1
|
Barros CA, Vieira TCRG. Lactoferrin as a Candidate Multifunctional Therapeutic in Synucleinopathies. Brain Sci 2025; 15:380. [PMID: 40309834 PMCID: PMC12025589 DOI: 10.3390/brainsci15040380] [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: 03/14/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Lactoferrin (Lf) is a multifunctional glycoprotein with well-established antimicrobial, anti-inflammatory, and iron-binding properties. Emerging evidence suggests that Lf also plays a neuroprotective role, particularly in neurodegenerative disorders characterized by protein aggregation, such as Parkinson's disease (PD). Alpha-synuclein (aSyn) aggregation is a pathological hallmark of PD and other synucleinopathies, contributing to neuronal dysfunction and disease progression. Recent studies indicate that Lf may interfere with aSyn aggregation, iron chelation, and modulation of oxidative stress and neuroinflammation. Additionally, Lf's ability to cross the blood-brain barrier and its potential impact on the gut-brain axis highlight its promise as a therapeutic agent. This review explores Lf's mechanisms of action in synucleinopathies, its potential as a disease-modifying therapy, and innovative delivery strategies that could enhance its clinical applicability. By addressing the pathological and therapeutic dimensions of aSyn aggregation, we propose Lf as a compelling candidate for future research and clinical development in neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Tuane C. R. G. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro 21941-599, RJ, Brazil;
| |
Collapse
|
2
|
Chen L, Shen Q, Liu Y, Zhang Y, Sun L, Ma X, Song N, Xie J. Homeostasis and metabolism of iron and other metal ions in neurodegenerative diseases. Signal Transduct Target Ther 2025; 10:31. [PMID: 39894843 PMCID: PMC11788444 DOI: 10.1038/s41392-024-02071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/24/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
As essential micronutrients, metal ions such as iron, manganese, copper, and zinc, are required for a wide range of physiological processes in the brain. However, an imbalance in metal ions, whether excessive or insufficient, is detrimental and can contribute to neuronal death through oxidative stress, ferroptosis, cuproptosis, cell senescence, or neuroinflammation. These processes have been found to be involved in the pathological mechanisms of neurodegenerative diseases. In this review, the research history and milestone events of studying metal ions, including iron, manganese, copper, and zinc in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), will be introduced. Then, the upstream regulators, downstream effector, and crosstalk of mental ions under both physiologic and pathologic conditions will be summarized. Finally, the therapeutic effects of metal ion chelators, such as clioquinol, quercetin, curcumin, coumarin, and their derivatives for the treatment of neurodegenerative diseases will be discussed. Additionally, the promising results and limitations observed in clinical trials of these metal ion chelators will also be addressed. This review will not only provide a comprehensive understanding of the role of metal ions in disease development but also offer perspectives on their modulation for the prevention or treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yingjuan Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yunqi Zhang
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Liping Sun
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Ning Song
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
3
|
Ruggiero M, Cianciulli A, Calvello R, Lofrumento DD, Saponaro C, Filannino FM, Porro C, Panaro MA. Lactoferrin Attenuates Pro-Inflammatory Response and Promotes the Conversion into Neuronal Lineages in the Astrocytes. Int J Mol Sci 2025; 26:405. [PMID: 39796258 PMCID: PMC11720426 DOI: 10.3390/ijms26010405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Neurodegenerative diseases are characterized by progressive loss of neurons and persistent inflammation. Neurons are terminally differentiated cells, and lost neurons cannot be replaced since neurogenesis is restricted to only two neurogenic niches in the adult brain, whose neurogenic potential decreases with age. In this regard, the astrocytes reprogramming into neurons may represent a promising strategy for restoring the lost neurons and rebuilding neural circuits. To date, many anti-inflammatory agents have been shown to reduce neuroinflammation; however, their potential to restore neuronal loss was poorly investigated. This study investigates the anti-inflammatory effects of lactoferrin on DI-TNC1 astrocyte cell line and its ability to induce astrocyte reprogramming in a context of sustained inflammation. For this purpose, astrocytes were pre-treated with lactoferrin (4 μg/mL) for 24 h, then with lipopolysaccharide (LPS) (400 ng/mL), and examined 2, 9 and 16 days from treatment. The results demonstrate that lactoferrin attenuates astrocyte reactivity by reducing Toll-like receptor 4 (TLR4), Glial fibrillary acidic protein (GFAP) and IL-6 expression, as well as by upregulating Interleukin-10 (IL-10) cytokine and NRF2 expression. Moreover, lactoferrin promotes the reprogramming of reactive astrocytes into proliferative neuroblasts by inducing the overexpression of the Sex determining region Y/SRY-box 2 (SOX2) reprogramming transcription factor. Overall, this study highlights the potential effects of lactoferrin to attenuate neuroinflammation and improve neurogenesis, suggesting a future strategy for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Melania Ruggiero
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Antonia Cianciulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Rosa Calvello
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| | - Dario Domenico Lofrumento
- Department of Biological and Environmental Sciences and Technologies, Section of Human Anatomy, University of Salento, 73100 Lecce, Italy;
| | - Concetta Saponaro
- IRCCS Istituto Tumori Giovanni Paolo II, V.le O. Flacco, 65, 70124 Bari, Italy;
| | - Francesca Martina Filannino
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (F.M.F.); (C.P.)
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (F.M.F.); (C.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy; (M.R.); (A.C.); (R.C.)
| |
Collapse
|
4
|
Rascón-Cruz Q, Siqueiros-Cendón TS, Siañez-Estrada LI, Villaseñor-Rivera CM, Ángel-Lerma LE, Olivas-Espino JA, León-Flores DB, Espinoza-Sánchez EA, Arévalo-Gallegos S, Iglesias-Figueroa BF. Antioxidant Potential of Lactoferrin and Its Protective Effect on Health: An Overview. Int J Mol Sci 2024; 26:125. [PMID: 39795983 PMCID: PMC11719613 DOI: 10.3390/ijms26010125] [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: 12/05/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic diseases, including cardiovascular and neurodegenerative diseases and cancer, are significant global health challenges. Oxidative stress, characterized by an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, is a critical factor in the progression of these pathologies. Lactoferrin (Lf), a multifunctional iron-binding glycoprotein, has emerged as a promising therapeutic agent due to its potent antioxidant, anti-inflammatory, and iron-regulating properties. Lf plays a pivotal role in iron homeostasis by chelating iron, modulating its cellular uptake, and reducing ROS production, thereby mitigating oxidative stress-related tissue damage. Lf also demonstrates neuroprotective potential in diseases like Parkinson's and Alzheimer's, where it alleviates oxidative damage, regulates iron metabolism, and enhances antioxidant defenses. Furthermore, its ability to enhance endogenous antioxidant mechanisms, such as superoxide dismutase and glutathione peroxidase, underscores its systemic protective effects. Lf's anti-inflammatory and antimicrobial activities also contribute to its broad-spectrum protective role in chronic diseases. This review consolidates evidence of Lf's mechanisms in mitigating oxidative stress and highlights its therapeutic potential as a versatile molecule for preventing and managing chronic conditions linked to oxidative damage.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Blanca Flor Iglesias-Figueroa
- Laboratorio de Biotecnología I, Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Circuito Universitarios s/n Nuevo Campus Universitario, Chihuahua 31125, Mexico; (Q.R.-C.); (T.S.S.-C.); (L.I.S.-E.); (C.M.V.-R.); (L.E.Á.-L.); (J.A.O.-E.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.)
| |
Collapse
|
5
|
Eker F, Duman H, Ertürk M, Karav S. The potential of lactoferrin as antiviral and immune-modulating agent in viral infectious diseases. Front Immunol 2024; 15:1402135. [PMID: 39620218 PMCID: PMC11604709 DOI: 10.3389/fimmu.2024.1402135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Emerging infectious diseases are caused by unpredictable viruses with the dangerous potential to trigger global pandemics. These viruses typically initiate infection by utilizing the anionic structures of host cell surface receptors to gain entry. Lactoferrin (Lf) is a multifunctional glycoprotein with multiple properties such as antiviral, anti-inflammatory and antioxidant activities. Due to its cationic structure, Lf naturally interacts with certain host cell receptors, such as heparan sulfate proteoglycans, as well as viral particles and other receptors that are targeted by viruses. Therefore, Lf may interfere with virus-host cell interactions by acting as a receptor competitor for viruses. Herein we summarize studies in which this competition was investigated with SARS-CoV-2, Zika, Dengue, Hepatitis and Influenza viruses in vitro. These studies have demonstrated not only Lf's competitive properties, but also its potential intracellular impact on host cells, such as enhancing cell survival and reducing infection efficiency by inhibiting certain viral enzymes. In addition, the immunomodulatory effect of Lf is highlighted, as it can influence the activity of specific immune cells and regulate cytokine release, thereby enhancing the host's response to viral infections. Collectively, these properties promote the potential of Lf as a promising candidate for research in viral infectious diseases.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | | | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| |
Collapse
|
6
|
Yong SJ, Veerakumarasivam A, Teoh SL, Lim WL, Chew J. Lactoferrin Protects Against Rotenone-Induced Toxicity in Dopaminergic SH-SY5Y Cells through the Modulation of Apoptotic-Associated Pathways. J Mol Neurosci 2024; 74:88. [PMID: 39297981 DOI: 10.1007/s12031-024-02267-7] [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: 06/16/2024] [Accepted: 09/12/2024] [Indexed: 09/21/2024]
Abstract
Parkinson's disease (PD) is a common motor neurodegenerative disease that still lacks effective therapeutic options. Previous studies have reported that lactoferrin exhibited neuroprotective effects in cellular and animal models of PD, typically induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 1-methyl-4-phenylpyridinium (MPP+) synthetic toxin. However, the neuroprotective capacity of lactoferrin in the rotenone-induced cellular model of PD remains relatively less established. Unlike MPTP/MPP+, rotenone is a naturally occurring environmental toxin known to induce chronic toxicity and increase the risk of PD in humans. In this study, we constructed a cellular model of PD by differentiating SH-SY5Y neuroblastoma cells with retinoic acid into mature dopaminergic neurons with increased β-tubulin III and tyrosine hydroxylase expression, followed by 24 h of rotenone exposure. Using this cellular model of PD, we showed that lactoferrin (1-10 µg/ml) pre-treatment for 48 h decreased loss of cell viability, mitochondrial membrane potential impairment, reactive oxygen species generation and pro-apoptotic activities (pan-caspase activation and nuclear condensation) in cells exposed to rotenone (1 and 5 µM) using biochemical assays, Hoechst 33342 staining and immunocytochemical techniques. We further demonstrated that 48 h of lactoferrin (10 µg/ml) pre-treatment decreased Bax:Bcl2 ratio and p42/44 mitogen-activated protein kinase expression but increased pAkt expression in 5 µM rotenone-exposed cells. Our study demonstrates that lactoferrin neuroprotective capacity is present in the rotenone-induced cellular model of PD, further supporting lactoferrin as a potential PD therapeutic that warrants further studies.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, 47500, Bandar Sunway, Malaysia
| | - Abhi Veerakumarasivam
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, 47500, Bandar Sunway, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000, Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, 47500, Bandar Sunway, Malaysia.
| | - Jactty Chew
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Selangor Darul Ehsan, 47500, Bandar Sunway, Malaysia.
| |
Collapse
|
7
|
Eker F, Bolat E, Pekdemir B, Duman H, Karav S. Lactoferrin: neuroprotection against Parkinson's disease and secondary molecule for potential treatment. Front Aging Neurosci 2023; 15:1204149. [PMID: 37731953 PMCID: PMC10508234 DOI: 10.3389/fnagi.2023.1204149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023] Open
Abstract
Parkinson's disease (PD) is the second-most common neurodegenerative disease and is largely caused by the death of dopaminergic (DA) cells. Dopamine loss occurs in the substantia nigra pars compacta and leads to dysfunctions in motor functions. Death of DA cells can occur with oxidative stress and dysfunction of glial cells caused by Parkinson-related gene mutations. Lactoferrin (Lf) is a multifunctional glycoprotein that is usually known for its presence in milk, but recent research shows that Lf is also found in the brain regions. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a known mitochondrial toxin that disturbs the mitochondrial electron transport chain (ETC) system and increases the rate of reactive oxygen species. Lf's high affinity for metals decreases the required iron for the Fenton reaction, reduces the oxidative damage to DA cells caused by MPTP, and increases their surveillance rate. Several studies also investigated Lf's effect on neurons that are treated with MPTP. The results pointed out that Lf's protective effect can also be observed without the presence of oxidative stress; thus, several potential mechanisms are currently being researched, starting with a potential HSPG-Lf interaction in the cellular membrane of DA cells. The presence of Lf activity in the brain region also showed that lactoferrin initiates receptor-mediated transcytosis in the blood-brain barrier (BBB) with the existence of lactoferrin receptors in the endothelial cells. The existence of Lf receptors both in endothelial cells and DA cells created the idea of using Lf as a secondary molecule in the transport of therapeutic agents across the BBB, especially in nanoparticle development.
Collapse
Affiliation(s)
| | | | | | | | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| |
Collapse
|
8
|
Ianiro G, Rosa L, Bonaccorsi di Patti MC, Valenti P, Musci G, Cutone A. Lactoferrin: from the structure to the functional orchestration of iron homeostasis. Biometals 2023; 36:391-416. [PMID: 36214975 DOI: 10.1007/s10534-022-00453-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/25/2022] [Indexed: 11/02/2022]
Abstract
Iron is by far the most widespread and essential transition metal, possessing crucial biological functions for living systems. Despite chemical advantages, iron biology has forced organisms to face with some issues: ferric iron insolubility and ferrous-driven formation of toxic radicals. For these reasons, acquisition and transport of iron constitutes a formidable challenge for cells and organisms, which need to maintain adequate iron concentrations within a narrow range, allowing biological processes without triggering toxic effects. Higher organisms have evolved extracellular carrier proteins to acquire, transport and manage iron. In recent years, a renewed interest in iron biology has highlighted the role of iron-proteins dysregulation in the onset and/or exacerbation of different pathological conditions. However, to date, no resolutive therapy for iron disorders has been found. In this review, we outline the efficacy of Lactoferrin, a member of the transferrin family mainly secreted by exocrine glands and neutrophils, as a new emerging orchestrator of iron metabolism and homeostasis, able to counteract iron disorders associated to different pathologies, including iron deficiency and anemia of inflammation in blood, Parkinson and Alzheimer diseases in the brain and cystic fibrosis in the lung.
Collapse
Affiliation(s)
- Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | | | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, Pesche, Italy.
| |
Collapse
|
9
|
Kopaeva MY, Azieva AM, Cherepov AB, Zarayskaya IY. Lactoferrin Modulates Induction of Transcription Factor c-Fos in Neuronal Cultures. Int J Mol Sci 2023; 24:ijms24098373. [PMID: 37176079 PMCID: PMC10179438 DOI: 10.3390/ijms24098373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Lactoferrin (Lf) is a multifunctional protein from the transferrin family. Of particular interest is the ability of Lf to affect a wide range of neuronal processes by modulating the expression of genes involved in long-term neuroplasticity. The expression of the immediate early gene c-fos that is rapidly activated in response to external influences, and its product, transcription factor c-Fos, is widely used as a marker of long-term neuronal plasticity. The present study aims to examine the effect of human Lf on the induction of transcription factor c-Fos in the primary mouse neuronal cultures after stimulation and to determine the cellular localization of human Lf and its colocalization with induced c-Fos protein. Primary dissociated cultures of hippocampal cells were obtained from the brains of newborn C57BL/6 mice (P0-P1). On day 7 of culturing, human Lf was added to the medium. After 24 h (day 8 in culture), c-Fos protein was induced in cells by triple application of 50 mM KCl. c-Fos content was analyzed using the immunofluorescent method 2 h after stimulation. Stimulation promoted exogenous Lf translocation into the nuclei of cultured neuronal cells, which correlated with increased induction of transcription factor c-Fos and was accompanied by nuclear colocalization of these proteins. These results attest to the potential of Lf as a modulator of neuronal processes and open up new prospects in studying the mechanisms of the regulatory effects of lactoferrin on cell function.
Collapse
Affiliation(s)
- Marina Yu Kopaeva
- National Research Center "Kurchatov Institute", 1 Akademika Kurchatova Sq., 123182 Moscow, Russia
| | - Asya M Azieva
- National Research Center "Kurchatov Institute", 1 Akademika Kurchatova Sq., 123182 Moscow, Russia
| | - Anton B Cherepov
- National Research Center "Kurchatov Institute", 1 Akademika Kurchatova Sq., 123182 Moscow, Russia
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya St., 125315 Moscow, Russia
| | - Irina Yu Zarayskaya
- Research Institute of Normal Physiology Named after P.K. Anokhin, 8 Baltiyskaya St., 125315 Moscow, Russia
| |
Collapse
|
10
|
Chen L, Zhao Q, Du X, Chen X, Jiao Q, Jiang H. Effects of oxidative stress caused by iron overload on arachidonic acid metabolites in MES23.5 cells. J Biosci 2022. [DOI: 10.1007/s12038-022-00321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
11
|
Xu SF, Pang ZQ, Fan YG, Zhang YH, Meng YH, Bai CY, Jia MY, Chen YH, Wang ZY, Guo C. Astrocyte-specific loss of lactoferrin influences neuronal structure and function by interfering with cholesterol synthesis. Glia 2022; 70:2392-2408. [PMID: 35946355 DOI: 10.1002/glia.24259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/07/2022]
Abstract
Growing evidence indicates that circulating lactoferrin (Lf) is implicated in peripheral cholesterol metabolism disorders. It has emerged that the distribution of Lf changes in astrocytes of aging brains and those exhibiting neurodegeneration; however, its physiological and/or pathological role remains unknown. Here, we demonstrate that astrocyte-specific knockout of Lf (designated cKO) led to decreased body weight and cognitive abnormalities during early life in mice. Accordingly, there was a reduction in neuronal outgrowth and synaptic structure in cKO mice. Importantly, Lf deficiency in the primary astrocytes led to decreased sterol regulatory element binding protein 2 (Srebp2) activation and cholesterol production, and cholesterol content in cKO mice and/or in astrocytes was restored by exogenous Lf or a Srebp2 agonist. Moreover, neuronal dendritic complexity and total dendritic length were decreased after culture with the culture medium of the primary astrocytes derived from cKO mice and that this decrease was reversed after cholesterol supplementation. Alternatively, these alterations were associated with an activation of AMP-activated protein kinase (AMPK) and inhibition of SREBP2 nuclear translocation. These data suggest that astrocytic Lf might directly or indirectly control in situ cholesterol synthesis, which may be implicated in neurodevelopment and several neurological diseases.
Collapse
Affiliation(s)
- Shuang-Feng Xu
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Zhong-Qiu Pang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Yan-Hui Zhang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Yu-Han Meng
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Chen-Yang Bai
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Meng-Yu Jia
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Yan-Hong Chen
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Zhan-You Wang
- Health Sciences Institute, Key Laboratory of Major Chronic Diseases of Nervous System, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| |
Collapse
|
12
|
El-Fakharany EM, Abu-Serie MM, Habashy NH, Eltarahony M. Augmenting apoptosis-mediated anticancer activity of lactoperoxidase and lactoferrin by nanocombination with copper and iron hybrid nanometals. Sci Rep 2022; 12:13153. [PMID: 35915221 PMCID: PMC9343395 DOI: 10.1038/s41598-022-17357-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022] Open
Abstract
There is an urgent need in the medicinal fields to discover biocompatible nanoformulations with low cytotoxicity, which provide new strategies for promising therapies for several types of tumors. Bovine lactoperoxidase (LP) and lactoferrin (LF) have recently attracted attention in medicine for their antitumor activities with recognized safety pattern. Both LP and LF are suitable proteins to be coated or adsorbed to Cu and Fe nanometals for developing stable nanoformulations that boost immunity and strong anticancer effects. New nanometals of Cu and Fe NPs embedded in LP and LF forming novel nanocombinations of LP-CNPs and LF-FNPs had a spherical shape with an average nanosize of about 21 nm. The combination of LP-CNPs and LF-FNPs significantly exhibited the highest growth inhibitory efficacy, in terms of effectively lowering the half-maximal inhibitory concentration (IC50) values, against Caco-2, HepG2 and MCF7 cells comparing to nanometals, LP, LF and individual nanoproteins (LP-CNPs or LF-FNPs). The highest apoptotic effect of this nanocombination (LP-CNPs and LF-FNPs) was confirmed by the highest percentages of annexin-stained apoptotic cells and G0 population with the strongest alteration in the expression of two well-characterized apoptosis guards (p53 and Bcl-2) and the maximum suppression in the proliferation marker (Ki-67). Also, the in silico analysis predicted that LP-CNPs and LF-FNPs enhanced AMP-activated protein kinase (AMPK, p53 activator) activity and inhibited cancer migration-related proteases (cathepsin B and matrix metalloproteinase (MMP)-9). Our results offer for the first time that these novel nanocombinations of LP and LF were superior in their selectivity and apoptosis-mediating anticancer activity to Cu and Fe nanometals as well as the free form of these proteins or their individual nanoforms.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, 21934, Alexandria, Egypt.
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GE-BRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, 21934, Alexandria, Egypt.
| | - Noha H Habashy
- Biochemistry Department, Faculty of Science, Alexandria University, New Borg El-Arab, 21511, Alexandria, Egypt
| | - Marwa Eltarahony
- Environmental Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt
| |
Collapse
|
13
|
Schirmbeck GH, Sizonenko S, Sanches EF. Neuroprotective Role of Lactoferrin during Early Brain Development and Injury through Lifespan. Nutrients 2022; 14:2923. [PMID: 35889882 PMCID: PMC9322498 DOI: 10.3390/nu14142923] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 12/04/2022] Open
Abstract
Early adverse fetal environments can significantly disturb central nervous system (CNS) development and subsequently alter brain maturation. Nutritional status is a major variable to be considered during development and increasing evidence links neonate and preterm infant impaired brain growth with neurological and psychiatric diseases in adulthood. Breastfeeding is one of the main components required for healthy newborn development due to the many "constitutive" elements breastmilk contains. Maternal intake of specific nutrients during lactation may alter milk composition, thus affecting newborn nutrition and, potentially, brain development. Lactoferrin (Lf) is a major protein present in colostrum and the main protein in human milk, which plays an important role in the benefits of breastfeeding during postnatal development. It has been demonstrated that Lf has antimicrobial, as well as anti-inflammatory properties, and is potentially able to reduce the incidence of sepsis and necrotizing enterocolitis (NEC), which are particularly frequent in premature births. The anti-inflammatory effects of Lf can reduce birth-related pathologies by decreasing the release of pro-inflammatory factors and inhibiting premature cervix maturation (also related to commensal microbiome abnormalities) that could contribute to disrupting brain development. Pre-clinical evidence shows that Lf protects the developing brain from neuronal injury, enhances brain connectivity and neurotrophin production, and decreases inflammation in models of perinatal inflammatory challenge, intrauterine growth restriction (IUGR) and neonatal hypoxia-ischemia (HI). In this context, Lf can provide nutritional support for brain development and cognition and prevent the origin of neuropsychiatric diseases later in life. In this narrative review, we consider the role of certain nutrients during neurodevelopment linking to the latest research on lactoferrin with respect to neonatology. We also discuss new evidence indicating that early neuroprotective pathways modulated by Lf could prevent neurodegeneration through anti-inflammatory and immunomodulatory processes.
Collapse
Affiliation(s)
- Gabriel Henrique Schirmbeck
- Biochemistry Post-Graduate Program, Biochemistry Department, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil;
| | - Stéphane Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, 1205 Geneva, Switzerland;
| | - Eduardo Farias Sanches
- Division of Child Development and Growth, Department of Pediatrics, School of Medicine, University of Geneva, 1205 Geneva, Switzerland;
| |
Collapse
|
14
|
Li B, Zhang B, Liu X, Zheng Y, Han K, Liu H, Wu C, Li J, Fan S, Peng W, Zhang F, Liu X. The effect of lactoferrin in aging: role and potential. Food Funct 2021; 13:501-513. [PMID: 34928288 DOI: 10.1039/d1fo02750f] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aging is frequently accompanied by various types of physiological deterioration, which increases the risk of human pathologies. Global public health efforts to increase human lifespan have increasingly focused on lowering the risk of aging-related diseases, such as diabetes, neurodegenerative diseases, cardiovascular disease, and cancers. Dietary intervention is a promising approach to maintaining human health during aging. Lactoferrin (LF) is known for its physiologically pleiotropic properties. Anti-aging interventions of LF have proven to be safe and effective for various pharmacological activities, such as anti-oxidation, anti-cellular senescence, anti-inflammation, and anti-carcinogenic. Moreover, LF has a pivotal role in modulating the major signaling pathways that influence the longevity of organisms. Thus, LF is expected to be able to attenuate the process of aging and greatly ameliorate its effects.
Collapse
Affiliation(s)
- Bing Li
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Bo Zhang
- Henan Key Laboratory of Rare Earth Functional Materials, The Key Laboratory of Rare Earth Functional Materials and Applications, Zhoukou Normal University, Zhoukou 466001, Henan, PR China
| | - Xudong Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Yidan Zheng
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Kuntong Han
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Henan Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Changjing Wu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Jin Li
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Shuhua Fan
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Weifeng Peng
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Fuli Zhang
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| | - Xiaomeng Liu
- Institute of Neuroscience and Translational Medicine, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466001, Henan, PR China.
| |
Collapse
|
15
|
Ryskalin L, Biagioni F, Busceti CL, Polzella M, Lenzi P, Frati A, Ferrucci M, Fornai F. Lactoferrin Protects against Methamphetamine Toxicity by Modulating Autophagy and Mitochondrial Status. Nutrients 2021; 13:nu13103356. [PMID: 34684361 PMCID: PMC8537867 DOI: 10.3390/nu13103356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 01/18/2023] Open
Abstract
Lactoferrin (LF) was used at first as a vehicle to deliver non-soluble active compounds to the body, including the central nervous system (CNS). Nonetheless, it soon became evident that, apart from acting as a vehicle, LF itself owns active effects in the CNS. In the present study, the effects of LF are assessed both in baseline conditions, as well as to counteract methamphetamine (METH)-induced neurodegeneration by assessing cell viability, cell phenotype, mitochondrial status, and specific autophagy steps. In detail, cell integrity in baseline conditions and following METH administration was carried out by using H&E staining, Trypan blue, Fluoro Jade B, and WST-1. Western blot and immuno-fluorescence were used to assess the expression of the neurofilament marker βIII-tubulin. Mitochondria were stained using Mito Tracker Red and Green and were further detailed and quantified by using transmission electron microscopy. Autophagy markers were analyzed through immuno-fluorescence and electron microscopy. LF counteracts METH-induced degeneration. In detail, LF significantly attenuates the amount of cell loss and mitochondrial alterations produced by METH; and mitigates the dissipation of autophagy-related proteins from the autophagy compartment, which is massively induced by METH. These findings indicate a protective role of LF in the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Francesca Biagioni
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
| | - Carla L. Busceti
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
| | - Maico Polzella
- Aliveda Laboratories, Viale Karol Wojtyla, 19, 56042 Crespina Lorenzana, Italy;
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Alessandro Frati
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
- Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135 Rome, Italy
| | - Michela Ferrucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy; (L.R.); (P.L.); (M.F.)
- Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (C.L.B.); (A.F.)
- Correspondence:
| |
Collapse
|
16
|
Li YQ, Guo C. A Review on Lactoferrin and Central Nervous System Diseases. Cells 2021; 10:cells10071810. [PMID: 34359979 PMCID: PMC8307123 DOI: 10.3390/cells10071810] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Central nervous system (CNS) diseases are currently one of the major health issues around the world. Most CNS disorders are characterized by high oxidative stress levels and intense inflammatory responses in affected tissues. Lactoferrin (Lf), a multifunctional iron-binding glycoprotein, plays a significant role in anti-inflammatory, antibacterial, antiviral, reactive oxygen species (ROS) modulator, antitumor immunity, and anti-apoptotic processes. Previous studies have shown that Lf is abnormally expressed in a variety of neurological diseases, especially neurodegenerative diseases. Recently, the promotion of neurodevelopment and neuroprotection by Lf has attracted widespread attention, and Lf could be exploited both as an active therapeutic agent and drug nanocarrier. However, our understanding of the roles of Lf proteins in the initiation or progression of CNS diseases is limited, especially the roles of Lf in regulating neurogenesis. This review highlights recent advances in the understanding of the major pharmacological effects of Lf in CNS diseases, including neurodegenerative diseases, cerebrovascular disease, developmental delays in children, and brain tumors.
Collapse
Affiliation(s)
| | - Chuang Guo
- Correspondence: ; Tel.: +86-24-8365-6109
| |
Collapse
|
17
|
Naidu SAG, Wallace TC, Davies KJA, Naidu AS. Lactoferrin for Mental Health: Neuro-Redox Regulation and Neuroprotective Effects across the Blood-Brain Barrier with Special Reference to Neuro-COVID-19. J Diet Suppl 2021; 20:218-253. [PMID: 33977807 DOI: 10.1080/19390211.2021.1922567] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Overall mental health depends in part on the blood-brain barrier, which regulates nutrient transfer in-and-out of the brain and its central nervous system. Lactoferrin, an innate metal-transport protein, synthesized in the substantia nigra, particularly in dopaminergic neurons and activated microglia is vital for brain physiology. Lactoferrin rapidly crosses the blood-brain barrier via receptor-mediated transcytosis and accumulates in the brain capillary endothelial cells. Lactoferrin receptors are additionally present on glioma cells, brain micro-vessels, and neurons. As a regulator of neuro-redox, microglial lactoferrin is critical for protection/repair of neurons and healthy brain function. Iron imbalance and oxidative stress are common among patients with neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease, dementia, depression, and multiple sclerosis. As an endogenous iron-chelator, lactoferrin prevents iron accumulation and dopamine depletion in Parkinson's disease patients. Oral lactoferrin supplementation could modulate the p-Akt/PTEN pathway, reduce Aβ deposition, and ameliorate cognitive decline in Alzheimer's disease. Novel lactoferrin-based nano-therapeutics have emerged as effective drug-delivery systems for clinical management of neurodegenerative disorders. Recent emergence of the Coronavirus disease-2019 (COVID-19) pandemic, initially considered a respiratory illness, demonstrated a broader virulence spectrum with the ability to cross the blood-brain barrier and inflict a plethora of neuropathological manifestations in the brain - the Neuro-COVID-19. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections are widely reported in Parkinson's disease, Alzheimer's disease, dementia, and multiple sclerosis patients with aggravated clinical outcomes. Lactoferrin, credited with several neuroprotective benefits in the brain could serve as a potential adjuvant in the clinical management of Neuro-COVID-19.
Collapse
Affiliation(s)
- Sreus A G Naidu
- N-terminus Research Laboratory, Yorba Linda, California, USA
| | - Taylor C Wallace
- Department of Nutrition and Food Studies, George Mason University, Fairfax, Virginia, USA
- Think Healthy Group, Washington, District of Columbia, USA
| | - Kelvin J A Davies
- Division of Biogerontology, Leonard Davis School of Gerontology, The University of Southern California, Los Angeles, California, USA
- Division of Molecular & Computational Biology, Dornsife College of Letters, Arts, and Sciences, The University of Southern California, Los Angeles, California, USA
- Department Biochemistry & Molecular Medicine, Keck School of Medicine of USC, The University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
18
|
Zhang Z, Sun X, Wang K, Yu Y, Zhang L, Zhang K, Gu J, Yuan X, Song G. Hydrogen-saturated saline mediated neuroprotection through autophagy via PI3K/AKT/mTOR pathway in early and medium stages of rotenone-induced Parkinson's disease rats. Brain Res Bull 2021; 172:1-13. [PMID: 33838212 DOI: 10.1016/j.brainresbull.2021.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/20/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
Some cardiovascular symptoms in the early stage of Parkinson's disease (PD) were related to degeneration of the rostral ventrolateral medulla (RVLM) catecholaminergic neurons. To date, little is known about the effects of hydrogen water on early stage of PD. Here, protective actions of hydrogen-saturated saline (HS) on rotenone-induced PD rats, as well as its underlying mechanisms were investigated. HS was used to treat PD rats at three general stages; early, medium and late, which were represented by rotenone induced rats for 0, 7 and 14 days. HS treatment significantly alleviated the cardiovascular and motor symptoms in rotenone-induced PD rats, improved the survival number of RVLM catecholaminergic neurons and nigral dopamine neurons only in early and medium stages of PD rats. Decreased levels of reactive oxygen species (ROS) and alpha-synuclein (α-Syn), transformation of microtubule associated protein 1 light chain 3 (LC3)-I/II and degradation of sequestosome 1 (p62) were detected, as well as increased expression level of autophagy related protein 5 (ATG5) and B-cell lymphoma-2 interacting protein 1 (Beclin-1) in the RVLM and substantia nigra (SN) after HS treatment in early and medium stages of PD rats. In addition, phosphorylation levels of phosphatidylinositol-3-kinase (PI3K), protein kinase B (Akt) and mammalian rapamycin target protein (mTOR) decreased after HS treatment in early and medium stages of PD rats. The results suggested that HS treatment exerted beneficial effects in early and medium stages before motor impairments emerged but not in the late stage of rotenone-induced PD rats. It exerted neuroprotection with RVLM catecholaminergic neurons and nigral dopamine neurons, mediated in part by decreasing levels of ROS and α-Syn through increasing autophagy machinery which were partly via inhibiting PI3K-Akt-mTOR pathway.
Collapse
Affiliation(s)
- Zhaoqiang Zhang
- Department of Physiology, Basic Medical College of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Xiao Sun
- Department of Nephrology, Taian City Central Hospital, Taian, 271000, China
| | - Kun Wang
- Postdoctoral Workstation, Taian City Central Hospital, Taian, 271000, China
| | - Yang Yu
- Life Science Research Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Li Zhang
- Department of Electrocardiogram, Taian Traditional Chinese Medicine Hospital, Taian, 271000, China
| | - Keping Zhang
- Department of Physiology, Basic Medical College of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Jinglongfei Gu
- Department of Physiology, Basic Medical College of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Xiaofan Yuan
- Department of Physiology, Basic Medical College of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China
| | - Guohua Song
- Life Science Research Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, China.
| |
Collapse
|
19
|
Nara PL, Sindelar D, Penn MS, Potempa J, Griffin WST. Porphyromonas gingivalis Outer Membrane Vesicles as the Major Driver of and Explanation for Neuropathogenesis, the Cholinergic Hypothesis, Iron Dyshomeostasis, and Salivary Lactoferrin in Alzheimer's Disease. J Alzheimers Dis 2021; 82:1417-1450. [PMID: 34275903 PMCID: PMC8461682 DOI: 10.3233/jad-210448] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/22/2022]
Abstract
Porphyromonas gingivalis (Pg) is a primary oral pathogen in the widespread biofilm-induced "chronic" multi-systems inflammatory disease(s) including Alzheimer's disease (AD). It is possibly the only second identified unique example of a biological extremophile in the human body. Having a better understanding of the key microbiological and genetic mechanisms of its pathogenesis and disease induction are central to its future diagnosis, treatment, and possible prevention. The published literature around the role of Pg in AD highlights the bacteria's direct role within the brain to cause disease. The available evidence, although somewhat adopted, does not fully support this as the major process. There are alternative pathogenic/virulence features associated with Pg that have been overlooked and may better explain the pathogenic processes found in the "infection hypothesis" of AD. A better explanation is offered here for the discrepancy in the relatively low amounts of "Pg bacteria" residing in the brain compared to the rather florid amounts and broad distribution of one or more of its major bacterial protein toxins. Related to this, the "Gingipains Hypothesis", AD-related iron dyshomeostasis, and the early reduced salivary lactoferrin, along with the resurrection of the Cholinergic Hypothesis may now be integrated into one working model. The current paper suggests the highly evolved and developed Type IX secretory cargo system of Pg producing outer membrane vesicles may better explain the observed diseases. Thus it is hoped this paper can provide a unifying model for the sporadic form of AD and guide the direction of research, treatment, and possible prevention.
Collapse
Affiliation(s)
| | | | - Marc S. Penn
- Summa Heart Health and Vascular Institute, Akron, OH, USA
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases in the School of Dentistry, University of Louisville, Louisville, KY, USA
| | - W. Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
20
|
Fan Y, Jiang J, Song S, Chen X. The selective extraction of iron-binding glycoprotein lactoferrin via a “deferrization-restoring” SPE strategy. Sep Purif Technol 2020. [DOI: 10.1016/j.seppur.2020.117522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Elzoghby AO, Abdelmoneem MA, Hassanin IA, Abd Elwakil MM, Elnaggar MA, Mokhtar S, Fang JY, Elkhodairy KA. Lactoferrin, a multi-functional glycoprotein: Active therapeutic, drug nanocarrier & targeting ligand. Biomaterials 2020; 263:120355. [PMID: 32932142 PMCID: PMC7480805 DOI: 10.1016/j.biomaterials.2020.120355] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Recent progress in protein-based nanomedicine, inspired by the success of Abraxane® albumin-paclitaxel nanoparticles, have resulted in novel therapeutics used for treatment of challenging diseases like cancer and viral infections. However, absence of specific drug targeting, poor pharmacokinetics, premature drug release, and off-target toxicity are still formidable challenges in the clinic. Therefore, alternative protein-based nanomedicines were developed to overcome those challenges. In this regard, lactoferrin (Lf), a glycoprotein of transferrin family, offers a promising biodegradable well tolerated material that could be exploited both as an active therapeutic and drug nanocarrier. This review highlights the major pharmacological actions of Lf including anti-cancer, antiviral, and immunomodulatory actions. Delivery technologies of Lf to improve its pries and enhance its efficacy were also reviewed. Moreover, different nano-engineering strategies used for fabrication of drug-loaded Lf nanocarriers were discussed. In addition, the use of Lf for functionalization of drug nanocarriers with emphasis on tumor-targeted drug delivery was illustrated. Besides its wide application in oncology nano-therapeutics, we discussed the recent advances of Lf-based nanocarriers as efficient platforms for delivery of anti-parkinsonian, anti-Alzheimer, anti-viral drugs, immunomodulatory and bone engineering applications.
Collapse
Affiliation(s)
- Ahmed O Elzoghby
- Center for Engineered Therapeutics, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Harvard-MIT Division of Health Sciences & Technology (HST), Cambridge, MA, 02139, USA; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Damanhur University, Damanhur, 22516, Egypt
| | - Islam A Hassanin
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Mahmoud M Abd Elwakil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Manar A Elnaggar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo (AUC), New Cairo, 11835, Egypt
| | - Sarah Mokhtar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, 333, Taiwan; Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
22
|
González-Sánchez M, Bartolome F, Antequera D, Puertas-Martín V, González P, Gómez-Grande A, Llamas-Velasco S, Herrero-San Martín A, Pérez-Martínez D, Villarejo-Galende A, Atienza M, Palomar-Bonet M, Cantero JL, Perry G, Orive G, Ibañez B, Bueno H, Fuster V, Carro E. Decreased salivary lactoferrin levels are specific to Alzheimer's disease. EBioMedicine 2020; 57:102834. [PMID: 32586758 PMCID: PMC7378957 DOI: 10.1016/j.ebiom.2020.102834] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/15/2020] [Accepted: 05/29/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Evidences of infectious pathogens in Alzheimer's disease (AD) brains may suggest a deteriorated innate immune system in AD pathophysiology. We previously demonstrated reduced salivary lactoferrin (Lf) levels, one of the major antimicrobial proteins, in AD patients. METHODS To assess the clinical utility of salivary Lf for AD diagnosis, we examine the relationship between salivary Lf and cerebral amyloid-β (Aβ) load using amyloid-Positron-Emission Tomography (PET) neuroimaging, in two different cross-sectional cohorts including patients with different neurodegenerative disorders. FINDINGS The diagnostic performance of salivary Lf in the cohort 1 had an area under the curve [AUC] of 0•95 (0•911-0•992) for the differentiation of the prodromal AD/AD group positive for amyloid-PET (PET+) versus healthy group, and 0•97 (0•924-1) versus the frontotemporal dementia (FTD) group. In the cohort 2, salivary Lf had also an excellent diagnostic performance in the health control group versus prodromal AD comparison: AUC 0•93 (0•876-0•989). Salivary Lf detected prodromal AD and AD dementia distinguishing them from FTD with over 87% sensitivity and 91% specificity. INTERPRETATION Salivary Lf seems to have a very good diagnostic performance to detect AD. Our findings support the possible utility of salivary Lf as a new non-invasive and cost-effective AD biomarker. FUNDING Instituto de Salud Carlos III (FIS15/00780, FIS18/00118), FEDER, Comunidad de Madrid (S2017/BMD-3700; NEUROMETAB-CM), and CIBERNED (PI2016/01) to E.C.; Spanish Ministry of Economy and Competitiveness (SAF2017-85310-R) to J.L.C., and (PSI2017-85311-P) to M.A.; International Centre on ageing CENIE-POCTEP (0348_CIE_6_E) to M.A.; Instituto de Salud Carlos III (PIE16/00021, PI17/01799), to H.B.
Collapse
Affiliation(s)
- Marta González-Sánchez
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Neurology Service Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Bartolome
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain.
| | - Desiree Antequera
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
| | - Veronica Puertas-Martín
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Pilar González
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Adolfo Gómez-Grande
- Nuclear Medicine Service, Hospital Universitario 12 de Octubre, Madrid, Spain; Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
| | - Sara Llamas-Velasco
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Neurology Service Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alejandro Herrero-San Martín
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Neurology Service Hospital Universitario 12 de Octubre, Madrid, Spain
| | - David Pérez-Martínez
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Neurology Service Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Villarejo-Galende
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain; Neurology Service Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain, CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Spain
| | - Miriam Palomar-Bonet
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain, CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Spain
| | - Jose Luis Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Seville, Spain, CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Spain
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio, San Antonio, TX, USA
| | - Gorka Orive
- Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country, Vitoria, Spain; Networked Center for Biomedical Research in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Borja Ibañez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain, CIBER de Enfermedades Cardiovasculares, Madrid, Spain; IIS-Fundacion Jiménez Díaz Hospital, Madrid, Spain
| | - Hector Bueno
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain, CIBER de Enfermedades Cardiovasculares, Madrid, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Hospital Universitario 12 de Octubre Research Institute (imas12), Cardiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain; Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain.
| |
Collapse
|
23
|
Abdelhamid M, Jung CG, Zhou C, Abdullah M, Nakano M, Wakabayashi H, Abe F, Michikawa M. Dietary Lactoferrin Supplementation Prevents Memory Impairment and Reduces Amyloid-β Generation in J20 Mice. J Alzheimers Dis 2020; 74:245-259. [DOI: 10.3233/jad-191181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Mona Abdelhamid
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Aichi, Japan
| | - Cha-Gyun Jung
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Aichi, Japan
| | - Chunyu Zhou
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Aichi, Japan
| | - Mohammad Abdullah
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Aichi, Japan
| | - Manabu Nakano
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co, Ltd. Zama, Kanagawa, Japan
| | - Hiroyuki Wakabayashi
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co, Ltd. Zama, Kanagawa, Japan
| | - Fumiaki Abe
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co, Ltd. Zama, Kanagawa, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Aichi, Japan
| |
Collapse
|
24
|
Liu H, Wu H, Zhu N, Xu Z, Wang Y, Qu Y, Wang J. Lactoferrin protects against iron dysregulation, oxidative stress, and apoptosis in 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP)‐induced Parkinson’s disease in mice. J Neurochem 2019; 152:397-415. [DOI: 10.1111/jnc.14857] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 07/10/2019] [Accepted: 08/01/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Huiying Liu
- School of Clinical Medicine Qingdao University Qingdao China
| | - Hao Wu
- School of Clinical Medicine Qingdao University Qingdao China
| | - Ning Zhu
- School of Clinical Medicine Qingdao University Qingdao China
| | - Zijie Xu
- School of Clinical Medicine Qingdao University Qingdao China
| | - Yue Wang
- School of Clinical Medicine Qingdao University Qingdao China
| | - Yan Qu
- Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders Department of Physiology Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders and State Key Disciplines: Physiology Medical College of Qingdao University Qingdao China
| | - Jun Wang
- Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders Department of Physiology Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders and State Key Disciplines: Physiology Medical College of Qingdao University Qingdao China
| |
Collapse
|
25
|
Li L, Lu J, Sun Y, Jin X. Acupuncture protects from 6-OHDA-induced neuronal damage by balancing the ratio of DMT1/Fpn1. Saudi J Biol Sci 2019; 26:1948-1955. [PMID: 31889778 PMCID: PMC6923499 DOI: 10.1016/j.sjbs.2019.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
Objective Acupuncture is a commonly used method to provide motor-symptomatic relief for patients with Parkinson s disease (PD). Our objective was to evaluate protective effects of acupuncture treatment and potential underlying mechanisms according to the “gut-brain axis” theory. Methods We employed a 6-OHDA-induced PD rat model. The effects of acupuncture on disease development were assessed by behavioural tests and immunohistochistry (IHC). ELISA, qPCR and western blot (WB) were employed to measure inflammatory parameters and Fe metabolism in the substantia nigra (SN), striatum, duodenum and blood, respectively. Results Our data show that acupuncture can significantly increase the expression of tyrosine hydroxylase (TH), compared with untreated and madopa treated rats (P < 0.01 and P < 0.05, respectively). Furthermore we could observe significantly decreased levels of pro-inflammatory markers in the duodenum and serum (P < 0.05), reduced deposition of Fe in the substantia nigra (P < 0.05) and but no change in transferrin expression after acupuncture treatment. The mRNA ratio of DMT1/Fpn1 in the SN of acupuncture treated rats (1.1) was comparable to that of the sham group (1.0) which differed both significantly from the untreated and madopa treated groups (P < 0.05). Furthermore, after acupuncture expression of α-synuclein was decreased in the duodenum. Conclusions Acupuncture can reduce iron accumulation in the SN and protect the loss of dopamine neurons by promoting balanced expression of the iron importer DMT1 and the iron exporter Fpn1. Furthermore CNS iron homeostasis may be affected by reduced systemic and intestinal inflammation.
Collapse
Affiliation(s)
- Lihong Li
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, China.,Department of Acupuncture, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Jun Lu
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yingying Sun
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaoqing Jin
- Department of Acupuncture, Zhejiang Hospital, Hangzhou 310013, China
| |
Collapse
|
26
|
Madi NM, El-Saka MH. The possible protective effect of lactoferrin on lipopolysaccharide-induced memory impairment in albino rats. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Affiliation(s)
- Nermin M. Madi
- Physiology Department, Faculty of Medicine, Tanta University, Egypt
| | | |
Collapse
|
27
|
Ndayisaba A, Kaindlstorfer C, Wenning GK. Iron in Neurodegeneration - Cause or Consequence? Front Neurosci 2019; 13:180. [PMID: 30881284 PMCID: PMC6405645 DOI: 10.3389/fnins.2019.00180] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Iron dyshomeostasis can cause neuronal damage to iron-sensitive brain regions. Neurodegeneration with brain iron accumulation reflects a group of disorders caused by iron overload in the basal ganglia. High iron levels and iron related pathogenic triggers have also been implicated in sporadic neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple system atrophy (MSA). Iron-induced dyshomeostasis within vulnerable brain regions is still insufficiently understood. Here, we summarize the modes of action by which iron might act as primary or secondary disease trigger in neurodegenerative disorders. In addition, available treatment options targeting brain iron dysregulation and the use of iron as biomarker in prodromal stages are critically discussed to address the question of cause or consequence.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Abstract
The key molecular events that provoke Parkinson's disease (PD) are not fully understood. Iron deposit was found in the substantia nigra pars compacta (SNpc) of PD patients and animal models, where dopaminergic neurons degeneration occurred selectively. The mechanisms involved in disturbed iron metabolism remain unknown, however, considerable evidence indicates that iron transporters dysregulation, activation of L-type voltage-gated calcium channel (LTCC) and ATP-sensitive potassium (KATP) channels, as well as N-methyl-D-aspartate (NMDA) receptors (NMDARs) contribute to this process. There is emerging evidence on the structural links and functional modulations between iron and α-synuclein, and the key player in PD which aggregates in Lewy bodies. Iron is believed to modulate α-synuclein synthesis, post-translational modification, and aggregation. Furthermore, glia, especially activated astroglia and microglia, are involved in iron deposit in PD. Glial contributions were largely dependent on the factors they released, e.g., neurotrophic factors, pro-inflammatory factors, lactoferrin, and those undetermined. Therefore, iron chelation using iron chelators, the extracts from many natural foods with iron chelating properties, may be an effective therapy for prevention and treatment of the disease.
Collapse
|
29
|
Xu SF, Zhang YH, Wang S, Pang ZQ, Fan YG, Li JY, Wang ZY, Guo C. Lactoferrin ameliorates dopaminergic neurodegeneration and motor deficits in MPTP-treated mice. Redox Biol 2018; 21:101090. [PMID: 30593976 PMCID: PMC6307097 DOI: 10.1016/j.redox.2018.101090] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
Brain iron accumulation is common in patients with Parkinson's disease (PD). Iron chelators have been investigated for their ability to prevent neurodegenerative diseases with features of iron overload. Given the non-trivial side effects of classical iron chelators, lactoferrin (Lf), a multifunctional iron-binding globular glycoprotein, was screened to identify novel neuroprotective pathways against dopaminergic neuronal impairment. We found that Lf substantially ameliorated PD-like motor dysfunction in the subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. We further showed that Lf could alleviate MPTP-triggered apoptosis of DA neurons, neuroinflammation, and histological alterations. As expected, we also found that Lf suppressed MPTP-induced excessive iron accumulation and the upregulation of divalent metal transporter (DMT1) and transferrin receptor (TFR), which is the main intracellular iron regulation protein, and subsequently improved the activity of several antioxidant enzymes. We probed further and determined that the neuroprotection provided by Lf was involved in the upregulated levels of brain-derived neurotrophic factor (BDNF), hypoxia-inducible factor 1α (HIF-1α) and its downstream protein, accompanied by the activation of extracellular regulated protein kinases (ERK) and cAMP response element binding protein (CREB), as well as decreased phosphorylation of c-Jun N-terminal kinase (JNK) and mitogen activated protein kinase (MAPK)/P38 kinase in vitro and in vivo. Our findings suggest that Lf may be an alternative safe drug in ameliorating MPTP-induced brain abnormalities and movement disorder.
Collapse
Affiliation(s)
- Shuang-Feng Xu
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Yan-Hui Zhang
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Shan Wang
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Zhong-Qiu Pang
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China
| | - Jia-Yi Li
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China; Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China; Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, BMC A10, 22184 Lund, Sweden
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China; Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China.
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, No.195, Chuangxin Road, Hunnan District, Shenyang 110169, China.
| |
Collapse
|
30
|
Hassel B, De Souza GA, Stensland ME, Ivanovic J, Voie Ø, Dahlberg D. The proteome of pus from human brain abscesses: host-derived neurotoxic proteins and the cell-type diversity of CNS pus. J Neurosurg 2018; 129:829-837. [DOI: 10.3171/2017.4.jns17284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVEWhat determines the extent of tissue destruction during brain abscess formation is not known. Pyogenic brain infections cause destruction of brain tissue that greatly exceeds the area occupied by microbes, as seen in experimental studies, pointing to cytotoxic factors other than microbes in pus. This study examined whether brain abscess pus contains cytotoxic proteins that might explain the extent of tissue destruction.METHODSPus proteins from 20 human brain abscesses and, for comparison, 7 subdural empyemas were analyzed by proteomics mass spectrometry. Tissue destruction was determined from brain abscess volumes as measured by MRI.RESULTSBrain abscess volume correlated with extracellular pus levels of antibacterial proteins from neutrophils and macrophages: myeloperoxidase (r = 0.64), azurocidin (r = 0.61), lactotransferrin (r = 0.57), and cathelicidin (r = 0.52) (p values 0.002–0.018), suggesting an association between leukocytic activity and tissue damage. In contrast, perfringolysin O, a cytotoxic protein from Streptococcus intermedius that was detected in 16 patients, did not correlate with abscess volume (r = 0.12, p = 0.66). The median number of proteins identified in each pus sample was 870 (range 643–1094). Antibiotic or steroid treatment prior to pus evacuation did not reduce the number or levels of pus proteins. Some of the identified proteins have well-known neurotoxic effects, e.g., eosinophil cationic protein and nonsecretory ribonuclease (also known as eosinophil-derived neurotoxin). The cellular response to brain infection was highly complex, as reflected by the presence of proteins that were specific for neutrophils, eosinophils, macrophages, platelets, fibroblasts, or mast cells in addition to plasma and erythrocytic proteins. Other proteins (neurofilaments, myelin basic protein, and glial fibrillary acidic protein) were specific for brain cells and reflected damage to neurons, oligodendrocytes, and astrocytes, respectively. Pus from subdural empyemas had significantly higher levels of plasma proteins and lower levels of leukocytic proteins than pus from intracerebral abscesses, suggesting greater turnover of the extracellular fluid of empyemas and washout of pus constituents.CONCLUSIONSBrain abscess pus contains leukocytic proteins that are neurotoxic and likely participate actively in the excessive tissue destruction inherent in brain abscess formation. These findings underscore the importance of rapid evacuation of brain abscess pus.
Collapse
Affiliation(s)
- Bjørnar Hassel
- 1Department of Complex Neurology and Neurohabilitation,
- 2Norwegian Defence Research Establishment (FFI), Kjeller, Norway; and
| | - Gustavo Antonio De Souza
- 3Institute of Immunology and Centre for Immune Regulation, and
- 4The Brain Institute, Universidade Federal do Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | - Jugoslav Ivanovic
- 5Department of Neurosurgery, Oslo University Hospital, University of Oslo
| | - Øyvind Voie
- 2Norwegian Defence Research Establishment (FFI), Kjeller, Norway; and
| | - Daniel Dahlberg
- 5Department of Neurosurgery, Oslo University Hospital, University of Oslo
| |
Collapse
|
31
|
Yan F, Chen Y, Li M, Wang Y, Zhang W, Chen X, Ye Q. Gastrointestinal nervous system α-synuclein as a potential biomarker of Parkinson disease. Medicine (Baltimore) 2018; 97:e11337. [PMID: 29995769 PMCID: PMC6076112 DOI: 10.1097/md.0000000000011337] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Lewy bodies (LB) play an essential role in the development, survival, and function maintenance of midbrain dopaminergic (DA) neurons in Parkinson disease (PD). Alpha-synuclein (α-synuclein) is the major component of Lewy bodies and is a potential target for Parkinson's disease (PD) therapies. α-synuclein can be detected in the gastrointestinal (GI) nervous system, but whether there is any association between altered α-synuclein expression in the GI nervous system and the onset of PD is not known. The answer to this question presents the opportunity for a promising biomarker in the pre-clinical diagnosis of PD. As such, this study aimed to measure the α-synuclein level in the GI nervous system of Parkinson's disease patients.The protein levels of α-synuclein in the GI nervous system of 31 PD patients (PD group) and 32 patients without PD or Parkinsonism-plus syndrome (control group) were evaluated via immunohistochemical staining. The χ test was performed to evaluate the differences between the PD group and control group. In addition to the distribution of α-synuclein positive protein, regional distribution of the protein in the stomach was also evaluated across groups.Alpha synuclein overexpression was found in the GI nervous tissue of PD patients. The PD group included 17 positive results and 14 negative results. The control group exhibited 7 positive results and 24 negative results. The χ test showed that χ = 7.255, P = .01. The distribution of these positive cases in the gastrointestinal system, the χ test showed that P = .949. The 21 stomach tissues had 7 α-synuclein positive protein tissues, while the body of stomach (4 α-synuclein positive protein) was higher than in other regions.Aberrant expression of α-synuclein was detected in the GI tissues of PD patients, though the distribution of α-synuclein in the gastrointestinal tract had no specificity. Gastrointestinal mucous biopsy could be regarded as a potential opportunity for the early-stage diagnostic exploration of PD, through the detection of α-synuclein inclusions.
Collapse
Affiliation(s)
- Fudong Yan
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Hospital of Fujian Shunchang
| | - Ying Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
| | - Min Li
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
| | - Yingqing Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
| | - Wenmin Zhang
- Department of Pathology, School of Basic Medical Sciences
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
32
|
Xu H, Liu X, Xia J, Yu T, Qu Y, Jiang H, Xie J. Activation of NMDA receptors mediated iron accumulation via modulating iron transporters in Parkinson's disease. FASEB J 2018; 32:fj201800060RR. [PMID: 29897813 DOI: 10.1096/fj.201800060rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increasing evidence has confirmed that nigral iron accumulation and activation of NMDA receptors (NRs) contribute to the neurodegeneration of dopamine (DA) neurons in Parkinson's disease (PD). Earlier work indicated that activation of NRs participated in iron metabolism in the hippocampus. However, the relationship between activation of NRs and iron accumulation in DA neurons of the substantia nigra in PD was unknown. In this study, our results showed that NRs inhibitors MK-801 and AP5 protected nigrostriatal projection system and reduced nigral iron levels of 6-hydroxydopamine (6-OHDA)-induced PD rats. In vitro studies demonstrated that NMDA treatment increased the expression of iron importer divalent metal transporter 1 (DMT1) and decreased the expression of iron exporter ferropotin 1 (Fpn1), which were dependent on iron regulatory protein 1 (IRP1). This led to increased intracellular iron levels and intensified the decrease in mitochondrial transmembrane potential in MES23.5 dopaminergic neurons. In addition, we reported that MK801 and neuronal nitric oxide synthase inhibitor could antagonize 6-OHDA-induced up-regulation of IRP1 and DMT1 and down-regulation of Fpn1, thus attenuating 6-OHDA-induced iron accumulation in MES23.5 cells. This suggested that 6-OHDA-induced activation of NRs might modulate the expression of DMT1 and Fpn1 via the neuronal nitric oxide synthase-IRP1 pathway.-Xu, H., Liu, X., Xia, J., Yu, T., Qu, Y., Jiang, H., Xie, J., Activation of NMDA receptors mediated iron accumulation via modulating iron transporters in Parkinson's disease.
Collapse
Affiliation(s)
- Huamin Xu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Xiaodong Liu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Jianjian Xia
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Tianshu Yu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Yanan Qu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, and Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Erythropoietin and Nrf2: key factors in the neuroprotection provided by apo-lactoferrin. Biometals 2018; 31:425-443. [PMID: 29748743 DOI: 10.1007/s10534-018-0111-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/03/2018] [Indexed: 02/06/2023]
Abstract
Among the properties of lactoferrin (LF) are bactericidal, antianemic, immunomodulatory, antitumour, antiphlogistic effects. Previously we demonstrated its capacity to stabilize in vivo HIF-1-alpha and HIF-2-alpha, which are redox-sensitive multiaimed transcription factors. Various tissues of animals receiving recombinant human LF (rhLF) responded by expressing the HIF-1-alpha target genes, hence such proteins as erythropoietin (EPO), ceruloplasmin, etc. were synthesized in noticeable amounts. Among organs in which EPO synthesis occurred were brain, heart, spleen, liver, kidneys and lungs. Other researchers showed that EPO can act as a protectant against severe brain injury and status epilepticus in rats. Therefore, we tried rhLF as a protector against the severe neurologic disorders developed in rats, such as the rotenone-induced model of Parkinson's disease and experimental autoimmune encephalomyelitis as a model of multiple sclerosis, and observed its capacity to mitigate the grave symptoms. Moreover, an intraperitoneal injection of rhLF into mice 1 h after occlusion of the medial cerebral artery significantly diminished the necrosis area measured on the third day in the ischaemic brain. During this period EPO was synthesized in various murine tissues. It was known that EPO induces nuclear translocation of Nrf2, which, like HIF-1-alpha, is a transcription factor. In view that under conditions of hypoxia both factors demonstrate a synergistic protective effect, we suggested that LF activates the Keap1/Nrf2 signaling pathway, an important link in proliferation and differentiation of normal and malignant cells. J774 macrophages were cultured for 3 days without or in the presence of ferric and ferrous ions (RPMI-1640 and DMEM/F12, respectively). Then cells were incubated with rhLF or Deferiprone. Confocal microscopy revealed nuclear translocation of Nrf2 (the key event in Keap1/Nrf2 signaling) induced by apo-rhLF (iron-free, RPMI-1640). The reference compound Deferiprone (iron chelator) had the similar effect. Upon iron binding (in DMEM/F12) rhLF did not activate the Keap1/Nrf2 pathway. Added to J774, apo-rhLF enhanced transcription of Nrf2-dependent genes coding for glutathione S-transferase P and heme oxygenase-1. Western blotting revealed presence of Nrf2 in mice brain after 6 days of oral administration of apo-rhLF, but not Fe-rhLF or equivalent amount of PBS. Hence, apo-LF, but not holo-LF, induces the translocation of Nrf2 from cytoplasm to the nucleus, probably due to its capacity to induce EPO synthesis.
Collapse
|
34
|
Xu H, Wang Y, Song N, Wang J, Jiang H, Xie J. New Progress on the Role of Glia in Iron Metabolism and Iron-Induced Degeneration of Dopamine Neurons in Parkinson's Disease. Front Mol Neurosci 2018; 10:455. [PMID: 29403352 PMCID: PMC5780449 DOI: 10.3389/fnmol.2017.00455] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 12/26/2017] [Indexed: 12/26/2022] Open
Abstract
It is now increasingly appreciated that glial cells play a critical role in the regulation of iron homeostasis. Impairment of these properties might lead to dysfunction of iron metabolism and neurodegeneration of neurons. We have previously shown that dysfunction of glia could cause iron deposit and enhance iron-induced degeneration of dopamine (DA) neurons in Parkinson’s disease (PD). There also has been a substantial growth of knowledge regarding the iron metabolism of glia and their effects on iron accumulation and degeneration of DA neurons in PD in recent years. Here, we attempt to describe the role of iron metabolism of glia and the effect of glia on iron accumulation and degeneration of DA neurons in the substantia nigra of PD. This could provide evidence to reveal the mechanisms underlying nigral iron accumulation of DA neurons in PD and provide the basis for discovering new potential therapeutic targets for PD.
Collapse
Affiliation(s)
- Huamin Xu
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Youcui Wang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Ning Song
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Jun Wang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Hong Jiang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| | - Junxia Xie
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
35
|
Song N, Wang J, Jiang H, Xie J. Astroglial and microglial contributions to iron metabolism disturbance in Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864:967-973. [PMID: 29317336 DOI: 10.1016/j.bbadis.2018.01.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/24/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
Abstract
Understandings of the disturbed iron metabolism in Parkinson's disease (PD) are largely from the perspectives of neurons. Neurodegenerative processes in PD trigger universal and conserved astroglial dysfunction and microglial activation. In this review, we start with astroglia and microglia in PD with an emphasis on their roles in spreading α-synuclein pathology, and then focus on their contributions in iron metabolism under normal conditions and the diseased state of PD. Elevated iron in the brain regions affects glial features, meanwhile, glial effects on neuronal iron metabolism are largely dependent on their releasing factors. These advances might be valuable for better understanding and modulating iron metabolism disturbance in PD.
Collapse
Affiliation(s)
- Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China.
| | - Jun Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
36
|
Abu-Serie MM, El-Fakharany EM. Efficiency of novel nanocombinations of bovine milk proteins (lactoperoxidase and lactoferrin) for combating different human cancer cell lines. Sci Rep 2017; 7:16769. [PMID: 29196676 PMCID: PMC5711920 DOI: 10.1038/s41598-017-16962-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/19/2017] [Indexed: 02/06/2023] Open
Abstract
Bovine lactoperoxidase (LPO) and lactoferrin (LF) are suitable proteins to be loaded or adsorbed to chitosan nanoparticles (NPs) for preparing stable nanoformulations with potent anticancer activity. In the present study, nanocombinations of LPO and LF revealed improvement in their stability and activity compared to single (free or nanoformulated) bovine proteins. The coating or loading of LPO-loaded NPs with LF resulted in the highest synergistic cytotoxicity effect against Caco-2, HepG-2, MCF-7 and PC-3 cells in comparison with other NPs and free proteins without causing toxicity toward normal cells. This synergistic improvement in the anticancer activity was apoptosis-dependent that was confirmed by severe alterations in cellular morphology, high percentage of annexin-stained cells and sub-G1 populations as well as nuclear staining with orange fluorescence of treated cancer cells. Additionally, significant alterations in the expression of well characterized cellular proliferation and apoptosis guards (NF-κB, Bcl-2 and p53) in these NPs-treated cancer cells compared to 5-fluorouracil (5-FU) treated cells. Our findings provide for the first time that these new synergistic nanoformulated forms of LPO and LF were superior in their selective apoptosis-mediating anticancer effect than free form of these proteins and 5-FU. LF coating or loading of LPO-loaded NPs present as promising therapy for cancer.
Collapse
Affiliation(s)
- Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City), New Borg EL-Arab, 21934, Alexandria, Egypt.
| | - Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City), New Borg EL-Arab, 21934, Alexandria, Egypt.
| |
Collapse
|
37
|
Guo C, Yang ZH, Zhang S, Chai R, Xue H, Zhang YH, Li JY, Wang ZY. Intranasal Lactoferrin Enhances α-Secretase-Dependent Amyloid Precursor Protein Processing via the ERK1/2-CREB and HIF-1α Pathways in an Alzheimer's Disease Mouse Model. Neuropsychopharmacology 2017; 42:2504-2515. [PMID: 28079060 PMCID: PMC5686501 DOI: 10.1038/npp.2017.8] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/03/2017] [Accepted: 01/07/2017] [Indexed: 02/07/2023]
Abstract
Growing evidence suggests that lactoferrin (Lf), an iron-binding glycoprotein, is a pleiotropic functional nutrient. In addition, Lf was recently implicated as a neuroprotective agent. These properties make Lf a valuable therapeutic candidate for the treatment of Alzheimer's disease (AD). However, the mechanisms regulating the physiological roles of Lf in the pathologic condition of AD remain unknown. In the present study, an APPswe/PS1DE9 transgenic mouse model of AD was used. We explored whether intranasal human Lf (hLf) administration could reduce β-amyloid (Aβ) deposition and ameliorate cognitive decline in this AD model. We found that hLf promoted the non-amyloidogenic metabolism of amyloid precursor protein (APP) processing through activation of α-secretase a-disintegrin and metalloprotease10 (ADAM10), resulting in enhanced cleavage of the α-COOH-terminal fragment of APP and the corresponding elevation of the NH2-terminal APP product, soluble APP-α (sAPPα), which consequently reduced Aβ generation and improved spatial cognitive learning ability in AD mice. To gain insight into the molecular mechanism by which Lf modulates APP processing, we evaluated the involvement of the critical molecules for APP cleavage and the signaling pathways in N2a cells stably transfected with Swedish mutant human APP (APPsw N2a cells). The results show that the ERK1/2-CREB and HIF-1α signaling pathways were activated by hLf treatment, which is responsible for the expression of induced ADAM10. Additional tests were performed before suggesting the potential use of hLf as an antioxidant and anti-inflammatory. These findings provide new insights into the sources and mechanisms by which hLf inhibits the cognitive decline that occurs in AD via activation of ADAM10 expression in an ERK1/2-CREB and HIF-1α-dependent manner.
Collapse
Affiliation(s)
- Chuang Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China,College of Life and Health Sciences, Northeastern University, Shenyang 110819, China, Tel/Fax: +86 24 22529997, E-mail: or
| | - Zhao-Hui Yang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shuai Zhang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Rui Chai
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Han Xue
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yan-Hui Zhang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zhan-You Wang
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, China,College of Life and Health Sciences, Northeastern University, Shenyang 110819, China, Tel/Fax: +86 24 22529997, E-mail: or
| |
Collapse
|