1
|
El-Shiekh RA, Mohamed AF, Mandour AA, Adel IM, Atwa AM, Elgindy AM, Esmail MM, Senna MM, Ebid N, Mustafa AM. Hesperidin in Chronic Fatigue Syndrome: An Integrated Analysis of Traditional Pharmacology and Machine Learning-Based Therapeutic Predictions. Chem Biodivers 2025:e202403506. [PMID: 40234200 DOI: 10.1002/cbdv.202403506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/17/2025]
Abstract
Hesperidin, a bioflavonoid abundantly found in citrus fruits, offers a myriad of health benefits. With the food industry extensively utilizing citrus fruits, particularly for juice production, substantial quantities of by-products such as peels, seeds, cells, and membrane residues accumulate. Remarkably, these by-products serve as a valuable source of hesperidin. Consequently, the extraction of hesperidin from these by-products has garnered significant scientific interest, aiming to harness its potential as a natural antioxidant. By shedding light on these aspects, this review provides a comprehensive review of hesperidin's role in enhancing human well-being, particularly in the context of chronic fatigue syndrome (CFS). By synthesizing current research, we elucidate the compound's antioxidant, anti-inflammatory, and neuroprotective effects, which may mitigate symptoms associated with CFS. Furthermore, we introduce machine learning methodologies to predict hesperidin's efficacy in clinical settings, offering a novel perspective on personalized nutrition strategies. Our findings underscore the need for further empirical studies to validate these predictions and explore hesperidin's mechanisms of action. This review not only bridges the gap between nutrition science and pharmacology but also highlights the promising future of hesperidin as a nutraceutical in combating chronic health conditions.
Collapse
Affiliation(s)
- Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Asmaa A Mandour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo, Egypt
| | - Islam M Adel
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed M Atwa
- College of Pharmacy, Al-Ayen Iraqi University, AUIQ, An Nasiriyah, Iraq
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Ali M Elgindy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Manar M Esmail
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Mohamed Magdy Senna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Nouran Ebid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Aya M Mustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
2
|
Utpal BK, Sutradhar B, Zehravi M, Sweilam SH, Durgawale TP, Arjun UVNV, Shanmugarajan TS, Kannan SP, Prasad PD, Usman MRM, Reddy KTK, Sultana R, Alshehri MA, Rab SO, Suliman M, Emran TB. Cellular stress response and neuroprotection of flavonoids in neurodegenerative diseases: Clinical insights into targeted therapy and molecular signaling pathways. Brain Res 2025; 1847:149310. [PMID: 39537124 DOI: 10.1016/j.brainres.2024.149310] [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: 09/14/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases (NDs) are caused by the gradual decline of neuronal structure and function, which presents significant challenges in treatment. Cellular stress responses significantly impact the pathophysiology of these disorders, often exacerbating neuronal damage. Plant-derived flavonoids have demonstrated potential as neuroprotective agents due to their potent anti-inflammatory, anti-apoptotic, and antioxidant properties. This review provides an in-depth analysis of the molecular processes and clinical insights that cause the neuroprotective properties of flavonoids in NDs. By controlling essential signaling pathways such as Nrf2/ARE, MAPK, and PI3K/Akt, flavonoids can lower cellular stress and improve neuronal survival. The study discusses the challenges of implementing these discoveries in clinical practice and emphasizes the therapeutic potential of specific flavonoids and their derivatives. Flavonoids are identified as potential therapeutic agents for NDs, potentially slowing progression by regulating cellular stress and improving neuroprotection despite their potential medicinal uses and clinical challenges. The study designed a strategy to identify literature published in prestigious journals, utilizing search results from PubMed, Scopus, and WOS. We selected and investigated original studies, review articles, and research reports published until 2024. It suggests future research and therapeutic approaches to effectively utilize the neuroprotective properties of flavonoids.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Baishakhi Sutradhar
- Department of Microbiology, Gono University (Bishwabidyalay), Nolam, Mirzanagar, Savar, Dhaka 1344, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| | - Trupti Pratik Durgawale
- Department of Pharmaceutical Chemistry, KVV's Krishna Institute of Pharmacy, Karad, Maharashtra, India
| | - Uppuluri Varuna Naga Venkata Arjun
- Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai 600117, Tamil Nadu, India
| | - Thukani Sathanantham Shanmugarajan
- Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai 600117, Tamil Nadu, India
| | - Shruthi Paramasivam Kannan
- Vels Institute of Science, Technology and Advanced Studies (VISTAS), PV Vaithiyalingam Rd, Velan Nagar, Krishna Puram, Pallavaram, Chennai 600117, Tamil Nadu, India
| | - P Dharani Prasad
- Department of Pharmacology, Mohan Babu University, MB School of Pharmaceutical Sciences, (Erstwhile, Sree Vidyaniketan College of Pharmacy), Tirupati, Andhra Pradesh 517102, India
| | - Md Rageeb Md Usman
- Department of Pharmacognosy, Smt. Sharadchandrika Suresh Patil College of Pharmacy, Chopda, Maharashtra, India
| | - Konatham Teja Kumar Reddy
- Department of Pharmacy, University College of Technology, Osmania University, Amberpet, Hyderabad, Telangana 500007, India
| | - Rokeya Sultana
- Department of Pharmacognosy, Yenepoya Pharmacy College and Research Centre, Yenepoya (deemed to be University), Mangalore, Karnataka, India
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1207, Bangladesh; Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
3
|
Yankuzo HM, Sulaiman I, Muhammad SA, Raji AA, Uthman YA, Imam MU. Brown rice attenuates iron-induced Parkinson's disease phenotypes in male wild-type drosophila: insights into antioxidant and iron metabolism modulation. Appl Physiol Nutr Metab 2025; 50:1-13. [PMID: 39588846 DOI: 10.1139/apnm-2024-0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Parkinson's disease (PD) is a progressive movement disorder associated with brain iron (Fe) accumulation and free radicals. Brown rice (BR) is antioxidant-rich and has been shown to ameliorate oxidative stress-induced damage. The aim of this study was to investigate the effects of BR compared to white rice (WR) on Fe-induced PD in a fruit fly model. Three-day-old male adult flies were divided into two groups: one on a normal diet and the other on Fe-diet (1 mmol/L) for 10 days to induce PD. After 10 days, the Fe-fed flies were redistributed into four groups: one on normal diet (Fe group), while the others were treated with BR (Fe + BR group), WR (Fe + WR group), or L-3,4-dihydroxyphenylalanine (L-dopa) (Fe + L-dopa group) for 5 days. Similarly, the flies initially on a normal diet were separated into four groups: one on normal diet (Control group), while the others were treated with BR (BR group), WR (WR group), or L-dopa (L-dopa group) for 5 days. Finally, Fe levels, dopamine, malonaldehyde (MDA), and antioxidant enzymes were measured, and the mRNA levels of antioxidant and Fe metabolism genes were assessed. BR significantly improved motor and cognitive functions, decreased fly head MDA and Fe levels, and increased antioxidant enzyme levels in comparison to the Fe and WR groups. Similarly, BR upregulated the mRNA levels of antioxidant genes: catalase, GPx, Nrf2, and DJ-1. The results suggest that BR could potentially reduce morbidities associated with PD possibly due to its bioactive compounds compared to WR.
Collapse
Affiliation(s)
- Hassan Muhammad Yankuzo
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Ismail Sulaiman
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Biochemistry and Molecular Biology, Faculty of Chemical and Life Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Suleiman Alhaji Muhammad
- Department of Biochemistry and Molecular Biology, Faculty of Chemical and Life Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Abdullahi Abdullahi Raji
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Yaaqub Abiodun Uthman
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
| | - Mustapha Umar Imam
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Usmanu Danfodiyo University Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Nigeria
| |
Collapse
|
4
|
Genç B, Karaman M. Effect of Ascorbic Acid on Oxidative Stress Parameters of Fruit Fly in the Presence of Fe (II). Biol Trace Elem Res 2024; 202:3810-3815. [PMID: 37980318 DOI: 10.1007/s12011-023-03960-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
Excessive iron intake causes lipid peroxidation and increases oxidative stress. Ascorbic acid is a natural compound with antioxidant and therapeutic properties. In this study, the effect of AA on oxidative stress parameters in fruit flies in the presence of iron was investigated. Third instar larvae were exposed to Fe and/or AA. Antioxidant enzyme activities, MDA, and GSH levels were determined in adult heads developing from the larvae. The mRNA levels of the enzymes were also analyzed. Iron treatment caused inhibition of SOD, CAT, and GPx enzymes, and only a decrease in the mRNA level of CAT. However, AA treatment together with iron prevented the inhibition. In addition, iron treatment increased the MDA level and decreased the GSH level. AA treatment together with iron ameliorated the changes in MDA and GSH levels. The results showed that AA can interfere with the iron-induced changes. Considering the potential of AA to ameliorate iron-induced changes, further studies are needed to elucidate the effect of AA on different mechanisms.
Collapse
Affiliation(s)
- Berna Genç
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey
| | - Melike Karaman
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey.
| |
Collapse
|
5
|
Kamal N, Abdallah MS, Abdel Wahed E, Sabri NA, Fahmy SF. Evaluation of the Effect of Loratadine versus Diosmin/Hesperidin Combination on Vinca Alkaloids-Induced Neuropathy: A Randomized Controlled Clinical Trial. Pharmaceuticals (Basel) 2024; 17:609. [PMID: 38794179 PMCID: PMC11124025 DOI: 10.3390/ph17050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Neurological injury is a crucial problem that interferes with the therapeutic use of vinca alkaloids as well as the quality of patient life. This study was conducted to assess the impact of using loratadine or diosmin/hesperidin on neuropathy induced by vinca alkaloids. Patients were randomized into one of three groups as follows: group 1 was the control group, group 2 received 450 mg diosmin and 50 mg hesperidin combination orally twice daily, and group 3 received loratadine 10 mg orally once daily. Subjective scores (numeric pain rating scale, douleur neuropathique 4, and functional assessment of cancer therapy/gynecologic oncology group-neurotoxicity (FACT/GOG-Ntx) scores), neuroinflammation biomarkers, adverse drug effects, quality of life, and response to chemotherapy were compared among the three groups. Both diosmin/hesperidin and loratadine improved the results of the neurotoxicity subscale in the FACT/GOG-Ntx score (p < 0.001, p < 0.01 respectively) and ameliorated the upsurge in neuroinflammation serum biomarkers. They also reduced the incidence and timing of paresthesia (p = 0.001 and p < 0.001, respectively) and dysuria occurrence (p = 0.042). Both loratadine and diosmin/hesperidin attenuated the intensity of acute neuropathy triggered by vinca alkaloids. Furthermore, they did not increase the frequency of adverse effects or interfere with the treatment response.
Collapse
Affiliation(s)
- Noha Kamal
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Sadat City (USC), Sadat City 32897, Egypt;
| | - Mahmoud S. Abdallah
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Sadat City (USC), Sadat City 32897, Egypt;
- Department of PharmD, Faculty of Pharmacy, Jadara University, Irbid 21110, Jordan
| | - Essam Abdel Wahed
- Hematology and Bone Marrow Transplantation Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt;
| | - Nagwa A. Sabri
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Cairo 11566, Egypt; (N.A.S.); (S.F.F.)
| | - Sarah Farid Fahmy
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, African Union Organization Street, Cairo 11566, Egypt; (N.A.S.); (S.F.F.)
| |
Collapse
|
6
|
Adedara AO, Bressan GN, Dos Santos MM, Fachinetto R, Abolaji AO, Barbosa NV. Antioxidant responses driven by Hesperetin and Hesperidin counteract Parkinson's disease-like phenotypes in Drosophila melanogaster. Neurotoxicology 2024; 101:117-127. [PMID: 38423185 DOI: 10.1016/j.neuro.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
The study investigated the protective effects of Hesperetin (HSP) and Hesperidin (HSD) on 1 methyl, 4 phenyl, 1,2,3,6 tetrahydropyridine hydrochloride (MPTP)-induced Parkinsonism in Drosophila melanogaster (D. melanogaster). After a lifespan study to select exposure time and concentrations, flies were co-exposed to MPTP (0.4 mg/g diet), Hesperetin (0.2 and 0.4 mg/g diet), and Hesperidin (0.1 and 0.4 mg/g) for 7 days. In addition to in vivo parameters, we assayed some markers of oxidative stress and antioxidant status (lipid peroxidation, protein carbonylation, thiol content, hydrogen peroxide, and nitrate/nitrite levels, mRNA expression of Keap-1 (Kelch-like ECH associated protein 1), /Nrf2 (Nuclear factor erythroid 2 related factor 2), catalase, and glutathione-S-transferase (GST) activities), and cholinergic (acetyl cholinesterase activity (AChE) and dopaminergic signaling content and the mRNA expression of tyrosine hydroxylase (TH), monoamine oxidase (MAO-like) activity). In addition to increasing the lifespan of flies, we found that both flavonoids counteracted the adverse effects of MPTP on survival, offspring emergence, and climbing ability of flies. Both flavonoids also reduced the oxidative damage on lipids and proteins and reestablished the basal levels of pro-oxidant species and activities of antioxidant enzymes in MPTP-exposed flies. These responses were accompanied by the normalization of the mRNA expression of Keap1/Nrf2 disrupted in flies exposed to MPTP. MPTP exposure also elicited changes in mRNA expression and content of TH as well as in MAO and AChE activity, which were reversed by HST and HSD. By efficiently hindering the oxidative stress in MPTP-exposed flies, our findings support the promising role of Hesperetin and Hesperidin as adjuvant therapy to manage Parkinsonism induced by chemicals such as MPTP.
Collapse
Affiliation(s)
- Adeola Oluwatosin Adedara
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil; Drosophila Laboratory, Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Science, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Getúlio Nicola Bressan
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Matheus Mulling Dos Santos
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Roselei Fachinetto
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil
| | - Amos Olalekan Abolaji
- Drosophila Laboratory, Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Science, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.
| | - Nilda Vargas Barbosa
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
7
|
Tavan M, Hanachi P, de la Luz Cádiz-Gurrea M, Segura Carretero A, Mirjalili MH. Natural Phenolic Compounds with Neuroprotective Effects. Neurochem Res 2024; 49:306-326. [PMID: 37940760 DOI: 10.1007/s11064-023-04046-z] [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: 08/01/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 11/10/2023]
Abstract
Neurodegenerative disorders are characterized by mitochondrial dysfunction and subsequently oxidative stress, inflammation, and apoptosis that contribute to neuronal cytotoxicity and degeneration. Huntington's (HD), Alzheimer's (AD), and Parkinson's (PD) diseases are three of the major neurodegenerative diseases. To date, researchers have found various natural phytochemicals that could potentially be used to treat neurodegenerative diseases. Particularly, the application of natural phenolic compounds has gained significant traction in recent years, driven by their various biological activities and therapeutic efficacy in human health. Polyphenols, by modulating different cellular functions, play an important role in neuroprotection and can neutralize the effects of oxidative stress, inflammation, and apoptosis in animal models. This review focuses on the current state of knowledge on phenolic compounds, including phenolic acids, flavonoids, stilbenes, and coumarins, as well as their beneficial effects on human health. We further provide an overview of the therapeutic potential and mechanisms of action of natural dietary phenolics in curing neurodegenerative diseases in animal models.
Collapse
Affiliation(s)
- Mansoureh Tavan
- Department of Agriculture, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, 1983969411, Iran.
- Department of Biotechnology, Faculty of Biological Science, Alzahra University, Tehran, Iran.
| | - Parichehr Hanachi
- Department of Biotechnology, Faculty of Biological Science, Alzahra University, Tehran, Iran
| | | | | | - Mohammad Hossein Mirjalili
- Department of Agriculture, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, 1983969411, Iran
| |
Collapse
|
8
|
Musachio EAS, Pires RG, Fernandes EJ, Andrade S, Meichtry LB, Janner DE, Meira GM, Ribeiro EE, Barbisan F, da Cruz IBM, Prigol M. The Amazonian Camu-Camu Fruit Modulates the Development of Drosophila melanogaster and the Neural Function of Adult Flies under Oxidative Stress Conditions. Antioxidants (Basel) 2024; 13:102. [PMID: 38247526 PMCID: PMC11154359 DOI: 10.3390/antiox13010102] [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: 10/09/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Camu-camu (Myrciaria dubia) is known for its antioxidant properties, although little is known about its developmental safety effects, particularly on adult neural function under basal redox and oxidative stress conditions. Therefore, this study sought to address this gap by conducting three complementary protocols using Drosophila melanogaster to investigate these effects. The initial assays revealed that second-stage larvae consumed diets supplemented with various concentrations of camu-camu uniformly, establishing a 50% lethal concentration at 4.799 mg/mL. Hence, non-lethal (0.1, 0.5, and 1 mg/mL) and sub-lethal (5 and 10 mg/mL) concentrations were then chosen to evaluate the effects of camu-camu on preimaginal development and adult neural function. Our observations showed that camu-camu impacts the expression of antioxidant enzymes, reactive species, and lipoperoxidation. Notably, sub-lethal concentrations decreased preimaginal viability and locomotor activity, negatively influenced geotaxis and acetylcholinesterase activity, and increased reactive species, catalase, and glutathione S-transferase activity in flies. Additionally, the protective effects of camu-camu against oxidative stress induced by iron (20 mM) were assessed. Flies supplemented with 0.5 mg/mL of camu-camu during the larval period showed improved neural viability and function, and this supplementation was found to protect against oxidative stress. These findings are instrumental in evaluating the safety and efficacy of commercial supplements based on camu-camu, offering significant insights for future research and application.
Collapse
Affiliation(s)
- Elize Aparecida Santos Musachio
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Rafaela Garay Pires
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Eliana Jardim Fernandes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Stefani Andrade
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Luana Barreto Meichtry
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Dieniffer Espinosa Janner
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| | - Graziela Moro Meira
- Laboratory of Biogenomics, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil; (G.M.M.); (F.B.)
| | - Euler Esteves Ribeiro
- Center for Research, Teaching and Technological Development-GERONTEC, Open University Foundation for the Elderly, Manaus 69029-040, AM, Brazil;
| | - Fernanda Barbisan
- Laboratory of Biogenomics, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil; (G.M.M.); (F.B.)
- Graduate Program in Gerontology, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Ivana Beatrice Mânica da Cruz
- Laboratory of Biogenomics, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil; (G.M.M.); (F.B.)
- Graduate Program in Gerontology, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Itaqui 97650-000, RS, Brazil; (E.A.S.M.); (R.G.P.); (E.J.F.); (S.A.); (L.B.M.); (D.E.J.); (M.P.)
| |
Collapse
|
9
|
Singh S, Chib S, Akhtar MJ, Kumar B, Chawla PA, Bhatia R. Paradigms and Success Stories of Natural Products in Drug Discovery Against Neurodegenerative Disorders (NDDs). Curr Neuropharmacol 2024; 22:992-1015. [PMID: 36606589 PMCID: PMC10964107 DOI: 10.2174/1570159x21666230105110834] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 01/07/2023] Open
Abstract
Neurodegenerative disorders (NDDs) are multifaceted complex disorders that have put a great health and economic burden around the globe nowadays. The multi-factorial nature of NDDs has presented a great challenge in drug discovery and continuous efforts are in progress in search of suitable therapeutic candidates. Nature has a great wealth of active principles in its lap that has cured the human population since ancient times. Natural products have revealed several benefits over conventional synthetic medications and scientists have shifted their vision towards exploring the therapeutic potentials of natural products in the past few years. The structural mimicking of natural compounds to endogenous ligands has presented them as a potential therapeutic candidate to prevent the development of NDDs. In the presented review, authors have summarized demographical facts about various NDDs including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and various types of sclerosis in the brain. The significant findings of new active principles of natural origin along with their therapeutic potentials on NDDs have been included. Also, a description of clinical trials and patents on natural products has been enlisted in this compilation. Although natural products have shown promising success in drug discovery against NDDs, still their use is associated with several ethical issues which need to be solved in the upcoming time.
Collapse
Affiliation(s)
- Sukhwinder Singh
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
| | - Shivani Chib
- Department of Pharmacology, ISF College of Pharmacy Moga, Punjab, 142001, India
| | - Md. Jawaid Akhtar
- Department of Pharmaceutical Chemistry, College of Pharmacy, National University of Science and Technology, PO620, PC 130 Azaiba, Bousher, Muscat, Oman
| | - Bhupinder Kumar
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, Garhwal, Uttarakhand, 246174, India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy Moga, Punjab, 142001, India
| |
Collapse
|
10
|
Chaudhary R, Singh R. Therapeutic Viewpoint on Rat Models of Locomotion Abnormalities and Neurobiological Indicators in Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:488-503. [PMID: 37202886 DOI: 10.2174/1871527322666230518111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/11/2022] [Accepted: 12/02/2022] [Indexed: 05/20/2023]
Abstract
BACKGROUND Locomotion problems in Parkinson's syndrome are still a research and treatment difficulty. With the recent introduction of brain stimulation or neuromodulation equipment that is sufficient to monitor activity in the brain using electrodes placed on the scalp, new locomotion investigations in patients having the capacity to move freely have sprung up. OBJECTIVE This study aimed to find rat models and locomotion-connected neuronal indicators and use them all over a closed-loop system to enhance the future and present treatment options available for Parkinson's disease. METHODS Various publications on locomotor abnormalities, Parkinson's disease, animal models, and other topics have been searched using several search engines, such as Google Scholar, Web of Science, Research Gate, and PubMed. RESULTS Based on the literature, we can conclude that animal models are used for further investigating the locomotion connectivity deficiencies of many biological measuring devices and attempting to address unanswered concerns from clinical and non-clinical research. However, translational validity is required for rat models to contribute to the improvement of upcoming neurostimulation-based medicines. This review discusses the most successful methods for modelling Parkinson's locomotion in rats. CONCLUSION This review article has examined how scientific clinical experiments lead to localised central nervous system injuries in rats, as well as how the associated motor deficits and connection oscillations reflect this. This evolutionary process of therapeutic interventions may help to improve locomotion- based treatment and management of Parkinson's syndrome in the upcoming years.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
- Department of Pharmacology, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda 151401, India
| |
Collapse
|
11
|
Bansal K, Singh V, Singh S, Mishra S. Neuroprotective Potential of Hesperidin as Therapeutic Agent in the Treatment of Brain Disorders: Preclinical Evidence-based Review. Curr Mol Med 2024; 24:316-326. [PMID: 36959141 DOI: 10.2174/1566524023666230320144722] [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: 10/03/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 03/25/2023]
Abstract
Neurodegenerative disorders (NDs) are progressive morbidities that represent a serious health issue in the aging world population. There is a contemporary upsurge in worldwide interest in the area of traditional remedies and phytomedicines are widely accepted by researchers due to their health-promoted effects and fewer side effects. Hesperidin, a flavanone glycoside present in the peels of citrus fruits, possesses various biological activities including anti-inflammatory and antioxidant actions. In various preclinical studies, hesperidin has provided significant protective actions in a variety of brain disorders such as Alzheimer's disease, epilepsy, Parkinson's disease, multiple sclerosis, depression, neuropathic pain, etc. as well as their underlying mechanisms. The findings indicate that the neuroprotective effects of hesperidin are mediated by modulating antioxidant defence activities and neural growth factors, diminishing apoptotic and neuro-inflammatory pathways. This review focuses on the potential role of hesperidin in managing and treating diverse brain disorders.
Collapse
Affiliation(s)
- Keshav Bansal
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Vanshita Singh
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Sakshi Singh
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Samiksha Mishra
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| |
Collapse
|
12
|
Ogunsuyi OB, Aro OP, Oboh G, Olagoke OC. Curcumin improves the ability of donepezil to ameliorate memory impairment in Drosophila melanogaster: involvement of cholinergic and cnc/Nrf2-redox systems. Drug Chem Toxicol 2023; 46:1035-1043. [PMID: 36069210 DOI: 10.1080/01480545.2022.2119995] [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: 02/24/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 11/03/2022]
Abstract
One of the well-established models for examining neurodegeneration and neurotoxicity is the Drosophila melanogaster model of aluminum-induced toxicity. Anti-cholinesterase drugs have been combined with other neuroprotective agents to improve Alzheimer's disease management, but there is not much information on the combination of anti-cholinesterases with dietary polyphenols to combat memory impairment. Here, we assess how curcumin influences some of the critical therapeutic effects of donepezil (a cholinesterase inhibitor) in AlCl3-treated Drosophila melanogaster. Harwich strain flies were exposed to 40 mM AlCl3 - alone or in combination with curcumin (1 mg/g) and/or donepezil (12.5 µg/g and 25 µg/g) - for seven days. The flies' behavioral evaluations (memory index and locomotor performance) were analyzed. Thereafter, the flies were processed into homogenates for the quantification of acetylcholinesterase (AChE), catalase, total thiol, and rate of lipid peroxidation, as well as the mRNA levels of acetylcholinesterase (ACE1) and cnc/NRF2. Results showed that AlCl3-treated flies presented impaired memory and increased activities of acetylcholinesterase and lipid peroxidation, while there were decrease in total thiol levels and catalase activity when compared to the control. Also, the expression of ACE1 was significantly increased while that of cnc/NRF2 was significantly decreased. However, combinations of curcumin and donepezil, especially at lower dose of donepezil, significantly improved the memory index and biochemical parameters compared to donepezil alone. Thus, curcumin plus donepezil offers unique therapeutic effects during memory impairment in the D. melanogaster model of neurotoxicity.
Collapse
Affiliation(s)
- Opeyemi Babatunde Ogunsuyi
- Department of Biomedical Technology, Federal University of Technology, Akure, Nigeria
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Olayemi Philemon Aro
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Drosophila Research Lab, Functional Foods and Nutraceuticals Unit, Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Olawande Chinedu Olagoke
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
König A, Sadova N, Dornmayr M, Schwarzinger B, Neuhauser C, Stadlbauer V, Wallner M, Woischitzschläger J, Müller A, Tona R, Kofel D, Weghuber J. Combined acid hydrolysis and fermentation improves bioactivity of citrus flavonoids in vitro and in vivo. Commun Biol 2023; 6:1083. [PMID: 37880345 PMCID: PMC10600125 DOI: 10.1038/s42003-023-05424-7] [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: 03/03/2023] [Accepted: 10/05/2023] [Indexed: 10/27/2023] Open
Abstract
Many bioactive plant compounds, known as phytochemicals, have the potential to improve health. Unfortunately, the bioavailability and bioactivity of phytochemicals such as polyphenolic flavonoids are reduced due to conjugation with sugar moieties. Here, we combine acid hydrolysis and tailored fermentation by lactic acid bacteria (Lactiplantibacillus plantarum) to convert the biologically less active flavonoid glycosides hesperidin and naringin into the more active aglycones hesperetin and naringenin. Using a comprehensive approach, we identify the most effective hydrolysis and fermentation conditions to increase the concentration of the aglycones in citrus extracts. The higher cellular transport and bioactivity of the biotransformed citrus extract are also demonstrated in vitro and in vivo. Superior antioxidant, anti-inflammatory and cell migration activities in vitro, as well as intestinal barrier protecting and antioxidant activities in Drosophila melanogaster are identified. In conclusion, the presented biotransformation approach improves the bioactivity of flavonoids, clearly traced back to the increase in aglycone content.
Collapse
Affiliation(s)
- Alice König
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria
| | - Nadiia Sadova
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
| | - Marion Dornmayr
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria
| | - Bettina Schwarzinger
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria
| | - Cathrina Neuhauser
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
| | - Verena Stadlbauer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria
| | - Melanie Wallner
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria
| | - Jakob Woischitzschläger
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria
| | - Andreas Müller
- TriPlant AG, Industriestrasse 17, Buetzberg, 4922, Switzerland
| | - Rolf Tona
- TriPlant AG, Industriestrasse 17, Buetzberg, 4922, Switzerland
| | - Daniel Kofel
- TriPlant AG, Industriestrasse 17, Buetzberg, 4922, Switzerland
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, Wels, 4600, Austria.
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, Tulln, 3430, Austria.
| |
Collapse
|
14
|
Gustavsson J, Johansson J, Falahati F, Andersson M, Papenberg G, Avelar-Pereira B, Bäckman L, Kalpouzos G, Salami A. The iron-dopamine D1 coupling modulates neural signatures of working memory across adult lifespan. Neuroimage 2023; 279:120323. [PMID: 37582419 DOI: 10.1016/j.neuroimage.2023.120323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
Brain iron overload and decreased integrity of the dopaminergic system have been independently reported as brain substrates of cognitive decline in aging. Dopamine (DA), and iron are co-localized in high concentrations in the striatum and prefrontal cortex (PFC), but follow opposing age-related trajectories across the lifespan. DA contributes to cellular iron homeostasis and the activation of D1-like DA receptors (D1DR) alleviates oxidative stress-induced inflammatory responses, suggesting a mutual interaction between these two fundamental components. Still, a direct in-vivo study testing the iron-D1DR relationship and their interactions on brain function and cognition across the lifespan is rare. Using PET and MRI data from the DyNAMiC study (n=180, age=20-79, %50 female), we showed that elevated iron content was related to lower D1DRs in DLPFC, but not in striatum, suggesting that dopamine-rich regions are less susceptible to elevated iron. Critically, older individuals with elevated iron and lower D1DR exhibited less frontoparietal activations during the most demanding task, which in turn was related to poorer working-memory performance. Together, our findings suggest that the combination of elevated iron load and reduced D1DR contribute to disturbed PFC-related circuits in older age, and thus may be targeted as two modifiable factors for future intervention.
Collapse
Affiliation(s)
- Jonatan Gustavsson
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden.
| | - Jarkko Johansson
- Faculty of Medicine, Department of Radiation Sciences, Umeå University, Sweden; Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Farshad Falahati
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Micael Andersson
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden; Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Bárbara Avelar-Pereira
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden; Department of Psychiatry and Behavioural Sciences, School of Medicine, Stanford University, Stanford, California 94304, USA
| | - Lars Bäckman
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Grégoria Kalpouzos
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Alireza Salami
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden; Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden; Department of Integrative Medical Biology, Umeå University, Umeå, Sweden; Wallenberg Center for Molecular Medicine, Umeå University, Sweden
| |
Collapse
|
15
|
Li X, Huang W, Tan R, Xu C, Chen X, Li S, Liu Y, Qiu H, Cao H, Cheng Q. The benefits of hesperidin in central nervous system disorders, based on the neuroprotective effect. Biomed Pharmacother 2023; 159:114222. [PMID: 36628819 DOI: 10.1016/j.biopha.2023.114222] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Disorders of central nervous system (CNS) disorders are considered serious health issues. The most common CNS diseases include neurodegenerative diseases, mental disorders, demyelinating disease, ischemia-reperfusion injury, and neuroinflammation. As a natural phenolic compound, hesperidin is a flavanone glycoside with various biological effects. Increasing evidence show that the growth of CNS diseases is hindered by hesperidin. Here, we have reviewed the related literature on neuropharmacological mechanisms for the preventive and therapeutic effects of hesperidin on CNS diseases. Several cellular and animal models have been developed to evaluate the underlying neuropharmacological mechanisms of hesperidin. Additionally, clinical evidence has confirmed its neuroprotective function. Hesperidin exerts its neuroprotective properties by decreasing neuro-inflammatory and apoptotic pathways. Hesperidin function has been studied in preclinical models for CNS diseases, but little is known about its definite effect in humans. Hesperidin can effectively alleviate depression and improve cognition and memory. It is urgent to explore and discover clinical trials for further confirmation of the neuroprotective efficacy of hesperidin and to evaluate its safety profile.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rongrong Tan
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Caijuan Xu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Xi Chen
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Sixin Li
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Ying Liu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Huiwen Qiu
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China
| | - Hui Cao
- Department of Psychiatry, The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China; Department of Psychiatry, Brain Hospital of Hunan Province (The Second People's Hospital of Hunan Province), Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| |
Collapse
|
16
|
Bellavite P. Neuroprotective Potentials of Flavonoids: Experimental Studies and Mechanisms of Action. Antioxidants (Basel) 2023; 12:antiox12020280. [PMID: 36829840 PMCID: PMC9951959 DOI: 10.3390/antiox12020280] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Neurological and neurodegenerative diseases, particularly those related to aging, are on the rise, but drug therapies are rarely curative. Functional disorders and the organic degeneration of nervous tissue often have complex causes, in which phenomena of oxidative stress, inflammation and cytotoxicity are intertwined. For these reasons, the search for natural substances that can slow down or counteract these pathologies has increased rapidly over the last two decades. In this paper, studies on the neuroprotective effects of flavonoids (especially the two most widely used, hesperidin and quercetin) on animal models of depression, neurotoxicity, Alzheimer's disease (AD) and Parkinson's disease are reviewed. The literature on these topics amounts to a few hundred publications on in vitro and in vivo models (notably in rodents) and provides us with a very detailed picture of the action mechanisms and targets of these substances. These include the decrease in enzymes that produce reactive oxygen and ferroptosis, the inhibition of mono-amine oxidases, the stimulation of the Nrf2/ARE system, the induction of brain-derived neurotrophic factor production and, in the case of AD, the prevention of amyloid-beta aggregation. The inhibition of neuroinflammatory processes has been documented as a decrease in cytokine formation (mainly TNF-alpha and IL-1beta) by microglia and astrocytes, by modulating a number of regulatory proteins such as Nf-kB and NLRP3/inflammasome. Although clinical trials on humans are still scarce, preclinical studies allow us to consider hesperidin, quercetin, and other flavonoids as very interesting and safe dietary molecules to be further investigated as complementary treatments in order to prevent neurodegenerative diseases or to moderate their deleterious effects.
Collapse
|
17
|
Adedara AO, Otenaike TA, Olabiyi AA, Adedara IA, Abolaji AO. Neurotoxic and behavioral deficit in Drosophila melanogaster co-exposed to rotenone and iron. Metab Brain Dis 2023; 38:349-360. [PMID: 36308588 DOI: 10.1007/s11011-022-01104-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/10/2022] [Indexed: 02/03/2023]
Abstract
Exposure to environmental toxicants has been linked with the onset of different neurodegenerative diseases in animals and humans. Here, we evaluated the toxic effects of co-exposure to iron and rotenone at low concentrations in Drosophila melanogaster. Adult wild-type flies were orally exposed to rotenone (50.0 µM) and ferrous sulfate (FeSO4; 1.0 and 10.0 µM) through the diet for 10 days. Thereafter, we evaluated markers of oxidative damage (Hydrogen Peroxide (H2O2), Nitric Oxide (NO), Protein Carbonyl, and malondialdehyde (MDA)), antioxidant status (catalase, Glutathione S-Transferase (GST), Total Thiol (T-SH) and Non-protein Thiol (NPSH), neurotransmission (monoamine oxidase; MAO and acetylcholinesterase, AChE) and mitochondrial respiration. The results indicated that flies fed rotenone and FeSO4 had impaired locomotion, reduced survival rate, and AChE activity with a corresponding increase in MAO activity when compared with the control (p < 0.05). Furthermore, rotenone and FeSO4 significantly decreased the antioxidant status with a concurrent accumulation of NO, MDA, and H2O2. Additionally, the activity of complex 1 and mitochondria bioenergetic capacity was compromised in the flies. These findings suggest that the combination of rotenone and FeSO4 elicited a possible synergistic toxic response in the flies and therefore provided further insights on the use of D. melanogaster in toxicological studies.
Collapse
Affiliation(s)
- Adeola O Adedara
- Drosophila Research and Training Centre, A2 Ajao Dental Street, Salami Somade Estate, Basorun, Ibadan, Nigeria
- Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Programa de Pos-Graduaçao em Bioquímica Toxicologica, Centro de Ciencias Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Titilayomi A Otenaike
- Drosophila Research and Training Centre, A2 Ajao Dental Street, Salami Somade Estate, Basorun, Ibadan, Nigeria
| | - Ayodeji A Olabiyi
- Programa de Pos-Graduaçao em Bioquímica Toxicologica, Centro de Ciencias Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, Brazil
- Department of Medical Biochemistry, Afe Babalola University, Ado Ekiti, Nigeria
| | - Isaac A Adedara
- Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Amos O Abolaji
- Drosophila Research and Training Centre, A2 Ajao Dental Street, Salami Somade Estate, Basorun, Ibadan, Nigeria.
- Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| |
Collapse
|
18
|
Camilo CJ, Leite DOD, da S Mendes JW, Dantas AR, de Carvalho NKG, Castro JWG, Salazar GJT, Ferreira MKA, de Meneses JEA, da Silva AW, Dos Santos HS, Tavares JF, Silva JPRE, Rodrigues FFG, Cheon C, Kim B, da Costa JGM. Analysis toxicity by different methods and anxiolytic effect of the aqueous extract Lippia sidoides Cham. Sci Rep 2022; 12:20626. [PMID: 36450779 PMCID: PMC9712538 DOI: 10.1038/s41598-022-23999-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Lippia sidoides Cham. (Verbenaceae) is a species often mentioned in traditional medicine due to the medicinal properties attributed to its leaves, which include antibacterial, antifungal, acaricidal and antioxidant. Several of these actions have been scientifically proven, according to reports in the literature; however, little is known about toxicological aspects of this plant. This work included studies to determine the chemical composition and toxicity tests, using several methods aiming to evaluate the safety for use of the aqueous extract of L. sidoides leaves, in addition, the anxiolytic effect on adult zebrafish was investigated, thus contributing to the pharmacological knowledge and traditional medicine concerning the specie under study. The chemical profile was determined by liquid chromatography coupled to mass spectrometry-HPLC/MS with electrospray ionization. Toxicity was evaluated by zebrafish, Drosophila melanogaster, blood cells, and Artemia salina models. 12 compounds belonging to the flavonoid class were identified. In the toxicity assays, the observed results showed low toxicity of the aqueous extract in all tests performed. In the analysis with zebrafish, the highest doses of the extract were anxiolytic, neuromodulating the GABAa receptor. The obtained results support the safe use of the aqueous extract of L. sidoides leaves for the development of new drugs and for the use by populations in traditional medicine.
Collapse
Affiliation(s)
- Cicera J Camilo
- Postgraduate Program in Ethnobiology and Nature Conservation, Federal Rural University of Pernambuco, R. Dr. Miguel, Parnamirim, PE, 56163-000, Brazil
| | - Débora O D Leite
- Northeast Biotechnology Network-RENORBIO, Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, 60714-903, Brazil
| | - Johnatan W da S Mendes
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil
| | - Alexandro R Dantas
- Natural Products Research Laboratory, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil
| | - Natália K G de Carvalho
- Natural Products Research Laboratory, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil
| | - José W G Castro
- Graduate Program in Biological Diversity and Natural Resources, Regional University of Cariri, Crato, Brazil
| | - Gerson J T Salazar
- Postgraduate Program in Ethnobiology and Nature Conservation, Federal Rural University of Pernambuco, R. Dr. Miguel, Parnamirim, PE, 56163-000, Brazil
| | | | | | - Antonio Wlisses da Silva
- Northeast Biotechnology Network-RENORBIO, Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, 60714-903, Brazil
| | - Helcio S Dos Santos
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil
| | - Josean F Tavares
- Multiuser Laboratory of Characterization and Analysis, Federal University of Paraíba, João Pessoa, 58051-900, Brazil
| | - Joanda P R E Silva
- Multiuser Laboratory of Characterization and Analysis, Federal University of Paraíba, João Pessoa, 58051-900, Brazil
| | - Fabiola F G Rodrigues
- Graduate Program in Biological Diversity and Natural Resources, Regional University of Cariri, Crato, Brazil
| | - Chunhoo Cheon
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Kyungheedae-Ro 26 Dongdaemun-Gu, Seoul, 05254, South Korea
| | - Bonglee Kim
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Kyungheedae-Ro 26 Dongdaemun-Gu, Seoul, 05254, South Korea.
| | - José Galberto Martins da Costa
- Northeast Biotechnology Network-RENORBIO, Graduate Program in Biotechnology, State University of Ceará, Fortaleza, Ceará, 60714-903, Brazil.
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil.
- Natural Products Research Laboratory, Regional University of Cariri, Crato, Ceará, 63105-00, Brazil.
| |
Collapse
|
19
|
Tonk-Rügen M, Vilcinskas A, Wagner AE. Insect Models in Nutrition Research. Biomolecules 2022; 12:1668. [PMID: 36421682 PMCID: PMC9687203 DOI: 10.3390/biom12111668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 09/08/2024] Open
Abstract
Insects are the most diverse organisms on earth, accounting for ~80% of all animals. They are valuable as model organisms, particularly in the context of genetics, development, behavior, neurobiology and evolutionary biology. Compared to other laboratory animals, insects are advantageous because they are inexpensive to house and breed in large numbers, making them suitable for high-throughput testing. They also have a short life cycle, facilitating the analysis of generational effects, and they fulfil the 3R principle (replacement, reduction and refinement). Many insect genomes have now been sequenced, highlighting their genetic and physiological similarities with humans. These factors also make insects favorable as whole-animal high-throughput models in nutritional research. In this review, we discuss the impact of insect models in nutritional science, focusing on studies investigating the role of nutrition in metabolic diseases and aging/longevity. We also consider food toxicology and the use of insects to study the gut microbiome. The benefits of insects as models to study the relationship between nutrition and biological markers of fitness and longevity can be exploited to improve human health.
Collapse
Affiliation(s)
- Miray Tonk-Rügen
- Institute of Nutritional Science, Justus Liebig University, Wilhelmstrasse 20, 35392 Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich-Buff-Ring 26-32, 35392 Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch of Bioresources, Ohlebergsweg 12, 35392 Giessen, Germany
| | - Anika E. Wagner
- Institute of Nutritional Science, Justus Liebig University, Wilhelmstrasse 20, 35392 Giessen, Germany
| |
Collapse
|
20
|
Bianchini MC, Soares LFW, Sousa JMFM, Ramborger BP, Gayer MC, Bridi JC, Roehrs R, Pinton S, Aschner M, Ávila DS, Puntel RL. MeHg exposure impairs both the catecholaminergic and cholinergic systems resulting in motor and non-motor behavioral changes in Drosophila melanogaster. Chem Biol Interact 2022; 365:110121. [PMID: 35995257 DOI: 10.1016/j.cbi.2022.110121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
Human exposure to the natural environmental contaminant methylmercury (MeHg) has been associated to adverse health effects. Importantly, the mechanisms by which this organomercurial exerts its neurotoxicity have yet to be fully clarified. Therefore, the aim of this study was to evaluate whether exposure to MeHg alters dopamine (DA) and octopamine (OA) levels, acetylcholinesterase (AChE) activity and impacts both motor and non-motor behaviours. We studied the effect of MeHg by feeding 1-2 d old flies (male and females) with 25 and 50 μM MeHg for 4 d and determined effects on survival, motor and non-motor behaviours, oxidative stress, AChE and tyrosine hydroxylase (TH) activities, as well as DA and OA levels. We found that Drosophila melanogaster (D. melanogaster) exposed to MeHg showed a reduction in survival rate, associated with the inhibition of AChE and TH activities in head of flies and decreased DA and OA levels. These changes were accompanied by behavioural alterations, such as locomotor deficit and increased grooming behaviour, in addition to an increase in oxidative stress markers both in head and in body of flies, and an increase in glutathione-S-transferase (GST) activity in head of flies. Collectively, our data support the hypothesis that MeHg neurotoxicity is associated with altered OA and DA levels, AChE inhibition, which may serve, at least in part, as the underpinnings of both motor and non-motor behavioural changes.
Collapse
Affiliation(s)
- Matheus C Bianchini
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Luiz F W Soares
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - João M F M Sousa
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Bruna P Ramborger
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Mateus C Gayer
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Jessika C Bridi
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Rafael Roehrs
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Simone Pinton
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, 10461, NY, United States
| | - Daiana S Ávila
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil
| | - Robson L Puntel
- Universidade Federal do Pampa - Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), Uruguaiana, RS, Brazil.
| |
Collapse
|
21
|
Barberino RS, Silva RLS, Palheta Junior RC, Smitz JEJ, Matos MHT. Protective Effects of Antioxidants on Cyclophosphamide-Induced Ovarian Toxicity. Biopreserv Biobank 2022; 21:121-141. [PMID: 35696235 DOI: 10.1089/bio.2021.0159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The most common limitation of anticancer chemotherapy is the injury to normal cells. Cyclophosphamide, which is one of the most widely used alkylating agents, can cause premature ovarian insufficiency and infertility since the ovarian follicles are extremely sensitive to their effects. Although little information is available about the pathogenic mechanism of cyclophosphamide-induced ovarian damage, its toxicity is attributed to oxidative stress, inflammation, and apoptosis. The use of compounds with antioxidant and cytoprotective properties to protect ovarian function from deleterious effects during chemotherapy would be a significant advantage. Thus, this article reviews the mechanism by which cyclophosphamide exerts its toxic effects on the different cellular components of the ovary, and describes 24 cytoprotective compounds used to ameliorate cyclophosphamide-induced ovarian injury and their possible mechanisms of action. Understanding these mechanisms is essential for the development of efficient and targeted pharmacological complementary therapies that could protect and prolong female fertility.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Regina Lucia S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta Junior
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, Free University Brussels-VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| |
Collapse
|
22
|
Chen J, Xu J, Huang P, Luo Y, Shi Y, Ma P. The potential applications of traditional Chinese medicine in Parkinson's disease: A new opportunity. Biomed Pharmacother 2022; 149:112866. [PMID: 35367767 DOI: 10.1016/j.biopha.2022.112866] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
Parkinson's disease (PD) presents a common challenge for people all over the world and has become a major research hotspot due to the large population affected by the illness and the difficulty of clinical treatment. The prevalence of PD is increasing every year, the pathogenesis is complex, and the current treatment is ineffective. Therefore, it has become imperative to find effective drugs for PD. With the advantages of low cost, high safety and high biological activity, Chinese medicine has great advantages in the prevention and treatment of PD. This review systematically summarizes the potential of Chinese medicine for the treatment of PD, showing that Chinese medicine can exert anti-PD effects through various pathways, such as anti-inflammatory and antioxidant pathways, reducing mitochondrial dysfunction, inhibiting endoplasmic reticulum stress and iron death, and regulating intestinal flora. These mainly involve HMGB1/TLR4, PI3K/Akt, NLRP3/ caspase-1/IL-1β, Nrf2/HO-1, SIRT1/Akt1, PINK1/parkin, Bcl-2/Bax, BDNF-TrkB and other signaling pathways. In sum, based on modern phytochemistry, pharmacology and genomic proteomics, Chinese medicine is likely to be a potential candidate for PD treatment, which requires more clinical trials to further elucidate its importance in the treatment of PD.
Collapse
Affiliation(s)
- Jiaxue Chen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingke Xu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ping Huang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yining Luo
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuanshu Shi
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ping Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
23
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
24
|
Improvement of non-motor and motor behavioral alterations associated with Parkinson-like disease in Drosophila melanogaster: comparative effects of treatments with hesperidin and L-dopa. Neurotoxicology 2022; 89:174-183. [DOI: 10.1016/j.neuro.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 12/15/2021] [Accepted: 02/10/2022] [Indexed: 11/19/2022]
|
25
|
Gulcan HO. Selected natural and synthetic agents effective against Parkinson's disease with diverse mechanisms. Curr Top Med Chem 2021; 22:199-208. [PMID: 34844541 DOI: 10.2174/1568026621666211129141316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/08/2021] [Accepted: 11/28/2021] [Indexed: 11/22/2022]
Abstract
Similar to other neurodegenerative diseases, Parkinson's disease (PD) has been extensively investigated with respect to its neuropathological background and possible treatment options. Since the symptomatic outcomes are generally related to dopamine deficiency, the current treatment strategies towards PD mainly employ dopaminergic agonists as well as the compounds acting on dopamine metabolism. These drugs do not provide disease modifying properties; therefore alternative drug discovery studies focus on targets involved in the progressive neurodegenerative character of PD. This study has aimed to present the pathophysiology of PD concomitant to the representation of drugs and promising molecules displaying activity against the validated and non-validated targets of PD.
Collapse
Affiliation(s)
- Hayrettin Ozan Gulcan
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, 99520, T.R. North Cyprus, via Mersin 10. Turkey
| |
Collapse
|
26
|
Oyebode OT, Abolaji AO, Faleke HO, Olorunsogo OO. Methanol fraction of Ficus mucoso (welw) prevents iron-induced oxidative damage and alters mitochondrial dysfunction in Drosophila melanogaster. Drug Chem Toxicol 2021; 45:2644-2652. [PMID: 34592861 DOI: 10.1080/01480545.2021.1979997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The present study investigated the antioxidant and cyto-/mito-protective roles of Methanol Fraction of Ficus mucoso (MFFM) in iron-induced oxidative damage in Drosophila melanogaster. At first, 10-day survival rates were carried out separately on FeSO4 and MFFM, respectively, after which ameliorative effects of MFFM were investigated on FeSO4-induced toxicity for 5 days using biochemical and behavioral markers. Additionally, mitochondria were isolated from treated D. melanogaster to assess mitochondrial Permeability Transition (mPT) pore opening. The results showed that FeSO4 significantly reduced survival rate, total thiol level and activities of catalase and glutathione-S-transferase in D. melanogaster. In addition, treatment with FeSO4 caused increased generation of H2O2, NO (nitrite/nitrates) and acetylcholinesterase (AChE) activity compared with control (p < 0.05). Conversely, MFFM restored FeSO4-induced inhibition of glutathione-S-transferase and catalase activities, as well as glutathione and total thiol levels. FeSO4-induced elevation of AChE activity as well as H2O2 and NO (nitrites/nitrates) levels were ameliorated by MFFM with improved climbing activity. Interestingly, MFFM prevented FeSO4-induced mitochondrial Permeability Transition (mPT) pore opening, and elevated mitochondrial ATPase activity and mitochondrial lipid peroxides generation in D. melanogaster. Taken together, our results demonstrated that iron impaired anti-stress defence capacity, altered behavioral functions, increased generation of mitochondrial malondialdehyde and activated opening of the mPT pore in D. melanogaster. Conversely, methanol fraction of F. mucoso protected against iron-induced cyto-/mito-toxic effects. F. mucoso possibly contain bioactive agents which might be useful in the management of disorders associated with oxidative stress induced by iron and or related metals.
Collapse
Affiliation(s)
- Olubukola T Oyebode
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Amos O Abolaji
- Molecular Drug Metabolism and Toxicology, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Hammed O Faleke
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria.,Molecular Drug Metabolism and Toxicology, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| | - Olufunso O Olorunsogo
- Laboratories for Biomembrane Research and Biotechnology, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
27
|
Musachio EAS, de Freitas Couto S, Poetini MR, Bortolotto VC, Dahleh MMM, Janner DE, Araujo SM, Ramborger BP, Rohers R, Guerra GP, Prigol M. Bisphenol A exposure during the embryonic period: Insights into dopamine relationship and behavioral disorders in Drosophila melanogaster. Food Chem Toxicol 2021; 157:112526. [PMID: 34461193 DOI: 10.1016/j.fct.2021.112526] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/06/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022]
Abstract
Environmental factors are involved in the pathogenesis of neurodevelopmental disorders in addition to genetic factors. In this sense, we demonstrated here that the embryonic exposure of Drosophila melanogaster to Bisphenol A (BPA) 1 mM resulted in changes in development, behavior, and biochemical markers punctuated below. BPA did not alter the oviposition and viability of the eggs, however, it was evidenced a decrease in the rate of pupal eclosion and life span of the hatched flies of the generation filial 1 (F1). F1 flies also developed behavioral changes such as incompatibility in the social interaction between them, and hyperactivity demonstrated by increased locomotion in open field tests, increased grooming, and aggression episodes. Furthermore, decreases in dopamine levels and tyrosine hydroxylase activity have also been observed in flies' heads, possibly related to oxidative damage. Through analyzes of oxidative stress biomarkers, carried out on samples of flies' heads, we observed an increase in malondialdehyde and reactive species, decrease in the activity of the superoxide dismutase and catalase, which possibly culminated in the reduction of cell viability. Thus, it is important to emphasize that BPA developed atypical behaviors in Drosophila melanogaster, reinforce the importance of the environmental factor in the development of neurobehavioral diseases.
Collapse
Affiliation(s)
- Elize Aparecida Santos Musachio
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Shanda de Freitas Couto
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil; Department of Nutrition, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Marcia Rósula Poetini
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Mustafa Munir Mustafa Dahleh
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Dieniffer Espinosa Janner
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Stífani Machado Araujo
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Bruna Piaia Ramborger
- Interdisciplinary Group of Research in Teaching Practice, Federal University of Pampa, Uruguaiana Campus, RS, Brazil
| | - Rafael Rohers
- Interdisciplinary Group of Research in Teaching Practice, Federal University of Pampa, Uruguaiana Campus, RS, Brazil
| | - Gustavo Petri Guerra
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil; Department of Nutrition, Federal University of Pampa, Itaqui Campus, RS, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules, Federal University of Pampa, Itaqui Campus, RS, Brazil; Department of Nutrition, Federal University of Pampa, Itaqui Campus, RS, Brazil.
| |
Collapse
|
28
|
Walczak-Nowicka ŁJ, Herbet M. Acetylcholinesterase Inhibitors in the Treatment of Neurodegenerative Diseases and the Role of Acetylcholinesterase in their Pathogenesis. Int J Mol Sci 2021; 22:9290. [PMID: 34502198 PMCID: PMC8430571 DOI: 10.3390/ijms22179290] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022] Open
Abstract
Acetylcholinesterase (AChE) plays an important role in the pathogenesis of neurodegenerative diseases by influencing the inflammatory response, apoptosis, oxidative stress and aggregation of pathological proteins. There is a search for new compounds that can prevent the occurrence of neurodegenerative diseases and slow down their course. The aim of this review is to present the role of AChE in the pathomechanism of neurodegenerative diseases. In addition, this review aims to reveal the benefits of using AChE inhibitors to treat these diseases. The selected new AChE inhibitors were also assessed in terms of their potential use in the described disease entities. Designing and searching for new drugs targeting AChE may in the future allow the discovery of therapies that will be effective in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8bStreet, 20-090 Lublin, Poland;
| |
Collapse
|
29
|
Nutraceutical Strategy to Counteract Eye Neurodegeneration and Oxidative Stress in Drosophila melanogaster Fed with High-Sugar Diet. Antioxidants (Basel) 2021; 10:antiox10081197. [PMID: 34439445 PMCID: PMC8388935 DOI: 10.3390/antiox10081197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022] Open
Abstract
Aberrant production of reactive oxygen species (ROS) is a common feature of damaged retinal neurons in diabetic retinopathy, and antioxidants may exert both preventive and therapeutic action. To evaluate the beneficial and antioxidant properties of food supplementation with Lisosan G, a powder of bran and germ of grain (Triticum aestivum) obtained by fermentation with selected lactobacillus and natural yeast strains, we used an in vivo model of hyperglycemia-induced retinal damage, the fruit fly Drosophila melanogaster fed with high-sucrose diet. Lisosan G positively affected the visual system of hyperglycemic flies at structural/functional level, decreased apoptosis, and reactivated protective autophagy at the retina internal network. Also, in high sucrose-fed Drosophila, Lisosan G reduced the levels of brain ROS and retina peroxynitrite. The analysis of oxidative stress-related metabolites suggested 7,8-dihydrofolate, uric acid, dihydroorotate, γ-L-glutamyl-L-cysteine, allantoin, cysteinyl-glycine, and quinolate as key mediators of Lisosan G-induced inhibition of neuronal ROS, along with the upregulation of glutathione system. Of note, Lisosan G may impact oxidative stress and the ensuing retinal cell death, also independently from autophagy, although the autophagy-ROS cross-talk is critical. This study demonstrated that the continuous supplementation with the alimentary integrator Lisosan G exerts a robust and multifaceted antioxidant effect on retinal neurons, thus providing efficacious neuroprotection of hyperglycemic eye.
Collapse
|
30
|
Silva NC, Poetini MR, Bianchini MC, Almeida FP, Dahle MMM, Araujo SM, Bortolotto VC, Musachio EAS, Ramborger BP, Novo DLR, Roehrs R, Mesko MF, Prigol M, Puntel RL. Protective effect of gamma-oryzanol against manganese-induced toxicity in Drosophila melanogaster. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:17519-17531. [PMID: 33403631 DOI: 10.1007/s11356-020-11848-z] [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: 09/23/2020] [Accepted: 11/25/2020] [Indexed: 06/12/2023]
Abstract
Manganese (Mn) is an essential element that, in excess, seems to be involved in the development of different neurodegenerative conditions. Gamma-oryzanol (Ory) was previously reported to possess antioxidant and neuroprotective properties. Thus, we conducted this study to test the hypothesis that Ory can also protect flies in an Mn intoxication model. Adult wild-type flies were fed over 10 days with Mn (5 mM) and/or Ory (25 μM). Flies treated with Mn had a decrease in locomotor activity and a higher mortality rate compared to those in controls. Mn-treated flies also had a significant increase in acetylcholinesterase (AChE) activity, in Mn accumulation and in oxidative stress markers. Moreover, flies treated with Mn exhibited a significant decrease in dopamine levels and in tyrosine hydroxylase activity, as well as in mitochondrial and cellular viability. Particularly important, Ory protected against mortality and avoided locomotor and biochemical changes associated with Mn exposure. However, Ory did not prevent the accumulation of Mn. The present results support the notion that Ory effectively attenuates detrimental changes associated with Mn exposure in Drosophila melanogaster, reinforcing its neuroprotective action/potential.
Collapse
Affiliation(s)
- Neicí Cáceres Silva
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Márcia Rósula Poetini
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Matheus Chimelo Bianchini
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil
| | - Francielli Polet Almeida
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Mustafá Munir Mustafa Dahle
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Stífani Machado Araujo
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Vandreza Cardoso Bortolotto
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Elize Aparecida Santos Musachio
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil
| | - Bruna Piaia Ramborger
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil
| | - Diogo La Rosa Novo
- Universidade Federal de Pelotas, Campus Universitário, S/N - Prédio/Bloco: 30 e 32, Capão do Leão, RS, CEP 96160-000, Brazil
| | - Rafael Roehrs
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil
| | - Marcia Foster Mesko
- Universidade Federal de Pelotas, Campus Universitário, S/N - Prédio/Bloco: 30 e 32, Capão do Leão, RS, CEP 96160-000, Brazil
| | - Marina Prigol
- Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Programa de Pós-Graduação em Bioquímica (PPGBioq), Rua Joaquim de Sá Britto, s/n - Bairro Promorar, Itaqui, RS, CEP 97650-000, Brazil.
| | - Robson Luiz Puntel
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil.
| |
Collapse
|
31
|
Poetini MR, Musachio EAS, Araujo SM, Almeida FP, Dahleh MMM, Bortolotto VC, Janner DE, Pinheiro FC, Ramborger BP, Roehrs R, La Rosa Novo D, Mesko MF, Guerra GP, Prigol M. Iron overload during the embryonic period develops hyperactive like behavior and dysregulation of biogenic amines in Drosophila melanogaster. Dev Biol 2021; 475:80-90. [PMID: 33741348 DOI: 10.1016/j.ydbio.2021.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
Iron (Fe) is used in various cellular functions, and a constant balance between its uptake, transport, storage, and use is necessary to maintain its homeostasis in the body. Changes in Fe metabolism with a consequent overload of this metal are related to neurological changes and cover a broad spectrum of diseases, mainly when these changes occur during the embryonic period. This work aimed to evaluate the effect of exposure to Fe overload during the embryonic period of Drosophila melanogaster. Progenitor flies (male and female) were exposed to ferrous sulfate (FeSO4) for ten days in concentrations of 0.5, 1, and 5 mM. After mating and oviposition, the progenitors were removed and the treatment bottles preserved, and the number of daily hatches and cumulative hatching of the first filial generation (F1) were counted. Subsequently, F1 flies (separated by sex) were subjected to behavioral tests such as negative geotaxis test, open field test, grooming, and aggression test. They have evaluated the levels of dopamine (DA), serotonin (5-HT), octopamine (OA), tryptophan and tyrosine hydroxylase (TH), acetylcholinesterase, reactive species, and the levels of Fe in the progenitor flies and F1. The Fe levels of F1 flies are directly proportional to what is incorporated during the period of embryonic development; we also observed a delay in hatching and a reduction in the number of the hatch of F1 flies exposed during the embryonic period to the 5mM Fe diet, a fact that may be related to the reduction of the cell viability of the ovarian tissue of progenitor flies. The flies exposed to Fe (1 and 5 mM) showed an increase in locomotor activity (hyperactivity) and a significantly higher number of repetitive movements. In addition to a high number of aggressive encounters when compared to control flies. We can also observe an increase in the levels of biogenic amines DA and 5-HT and an increase in TH activity in flies exposed to Fe (1 and 5 mM) compared to the control group. We conclude that the hyperactive-like behavior demonstrated in both sexes by F1 flies exposed to Fe may be associated with a dysregulation in the levels of DA and 5-HT since Fe is a cofactor of TH, which had its activity increased in this study. Therefore, more attention is needed during the embryonic development period for exposure to Fe overload.
Collapse
Affiliation(s)
- Márcia Rósula Poetini
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Stífani Machado Araujo
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Francielli Polet Almeida
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Mustafa Munir Mustafa Dahleh
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Dieniffer Espinosa Janner
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Franciane Cabral Pinheiro
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Bruna Piaia Ramborger
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil
| | - Rafael Roehrs
- Universidade Federal do Pampa, Campus Uruguaiana, Programa de Pós-Graduação em Bioquímica (PPGBioq), BR-472 Km 7, Uruguaiana, RS, CEP 97500-970, Brazil
| | - Diogo La Rosa Novo
- Universidade Federal de Pelotas, Campus Universitário, S/N - Prédio/Bloco: 30 e 32, Capão do Leão, Rio Grande do Sul, CEP 96160-000, Brazil
| | - Márcia Foster Mesko
- Universidade Federal de Pelotas, Campus Universitário, S/N - Prédio/Bloco: 30 e 32, Capão do Leão, Rio Grande do Sul, CEP 96160-000, Brazil
| | - Gustavo Petri Guerra
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Marina Prigol
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas- LaftamBio Pampa, Universidade Federal do Pampa (UNIPAMPA), Campus Itaqui, Rua Joaquim de Sá Britto, s/n, Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil.
| |
Collapse
|
32
|
Sandner G, König A, Wallner M, Weghuber J. Alternative model organisms for toxicological fingerprinting of relevant parameters in food and nutrition. Crit Rev Food Sci Nutr 2021; 62:5965-5982. [PMID: 33683153 DOI: 10.1080/10408398.2021.1895060] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In the field of (food) toxicology, there is a strong trend of replacing animal trials with alternative methods for the assessment of adverse health effects in humans. The replacement of animal trials is not only driven by ethical concerns but also by the number of potential testing substances (food additives, packaging material, contaminants, and toxicants), which is steadily increasing. In vitro 2D cell culture applications in combination with in silico modeling might provide an applicable first response. However, those systems lack accurate predictions of metabolic actions. Thus, alternative in vivo models could fill the gap between cell culture and animal trials. In this review, we highlight relevant studies in the field and spotlight the applicability of alternative models, including C. elegans, D. rerio, Drosophila, HET-CAM and Lab-on-a-chip.
Collapse
Affiliation(s)
- Georg Sandner
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria
| | - Alice König
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria.,FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Melanie Wallner
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria.,FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria.,FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Tulln, Austria
| |
Collapse
|
33
|
Silva B, Biluca FC, Gonzaga LV, Fett R, Dalmarco EM, Caon T, Costa ACO. In vitro anti-inflammatory properties of honey flavonoids: A review. Food Res Int 2021; 141:110086. [PMID: 33641965 DOI: 10.1016/j.foodres.2020.110086] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022]
Abstract
Honey is a natural ready-to-eat product rich in flavonoids, which is known by the wound healing properties due to both antibacterial and antioxidant activity. Flavonoids mitigate inflammatory processes, and thus it could currently support studies of anti-inflammatory potential of honeys. In this review, in vitro anti-inflammatory properties of flavonoids found in honey were prioritized. Mechanistic information of specific isolated flavonoids as modulators of inflammatory processes are summarized aiming to stimulate studies regarding the action of honey in inflammatory events. Lastly, a structure-activity relationship (SAR) of flavonoids was also included. Flavonoids found in honey have demonstrated antioxidant properties and ability to inhibit pro-inflammatory enzymes such as COX, LOX, iNOS, and pro-inflammatory mediators, including nitric oxide, cytokines and chemokines. Transcriptional factors such as NF-κB are also modulated by flavonoids, controlling the expression of several inflammatory mediators. SAR studies demonstrate the effect of flavonoids in the prevention of inflammatory cascades. Despite the promising reports of in vitro anti-inflammatory activity, well-designed clinical trials need yet to be performed to confirm the benefits of honeys from different botanical sources in diseases that include episodes of inflammation.
Collapse
Affiliation(s)
- Bibiana Silva
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, SC, Brazil.
| | - Fabíola Carina Biluca
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Luciano Valdemiro Gonzaga
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Roseane Fett
- Department of Food Science and Technology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | | | - Thiago Caon
- Department of Pharmaceutical Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | | |
Collapse
|
34
|
Maan G, Sikdar B, Kumar A, Shukla R, Mishra A. Role of Flavonoids in Neurodegenerative Diseases: Limitations and Future Perspectives. Curr Top Med Chem 2021; 20:1169-1194. [PMID: 32297582 DOI: 10.2174/1568026620666200416085330] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Flavonoids, a group of natural dietary polyphenols, are known for their beneficial effects on human health. By virtue of their various pharmacological effects, like anti-oxidative, antiinflammatory, anti-carcinogenic and neuroprotective effects, flavonoids have now become an important component of herbal supplements, pharmaceuticals, medicinals and cosmetics. There has been enormous literature supporting neuroprotective effect of flavonoids. Recently their efficacy in various neurodegenerative diseases, like Alzheimer's disease and Parkinson diseases, has received particular attention. OBJECTIVE The mechanism of flavanoids neuroprotection might include antioxidant, antiapoptotic, antineuroinflammatory and modulation of various cellular and intracellular targets. In in-vivo systems, before reaching to brain, they have to cross barriers like extensive first pass metabolism, intestinal barrier and ultimately blood brain barrier. Different flavonoids have varied pharmacokinetic characteristics, which affect their pharmacodynamic profile. Therefore, brain accessibility of flavonoids is still debatable. METHODS This review emphasized on current trends of research and development on flavonoids, especially in neurodegenerative diseases, possible challenges and strategies to encounter using novel drug delivery system. RESULTS Various flavonoids have elicited their therapeutic potential against neurodegenerative diseases, however by using nanotechnology and novel drug delivery systems, the bioavailability of favonoids could be enhanced. CONCLUSION This study bridges a significant opinion on medicinal chemistry, ethanopharmacology and new drug delivery research regarding use of flavonoids in management of neurodegeneration.
Collapse
Affiliation(s)
- Gagandeep Maan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow-226002, U.P., India
| | - Biplab Sikdar
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow-226002, U.P., India
| | - Ashish Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow-226002, U.P., India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow-226002, U.P., India
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli (NIPER-R), Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow-226002, U.P., India
| |
Collapse
|
35
|
Devi S, Kumar V, Singh SK, Dubey AK, Kim JJ. Flavonoids: Potential Candidates for the Treatment of Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9020099. [PMID: 33498503 PMCID: PMC7909525 DOI: 10.3390/biomedicines9020099] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative disorders, such as Parkinson's disease (PD), Alzheimer's disease (AD), Amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), are the most concerning disorders due to the lack of effective therapy and dramatic rise in affected cases. Although these disorders have diverse clinical manifestations, they all share a common cellular stress response. These cellular stress responses including neuroinflammation, oxidative stress, proteotoxicity, and endoplasmic reticulum (ER)-stress, which combats with stress conditions. Environmental stress/toxicity weakened the cellular stress response which results in cell damage. Small molecules, such as flavonoids, could reduce cellular stress and have gained much attention in recent years. Evidence has shown the potential use of flavonoids in several ways, such as antioxidants, anti-inflammatory, and anti-apoptotic, yet their mechanism is still elusive. This review provides an insight into the potential role of flavonoids against cellular stress response that prevent the pathogenesis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Shweta Devi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, India;
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
- Correspondence: (V.K.); (J.-J.K.); Tel.: +82-10-9668-3464 (J.-J.K.); Fax: +82-53-801-3464 (J.-J.K.)
| | | | | | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea
- Correspondence: (V.K.); (J.-J.K.); Tel.: +82-10-9668-3464 (J.-J.K.); Fax: +82-53-801-3464 (J.-J.K.)
| |
Collapse
|
36
|
Jayapalan JJ, Subramanian P, Kani A, Hiji J, Najjar SG, Abdul-Rahman PS, Hashim OH. Hesperidin modulates the rhythmic proteomic profiling in Drosophila melanogaster under oxidative stress. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 105:e21738. [PMID: 32924199 DOI: 10.1002/arch.21738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 06/11/2023]
Abstract
The circadian clock regulates vital aspects of physiology including protein synthesis and oxidative stress response. In this investigation, we performed a proteome-wide scrutiny of rhythmic protein accrual in Drosophila melanogaster on exposure to rotenone, rotenone + hesperidin and hesperidin in D. melanogaster. Total protein from fly samples collected at 6 h intervals over the 24 h period was subjected to two-dimensional gel electrophoresis and mass spectrometry. Bioinformatics tool, Protein ANalysis THrough Evolutionary Relationships classification system was used to the determine the biological processes of the proteins of altered abundance. Conspicuous variations in the proteome (151 proteins) of the flies exposed to oxidative stress (by rotenone treatment) and after alleviating oxidative stress (by hesperidin treatment) were observed during the 24 h cycle. Significantly altered levels of abundance of a wide variety of proteins under oxidative stress (rotenone treatment) and under alleviation of oxidative stress (rotenone + hesperidin treatment) and hesperidin (alone) treatment were observed. These proteins are involved in metabolism, muscle activity, heat shock response, redox homeostasis, protein synthesis/folding/degradation, development, ion-channel/cellular transport, and gustatory and olfactory function of the flies. Our data indicates that numerous cellular processes are involved in the temporal regulation of proteins and widespread modulations happen under rotenone treatment and, action of hesperidin could also be seen on these categories of proteins.
Collapse
Affiliation(s)
- Jaime J Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Perumal Subramanian
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Akshaya Kani
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Jumriah Hiji
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sara G Najjar
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Puteri S Abdul-Rahman
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Onn H Hashim
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- University of Malaya Centre for Proteomics Research (UMCPR), Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Oxidative Stress in Parkinson's Disease: Potential Benefits of Antioxidant Supplementation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2360872. [PMID: 33101584 PMCID: PMC7576349 DOI: 10.1155/2020/2360872] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) occurs in approximately 1% of the population over 65 years of age and has become increasingly more common with advances in age. The number of individuals older than 60 years has been increasing in modern societies, as well as life expectancy in developing countries; therefore, PD may pose an impact on the economic, social, and health structures of these countries. Oxidative stress is highlighted as an important factor in the genesis of PD, involving several enzymes and signaling molecules in the underlying mechanisms of the disease. This review presents updated data on the involvement of oxidative stress in the disease, as well as the use of antioxidant supplements in its therapy.
Collapse
|
38
|
Elyasi L, Jahanshahi M, Jameie SB, Hamid Abadi HG, Nikmahzar E, Khalili M, Jameie M, Jameie M. 6-OHDA mediated neurotoxicity in SH-SY5Y cellular model of Parkinson disease suppressed by pretreatment with hesperidin through activating L-type calcium channels. J Basic Clin Physiol Pharmacol 2020; 32:11-17. [PMID: 32918805 DOI: 10.1515/jbcpp-2019-0270] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/11/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Parkinson's disease (PD) is a neurological condition with selective progressive degeneration of dopaminergic neurons. Routine therapies are symptomatic and palliative. Although, hesperidin (Hsd) is known for its neuroprotective effects, its exact cellular mechanism is still a mystery. Considering the important role of calcium (Ca2+) in cellular mechanisms of neurodegenerative diseases, the present study aimed to investigate the possible effects of Hsd on Ca2+ channels in cellular model of PD and the possible association between the selective vulnerability of neurons in cellular models of PD and expression of the physiological phenotype that changes Ca2+ homeostasis. METHODS SH-SY5Y cell line was used in this study; cell damage was induced by 150 µM 6-OHDA and the cells' viability was examined using MTT assay. Intracellular calcium, reactive oxygen species (ROS) and mitochondrial membrane potential were determined by the fluorescence spectrophotometry method. The expressions of calcium channel receptors were determined by gel electrophoresis and immunoblotting. RESULTS Loss of cell viability and mitochondrial membrane potential were confirmed in 6-OHDA treated cells. In addition, intracellular ROS and calcium levels, calcium channel receptors significantly increased in 6-OHDA-treated cells. Incubation of SH-SY5Y cells with hesperidin showed a protective effect, reduced the biochemical markers of cell damage/death, and balanced calcium hemostasis. CONCLUSIONS Based on our findings, it seems that hesperidin could suppress the progression of the cellular model of PD via acting on intracellular calcium homeostasis. Further studies are needed to confirm the potential benefits of preventive and therapeutic effects of stabilizing cellular calcium homeostasis in neurodegenerative disease.
Collapse
Affiliation(s)
- Leila Elyasi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - S B Jameie
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
| | - Hatef Ghasemi Hamid Abadi
- Immunogenetic Research Center, Department of Anatomy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Emsehgol Nikmahzar
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Masoumeh Khalili
- Neuroscience Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Melika Jameie
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mana Jameie
- Neuroscience Research Center (NRC), Iran University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Jaiswal P, Mandal M, Mishra A. Effect of hesperidin on fluoride-induced neurobehavioral and biochemical changes in rats. J Biochem Mol Toxicol 2020; 34:e22575. [PMID: 32627286 DOI: 10.1002/jbt.22575] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/24/2020] [Accepted: 06/23/2020] [Indexed: 01/15/2023]
Abstract
Fluoride is the second largest contaminant of drinking water. Fluoride toxicity is a major concern in the endemic areas where a high amount of fluoride is present in ground water. Oxidative stress has been proposed to be one of the mechanisms of fluoride-induced toxicity. Antioxidant-rich food has been found to alleviate fluoride-induced toxicity. Therefore, in this study, we have examined the effect of hesperidin on fluoride-induced neurobehavioral changes in rats. In the current study, male Sprague-Dawley rats were exposed to sodium fluoride through drinking water (120 ppm). Hesperidin (200 mg kg-1 d-1 ; per os) was administered either alone or in combination with fluoride-containing drinking water. Bisphinol A diglycidyl ether (BADGE) was used as peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist and was administered (10 mg kg-1 d-1 ; intraperitoneal injection) with/without hesperidin along with fluoride-containing drinking water. The behavioral changes in the animals were assessed by analyzing rotarod test, novel object recognition test, and forced swim test (FST). After 8 weeks, animals were killed to isolate blood and brain for monitoring biochemical changes. The 8-week exposure of fluoride resulted in motor impairment as observed with reduced fall time in rotarod test, memory impairment as observed with reduced preference index in novel object recognition test, and depression-like behavior as observed with reduced mobility index in the FST. Treatment with hesperidin improved neurobehavioral impairment along with restoration in brain biochemical changes (ie, acetylcholinesterase activity and antioxidant and oxidative stress parameters). The protective effect of hesperidin was reversed by coadministration of BADGE. The neuroprotective effect of hesperidin appears to be contributed through PPAR-γ receptor.
Collapse
Affiliation(s)
- Pawan Jaiswal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research - Raebareli (NIPER-R), Lucknow, India
| | - Mukesh Mandal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research - Raebareli (NIPER-R), Lucknow, India
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research - Raebareli (NIPER-R), Lucknow, India
| |
Collapse
|
40
|
Johnson SL, Iannucci J, Seeram NP, Grammas P. Inhibiting thrombin improves motor function and decreases oxidative stress in the LRRK2 transgenic Drosophila melanogaster model of Parkinson's disease. Biochem Biophys Res Commun 2020; 527:532-538. [PMID: 32423817 DOI: 10.1016/j.bbrc.2020.04.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease characterized by the presence of tremors, loss of dopaminergic neurons and accumulation of α-synuclein. While there is no single direct cause of PD, genetic mutations, exposure to pesticides, diet and traumatic brain injury have been identified as risk factors. Increasing evidence suggests that oxidative stress and neuroinflammation contribute to the pathogenesis of neuronal injury in neurodegenerative diseases such as PD and Alzheimer's disease (AD). We have previously documented that the multifunctional inflammatory mediator thrombin contributes to oxidative stress and neuroinflammation in AD. Here, for the first time, we explore the role of thrombin in a transgenic PD model, the LRRK2 mutant Drosophila melanogaster. Transgenic flies were treated with the direct thrombin inhibitor dabigatran for 7 days and locomotor activity and indices of oxidative stress evaluated. Our data show that dabigatran treatment significantly (p < 0.05) improved climbing activity, a measurement of locomotor ability, in male but had no effect on locomotor performance in female flies. Dabigatran treatment had no effect on tyrosine hydroxylase levels. Analysis of oxidative stress in male flies showed that dabigatran was able to significantly (p < 0.01) lower reactive oxygen species levels. Furthermore, Western blot analysis showed that the pro-oxidant proteins iNOS and NOX4 are elevated in LRRK2 male flies compared to wildtype and that treatment with dabigatran reduced expression of these proteins. Our results indicate that dabigatran treatment could improve motor function in PD by reducing oxidative stress. These data suggest that targeting thrombin may improve oxidative stress related pathologies in PD.
Collapse
Affiliation(s)
- Shelby L Johnson
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Jaclyn Iannucci
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Navindra P Seeram
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Paula Grammas
- The George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| |
Collapse
|
41
|
Adedayo BC, Ogunsuyi OB, Akinniyi ST, Oboh G. Effect ofAndrographis paniculataandPhyllanthus amarusleaf extracts on selected biochemical indices inDrosophila melanogastermodel of neurotoxicity. Drug Chem Toxicol 2020; 45:407-416. [DOI: 10.1080/01480545.2019.1708377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Bukola Christiana Adedayo
- Functional Foods and Nutraceuticals Unit of Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Opeyemi Babatunde Ogunsuyi
- Functional Foods and Nutraceuticals Unit of Biochemistry Department, Federal University of Technology, Akure, Nigeria
- Department of Biomedical Technology, Federal University of Technology, Akure, Nigeria
| | - Stephanie Tolulope Akinniyi
- Functional Foods and Nutraceuticals Unit of Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Functional Foods and Nutraceuticals Unit of Biochemistry Department, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
42
|
Save SS, Rachineni K, Hosur RV, Choudhary S. Natural compound safranal driven inhibition and dis-aggregation of α-synuclein fibrils. Int J Biol Macromol 2019; 141:585-595. [PMID: 31505208 DOI: 10.1016/j.ijbiomac.2019.09.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/25/2019] [Accepted: 09/06/2019] [Indexed: 10/26/2022]
Abstract
Self-assembly of α-synuclein (α-Syn) is linked with a variety of neurodegenerative diseases collectively called as α-synucleiopathies. Therefore, discovering suitable inhibitors for this self-association process of α-Syn is a subject of intense research. In this background, we have demonstrated here that the natural compound, Safranal, delays/inhibits α-Syn fibrillation/aggregation, and we have also characterized its mode of action. The α-Syn fibrillation/aggregation kinetics studies in combination with TEM studies demonstrated that Safranal effectively inhibits α-Syn fibrillation/aggregation. NMR studies revealed that Safranal binds with α-Syn and stabilizes the monomeric protein. ANS fluorescence and CD measurements indicated that Safranal binds to the hydrophobic residues of the protein and causes delay in the formation of β-sheet rich structures which are crucial for the fibrillation to occur. The results obtained from fluorescence quenching, NMR and ANS binding assays, when analysed taking into consideration the molecular structure of Safranal provide valuable insights into the mechanism of inhibition of α-Syn fibrillation/aggregation. We infer that inhibition of α-Syn fibrillation/aggregation is primarily driven by hydrophobic interactions between Safranal and the protein. Further, Safranal is also seen to dis-aggregates pre-formed α-Syn fibrils. These findings implicate that Safranal could become a potent therapeutic intervention in Parkinson's disease and other protein aggregation related disorders.
Collapse
Affiliation(s)
- Shreyada S Save
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidhyanagri Campus, Kalina, Mumbai 400098, India
| | - Kavitha Rachineni
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidhyanagri Campus, Kalina, Mumbai 400098, India
| | - Ramakrishna V Hosur
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidhyanagri Campus, Kalina, Mumbai 400098, India
| | - Sinjan Choudhary
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidhyanagri Campus, Kalina, Mumbai 400098, India.
| |
Collapse
|
43
|
da Silva LM, Pezzini BC, Somensi LB, Bolda Mariano LN, Mariott M, Boeing T, dos Santos AC, Longo B, Cechinel-Filho V, de Souza P, de Andrade SF. Hesperidin, a citrus flavanone glycoside, accelerates the gastric healing process of acetic acid-induced ulcer in rats. Chem Biol Interact 2019; 308:45-50. [PMID: 31095933 DOI: 10.1016/j.cbi.2019.05.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/25/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
|
44
|
Hu X, Fu W, Yang X, Mu Y, Gu W, Zhang M. Effects of cadmium on fecundity and defence ability of Drosophila melanogaster. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 171:871-877. [PMID: 30665104 DOI: 10.1016/j.ecoenv.2019.01.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 06/09/2023]
Abstract
Cadmium (chemical symbol, Cd) is an extremely common pollutant that poses a toxicity threat to organisms. Therefore, we tested Drosophila melanogaster fecundity, Cd accumulation, and activity of two enzymes following Cd stress and used quantitative real-time polymerase chain reaction (qPCR) to quantify the mRNA expression levels of several genes involved in fecundity and defence. D. melanogaster was placed in a medium containing different concentrations of Cd (13, 26, and 52 mg L-1), following which, inductively coupled plasma atomic emission spectroscopy showed that Cd accumulation in Drosophila increased with the increase in its dietary intake. We also observed that Cd at these concentrations significantly prolonged the mating latency in females and reduced the number of eggs laid. However, the same Cd concentrations did not affect male fecundity. Acetylcholinesterase activity was only detected at 52 mg L-1 Cd in both sexes, whereas glutathione S-transferase activity was inhibited at 26 and 52 mg L-1 Cd in females. The results of qPCR indicated that exposure to 13-52 mg L-1 Cd affected the expression of reproduction-related genes, including downregulation of enok and upregulation of dally and dpp. The same level of exposure also induced transcriptional responses from three defence-related genes (hsp70, gstd2, and gstd6). Taken together, the results revealed that Cd exposure might negatively affect the expression of genes associated with D. melanogaster reproduction and trigger the transcription of defence-related genes. We suggest that further analyses of fecundity and defence responses may help develop indicators of Cd toxicity and improve our understanding of antitoxin defences.
Collapse
Affiliation(s)
- Xiaoyu Hu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Weili Fu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Xingran Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yun Mu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Wei Gu
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Min Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
45
|
Natural Compounds for the Management of Parkinson's Disease and Attention-Deficit/Hyperactivity Disorder. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4067597. [PMID: 30596091 PMCID: PMC6282143 DOI: 10.1155/2018/4067597] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/31/2018] [Accepted: 11/11/2018] [Indexed: 11/18/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder with an unknown aetiology. The pathogenic mechanisms include oxidative stress, mitochondrial dysfunction, protein dysfunction, inflammation, autophagy, apoptosis, and abnormal deposition of α-synuclein. Currently, the existing pharmacological treatments for PD cannot improve fundamentally the degenerative process of dopaminergic neurons and have numerous side effects. On the other hand, attention-deficit/hyperactivity disorder (ADHD) is the most common neurodevelopmental disorder of childhood and is characterised by hyperactivity, impulsivity, and inattention. The aetiology of ADHD remains unknown, although it has been suggested that its pathophysiology involves abnormalities in several brain regions, disturbances of the catecholaminergic pathway, and oxidative stress. Psychostimulants and nonpsychostimulants are the drugs prescribed for the treatment of ADHD; however, they have been associated with increased risk of substance use and have several side effects. Today, there are very few tools available to prevent or to counteract the progression of such neurological disorders. Thus, therapeutic approaches with high efficiency and fewer side effects are needed. This review presents a brief overview of the two neurological disorders and their current treatments, followed by a discussion of the natural compounds which have been studied as therapeutic agents and the mechanisms underlying the beneficial effects, in particular, the decrease in oxidative stress.
Collapse
|
46
|
Fu Z, Chen Z, Xie Q, Lei H, Xiang S. Hesperidin protects against IL-1β-induced inflammation in human osteoarthritis chondrocytes. Exp Ther Med 2018; 16:3721-3727. [PMID: 30233731 DOI: 10.3892/etm.2018.6616] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/25/2018] [Indexed: 12/24/2022] Open
Abstract
Hesperidin is a vitamin P flavonoid compound primarily present in citrus fruits, which possesses an anti-inflammatory effect. The functional role of hesperidin in interleukin (IL)-1β-stimulated human osteoarthritis (OA) chondrocytes is still unknown. In the present study, anti-inflammatory effects of hesperidin in IL-1β-stimulated chondrocytes were investigated. The results demonstrated that hesperidin treatment markedly decreased nitric oxide and prostaglandin E2 production and markedly downregulated inducible nitric oxide synthase and cyclooxygenase-2 expression in IL-1β-stimulated OA chondrocytes. In addition, hesperidin markedly reduced IL-1β-induced matrix metalloproteinase (MMP)-3 and MMP-13 expression in human OA chondrocytes. Furthermore, hesperidin markedly decreased the activation of nuclear factor (NF)-κB in human OA chondrocytes. In conclusion, it was revealed for the first time that hesperidin inhibited inflammatory responses in IL-1β-stimulated human chondrocytes, potentially through inhibiting the activation of the NF-κB signaling pathway. These data suggest that hesperidin may be a potential agent for the treatment of OA.
Collapse
Affiliation(s)
- Zhaozong Fu
- Department of Spine Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, P.R. China
| | - Zhongxian Chen
- Department of Spine Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, P.R. China
| | - Qinghua Xie
- Department of Spine Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, P.R. China
| | - Hongjun Lei
- Department of Spine Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, P.R. China
| | - Shanshan Xiang
- Department of Spine Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong 529030, P.R. China
| |
Collapse
|
47
|
Marelja Z, Leimkühler S, Missirlis F. Iron Sulfur and Molybdenum Cofactor Enzymes Regulate the Drosophila Life Cycle by Controlling Cell Metabolism. Front Physiol 2018; 9:50. [PMID: 29491838 PMCID: PMC5817353 DOI: 10.3389/fphys.2018.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/16/2018] [Indexed: 12/20/2022] Open
Abstract
Iron sulfur (Fe-S) clusters and the molybdenum cofactor (Moco) are present at enzyme sites, where the active metal facilitates electron transfer. Such enzyme systems are soluble in the mitochondrial matrix, cytosol and nucleus, or embedded in the inner mitochondrial membrane, but virtually absent from the cell secretory pathway. They are of ancient evolutionary origin supporting respiration, DNA replication, transcription, translation, the biosynthesis of steroids, heme, catabolism of purines, hydroxylation of xenobiotics, and cellular sulfur metabolism. Here, Fe-S cluster and Moco biosynthesis in Drosophila melanogaster is reviewed and the multiple biochemical and physiological functions of known Fe-S and Moco enzymes are described. We show that RNA interference of Mocs3 disrupts Moco biosynthesis and the circadian clock. Fe-S-dependent mitochondrial respiration is discussed in the context of germ line and somatic development, stem cell differentiation and aging. The subcellular compartmentalization of the Fe-S and Moco assembly machinery components and their connections to iron sensing mechanisms and intermediary metabolism are emphasized. A biochemically active Fe-S core complex of heterologously expressed fly Nfs1, Isd11, IscU, and human frataxin is presented. Based on the recent demonstration that copper displaces the Fe-S cluster of yeast and human ferredoxin, an explanation for why high dietary copper leads to cytoplasmic iron deficiency in flies is proposed. Another proposal that exosomes contribute to the transport of xanthine dehydrogenase from peripheral tissues to the eye pigment cells is put forward, where the Vps16a subunit of the HOPS complex may have a specialized role in concentrating this enzyme within pigment granules. Finally, we formulate a hypothesis that (i) mitochondrial superoxide mobilizes iron from the Fe-S clusters in aconitase and succinate dehydrogenase; (ii) increased iron transiently displaces manganese on superoxide dismutase, which may function as a mitochondrial iron sensor since it is inactivated by iron; (iii) with the Krebs cycle thus disrupted, citrate is exported to the cytosol for fatty acid synthesis, while succinyl-CoA and the iron are used for heme biosynthesis; (iv) as iron is used for heme biosynthesis its concentration in the matrix drops allowing for manganese to reactivate superoxide dismutase and Fe-S cluster biosynthesis to reestablish the Krebs cycle.
Collapse
Affiliation(s)
- Zvonimir Marelja
- Imagine Institute, Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Silke Leimkühler
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|