nevine Fathy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt.
| | - Mona A Kortam
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Governorate, Giza, Egypt
| |
Collapse
|
16
|
Hou W, Zhou Y, Rui J, Bai R, Bhasin AK, Ruan BH. Design and synthesis of novel tellurodibenzoic acid compounds as kidney-type glutaminase (KGA) inhibitors. Bioorg Med Chem Lett 2019; 29:1673-1676. [DOI: 10.1016/j.bmcl.2019.04.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022]
|
17
|
Soares ATG, Rodrigues LBL, Salgueiro WG, Dal Forno AHDC, Rodrigues CF, Sacramento M, Franco J, Alves D, Oliveira RDP, Pinton S, Ávila DS. Organoselenotriazoles attenuate oxidative damage induced by mitochondrial dysfunction in mev-1 Caenorhabditis elegans mutants. J Trace Elem Med Biol 2019; 53:34-40. [PMID: 30910204 DOI: 10.1016/j.jtemb.2019.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 01/12/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
Organic selenium compounds have several pharmacological activities already described, as anti-inflammatory and antitumor activities, which have been attributed to their antioxidant effects. Because they are promising in pharmacology, the synthesis of these compounds has increased significantly. As many new molecules are synthesized the use of a simple model like Caenorhabditis elegans is highly advantageous for initial evaluation of the toxicity and therapeutic potential of these molecules. The objective of this study was to evaluate the toxicity and antioxidant capacity of a series of selenotriazoles compounds in C. elegans. The animals were exposed to the compounds in liquid medium for only 30 min at the first larval stage (L1). The compounds had no toxic effects at the concentrations tested. Treatment with selenotriazoles (10 μM) partially reversed the stress induced by the pesticide paraquat (1 mM). Se-Tz Ia compound partially increased the survival of worms treated with H2O2 (0.5 mM). The compounds also increased the longevity of mev-1 mutants, which have a reduced life span by the production of excessive reactive oxygen species (ROS) in the mitochondria caused by a mutation in complex II of the electron transport chain. In addition, the compounds reduced the levels of ROS determined by the fluorescent probe DCF-DA as well as also reduced catalase enzyme activity in these animals. Based on the results found, it is possible to conclude that the compounds have antioxidant activity mainly in oxidative stress condition generated by a mitochondrial dysfunction in C. elegans.
Collapse
Affiliation(s)
- Ana Thalita Gonçalves Soares
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil
| | - Luiz Brasil Lopes Rodrigues
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil
| | - Willian Goulart Salgueiro
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil
| | - Ana Helena de Castro Dal Forno
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil
| | - Cristiane Freitas Rodrigues
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil
| | - Manoela Sacramento
- Programa de Pós-Graduação em Química (PPGQ), Laboratório de Síntese Orgânica Limpa-LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil; Programa de Pós-Graduação em Biotecnologia (PPGB), Grupo de Pesquisa em Neurobiotecnologia-GPN, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeferson Franco
- Interdisciplinary Center for Biotechnology Research, CIPBIOTEC, Universidade Federal do Pampa, Campus São Gabriel, 97.300-000, São Gabriel, RS, Brazil
| | - Diego Alves
- Programa de Pós-Graduação em Química (PPGQ), Laboratório de Síntese Orgânica Limpa-LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, UFPel, Pelotas, RS, Brazil; Programa de Pós-Graduação em Biotecnologia (PPGB), Grupo de Pesquisa em Neurobiotecnologia-GPN, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Riva de Paula Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Simone Pinton
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| | - Daiana S Ávila
- Programa de Pós-Graduação em Bioquímica, Laboratório de Bioquímica e Toxicologia em Caenorhabditis elegans (GBTOXce), Universidade Federal do Pampa, UNIPAMPA, Uruguaiana, RS 97500-970, Brazil.
| |
Collapse
|
18
|
Effect of Alkaloid Extract from African Jointfir ( Gnetum africanum) Leaves on Manganese-Induced Toxicity in Drosophila melanogaster. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8952646. [PMID: 30693067 PMCID: PMC6332884 DOI: 10.1155/2018/8952646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/01/2018] [Indexed: 11/17/2022]
Abstract
Metal-induced toxicity in fruit fly (Drosophila melanogaster) is one of the established models for studying neurotoxicity and neurodegenerative diseases. Phytochemicals, especially alkaloids, have been reported to exhibit neuroprotection. Here, we assessed the protective effect of alkaloid extract from African Jointfir (Gnetum africanum) leaf on manganese- (Mn-) induced toxicity in wild type fruit fly. Flies were exposed to 10 mM Mn, the alkaloid extract and cotreatment of Mn plus extract, respectively. The survival rate and locomotor performance of the flies were assessed 5 days posttreatment, at which point the flies were homogenized and assayed for acetylcholinesterase (AChE) activity, nitric oxide (NO), and reactive oxygen species (ROS) levels. Results showed that the extract significantly reverted Mn-induced reduction in the survival rate and locomotor performance of the flies. Furthermore, the extract counteracted the Mn-induced elevation in AChE activity, NO, and ROS levels. The alkaloid extract of the African Jointfir leaf may hence be a source of useful phytochemicals for the development of novel therapies for the management of neurodegeneration.
Collapse
|
19
|
Al-Massri KF, Ahmed LA, El-Abhar HS. Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel-induced neuropathy in rats: Role of Notch1 receptor and JAK/STAT signaling pathway. Behav Brain Res 2018; 360:303-311. [PMID: 30543902 DOI: 10.1016/j.bbr.2018.12.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/12/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
Abstract
Peripheral neuropathy is a common adverse effect observed during the use of paclitaxel (PTX) as chemotherapy. The present investigation was directed to estimate the modulatory effect of bone marrow derived mesenchymal stem cells (BM-MSCs) on pregabalin (PGB) treatment in PTX-induced peripheral neuropathy. Neuropathic pain was induced in rats by injecting PTX (2 mg/kg, i.p) 4 times every other day. Rats were then treated with PGB (30 mg/kg/day, p.o.) for 21 days with or without a single intravenous administration of BM-MSCs. At the end of experiment, behavioral and motor abnormalities were assessed. Animals were then sacrificed for measurement of total antioxidant capacity (TAC), nerve growth factor (NGF), nuclear factor kappa B p65 (NF-κB p65), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and active caspase-3 in the sciatic nerve. Moreover, protein expressions of Notch1 receptor, phosphorylated Janus kinase 2 (p-JAK2), phosphorylated signal transducer and activator of transcription 3 (p-STAT3), and phosphorylated p38 mitogen-activated protein kinase (p-p38-MAPK) were estimated. Finally, histological examinations were performed to assess severity of sciatic nerve damage and for estimation of BM-MSCs homing. Combined PGB/BM-MSCs therapy provided an additional improvement toward reducing PTX-induced oxidative stress, neuro-inflammation, and apoptotic markers. Interestingly, BM-MSCs therapy effectively prevented motor impairment observed by PGB treatment. Combined therapy also induced a significant increase in cell homing and prevented PTX-induced sciatic nerve damage in histological examination. The present study highlights a significant role for BM-MSCs in enhancing treatment potential of PGB and reducing its motor side effects when used as therapy in the management of peripheral neuropathy.
Collapse
Affiliation(s)
- Khaled F Al-Massri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
20
|
El-Hady WM, Galal AAA. Neurotoxic Outcomes of Subchronic Manganese Chloride Exposure via Contaminated Water in Adult Male Rats and the Potential Benefits of Ebselen. Biol Trace Elem Res 2018. [PMID: 29516356 DOI: 10.1007/s12011-018-1291-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The neurological effects of manganese (Mn) exposure on adults consuming contaminated water remain unclear. Accordingly, the current experiment was planned to explore the neurotoxic consequences of subchronic Mn exposure via contaminated water and to examine whether ebselen (Ebs) improved these outcomes. Rats exposed to oral MnCl2 (50 mg/kg body weight) for 30 successive days exhibited reduced rearing and ambulation. Furthermore, Mn administration increased brain Mn concentrations and induced superoxide dismutase, catalase, and glutathione depletion. Mn administration also increased lipid peroxidation biomarker levels. Additionally, Mn increased interleukin1-β and prostaglandin E2 levels and altered caspase-3 and Bcl-2 expression. Mn intoxication also induced marked gliosis, numerous vacuolations, and disoriented and pyknotic Purkinje cells as well as marked vascular congestion in brain tissue. Meanwhile, intraperitoneal administration of Ebs (15 mg/kg body weight) to Mn-intoxicated rats improved the behavioral performance and oxidative damage as well as inflammatory, apoptotic, and histopathological changes. The above results indicate that Ebs alleviated Mn neurotoxicity via its antioxidant, anti-inflammatory, and anti-apoptotic activities. Therefore, Ebs could represent a promising agent in the prevention of Mn-induced neurotoxicity.
Collapse
Affiliation(s)
- Walaa M El-Hady
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Azza A A Galal
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| |
Collapse
|
21
|
Al-Massri KF, Ahmed LA, El-Abhar HS. Pregabalin and lacosamide ameliorate paclitaxel-induced peripheral neuropathy via inhibition of JAK/STAT signaling pathway and Notch-1 receptor. Neurochem Int 2018; 120:164-171. [PMID: 30118739 DOI: 10.1016/j.neuint.2018.08.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/22/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
Anticonvulsant drugs such as pregabalin (PGB) and lacosamide (LCM), exhibit potent analgesic effects in diabetic neuropathy; however, their possible role/mechanisms in paclitaxel (PTX)-induced peripheral neuropathy have not been elucidated, which is the aim of the present study. Neuropathic pain was induced in rats by injecting PTX (2 mg/kg, i. p) on days 0, 2, 4 and 6. Forty eight hours after the last dose of PTX, rats were treated orally with 30 mg/kg/day of either PGB or LCM for 21 days. Both therapies improved thermal hyperalgesia and cold allodynia induced by PTX. Interestingly, LCM therapy showed no motor impairment that was observed upon using PGB, as demonstrated using rotarod test. Treatment with PGB or LCM restored the sciatic nerve content of the depleted total antioxidant capacity (TAC) and nerve growth factor (NGF), and lessened the elevated contents of nuclear factor kappa B p65 (NF-kB p65), tumor necrosis factor-α (TNF-α), and active caspase-3. On the molecular level, the drugs reduced the protein expression of Notch1 receptor, phosphorylated p38 mitogen-activated protein kinase (p-p38-MAPK), and the trajectory interleukin-6/phosphorylated janus kinase 2/phosphorylated signal transducer and activator of transcription 3 (IL-6/p-JAK2/p-STAT3). Therefore, the current study demonstrated a pivotal role for LCM in the management of PTX-induced peripheral neuropathy similar to PGB, but without motor adverse effects via the inhibition of oxidative stress, inflammation and apoptosis, as well as IL-6/JAK/STAT pathway and Notch1 receptor over-expression.
Collapse
Affiliation(s)
- Khaled F Al-Massri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
22
|
Gomes Silva AP, da Silva Araujo Santiago M, Maranho LA, de Oliveira RP, Constantino DHJ, Pereira CDS, da Silva RCB, Perobelli JE. Could male reproductive system be the main target of subchronic exposure to manganese in adult animals? Toxicology 2018; 409:1-12. [PMID: 29990519 DOI: 10.1016/j.tox.2018.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/28/2018] [Accepted: 07/05/2018] [Indexed: 01/28/2023]
Abstract
Manganese (Mn) is one of the most common chemical elements on Earth and an essential micronutrient in animal organism. However, in supraphysiological levels and long-term exposures, it is a potential toxicant. Although nervous system is the most studied in relation to Mn toxicity, other tissues can have their function impaired by Mn in high doses. The present study investigated the possible adverse effects of subchronic exposure to supraphysiologic level of Mn (5 mg/kg or 15 mg/kg, intraperitoneally) on reproductive, neurobehavioral, renal and hepatic parameters of male rats. For the first time, the vulnerability of these parameters to Mn was concomitantly investigated. While our results demonstrate that Mn treatments were not sufficient to produce a marked effect of neurotoxic, hepatotoxic or renal toxicity in adult rats, we found typical indicators of reproductive toxicity such as histopathological changes (major in testes and epididymis) and impaired sperm concentration and quality. Mn, under these experimental conditions, seems to exert reproductive toxicity by different testicular mechanisms, i.e. direct and indirect action on germ cells. On the other hand, exposure to Mn did not change the pattern of cognitive and emotional behaviors and the histological organization of kidneys of experimental rats. The liver showed a weight increasement and hidropic degeneration, probable due to the detoxification overload. In summary, for the first time it was demonstrated that adult male reproductive system was more sensitive to Mn toxicity than nervous, hepatic and renal systems, although nervous system is known as the main target tissue of this metal.
Collapse
Affiliation(s)
- Ana Priscila Gomes Silva
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | - Marcella da Silva Araujo Santiago
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | - Luciane Alves Maranho
- Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | - Rodolpho Pereira de Oliveira
- Laboratório de Psicobiologia da Esquizofrenia, Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | | | - Camilo Dias Seabra Pereira
- Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | - Regina Cláudia Barbosa da Silva
- Laboratório de Psicobiologia da Esquizofrenia, Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| | - Juliana Elaine Perobelli
- Laboratório de Toxicologia Experimental-LATOEX, Departamento de Ciências do Mar, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, SP, Brazil.
| |
Collapse
|
23
|
Shalaby HN, El-Tanbouly DM, Zaki HF. Topiramate mitigates 3-nitropropionic acid-induced striatal neurotoxicity via modulation of AMPA receptors. Food Chem Toxicol 2018; 118:227-234. [PMID: 29753867 DOI: 10.1016/j.fct.2018.05.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/01/2018] [Accepted: 05/08/2018] [Indexed: 01/10/2023]
Abstract
Prevalence of glutamate receptor subunit 2 (GluR2)-lacking alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors is a hallmark of excitotoxicity-related neurodegenerative diseases. Topiramate (TPM) is a structurally novel anticonvulsant with a well-known modulatory effects on AMPA/kainate subtypes of glutamate receptors. The present study aimed at investigating the neuroprotective potential of TPM on 3-nitropropionic acid (3-NP)-induced striatal neurodegeneration and Huntington's disease-like symptoms. Rats were injected with 3-NP (10 mg/kg/i.p.) for 14 days. TPM (50 mg/kg/p.o.) was given once a day, 1 h before 3-NP. TPM amended 3-NP induced changes in neurobehavioral performance, striatal neurotransmitters levels and histopathological injury. 3-NP control rats showed a significant ablation in the mRNA expression of Ca2+-impermeable Glu2R subunit along with an elevation in its regulatory protein (protein interacting with C kinase-1) PICK1, an effect that was largely reversed by TPM. TPM in addition, enhanced the phosphorylation of the protein kinase B/glycogen synthase kinase-3β/cAMP response element binding protein (Akt/GSK-3β/CREB) cue. Moreover, improvement in oxidative status, suppression of caspase-3 activity and restoration of striatal BDNF were noticed following treatment with TPM. The current study revealed that TPM boosted the neuroprotective (Akt/GSK-3β/CREB) pathway by its negative modulatory effect on AMPA glutamate receptors as well as its direct antioxidant property.
Collapse
Affiliation(s)
- Heba N Shalaby
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalia M El-Tanbouly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
24
|
Protective effect of vinpocetine against neurotoxicity of manganese in adult male rats. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:729-742. [PMID: 29671021 DOI: 10.1007/s00210-018-1498-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/05/2018] [Indexed: 01/10/2023]
Abstract
Manganese (Mn) is required for many essential biological processes as well as in the development and functioning of the brain. Extensive accumulation of Mn in the brain may cause central nervous system dysfunction known as manganism, a motor disorder associated with cognitive and neuropsychiatric deficits similar to parkinsonism. Vinpocetine, a synthetic derivative of the alkaloid vincamine, is used to improve the cognitive function in cerebrovascular diseases. It possesses antioxidant and antiinflammatory properties. The present work was designed to explore the potential neuroprotective mechanisms exerted by vinpocetine in the Mn-induced neurotoxicity in rats. Rats were allocated into four groups. First group was given saline. The other three groups were given MnCl2; two of them were treated with either L-dopa, the gold standard antiparkinsonian drug, or vinpocetine. Rats receiving MnCl2 exhibited lengthened catalepsy duration in the grid and bar tests, motor impairment in the open-field test and short-term memory deficit in the Y-maze test. Additionally, histological examination revealed structural alterations and degeneration in different brain regions. Besides, striatal monoamines and mitochondrial complex I contents were declined, apoptotic biomarker caspase-3 expression and acetylcholinesterase activity were elevated. Moreover, oxidative stress and inflammation were detected in the striata. L-dopa or vinpocetine exerted protective effects against MnCl2-induced neurotoxicity. It could be hypothesized that modulation of monoamines, upregulation of mitochondrial complex I, antioxidant, antiinflammatory, and antiapoptotic activities are significant mechanisms underlying the neuroprotective effect of vinpocetine in the Mn-induced neurotoxicity model in rats.
Collapse
|
25
|
Peres TV, Ong LK, Costa AP, Eyng H, Venske DKR, Colle D, Gonçalves FM, Lopes MW, Farina M, Aschner M, Dickson PW, Dunkley PR, Leal RB. Tyrosine hydroxylase regulation in adult rat striatum following short-term neonatal exposure to manganese. Metallomics 2017; 8:597-604. [PMID: 26790482 DOI: 10.1039/c5mt00265f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Manganese (Mn) is an essential trace element required for a range of physiological processes, but Mn can also be neurotoxic especially during development. Excess levels of Mn accumulate preferentially in the striatum and can induce a syndrome called manganism, characterized by an initial stage of psychiatric disorder followed by motor impairment. In the present study, we investigated the effects of Mn exposure on the developing dopaminergic system, specifically tyrosine hydroxylase (TH) protein and phosphorylation levels in the striatum of rats. Neonatal rats were exposed to Mn intraperitoneally (ip) from post-natal day 8 up to day 12 (PND8-12). Striatal tissue was analysed on PND14 or PND70, to detect either short-term or long-term effects induced by Mn exposure. There was a dose dependent increase in TH protein levels in the striatum at PND14, reaching significance at 20 mg kg(-1) Mn, and this correlated with an increase in TH phosphorylation at serines 40, 31 and 19. However, in the striatum at PND70, a time by which Mn levels were no longer elevated, there was a dose dependent decrease in TH protein levels, reaching significance at 20 mg kg(-1) Mn, and this correlated with TH phosphorylation at Ser40 and Ser19. There was however a significant increase in phosphorylation of TH at serine 31 at 20 mg kg(-1) Mn, which did not correlate with TH protein levels. Taken together our findings suggest that neonatal Mn exposure can have both short-term and long-term effects on the regulation of TH in the striatal dopaminergic system.
Collapse
Affiliation(s)
- Tanara V Peres
- Programa de Pós-graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil. and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lin K Ong
- School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Ana Paula Costa
- Programa de Pós-graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Helena Eyng
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Débora K R Venske
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Dirleise Colle
- Programa de Pós-graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Filipe M Gonçalves
- Programa de Pós-graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Mark W Lopes
- Programa de Pós-graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Marcelo Farina
- Programa de Pós-graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil. and Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil and Programa de Pós-graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Phillip W Dickson
- School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Peter R Dunkley
- School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Rodrigo B Leal
- Programa de Pós-graduação em Neurociências, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil. and Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil and Programa de Pós-graduação em Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
26
|
Caracelli I, Maganhi SH, de Oliveira Cardoso J, Cunha RL, Vega-Teijido MA, Zukerman-Schpector J, Tiekink ER. Crystallographic and docking (Cathepsins B, K, L and S) studies on bioactive halotelluroxetanes. Z KRIST-CRYST MATER 2017. [DOI: 10.1515/zkri-2017-2079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The molecular structures of the halotelluroxetanes p-MeOC6H4Te(X)[C(=C(H)X′)C(CH2)nO], X=X′=Cl and n=6 (1) and X=Cl, X′=Br and n=5 (4), show similar binuclear aggregates sustained by {· · ·Te–O}2 cores comprising covalent Te–O and secondary Te· · ·O interactions. The resulting C2ClO2(lone-pair) sets define pseudo-octahedral geometries. In each structure, C–X· · ·π(arene) interactions lead to supramolecular layers. Literature studies have shown these and related compounds (i.e. 2: X=X′=Cl and n=5; 3: X=X′=Br and n=5) to inhibit Cathepsins B, K, L and S to varying extents. Molecular docking calculations have been conducted on ligands (i.e. cations derived by removal of the tellurium-bound X atoms) 1′–3′ (note 3′=4′) enabling correlations between affinity for sub-sites and inhibition. The common feature of all docked complexes was the formation of a Te–S covalent bond with cysteine residues, the relative stability of the ligands with an E-configuration and the formation of a C–O· · ·π interaction with the phenyl ring; for 1′ the Te–S covalent bond was weak, a result correlating with its low inhibition profile. At the next level differences are apparent, especially with respect to the interactions formed by the organic-ligand-bound halides. While these atoms do not form specific interactions in Cathepsins B and K, in Cathepsin L, these halides are involved in C–O· · ·X halogen bonds.
Collapse
Affiliation(s)
- Ignez Caracelli
- BioMat, Departamento de Física , Universidade Federal de São Carlos , C.P. 676 , São Carlos, SP, 13565-905 , Brazil
| | - Stella H. Maganhi
- BioMat, Programa de Pós-graduação em Biotecnologia , Universidade Federal de São Carlos , C.P. 676 , São Carlos, SP, 13565-905 , Brazil
| | - Josiane de Oliveira Cardoso
- BioMat, Departamento de Física , Universidade Federal de São Carlos , C.P. 676 , São Carlos, SP, 13565-905 , Brazil
| | - Rodrigo L.O.R. Cunha
- Center of Natural Sciences and Humanities, Federal University of ABC , Santo André, São Paulo 09210-180 , Brazil
| | - Mauricio Angel Vega-Teijido
- Laboratório de Cristalografia, Estereodinâmica e Modelagem Molecular , Departamento de Química , Universidade Federal de São Carlos , C.P. 676 , São Carlos, SP, 13565-905 , Brazil
| | - Julio Zukerman-Schpector
- Laboratório de Cristalografia, Estereodinâmica e Modelagem Molecular , Departamento de Química , Universidade Federal de São Carlos , C.P. 676 , São Carlos, SP, 13565-905 , Brazil
| | - Edward R.T. Tiekink
- Research Centre for Crystalline Materials, School of Science and Technology , Sunway University , 47500 Bandar Sunway , Selangor Darul Ehsan , Malaysia
| |
Collapse
|
27
|
Chi L, Gao B, Bian X, Tu P, Ru H, Lu K. Manganese-induced sex-specific gut microbiome perturbations in C57BL/6 mice. Toxicol Appl Pharmacol 2017; 331:142-153. [PMID: 28610994 PMCID: PMC5653225 DOI: 10.1016/j.taap.2017.06.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 06/07/2017] [Accepted: 06/10/2017] [Indexed: 12/26/2022]
Abstract
Overexposure to manganese (Mn) leads to toxic effects, such as promoting the development of Parkinson's-like neurological disorders. The gut microbiome is deeply involved in immune development, host metabolism, and xenobiotics biotransformation, and significantly influences central nervous system (CNS) via the gut-brain axis, i.e. the biochemical signaling between the gastrointestinal tract and the CNS. However, it remains unclear whether Mn can affect the gut microbiome and its metabolic functions, particularly those linked to neurotoxicity. In addition, sex-specific effects of Mn have been reported, with no mechanism being identified yet. Recently, we have shown that the gut microbiome is largely different between males and females, raising the possibility that differential gut microbiome responses may contribute to sex-selective toxicity of Mn. Here, we applied high-throughput sequencing and gas chromatography-mass spectrometry (GC-MS) metabolomics to explore how Mn2+ exposure affects the gut microbiome and its metabolism in C57BL/6 mice. Mn2+ exposure perturbed the gut bacterial compositions, functional genes and fecal metabolomes in a highly sex-specific manner. In particular, bacterial genes and/or key metabolites of neurotransmitter synthesis and pro-inflammatory mediators are significantly altered by Mn2+ exposure, which can potentially affect chemical signaling of gut-brain interactions. Likewise, functional genes involved in iron homeostasis, flagellar motility, quorum sensing, and Mn transportation/oxidation are also widely changed by Mn2+ exposure. Taken together, this study has demonstrated that Mn2+ exposure perturbs the gut microbiome and its metabolic functions, which highlights the potential role of the gut microbiome in Mn2+ toxicity, particularly its sex-specific toxic effects.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States
| | - Bei Gao
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, United States
| | - Xiaoming Bian
- Department of Environmental Health Science, University of Georgia, Athens, GA 30602, United States
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States
| | - Hongyu Ru
- Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC 27607, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, 27599, United States.
| |
Collapse
|
28
|
Marreilha dos Santos AP, Andrade V, Aschner M. Neuroprotective and Therapeutic Strategies for Manganese-Induced Neurotoxicity. CLINICAL PHARMACOLOGY AND TRANSLATIONAL MEDICINE 2017; 1:54-62. [PMID: 30854510 PMCID: PMC6402347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Manganese (Mn) is an essential element required for growth, development and general maintenance of health. However, chronic or high occupational and environmental exposure to excessive levels of Mn has long been known to lead to a progressive neurological disorder similar to Parkinsonism. Manganism patients display a variety of symptoms, including mental, cognitive and behavioural impediments, as well as motor dysfunctions that are associated with basal ganglia dysfunction. Taking into account the pharmacokinetics and Mn-related toxicity mechanisms, several neuroprotective compounds and therapeutic approaches have been investigated to assess their efficacy in mitigating its neurotoxicity. Here, we will briefly address some of the toxic mechanisms of Mn, followed by neuroprotective strategies and therapeutic approaches aiming to reduce or treat Mn induced neurotoxicity. Natural and synthetic antioxidants, anti-inflammatory compounds, ATP/ADP ratio protectors and glutamate protectors have been introduced in view of decreasing Mn-induced neurotoxicity. In addition, the efficacy and mechanisms of several therapeutic interventions such as levodopa, ethylene-diamine-tetraacetic acid (EDTA) and para-aminosalicylic acid (PAS), aimed at ameliorating Mn neurotoxic symptoms in humans, will be reviewed.
Collapse
Affiliation(s)
- AP Marreilha dos Santos
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - V Andrade
- Institute of Medicine Research (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon. Portugal
| | - M Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, USA
| |
Collapse
|
29
|
Su C, Chen K, Zou Y, Shen Y, Xia B, Liang G, Lv Y, Wang F, Huang D, Yang X. Chronic exposure to manganese sulfate leads to adverse dose-dependent effects on the neurobehavioral ability of rats. ENVIRONMENTAL TOXICOLOGY 2016; 31:1571-1579. [PMID: 26097037 DOI: 10.1002/tox.22161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 05/31/2015] [Accepted: 05/31/2015] [Indexed: 06/04/2023]
Abstract
Manganese sulfate is the main combustion product of methylcyclopentadienyl manganese tricarbonyl (MMT). Currently, little is known about the neurobehavioral consequences of chronic manganese sulfate exposure. In this study, rats were treated with 0, 5.0, 10.0, and 20.0 mg/kg MnSO4 ·H2 O for 24 consecutive weeks via intraperitoneal injection. During the treatment period, spatial learning-memory ability was measured using the Morris water maze (MWM). At the end of the exposure period, spontaneous motor behavior and emotional status, hippocampal histologic changes, and Hsp70 mRNA levels were measured using the open-field test (OFT), hematoxylin-eosin staining and real-time quantitative PCR (RT-PCR), respectively. A dose-dependent decrease was noted in the spatial learning-memory ability and the spontaneous activities of rats (P < 0.05), and negative emotions differed significantly between the exposed groups and the control group (P < 0.05). Moreover, overt morphological changes in the hippocampuses of the exposed rats were detected. Cellular degeneration and death were also found. The Hsp70 mRNA levels of the hippocampal areas in the 20.0 mg/kg group (1.567 ± 0.236) were significantly increased compared with the control group (P < 0.05). These results suggest that chronic exposure to manganese sulfate can have adverse dose-dependent effects on rats' neurobehavioral ability, and the mechanism of abnormal hippocampal Hsp70 expression needs to be further explored. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1571-1579, 2016.
Collapse
Affiliation(s)
- Cheng Su
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Kangcheng Chen
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Yunfeng Zou
- Department of Toxicology, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Yuefei Shen
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bing Xia
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Guiqiang Liang
- Department of Toxicology, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Yingnan Lv
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Fenfen Wang
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Damin Huang
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, Guangxi Medical University, School of Public Health, Nanning, Guangxi, China.
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
30
|
Parmalee NL, Aschner M. Manganese and aging. Neurotoxicology 2016; 56:262-268. [PMID: 27293182 DOI: 10.1016/j.neuro.2016.06.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/04/2016] [Accepted: 06/05/2016] [Indexed: 12/11/2022]
Abstract
Manganese (Mn) is an essential metal that is required as a cofactor for many enzymes and is necessary for optimal biological function. Mn is abundant in the earth's crust and is present in soil and well water. Mn is also found in industrial settings, including mining, welding, and battery manufacture. Mn is also present in infant formula, parenteral nutrition, as well as pesticides and gasoline additives. A sufficient amount of Mn is obtained from most diets, and Mn deficiency is exceedingly rare. Excessive exposure to Mn in high doses can result in a condition known as manganism that results in psychological and emotional disturbances and motor symptoms that are reminiscent of Parkinson's disease, including gait disturbance, tremor, rigidity, and bradykinesia. Treatment for manganism is to remove the patient from Mn exposure, though symptoms are generally irreversible. The effects of exposure to Mn at lower doses are less clear. Little work has been done to evaluate the effects of chronic exposure to subclinical levels of Mn, especially in regard to lifelong exposures and the effects on the aging process. Mn is known to have effects on some of the same mechanistic processes that are altered in aging. This review will describe the general effects of Mn exposure and will focus on how Mn may be related to some of the mechanism of aging: neurogenesis, oxidative stress, and microglial activation and inflammation.
Collapse
Affiliation(s)
- Nancy L Parmalee
- Albert Einstein College of Medicine, Department of Molecular Pharmacology, 1300 Morris Park Avenue, Bronx, NY, United States.
| | - Michael Aschner
- Albert Einstein College of Medicine, Department of Molecular Pharmacology, 1300 Morris Park Avenue, Bronx, NY, United States.
| |
Collapse
|
31
|
Ahmed LA, Darwish HA, Abdelsalam RM, Amin HA. Role of Rho Kinase Inhibition in the Protective Effect of Fasudil and Simvastatin Against 3-Nitropropionic Acid-Induced Striatal Neurodegeneration and Mitochondrial Dysfunction in Rats. Mol Neurobiol 2015; 53:3927-3938. [DOI: 10.1007/s12035-015-9303-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022]
|
32
|
Trindade C, Juchem ALM, de Albuquerque NRM, de Oliveira IM, Rosa RM, Guecheva TN, Saffi J, Henriques JAP. Antigenotoxic and antimutagenic effects of diphenyl ditelluride against several known mutagens in Chinese hamster lung fibroblasts. Mutagenesis 2015; 30:799-809. [DOI: 10.1093/mutage/gev037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
33
|
Chen P, Chakraborty S, Peres TV, Bowman AB, Aschner M. Manganese-induced Neurotoxicity: From C. elegans to Humans. Toxicol Res (Camb) 2014; 4:191-202. [PMID: 25893090 DOI: 10.1039/c4tx00127c] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Manganese (Mn) is one of the most abundant metals on the earth. It is required for normal cellular activities, but overexposure leads to toxicity. Neurons are more susceptible to Mn-induced toxicity than other cells, and accumulation of Mn in the brain results in Manganism that presents with Parkinson's disease (PD)-like symptoms. In the last decade, a number of Mn transporters have been identified, which improves our understanding of Mn transport in and out of cells. However, the mechanism of Mn-induced neurotoxicity is only partially uncovered, with further research needed to explore the whole picture of Mn-induced toxicity. In this review, we will address recent progress in Mn-induced neurotoxicity from C. elegans to humans, and explore future directions that will help understand the mechanisms of its neurotoxicity.
Collapse
Affiliation(s)
- Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sudipta Chakraborty
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tanara V Peres
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA ; Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt University Medical Center, Nashville TN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
34
|
Comparsi B, Meinerz DF, Dalla Corte CL, Prestes AS, Stefanello ST, Santos DB, Souza DD, Farina M, Dafre AL, Posser T, Franco JL, Rocha JBT. N-acetylcysteine does not protect behavioral and biochemical toxicological effect after acute exposure of diphenyl ditelluride. Toxicol Mech Methods 2014; 24:529-35. [DOI: 10.3109/15376516.2014.920449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
35
|
Signaling mechanisms and disrupted cytoskeleton in the diphenyl ditelluride neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:458601. [PMID: 25050142 PMCID: PMC4090446 DOI: 10.1155/2014/458601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/26/2014] [Indexed: 01/14/2023]
Abstract
Evidence from our group supports that diphenyl ditelluride (PhTe)2 neurotoxicity depends on modulation of signaling pathways initiated at the plasma membrane. The (PhTe)2-evoked signal is transduced downstream of voltage-dependent Ca2+ channels (VDCC), N-methyl-D-aspartate receptors (NMDA), or metabotropic glutamate receptors activation via different kinase pathways (protein kinase A, phospholipase C/protein kinase C, mitogen-activated protein kinases (MAPKs), and Akt signaling pathway). Among the most relevant cues of misregulated signaling mechanisms evoked by (PhTe)2 is the cytoskeleton of neural cells. The in vivo and in vitro exposure to (PhTe)2 induce hyperphosphorylation/hypophosphorylation of neuronal and glial intermediate filament (IF) proteins (neurofilaments and glial fibrillary acidic protein, resp.) in different brain structures of young rats. Phosphorylation of IFs at specific sites modulates their association/disassociation and interferes with important physiological roles, such as axonal transport. Disrupted cytoskeleton is a crucial marker of neurodegeneration and is associated with reactive astrogliosis and apoptotic cell death. This review focuses the current knowledge and important results on the mechanisms of (PhTe)2 neurotoxicity with special emphasis on the cytoskeletal proteins and their differential regulation by kinases/phosphatases and Ca2+-mediated mechanisms in developmental rat brain. We propose that the disrupted cytoskeletal homeostasis could support brain damage provoked by this neurotoxicant.
Collapse
|
36
|
Manganese-induced atypical parkinsonism is associated with altered Basal Ganglia activity and changes in tissue levels of monoamines in the rat. PLoS One 2014; 9:e98952. [PMID: 24896650 PMCID: PMC4045849 DOI: 10.1371/journal.pone.0098952] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/09/2014] [Indexed: 01/08/2023] Open
Abstract
Manganese neurotoxicity is associated with motor and cognitive disturbances known as Manganism. However, the mechanisms underlying these deficits remain unknown. Here we investigated the effects of manganese intoxication on motor and non-motor parkinsonian-like deficits such as locomotor activity, motor coordination, anxiety and “depressive-like” behaviors. Then, we studied the impact of this intoxication on the neuronal activity, the globus pallidus (GP) and subthalamic nucleus (STN). At the end of experiments, post-mortem tissue level of the three monoamines (dopamine, norepinephrine and serotonin) has been determined. The experiments were carried out in adult Sprague-Dawley rats, daily treated with MnCl2 (10 mg/kg/, i.p.) for 5 weeks. We show that manganese progressively reduced locomotor activity as well as motor coordination in parallel with the manifestation of anxiety and “depressive-like” behaviors. Electrophysiological results show that, while majority of GP and STN neurons discharged regularly in controls, manganese increased the number of GP and STN neurons discharging irregularly and/or with bursts. Biochemical results show that manganese significantly decreased tissue levels of norepinephrine and serotonin with increased metabolism of dopamine in the striatum. Our data provide evidence that manganese intoxication is associated with impaired neurotransmission of monoaminergic systems, which is at the origin of changes in basal ganglia neuronal activity and the manifestation of motor and non-motor deficits similar to those observed in atypical Parkinsonism.
Collapse
|
37
|
Yang X, Bao Y, Fu H, Li L, Ren T, Yu X. Selenium protects neonates against neurotoxicity from prenatal exposure to manganese. PLoS One 2014; 9:e86611. [PMID: 24466170 PMCID: PMC3899298 DOI: 10.1371/journal.pone.0086611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 12/13/2013] [Indexed: 12/11/2022] Open
Abstract
Manganese (Mn) exposure can affect brain development. Whether Selenium (Se) can protect neonates against neurotoxicity from Mn exposure remains unclear. We investigated this issue in 933 mother-newborn pairs in Shanghai, China, from 2008 through 2009. Umbilical cord serum concentrations of Mn and Se were measured and Neonatal Behavioral Neurological Assessment (NBNA) tests were conducted. The scores <37 were defined as the low NBNA. The median concentrations of cord serum Mn and Se were 4.0 µg/L and 63.1 µg/L, respectively. After adjusting for potential confounders, the interaction between Se and Mn was observed. Cord blood Mn levels had different effects on NBNA scores stratified by different cord blood Se levels. With Se
Collapse
Affiliation(s)
- Xin Yang
- MOE-Shanghai Key Lab of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - YiXiao Bao
- Department of Pediatrics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - HuanHuan Fu
- MOE-Shanghai Key Lab of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - LuanLuan Li
- MOE-Shanghai Key Lab of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - TianHong Ren
- MOE-Shanghai Key Lab of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - XiaoDan Yu
- MOE-Shanghai Key Lab of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Wollenhaupt SGN, Soares AT, Salgueiro WG, Noremberg S, Reis G, Viana C, Gubert P, Soares FA, Affeldt RF, Lüdtke DS, Santos FW, Denardin CC, Aschner M, Avila DS. Seleno- and telluro-xylofuranosides attenuate Mn-induced toxicity in C. elegans via the DAF-16/FOXO pathway. Food Chem Toxicol 2013; 64:192-9. [PMID: 24296137 DOI: 10.1016/j.fct.2013.11.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 11/04/2013] [Accepted: 11/22/2013] [Indexed: 01/12/2023]
Abstract
Organochalcogens are promising pharmacological agents that possess significant biological activities. Nevertheless, because of the complexity of mammalian models, it has been difficult to determine the molecular pathways and specific proteins that are modulated in response to treatments with these compounds. The nematode worm Caenorhabditis elegans is an alternative experimental model that affords easy genetic manipulations, green fluorescent protein tagging and in vivo live analysis of toxicity. Abundant evidence points to oxidative stress in mediating manganese (Mn)-induced toxicity. In this study we challenged worms with Mn, and investigated the efficacy of inedited selenium- and tellurium-xylofuranosides in reversing and/or protecting the worms from Mn-induced toxicity. In addition, we investigated their putative mechanism of action. First, we determined the lethal dose 50% (LD50) and the effects of the xylofuranosides on various toxic parameters. This was followed by studies on the ability of xylofuranosides to afford protection against Mn-induced toxicity. Both Se- and Te-xylofuranosides increased the expression of superoxide dismutase (SOD-3). Furthermore, we observed that the xylofuranosides induced nuclear translocation of the transcription factor DAF-16/FOXO, which in the worm is known to regulate stress responsiveness, aging and metabolism. These findings suggest that xylofuranosides attenuate toxicity Mn-induced, by regulating the DAF-16/FOXO signaling pathway.
Collapse
Affiliation(s)
- Suzi G N Wollenhaupt
- Laboraterio do Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Ana Thalita Soares
- Laboraterio do Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Willian G Salgueiro
- Laboraterio do Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Simone Noremberg
- Departamento de Química, Universidade Federal de Santa Maria - UFSM, CEP 97105-900, Santa Maria, RS, Brazil
| | - Gabriel Reis
- Departamento de Química, Universidade Federal de Santa Maria - UFSM, CEP 97105-900, Santa Maria, RS, Brazil
| | - Carine Viana
- Departamento de Química, Universidade Federal de Santa Maria - UFSM, CEP 97105-900, Santa Maria, RS, Brazil
| | - Priscila Gubert
- Departamento de Química, Universidade Federal de Santa Maria - UFSM, CEP 97105-900, Santa Maria, RS, Brazil
| | - Felix A Soares
- Departamento de Química, Universidade Federal de Santa Maria - UFSM, CEP 97105-900, Santa Maria, RS, Brazil
| | - Ricardo F Affeldt
- Instituto de Química, Universidade Federal do Rio Grande do Sul - UFRGS, CEP 91501-970, Porto Alegre, RS, Brazil
| | - Diogo S Lüdtke
- Instituto de Química, Universidade Federal do Rio Grande do Sul - UFRGS, CEP 91501-970, Porto Alegre, RS, Brazil
| | - Francielli W Santos
- Laboratório de Biotecnologia da Reprodução (Biotech), Campus Uruguaiana, Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Cristiane C Denardin
- Laboraterio do Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil
| | - Michael Aschner
- Division of Clinical Pharmacology and Pediatric Toxicology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Daiana S Avila
- Laboraterio do Grupo de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans (GBToxCe), Universidade Federal do Pampa - UNIPAMPA, CEP 97500-970, Uruguaiana, RS, Brazil.
| |
Collapse
|
39
|
Brain deposition and neurotoxicity of manganese in adult mice exposed via the drinking water. Arch Toxicol 2013; 88:47-64. [PMID: 23832297 DOI: 10.1007/s00204-013-1088-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 06/20/2013] [Indexed: 12/27/2022]
Abstract
Natural leaching processes and/or anthropogenic contamination can result in ground water concentrations of the essential metal manganese (Mn) that far exceed the current regulatory standards. Neurological consequences of Mn drinking water (DW) overexposure to experimental animals, i.e., mice, including its brain deposition/distribution and behavioral effects are understudied. Adult male C57BL/6 mice were exposed to Mn via the DW for 8 weeks. After 5 weeks of Mn exposure, magnetic resonance imaging revealed significant Mn deposition in all examined brain regions; the degree of Mn deposition did not increase further a week later. Behaviorally, early hyperactivity and more time spent in the center of the arenas in an open field test, decreased forelimb grip strength and less time swimming in a forced swim test were observed after 6 weeks of Mn DW exposure. Eight-week Mn DW exposure did not alter striatal dopamine, its metabolites, or the expression of key dopamine homeostatic proteins, but it significantly increased striatal 5-hydroxyindoleacetic acid (a serotonin metabolite) levels, without affecting the levels of serotonin itself. Increased expression (mRNA) of glial fibrillary acidic protein (GFAP, an astrocyte activation marker), heme oxygenase-1 and inducible nitric oxide synthase (oxidative and nitrosative stress markers, respectively) were observed 8 weeks post-Mn DW exposure in the substantia nigra. Besides mRNA increases, GFAP protein expression was increased in the substantia nigra pars reticulata. In summary, the neurobehavioral deficits, characterized by locomotor and emotional perturbations, and nigral glial activation associated with significant brain Mn deposition are among the early signs of Mn neurotoxicity caused by DW overexposure.
Collapse
|
40
|
Colle D, Santos DB, Moreira ELG, Hartwig JM, dos Santos AA, Zimmermann LT, Hort MA, Farina M. Probucol increases striatal glutathione peroxidase activity and protects against 3-nitropropionic acid-induced pro-oxidative damage in rats. PLoS One 2013; 8:e67658. [PMID: 23799154 PMCID: PMC3683065 DOI: 10.1371/journal.pone.0067658] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/21/2013] [Indexed: 01/08/2023] Open
Abstract
Huntington's disease (HD) is an autosomal dominantly inherited neurodegenerative disease characterized by symptoms attributable to the death of striatal and cortical neurons. The molecular mechanisms mediating neuronal death in HD involve oxidative stress and mitochondrial dysfunction. Administration of 3-nitropropionic acid (3-NP), an irreversible inhibitor of the mitochondrial enzyme succinate dehydrogenase, in rodents has been proposed as a useful experimental model of HD. This study evaluated the effects of probucol, a lipid-lowering agent with anti-inflammatory and antioxidant properties, on the biochemical parameters related to oxidative stress, as well as on the behavioral parameters related to motor function in an in vivo HD model based on 3-NP intoxication in rats. Animals were treated with 3.5 mg/kg of probucol in drinking water daily for 2 months and, subsequently, received 3-NP (25 mg/kg i.p.) once a day for 6 days. At the end of the treatments, 3-NP-treated animals showed a significant decrease in body weight, which corresponded with impairment on motor ability, inhibition of mitochondrial complex II activity and oxidative stress in the striatum. Probucol, which did not rescue complex II inhibition, protected against behavioral and striatal biochemical changes induced by 3-NP, attenuating 3-NP-induced motor impairments and striatal oxidative stress. Importantly, probucol was able to increase activity of glutathione peroxidase (GPx), an enzyme important in mediating the detoxification of peroxides in the central nervous system. The major finding of this study was that probucol protected against 3-NP-induced behavioral and striatal biochemical changes without affecting 3-NP-induced mitochondrial complex II inhibition, indicating that long-term probucol treatment resulted in an increased resistance against neurotoxic events (i.e., increased oxidative damage) secondary to mitochondrial dysfunction. These data appeared to be of great relevance when extrapolated to human neurodegenerative processes involving mitochondrial dysfunction and indicates that GPx is an important molecular target involved in the beneficial effects of probucol.
Collapse
Affiliation(s)
- Dirleise Colle
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- * E-mail: marcelo.farina@.ufsc.br (MF); (DC)
| | - Danúbia Bonfanti Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Eduardo Luiz Gasnhar Moreira
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Juliana Montagna Hartwig
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Alessandra Antunes dos Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Luciana Teixeira Zimmermann
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Mariana Appel Hort
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- * E-mail: marcelo.farina@.ufsc.br (MF); (DC)
| |
Collapse
|
41
|
Meinerz DF, Comparsi B, Allebrandt J, Mariano DOC, Dos Santos DB, Zemolin APP, Farina M, Dafre LA, Rocha JBT, Posser T, Franco JL. Sub-acute administration of (S)-dimethyl 2-(3-(phenyltellanyl) propanamido) succinate induces toxicity and oxidative stress in mice: unexpected effects of N-acetylcysteine. SPRINGERPLUS 2013; 2:182. [PMID: 23658858 PMCID: PMC3644195 DOI: 10.1186/2193-1801-2-182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/16/2013] [Indexed: 12/21/2022]
Abstract
The organic tellurium compound (S)-dimethyl 2-(3-(phenyltellanyl) propanamide) succinate (TeAsp) exhibits thiol-peroxidase activity that could potentially offer protection against oxidative stress. However, data from the literature show that tellurium is a toxic agent to rodents. In order to mitigate such toxicity, N-acetylcysteine (NAC) was administered in parallel with TeAsp during 10 days. Mice were separated into four groups receiving daily injections of (A) vehicle (PBS 2.5 ml/kg, i.p. and DMSO 1 ml/kg, s.c.), (B) NAC (100 mg/kg, i.p. and DMSO s.c.), (C) PBS i.p. and TeAsp (92.5 μmol/kg, s.c), or (D) NAC plus TeAsp. TeAsp treatment started on the fourth day. Vehicle or NAC-treated animals showed an increase in body weight whereas TeAsp caused a significant reduction. Contrary to expected, NAC co-administration potentiated the toxic effect of TeAsp, causing a decrease in body weight. Vehicle, NAC or TeAsp did not affect the exploratory and motor activity in the open-field test at the end of the treatment, while the combination of NAC and TeAsp produced a significant decrease in these parameters. No DNA damage or alterations in cell viability were observed in leukocytes of treated animals. Treatments produced no or minor effects on the activities of antioxidant enzymes catalase, glutathione peroxidase and glutathione reductase, whereas the activity of the thioredoxin reductase was decreased in the brain and increased the liver of the animals in the groups receiving TeAsp or TeAsp plus NAC. In conclusion, the toxicity of TeAsp was potentiated by NAC and oxidative stress appears to play a central role in this process.
Collapse
Affiliation(s)
- Daiane F Meinerz
- Departamento de Química, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS CEP 97105-900 Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Souza ACG, Sari MHM, Pinton S, Luchese C, Neto JSS, Nogueira CW. 2-Phenylethynyl-butyltellurium attenuates amyloid-β peptide(25-35)-induced learning and memory impairments in mice. J Neurosci Res 2013; 91:848-53. [DOI: 10.1002/jnr.23211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Revised: 01/07/2013] [Accepted: 01/07/2013] [Indexed: 01/13/2023]
Affiliation(s)
- Ana Cristina Guerra Souza
- Departamento de Química; Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; Santa Maria; Brasil
| | | | - Simone Pinton
- Departamento de Química; Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; Santa Maria; Brasil
| | - Cristiane Luchese
- Mestrado em Nanociências; Centro de Ciências Tecnológicas; Centro Universitário Franciscano; Santa Maria; Brazil
| | - José Sebastião Santos Neto
- Departamento de Química; Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; Santa Maria; Brasil
| | - Cristina Wayne Nogueira
- Departamento de Química; Centro de Ciências Naturais e Exatas; Universidade Federal de Santa Maria; Santa Maria; Brasil
| |
Collapse
|
43
|
Farina M, Avila DS, da Rocha JBT, Aschner M. Metals, oxidative stress and neurodegeneration: a focus on iron, manganese and mercury. Neurochem Int 2012; 62:575-94. [PMID: 23266600 DOI: 10.1016/j.neuint.2012.12.006] [Citation(s) in RCA: 357] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Essential metals are crucial for the maintenance of cell homeostasis. Among the 23 elements that have known physiological functions in humans, 12 are metals, including iron (Fe) and manganese (Mn). Nevertheless, excessive exposure to these metals may lead to pathological conditions, including neurodegeneration. Similarly, exposure to metals that do not have known biological functions, such as mercury (Hg), also present great health concerns. This review focuses on the neurodegenerative mechanisms and effects of Fe, Mn and Hg. Oxidative stress (OS), particularly in mitochondria, is a common feature of Fe, Mn and Hg toxicity. However, the primary molecular targets triggering OS are distinct. Free cationic iron is a potent pro-oxidant and can initiate a set of reactions that form extremely reactive products, such as OH. Mn can oxidize dopamine (DA), generating reactive species and also affect mitochondrial function, leading to accumulation of metabolites and culminating with OS. Cationic Hg forms have strong affinity for nucleophiles, such as -SH and -SeH. Therefore, they target critical thiol- and selenol-molecules with antioxidant properties. Finally, we address the main sources of exposure to these metals, their transport mechanisms into the brain, and therapeutic modalities to mitigate their neurotoxic effects.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | | | | | | |
Collapse
|
44
|
Moberly AH, Czarnecki LA, Pottackal J, Rubinstein T, Turkel DJ, Kass MD, McGann JP. Intranasal exposure to manganese disrupts neurotransmitter release from glutamatergic synapses in the central nervous system in vivo. Neurotoxicology 2012; 33:996-1004. [PMID: 22542936 PMCID: PMC3432160 DOI: 10.1016/j.neuro.2012.04.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/29/2012] [Accepted: 04/14/2012] [Indexed: 11/28/2022]
Abstract
Chronic exposure to aerosolized manganese induces a neurological disorder that includes extrapyramidal motor symptoms and cognitive impairment. Inhaled manganese can bypass the blood-brain barrier and reach the central nervous system by transport down the olfactory nerve to the brain's olfactory bulb. However, the mechanism by which Mn disrupts neural function remains unclear. Here we used optical imaging techniques to visualize exocytosis in olfactory nerve terminals in vivo in the mouse olfactory bulb. Acute Mn exposure via intranasal instillation of 2-200 μg MnCl(2) solution caused a dose-dependent reduction in odorant-evoked neurotransmitter release, with significant effects at as little as 2 μg MnCl(2) and a 90% reduction compared to vehicle controls with a 200 μg exposure. This reduction was also observed in response to direct electrical stimulation of the olfactory nerve layer in the olfactory bulb, demonstrating that Mn's action is occurring centrally, not peripherally. This is the first direct evidence that Mn intoxication can disrupt neurotransmitter release, and is consistent with previous work suggesting that chronic Mn exposure limits amphetamine-induced dopamine increases in the basal ganglia despite normal levels of dopamine synthesis (Guilarte et al., J Neurochem 2008). The commonality of Mn's action between glutamatergic neurons in the olfactory bulb and dopaminergic neurons in the basal ganglia suggests that a disruption of neurotransmitter release may be a general consequence wherever Mn accumulates in the brain and could underlie its pleiotropic effects.
Collapse
Affiliation(s)
- Andrew H Moberly
- Behavioral Neuroscience Section, Department of Psychology, Rutgers, The State University of New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Ibrahim M, Hassan W, Meinerz DF, dos Santos M, V. Klimaczewski C, M. Deobald A, Costa MS, Nogueira CW, Barbosa NBV, Rocha JBT. Antioxidant properties of diorganoyl diselenides and ditellurides: modulation by organic aryl or naphthyl moiety. Mol Cell Biochem 2012; 371:97-104. [DOI: 10.1007/s11010-012-1426-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 08/03/2012] [Indexed: 01/22/2023]
|
46
|
Avila DS, Benedetto A, Au C, Manarin F, Erikson K, Soares FA, Rocha JBT, Aschner M. Organotellurium and organoselenium compounds attenuate Mn-induced toxicity in Caenorhabditis elegans by preventing oxidative stress. Free Radic Biol Med 2012; 52:1903-10. [PMID: 22406322 PMCID: PMC3341511 DOI: 10.1016/j.freeradbiomed.2012.02.044] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 02/24/2012] [Accepted: 02/24/2012] [Indexed: 01/14/2023]
Abstract
Organochalcogens have been widely studied given their antioxidant activity, which confers neuroprotection, antiulcer, and antidiabetic properties. Given the complexity of mammalian models, understanding the cellular and molecular effects of organochalcogens has been hampered. The nematode worm Caenorhabditis elegans is an alternative experimental model that affords easy genetic manipulations, green fluorescent protein tagging, and in vivo live analysis of toxicity. We previously showed that manganese (Mn)-exposed worms exhibit oxidative-stress-induced neurodegeneration and life-span reduction. Here we use Mn-exposed worms as a model for an oxidatively challenged organism to investigate the underlying mechanisms of organochalcogen antioxidant properties. First, we recapitulate in C. elegans the effects of organochalcogens formerly observed in mice, including their antioxidant activity. This is followed by studies on the ability of these compounds to afford protection against Mn-induced toxicity. Diethyl-2-phenyl-2-tellurophenyl vinyl phosphonate (DPTVP) was the most efficacious compound, fully reversing the Mn-induced reduction in survival and life span. Ebselen was also effective, reversing the Mn-induced reduction in survival and life span, but to a lesser extent compared with DPTVP. DPTVP also lowered Mn-induced increases in oxidant levels, indicating that the increased survival associated with exposure to this compound is secondary to a decrease in oxidative stress. Furthermore, DPTVP induced nuclear translocation of the transcriptional factor DAF-16/FOXO, which regulates stress responsiveness and aging in worms. Our findings establish that the organochalcogens DPTVP and ebselen act as antiaging agents in a model of Mn-induced toxicity and aging by regulating DAF-16/FOXO signaling and attenuating oxidative stress.
Collapse
|
47
|
Souza ACG, Acker CI, Gai BM, Neto JSDS, Nogueira CW. 2-Phenylethynyl-butyltellurium improves memory in mice. Neurochem Int 2012; 60:409-14. [DOI: 10.1016/j.neuint.2012.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/19/2011] [Accepted: 01/11/2012] [Indexed: 01/10/2023]
|
48
|
Martins EN, Pessano NTC, Leal L, Roos DH, Folmer V, Puntel GO, Rocha JBT, Aschner M, Ávila DS, Puntel RL. Protective effect of Melissa officinalis aqueous extract against Mn-induced oxidative stress in chronically exposed mice. Brain Res Bull 2011; 87:74-9. [PMID: 22020131 DOI: 10.1016/j.brainresbull.2011.10.003] [Citation(s) in RCA: 229] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/10/2011] [Indexed: 10/16/2022]
Abstract
Manganese (Mn) is an essential element for biological systems; however occupational exposure to high levels of this metal may lead to neurodegenerative disorders, resembling Parkinson's disease (PD). While its mechanisms of neurotoxicity have yet to be fully understood, oxidative stress plays a critical role. Thus, the main goal of this study was to investigate the efficacy of aqueous extract of Melissa officinalis in attenuating Mn-induced brain oxidative stress in mice. Sixteen male mice were randomly divided into two groups and treated for 3 months: the first group consumed tap water (control group) and the second group was treated with Mn (50 mg/kg/day for habituation during the first 15 days followed by 100 mg/kg/day for additional 75 days) in the drinking water. After 3 months both groups were sub divided (n=4 per group) and treated for additional 3 months with Mn and/or M. officinalis in the drinking water. The first group (control) was treated with water and served as control; the second group (M. officinalis) was treated with M. officinalis (100 mg/kg/day); the third group was treated with Mn (100 mg/kg/day); the fourth group (Mn+M. officinalis) was treated with both Mn and M. officinalis (100 mg/kg/day each). Mn-treated mice showed a significant increase in thiobarbituric acid reactive species (TBARS) levels (a marker of oxidative stress) in both the hippocampus and striatum. These changes were accompanied by a decrease in total thiol content in the hippocampus and a significant increase in antioxidant enzyme activity (superoxide dismutase and catalase) in the hippocampus, striatum, cortex and cerebellum. Co-treatment with M. officinalis aqueous extract in Mn-treated mice significantly inhibited the antioxidant enzyme activities and attenuated the oxidative damage (TBARS and decreased total thiol levels). These results establish that M. officinalis aqueous extract possesses potent antioxidative properties, validating its efficacy in attenuating Mn-induced oxidative stress in the mouse brain.
Collapse
Affiliation(s)
- Eduarda N Martins
- Laboratório de Bioquímica e Toxicologia de Produtos Naturais e Sintéticos, Universidade Federal do Pampa, BR 472 Km 585 Sala 403, CEP 97500-970 Uruguaiana, RS, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Heimfarth L, Loureiro SO, Reis KP, de Lima BO, Zamboni F, Gandolfi T, Narvaes R, da Rocha JBT, Pessoa-Pureur R. Cross-Talk among Intracellular Signaling Pathways Mediates the Diphenyl Ditelluride Actions on the Hippocampal Cytoskeleton of Young Rats. Chem Res Toxicol 2011; 24:1754-64. [DOI: 10.1021/tx200307u] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Luana Heimfarth
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | | | - Karina Pires Reis
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | - Bárbara Ortiz de Lima
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | - Fernanda Zamboni
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | - Talita Gandolfi
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | - Rodrigo Narvaes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| | | | - Regina Pessoa-Pureur
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brasil
| |
Collapse
|
50
|
Heimfarth L, Loureiro SO, Reis KP, de Lima BO, Zamboni F, Lacerda S, Soska ÂK, Wild L, da Rocha JBT, Pessoa-Pureur R. Diphenyl ditelluride induces hypophosphorylation of intermediate filaments through modulation of DARPP-32-dependent pathways in cerebral cortex of young rats. Arch Toxicol 2011; 86:217-30. [DOI: 10.1007/s00204-011-0746-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/11/2011] [Indexed: 01/02/2023]
|