1
|
Xu SF, Cui JH, Liu X, Pang ZQ, Bai CY, Jiang C, Luan C, Li YP, Zhao Y, You YM, Guo C. Astrocytic lactoferrin deficiency augments MPTP-induced dopaminergic neuron loss by disturbing glutamate/calcium and ER-mitochondria signaling. Free Radic Biol Med 2024; 225:374-387. [PMID: 39406276 DOI: 10.1016/j.freeradbiomed.2024.10.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/05/2024] [Accepted: 10/12/2024] [Indexed: 10/20/2024]
Abstract
Increased levels of lactoferrin (Lf) are present in the aged brain and in the lesions of various neurodegenerative diseases, including Parkinson's disease (PD), and may contribute to the cascade of events involved in neurodevelopment and neuroprotection. However, whether Lf originates from astrocytes and functions within either the normal or pathological brain are unknown. Here, we employed mice with specific knockout of the astrocyte lactoferrin gene (named Lf-cKO) to explore its specific roles in the pathological process of PD. We observed a decrease in tyrosine hydroxylase-positive cells, mitochondrial dysfunction of residual dopaminergic neurons, and motor deficits in Lf-cKO mice, which were significantly aggravated after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. To further explore how astrocytic lactoferrin deficiency exacerbated PD-like manifestation in MPTP-treated mice, the critical molecules involved in endoplasmic reticulum (ER)-mitochondria contacts and signaling pathways were investigated. In vitro and in vivo models, we found an aberrant level of effects implicated in glutamate and calcium homeostasis, mitochondrial morphology and functions, mitochondrial dynamics, and mitochondria-associated ER membranes, accompanied by signs of oxidative stress and ER stress, which increase the fragility of dopaminergic neurons. These findings confirm the existence of astrocytic Lf and its influence on the fate of dopaminergic neurons by regulating glutamate/calcium metabolism and ER-mitochondria signaling. Our findings may be a promising target for the treatment of PD.
Collapse
Affiliation(s)
- Shuang-Feng Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Jun-He Cui
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Xin Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Zhong-Qiu Pang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chen-Yang Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chao Jiang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chuang Luan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yun-Peng Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yan Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yi-Ming You
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chuang Guo
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China.
| |
Collapse
|
2
|
Ferrari RR, Fantini V, Garofalo M, Di Gerlando R, Dragoni F, Rizzo B, Spina E, Rossi M, Calatozzolo C, Profka X, Ceroni M, Guaita A, Davin A, Gagliardi S, Poloni TE. A Map of Transcriptomic Signatures of Different Brain Areas in Alzheimer's Disease. Int J Mol Sci 2024; 25:11117. [PMID: 39456899 PMCID: PMC11508373 DOI: 10.3390/ijms252011117] [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: 08/20/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that progressively involves brain regions with an often-predictable pattern. Damage to the brain appears to spread and worsen with time, but the molecular mechanisms underlying the region-specific distribution of AD pathology at different stages of the disease are still under-investigated. In this study, a whole-transcriptome analysis was carried out on brain samples from the hippocampus (HI), temporal and parietal cortices (TC and PC, respectively), cingulate cortex (CG), and substantia nigra (SN) of six subjects with a definite AD diagnosis and three healthy age-matched controls in duplicate. The transcriptomic results showed a greater number of differentially expressed genes (DEGs) in the TC (1571) and CG (1210) and a smaller number of DEGs in the HI (206), PC (109), and SN (60). Furthermore, the GSEA showed a difference between the group of brain areas affected early (HI and TC) and the group of areas that were subsequently involved (PC, CG, and SN). Notably, in the HI and TC, there was a significant downregulation of shared DEGs primarily involved in synaptic transmission, while in the PC, CG, and SN, there was a significant downregulation of genes primarily involved in protein folding and trafficking. The course of AD could follow a definite time- and severity-related pattern that arises from protein misfolding, as observed in the PC, CG, and SN, and leads to synaptic impairment, as observed in the HI and TC. Therefore, a map of the molecular and biological processes involved in AD pathogenesis may be traced. This could aid in the discovery of novel biological targets in order to develop effective and well-timed therapeutic approaches.
Collapse
Affiliation(s)
- Riccardo Rocco Ferrari
- Department of Brain and Behavioral Sciences, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (V.F.); (E.S.); (A.G.)
| | - Valentina Fantini
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (V.F.); (E.S.); (A.G.)
- Laboratory of Translational Research, Azienda USL-IRCCS of Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Maria Garofalo
- Molecular Biology and Transcriptomic Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.G.); (R.D.G.); (F.D.); (B.R.); (S.G.)
| | - Rosalinda Di Gerlando
- Molecular Biology and Transcriptomic Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.G.); (R.D.G.); (F.D.); (B.R.); (S.G.)
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy
| | - Francesca Dragoni
- Molecular Biology and Transcriptomic Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.G.); (R.D.G.); (F.D.); (B.R.); (S.G.)
| | - Bartolo Rizzo
- Molecular Biology and Transcriptomic Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.G.); (R.D.G.); (F.D.); (B.R.); (S.G.)
| | - Erica Spina
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (V.F.); (E.S.); (A.G.)
| | - Michele Rossi
- Unity of Biostatistics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy;
| | - Chiara Calatozzolo
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (C.C.); (X.P.); (M.C.); (T.E.P.)
| | - Xhulja Profka
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (C.C.); (X.P.); (M.C.); (T.E.P.)
| | - Mauro Ceroni
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (C.C.); (X.P.); (M.C.); (T.E.P.)
| | - Antonio Guaita
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (V.F.); (E.S.); (A.G.)
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (C.C.); (X.P.); (M.C.); (T.E.P.)
| | - Annalisa Davin
- Laboratory of Neurobiology and Neurogenetics, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (V.F.); (E.S.); (A.G.)
| | - Stella Gagliardi
- Molecular Biology and Transcriptomic Unit, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy; (M.G.); (R.D.G.); (F.D.); (B.R.); (S.G.)
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Corso San Martino 10, 20081 Abbiategrasso, Italy; (C.C.); (X.P.); (M.C.); (T.E.P.)
- Department of Rehabilitation, ASP Golgi-Redaelli, Piazza E. Samek Lodovici 5, 20081 Abbiategrasso, Italy
| |
Collapse
|
3
|
Kaštelan S, Braš M, Pjevač N, Bakija I, Tomić Z, Pjevač Keleminić N, Gverović Antunica A. Tear Biomarkers and Alzheimer's Disease. Int J Mol Sci 2023; 24:13429. [PMID: 37686235 PMCID: PMC10488148 DOI: 10.3390/ijms241713429] [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: 07/07/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative brain disorder that represents the most common type of dementia. It poses a significant diagnostic challenge that requires timely recognition and treatment. Currently, there is no effective therapy for AD; however, certain medications may slow down its progression. The discovery of AD biomarkers, namely, magnetic resonance imaging, positron emission tomography and cerebrospinal fluid molecules (amyloid-β and tau) has advanced our understanding of this disease and has been crucial for identifying early neuropathologic changes prior to clinical changes and cognitive decline. The close interrelationship between the eye and the brain suggests that tears could be an interesting source of biomarkers for AD; however, studies in this area are limited. The identification of biomarkers in tears will enable the development of cost-effective, non-invasive methods of screening, diagnosis and disease monitoring. In order to use tears as a standard method for early and non-invasive diagnosis of AD, future studies need to be conducted on a larger scale.
Collapse
Affiliation(s)
- Snježana Kaštelan
- Department of Ophthalmology, Clinical Hospital Dubrava, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marijana Braš
- Centre for Palliative Medicine, Medical Ethics and Communication Skills, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Neda Pjevač
- Department of Medical Statistics, Epidemiology and Medical Informatics, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivana Bakija
- Department of Integrative Psychiatry, Psychiatry Hospital “Sveti Ivan”, 10090 Zagreb, Croatia
| | - Zora Tomić
- Health Centre of the Croatian Department of Internal Affairs, 10000 Zagreb, Croatia
| | - Nada Pjevač Keleminić
- Department of Family Medicine, Health Centre Zagreb-Centar, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Antonela Gverović Antunica
- Department of Ophthalmology, General Hospital Dubrovnik, University of Dubrovnik, 20000 Dubrovnik, Croatia
| |
Collapse
|
4
|
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 2023; 20:218-253. [PMID: 33977807 DOI: 10.1080/19390211.2021.1922567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
5
|
Islam F, Shohag S, Akhter S, Islam MR, Sultana S, Mitra S, Chandran D, Khandaker MU, Ashraf GM, Idris AM, Emran TB, Cavalu S. Exposure of metal toxicity in Alzheimer’s disease: An extensive review. Front Pharmacol 2022; 13:903099. [PMID: 36105221 PMCID: PMC9465172 DOI: 10.3389/fphar.2022.903099] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Metals serve important roles in the human body, including the maintenance of cell structure and the regulation of gene expression, the antioxidant response, and neurotransmission. High metal uptake in the nervous system is harmful because it can cause oxidative stress, disrupt mitochondrial function, and impair the activity of various enzymes. Metal accumulation can cause lifelong deterioration, including severe neurological problems. There is a strong association between accidental metal exposure and various neurodegenerative disorders, including Alzheimer’s disease (AD), the most common form of dementia that causes degeneration in the aged. Chronic exposure to various metals is a well-known environmental risk factor that has become more widespread due to the rapid pace at which human activities are releasing large amounts of metals into the environment. Consequently, humans are exposed to both biometals and heavy metals, affecting metal homeostasis at molecular and biological levels. This review highlights how these metals affect brain physiology and immunity and their roles in creating harmful proteins such as β-amyloid and tau in AD. In addition, we address findings that confirm the disruption of immune-related pathways as a significant toxicity mechanism through which metals may contribute to AD.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sheikh Shohag
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Shomaya Akhter
- Department of Genetic Engineering and Biotechnology, Faculty of Earth and Ocean Science, Bangabandhu Sheikh Mujibur Rahman Maritime University, Dhaka, Bangladesh
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Sharifa Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Subang Jaya, Malaysia
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Ghulam Md Ashraf, ; Abubakr M. Idris, ; Talha Bin Emran, ; Simona Cavalu,
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- *Correspondence: Ghulam Md Ashraf, ; Abubakr M. Idris, ; Talha Bin Emran, ; Simona Cavalu,
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- *Correspondence: Ghulam Md Ashraf, ; Abubakr M. Idris, ; Talha Bin Emran, ; Simona Cavalu,
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
- *Correspondence: Ghulam Md Ashraf, ; Abubakr M. Idris, ; Talha Bin Emran, ; Simona Cavalu,
| |
Collapse
|
6
|
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
|
7
|
Zhou HH, Wang G, Luo L, Ding W, Xu JY, Yu Z, Qin LQ, Wan Z. Dietary lactoferrin has differential effects on gut microbiota in young versus middle-aged APPswe/PS1dE9 transgenic mice but no effects on cognitive function. Food Nutr Res 2021; 65:5496. [PMID: 34776831 PMCID: PMC8559448 DOI: 10.29219/fnr.v65.5496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 04/11/2021] [Accepted: 05/25/2021] [Indexed: 11/20/2022] Open
Abstract
Background Existing evidence suggest that lactoferrin might be beneficial for Alzheimer’s disease, while precise mechanisms are not fully elucidated. Objective To determine the effects of lactoferrin intervention on cognitive function from APPswe/PS1dE9 (APP/PS1) mice, and potential mechanisms involved. Design Both the young and middle-aged male APP/PS1 mice were divided into the control and lactoferrin intervention groups with 16 weeks’ intervention. Results Lactoferrin had no effects on cognitive function for both the young and middle-aged mice, and no key markers involved in Aβ, tau pathology, neuro-inflammation and synaptic plasticity were altered after lactoferrin intervention. With regards to gut microbiota profiles, in the young APP/PS1 mice, lactoferrin elevated the α diversity index including ACE and Chao 1, and reduced the relative abundance of the genera Bacteroides and Alistipes and elevated Oscillibacter; in addition, Oscillibacter, Anaerotruncus, EF096579_g, EU454405_g, Mollicutes_RF39, EU474361_g, EU774448_g, and EF096976_g were specifically abundant via linear discriminant analysis with effect size (LEfSe) analysis. In the middle-aged APP/PS1 mice, the relative abundance of the phylum Proteobacteria, as well as the genera Oscillospira, Coprococcus, and Ruminococcus was significantly reduced post lactoferrin; additionally, S24_7, Bacteroidia, Bacteroidetes, and Methylobacterium were specific via LEfSe analysis in the lactoferrin group. Conclusions Dietary lactoferrin might be beneficial for gut microbiota homeostasis although it might have no effects on cognition.
Collapse
Affiliation(s)
- Huan-Huan Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Guiping Wang
- School of Physical Education, Soochow University, Suzhou, China.,Laboratory Animal Center, Medical College of Soochow University, Suzhou, China
| | - Lan Luo
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Wei Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Jia-Ying Xu
- School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Zengli Yu
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China.,School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
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
|
9
|
Reseco L, Atienza M, Fernandez-Alvarez M, Carro E, Cantero JL. Salivary lactoferrin is associated with cortical amyloid-beta load, cortical integrity, and memory in aging. ALZHEIMERS RESEARCH & THERAPY 2021; 13:150. [PMID: 34488875 PMCID: PMC8422723 DOI: 10.1186/s13195-021-00891-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/24/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Aging is associated with declining protective immunity and persistent low-grade inflammatory responses, which significantly contribute to Alzheimer's disease (AD) pathogenesis. Detecting aging-related cerebral vulnerability associated with deterioration of the immune system requires from non-invasive biomarkers able to detect failures in the brain-immunity connection. Reduced levels of salivary lactoferrin (sLF), an iron-binding protein with immunomodulatory activity, have been related to AD diagnosis. However, it remains unknown whether decreased sLF is associated with increased cortical amyloid-beta (Aβ) load and/or with loss of cortical integrity in normal aging. METHODS Seventy-four cognitively normal older adults (51 females) participated in the study. We applied multiple linear regression analyses to assess (i) whether sLF is associated with cortical Aβ load measured by 18F-Florbetaben (FBB)-positron emission tomography (PET), (ii) whether sLF-related variations in cortical thickness and cortical glucose metabolism depend on global Aβ burden, and (iii) whether such sLF-related cortical abnormalities moderate the relationship between sLF and cognition. RESULTS sLF was negatively associated with Aβ load in parieto-temporal regions. Moreover, sLF was related to thickening of the middle temporal cortex, increased FDG uptake in the posterior cingulate cortex, and poorer memory. These associations were stronger in individuals showing the highest Aβ burden. CONCLUSIONS sLF levels are sensitive to variations in cortical Aβ load, structural and metabolic cortical abnormalities, and subclinical memory impairment in asymptomatic older adults. These findings provide support for the use of sLF as a non-invasive biomarker of cerebral vulnerability in the general aging population.
Collapse
Affiliation(s)
- Lucia Reseco
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, 41013, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, 41013, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Marina Fernandez-Alvarez
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, 41013, Seville, Spain.,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
| | - Eva Carro
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain.,Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Jose L Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, Ctra. de Utrera Km 1, 41013, Seville, Spain. .,CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain.
| |
Collapse
|
10
|
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: 43] [Impact Index Per Article: 10.8] [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
|
11
|
Rand D, Ravid O, Atrakchi D, Israelov H, Bresler Y, Shemesh C, Omesi L, Liraz-Zaltsman S, Gosselet F, Maskrey TS, Schnaider Beeri M, Wipf P, Cooper I. Endothelial Iron Homeostasis Regulates Blood-Brain Barrier Integrity via the HIF2α-Ve-Cadherin Pathway. Pharmaceutics 2021; 13:pharmaceutics13030311. [PMID: 33670876 PMCID: PMC7997362 DOI: 10.3390/pharmaceutics13030311] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022] Open
Abstract
The objective of this study was to investigate the molecular response to damage at the blood-brain barrier (BBB) and to elucidate critical pathways that might lead to effective treatment in central nervous system (CNS) pathologies in which the BBB is compromised. We have used a human, stem-cell derived in-vitro BBB injury model to gain a better understanding of the mechanisms controlling BBB integrity. Chemical injury induced by exposure to an organophosphate resulted in rapid lipid peroxidation, initiating a ferroptosis-like process. Additionally, mitochondrial ROS formation (MRF) and increase in mitochondrial membrane permeability were induced, leading to apoptotic cell death. Yet, these processes did not directly result in damage to barrier functionality, since blocking them did not reverse the increased permeability. We found that the iron chelator, Desferal© significantly decreased MRF and apoptosis subsequent to barrier insult, while also rescuing barrier integrity by inhibiting the labile iron pool increase, inducing HIF2α expression and preventing the degradation of Ve-cadherin specifically on the endothelial cell surface. Moreover, the novel nitroxide JP4-039 significantly rescued both injury-induced endothelium cell toxicity and barrier functionality. Elucidating a regulatory pathway that maintains BBB integrity illuminates a potential therapeutic approach to protect the BBB degradation that is evident in many neurological diseases.
Collapse
Affiliation(s)
- Daniel Rand
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Orly Ravid
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Hila Israelov
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Yael Bresler
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Chen Shemesh
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Liora Omesi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
| | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Department of Pharmacology, The Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem 97905, Israel
- Department of Sports Therapy, Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono 55000, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), Artois University, UR 2465, F-62300 Lens, France;
| | - Taber S. Maskrey
- Department of Chemistry and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- School of Psychology, Interdisciplinary Center (IDC), Herzliya 4610101, Israel
| | - Peter Wipf
- Department of Chemistry and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (T.S.M.); (P.W.)
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer 52621, Israel; (D.R.); (O.R.); (D.A.); (H.I.); (Y.B.); (C.S.); (L.O.); (S.L.-Z.); (M.S.B.)
- School of Psychology, Interdisciplinary Center (IDC), Herzliya 4610101, Israel
- The Nehemia Rubin Excellence in Biomedical Research—The TELEM Program, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Correspondence:
| |
Collapse
|
12
|
Li F, Hearn M, Bennett LE. The role of microbial infection in the pathogenesis of Alzheimer's disease and the opportunity for protection by anti-microbial peptides. Crit Rev Microbiol 2021; 47:240-253. [PMID: 33555958 DOI: 10.1080/1040841x.2021.1876630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Its pathology is primarily characterized by extracellular deposits of amyloid β peptide and intracellular neurofibrillary tangles. Current rationales to explain the pathogenesis of AD include amyloid cascade, inflammation, infection defense and anti-microbial protection hypotheses. This review focuses on recent advances in the infection hypothesis, in particular on those pathogenic microbes that act systemically, via periodontal and gastro-intestinal infection routes. It is proposed that the evidence convincingly supports that pathogenic microbial infection is associated with, and is likely a causative trigger for, AD pathology. Microbes can drive AD pathology by two main pathways: either by directly infecting the brain and stimulating amyloid-mediated defence (causative trigger) or indirectly, by stimulating the pro-inflammatory effects of infection. In this context, it follows that anti-microbial/anti-infection therapies could be effective for regulating the pathology and symptoms of AD, depending on the stage of disease. As long-term administration of traditional antibiotic therapy is not recommended, alternative antibiotic agents such as anti-microbial peptides (AMPs), could be preferred for intervention and disease management of AD.
Collapse
Affiliation(s)
- Feijie Li
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - Milton Hearn
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - Louise E Bennett
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
13
|
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
|
14
|
Tsatsanis A, McCorkindale AN, Wong BX, Patrick E, Ryan TM, Evans RW, Bush AI, Sutherland GT, Sivaprasadarao A, Guennewig B, Duce JA. The acute phase protein lactoferrin is a key feature of Alzheimer's disease and predictor of Aβ burden through induction of APP amyloidogenic processing. Mol Psychiatry 2021; 26:5516-5531. [PMID: 34400772 PMCID: PMC8758478 DOI: 10.1038/s41380-021-01248-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/17/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023]
Abstract
Amyloidogenic processing of the amyloid precursor protein (APP) forms the amyloid-β peptide (Aβ) component of pathognomonic extracellular plaques of AD. Additional early cortical changes in AD include neuroinflammation and elevated iron levels. Activation of the innate immune system in the brain is a neuroprotective response to infection; however, persistent neuroinflammation is linked to AD neuropathology by uncertain mechanisms. Non-parametric machine learning analysis on transcriptomic data from a large neuropathologically characterised patient cohort revealed the acute phase protein lactoferrin (Lf) as the key predictor of amyloid pathology. In vitro studies showed that an interaction between APP and the iron-bound form of Lf secreted from activated microglia diverted neuronal APP endocytosis from the canonical clathrin-dependent pathway to one requiring ADP ribosylation factor 6 trafficking. By rerouting APP recycling to the Rab11-positive compartment for amyloidogenic processing, Lf dramatically increased neuronal Aβ production. Lf emerges as a novel pharmacological target for AD that not only modulates APP processing but provides a link between Aβ production, neuroinflammation and iron dysregulation.
Collapse
Affiliation(s)
- Andrew Tsatsanis
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK
| | - Andrew N. McCorkindale
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Charles Perkins Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW Australia
| | - Bruce X. Wong
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK ,grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Ellis Patrick
- grid.1013.30000 0004 1936 834XFaculty of Science, School of Mathematics and Statistics, University of Sydney, Camperdown, NSW Australia
| | - Tim M. Ryan
- grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Robert W. Evans
- grid.7728.a0000 0001 0724 6933School of Engineering and Design, Brunel University, London, UK
| | - Ashley I. Bush
- grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| | - Greg T. Sutherland
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Charles Perkins Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW Australia
| | - Asipu Sivaprasadarao
- grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK
| | - Boris Guennewig
- grid.1013.30000 0004 1936 834XFaculty of Medicine and Health, Brain and Mind Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW Australia
| | - James A. Duce
- grid.5335.00000000121885934The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.9909.90000 0004 1936 8403Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, West Yorkshire UK ,grid.1008.90000 0001 2179 088XMelbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC Australia
| |
Collapse
|
15
|
Olsen I, Singhrao SK. Low levels of salivary lactoferrin may affect oral dysbiosis and contribute to Alzheimer's disease: A hypothesis. Med Hypotheses 2020; 146:110393. [PMID: 33229194 DOI: 10.1016/j.mehy.2020.110393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
Recently it has been reported that reduced levels of salivary lactoferrin (LF) can be a plausible biomarker for amyloid beta (Aβ) accumulation in Alzheimer's disease (AD) brains. This could mean that reduced levels of salivary LF act as a trigger for oral dysbiosis and that low LF levels could change the oral microbiota. A chemical change in the composition of saliva has not yet been considered as a cause for microbial dysbiosis but does present an opportunity to view oral dysbiosis as a plausible contributory factor in the development of AD pathophysiology. Oral dysbiosis has largely been reported as a result of inadequate oral hygiene and dry mouth in elderly subjects. Here we discuss if the deficiency of LF in saliva and gingival fluid of AD patients can facilitate proliferation of oral pathogens, and as a result their spread elsewhere in the body. Additionally, we ask if LF in the AD brain could be overexposed as a result of chronic infection. Together these outcomes will indicate if reduced levels of salivary LF can act as a trigger of oral dysbiosis.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.
| | - Sim K Singhrao
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| |
Collapse
|
16
|
Kabir MT, Uddin MS, Zaman S, Begum Y, Ashraf GM, Bin-Jumah MN, Bungau SG, Mousa SA, Abdel-Daim MM. Molecular Mechanisms of Metal Toxicity in the Pathogenesis of Alzheimer’s Disease. Mol Neurobiol 2020; 58:1-20. [DOI: 10.1007/s12035-020-02096-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/25/2020] [Indexed: 12/24/2022]
|
17
|
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
|
18
|
Iron Pathophysiology in Alzheimer’s Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:67-104. [DOI: 10.1007/978-981-13-9589-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
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: 188] [Impact Index Per Article: 31.3] [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
|
20
|
Mohamed WA, Salama RM, Schaalan MF. A pilot study on the effect of lactoferrin on Alzheimer's disease pathological sequelae: Impact of the p-Akt/PTEN pathway. Biomed Pharmacother 2019; 111:714-723. [PMID: 30611996 DOI: 10.1016/j.biopha.2018.12.118] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/29/2018] [Accepted: 12/30/2018] [Indexed: 01/30/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases in which the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (PKB or Akt) pathway is deregulated in response to phosphatase and tensin homolog (PTEN) overexpression. Lactoferrin (LF), a multifunctional iron-binding glycoprotein, is involved in AD pathology; however, direct evidence of its impact upon AD remains unclear. To elucidate LF's role in AD, the possible protective mechanism post-LF administration for 3 months was investigated in AD patients by observing changes in the p-Akt/PTEN pathway. AD patients showed decreased serum acetylcholine (ACh), serotonin (5-HT), antioxidant and anti-inflammatory markers, and decreased expression of Akt in peripheral blood lymphocytes (PBL), as well as PI3K, and p-Akt levels in PBL lysate; all these parameters were significantly improved after daily LF administration for 3 months. Similarly, elevated serum amyloid β (Aβ) 42, cholesterol, oxidative stress markers, IL-6, heat shock protein (HSP) 90, caspase-3, and p-tau, as well as increased expression of tau, MAPK1 and PTEN in AD patients, were significantly reduced upon LF intake. Improvement in the aforementioned AD surrogate markers post-LF treatment was reflected in enhanced cognitive function assessed by the Mini-Mental State Examination (MMSE) and Alzheimer's Disease Assessment Scale-Cognitive Subscale 11-item (ADAS-COG 11) questionnaires as clinical endpoints. These results provide a basis for a possible protective mechanism of LF in AD through its ability to alleviate the AD pathological cascade and cognitive decline via modulation of the p-Akt/PTEN pathway, which affects the key players of inflammation and oxidative stress that are involved in AD pathology.
Collapse
Affiliation(s)
| | - Rania M Salama
- Pharmacology and Toxicology Department, Translational and Clinical Research Unit, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Mona F Schaalan
- Pharmacy Practice and Clinical Pharmacy Department, Translational and Clinical Research Unit, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| |
Collapse
|
21
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
22
|
Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C. Iron and Alzheimer's Disease: From Pathogenesis to Therapeutic Implications. Front Neurosci 2018; 12:632. [PMID: 30250423 PMCID: PMC6139360 DOI: 10.3389/fnins.2018.00632] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/22/2018] [Indexed: 12/28/2022] Open
Abstract
As people age, iron deposits in different areas of the brain may impair normal cognitive function and behavior. Abnormal iron metabolism generates hydroxyl radicals through the Fenton reaction, triggers oxidative stress reactions, damages cell lipids, protein and DNA structure and function, and ultimately leads to cell death. There is an imbalance in iron homeostasis in Alzheimer's disease (AD). Excessive iron contributes to the deposition of β-amyloid and the formation of neurofibrillary tangles, which in turn, promotes the development of AD. Therefore, iron-targeted therapeutic strategies have become a new direction. Iron chelators, such as desferoxamine, deferiprone, deferasirox, and clioquinol, have received a great deal of attention and have obtained good results in scientific experiments and some clinical trials. Given the limitations and side effects of the long-term application of traditional iron chelators, alpha-lipoic acid and lactoferrin, as self-synthesized naturally small molecules, have shown very intriguing biological activities in blocking Aβ-aggregation, tauopathy and neuronal damage. Despite a lack of evidence for any clinical benefits, the conjecture that therapeutic chelation, with a special focus on iron ions, is a valuable approach for treating AD remains widespread.
Collapse
Affiliation(s)
- Jun-Lin Liu
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yong-Gang Fan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Zheng-Sheng Yang
- Department of Dermatology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China.,Key Laboratory of Medical Cell Biology of Ministry of Education, Institute of Health Sciences, China Medical University, Shenyang, China
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
23
|
Farah R, Haraty H, Salame Z, Fares Y, Ojcius DM, Said Sadier N. Salivary biomarkers for the diagnosis and monitoring of neurological diseases. Biomed J 2018; 41:63-87. [PMID: 29866603 PMCID: PMC6138769 DOI: 10.1016/j.bj.2018.03.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/13/2018] [Accepted: 03/29/2018] [Indexed: 12/17/2022] Open
Abstract
Current research efforts on neurological diseases are focused on identifying novel disease biomarkers to aid in diagnosis, provide accurate prognostic information and monitor disease progression. With advances in detection and quantification methods in genomics, proteomics and metabolomics, saliva has emerged as a good source of samples for detection of disease biomarkers. Obtaining a sample of saliva offers multiple advantages over the currently tested biological fluids as it is a non-invasive, painless and simple procedure that does not require expert training or harbour undesirable side effects for the patients. Here, we review the existing literature on salivary biomarkers and examine their validity in diagnosing and monitoring neurodegenerative and neuropsychiatric disorders such as autism and Alzheimer's, Parkinson's and Huntington's disease. Based on the available research, amyloid beta peptide, tau protein, lactoferrin, alpha-synuclein, DJ-1 protein, chromogranin A, huntingtin protein, DNA methylation disruptions, and micro-RNA profiles provide display a reliable degree of consistency and validity as disease biomarkers.
Collapse
Affiliation(s)
- Raymond Farah
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Haraty
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Ziad Salame
- Research Department, Faculty of Dental Medicine, Lebanese University, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA, USA.
| | - Najwane Said Sadier
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
24
|
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
|
25
|
Li Y, Jiao Q, Xu H, Du X, Shi L, Jia F, Jiang H. Biometal Dyshomeostasis and Toxic Metal Accumulations in the Development of Alzheimer's Disease. Front Mol Neurosci 2017; 10:339. [PMID: 29114205 PMCID: PMC5660707 DOI: 10.3389/fnmol.2017.00339] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022] Open
Abstract
Biometal dyshomeostasis and toxic metal accumulation are common features in many neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease, and Huntington’s disease. The neurotoxic effects of metal imbalance are generally associated with reduced enzymatic activities, elevated protein aggregation and oxidative stress in the central nervous system, in which a cascade of events lead to cell death and neurodegeneration. Although the links between biometal imbalance and neurodegenerative disorders remain elusive, a major class of endogenous proteins involved in metal transport has been receiving increasing attention over recent decades. The abnormal expression of these proteins has been linked to biometal imbalance and to the pathogenesis of AD. Here, we present a brief overview of the physiological roles of biometals including iron, zinc, copper, manganese, magnesium and calcium, and provide a detailed description of their transporters and their synergistic involvement in the development of AD. In addition, we also review the published data relating to neurotoxic metals in AD, including aluminum, lead, cadmium, and mercury.
Collapse
Affiliation(s)
- Yong Li
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Qian Jiao
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Huamin Xu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Xixun Du
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Limin Shi
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Fengju Jia
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| | - Hong Jiang
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Abstract
Lactoferrin (Lf) is the major whey protein in milk, with multiple beneficial health effects including direct antimicrobial activities, anti-inflammatory effects, and iron homeostasis. Oral Lf supplementation in human preterm infants has been shown to reduce the incidence of sepsis and necrotizing enterocolitis. In preclinical models of antenatal stress and perinatal brain injury, bovine Lf protected the developing brain from neuronal loss, improved connectivity, increased neurotrophic factors, and decreased inflammation. It also supported brain development and cognition. Further, Lf can prevent preterm delivery by reducing proinflammatory factors and inhibiting premature cervix maturation. We review here the latest research on Lf in the field of neonatology.
Collapse
Affiliation(s)
- Theresa J Ochoa
- a Department of Pediatrics, Universidad Peruana Cayetano Heredia, Lima, Peru.,b Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Stéphane V Sizonenko
- c Division of Child Development and Growth, Department of Child and Adolescent, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
27
|
Martorell P, Llopis S, Gonzalez N, Ramón D, Serrano G, Torrens A, Serrano JM, Navarro M, Genovés S. A nutritional supplement containing lactoferrin stimulates the immune system, extends lifespan, and reduces amyloid β peptide toxicity in Caenorhabditis elegans. Food Sci Nutr 2016; 5:255-265. [PMID: 28265360 PMCID: PMC5332254 DOI: 10.1002/fsn3.388] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 04/06/2016] [Accepted: 04/21/2016] [Indexed: 11/23/2022] Open
Abstract
Lactoferrin is a highly multifunctional glycoprotein involved in many physiological functions, including regulation of iron absorption and immune responses. Moreover, there is increasing evidence for neuroprotective effects of lactoferrin. We used Caenorhabditis elegans as a model to test the protective effects, both on phenotype and transcriptome, of a nutraceutical product based on lactoferrin liposomes. In a dose‐dependent manner, the lactoferrin‐based product protected against acute oxidative stress and extended lifespan of C. elegans N2. Furthermore, Paralysis of the transgenic C. elegans strain CL4176, caused by Aβ1‐42 aggregates, was clearly ameliorated by treatment. Transcriptome analysis in treated nematodes indicated immune system stimulation, together with enhancement of processes involved in the oxidative stress response. The lactoferrin‐based product also improved the protein homeostasis processes, cellular adhesion processes, and neurogenesis in the nematode. In summary, the tested product exerts protection against aging and neurodegeneration, modulating processes involved in oxidative stress response, protein homeostasis, synaptic function, and xenobiotic metabolism. This lactoferrin‐based product is also able to stimulate the immune system, as well as improving reproductive status and energy metabolism. These findings suggest that oral supplementation with this lactoferrin‐based product could improve the immune system and antioxidant capacity. Further studies to understand the molecular mechanisms related with neuronal function would be of interest.
Collapse
Affiliation(s)
- Patricia Martorell
- Cell Biology Laboratory Food Biotechnology Department Biópolis SL Paterna, Valencia 46980 Spain
| | - Silvia Llopis
- Cell Biology Laboratory Food Biotechnology Department Biópolis SL Paterna, Valencia 46980 Spain
| | - Nuria Gonzalez
- Cell Biology Laboratory Food Biotechnology Department Biópolis SL Paterna, Valencia 46980 Spain
| | - Daniel Ramón
- Cell Biology Laboratory Food Biotechnology Department Biópolis SL Paterna, Valencia 46980 Spain
| | - Gabriel Serrano
- Research and Development Department Sesderma Laboratories Rafelbuñol, Valencia 46138 Spain
| | - Ana Torrens
- Research and Development Department Sesderma Laboratories Rafelbuñol, Valencia 46138 Spain
| | - Juan M Serrano
- Research and Development Department Sesderma Laboratories Rafelbuñol, Valencia 46138 Spain
| | - Maria Navarro
- Research and Development Department Sesderma Laboratories Rafelbuñol, Valencia 46138 Spain
| | - Salvador Genovés
- Cell Biology Laboratory Food Biotechnology Department Biópolis SL Paterna, Valencia 46980 Spain
| |
Collapse
|
28
|
van de Looij Y, Ginet V, Chatagner A, Toulotte A, Somm E, Hüppi PS, Sizonenko SV. Lactoferrin during lactation protects the immature hypoxic-ischemic rat brain. Ann Clin Transl Neurol 2014; 1:955-67. [PMID: 25574471 PMCID: PMC4284122 DOI: 10.1002/acn3.138] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 12/13/2022] Open
Abstract
Objective Lactoferrin (Lf) is an iron-binding glycoprotein secreted in maternal milk presenting anti-inflammatory and antioxidant properties. It shows efficient absorption into the brain from nutritional source. Brain injury frequently resulting from cerebral hypoxia-ischemia (HI) has a high incidence in premature infants with ensuing neurodevelopmental disabilities. We investigated the neuroprotective effect of maternal nutritional supplementation with Lf during lactation in a rat model of preterm HI brain injury using magnetic resonance imaging (MRI), brain gene, and protein expression. Methods Moderate brain HI was induced using unilateral common carotid artery occlusion combined with hypoxia (6%, 30 min) in the postnatal day 3 (P3) rat brain (24–28 weeks human equivalent). High-field multimodal MRI techniques were used to investigate the effect of maternal Lf supplementation through lactation. Expression of cytokine coding genes (TNF-α and IL-6), the prosurvival/antiapoptotic AKT protein and caspase-3 activation were also analyzed in the acute phase after HI. Results MRI analysis demonstrated reduced cortical injury in Lf rats few hours post-HI and in long-term outcome (P25). Lf reduced HI-induced modifications of the cortical metabolism and altered white matter microstructure was recovered in Lf-supplemented rats at P25. Lf supplementation significantly decreased brain TNF-α and IL-6 gene transcription, increased phosphorylated AKT levels and reduced activation of caspase-3 at 24 h post-injury. Interpretation Lf given through lactation to rat pups with cerebral HI injury shows neuroprotective effects on brain metabolism, and cerebral gray and white matter recovery. This nutritional intervention may be of high interest for the clinical field of preterm brain neuroprotection.
Collapse
Affiliation(s)
- Yohan van de Looij
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland ; Laboratory for Functional and Metabolic Imaging (LIFMET), Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Vanessa Ginet
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| | - Alexandra Chatagner
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| | - Audrey Toulotte
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| | - Emmanuel Somm
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| | - Petra S Hüppi
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| | - Stéphane V Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva Geneva, Switzerland
| |
Collapse
|
29
|
Kuo YC, Chou PR. Neuroprotection against degeneration of sk-N-mc cells using neuron growth factor-encapsulated liposomes with surface cereport and transferrin. J Pharm Sci 2014; 103:2484-97. [PMID: 25041794 DOI: 10.1002/jps.24081] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/14/2014] [Accepted: 06/16/2014] [Indexed: 11/06/2022]
Abstract
Liposomes with Cereport (RMP-7) and transferrin (Tf) (RMP-7/Tf/liposomes) were employed to target the blood-brain barrier (BBB) and to inhibit the degeneration of neurons insulted with fibrillar β-amyloid peptide 1-42 (Aβ1-42). Neuron growth factor (NGF)-encapsulated RMP-7/Tf/liposomes (RMP-7/Tf/NGF-liposomes) were used to permeate a monolayer of human brain-microvascular endothelial cells (HBMECs) regulated by human astrocytes (HAs) and to treat Aβ1-42 -attacked SK-N-MC cells. An increase in RMT-7 concentration increased the particle size, zeta potential, propidium iodide (PI) permeability, and NGF permeability, but decreased the cross-linking efficiency of RMT-7, viability of HBMECs and HAs, and transendothelial electrical resistance (TEER). In addition, an increase in Tf concentration enhanced the particle size, viability of HBMECs, HAs, and SK-N-MC cells, PI permeability, and NGF permeability, but reduced the zeta potential, cross-linking efficiency of RMT-7 and Tf, and TEER. RMP-7/Tf/NGF-liposomes can transport NGF across the BBB and improve the neuroprotection for Alzheimer's disease therapy in preclinical trials.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, 62102, Republic of China
| | | |
Collapse
|
30
|
Protection of SK-N-MC cells against β-amyloid peptide-induced degeneration using neuron growth factor-loaded liposomes with surface lactoferrin. Biomaterials 2014; 35:5954-64. [PMID: 24746790 DOI: 10.1016/j.biomaterials.2014.03.082] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/28/2014] [Indexed: 01/26/2023]
Abstract
A liposomal system with surface lactoferrin (Lf) was developed for delivering neuron growth factor (NGF) across the blood-brain barrier (BBB) and improving the viability of neuron-like SK-N-MC cells with deposited β-amyloid peptide (Aβ). The Lf-grafted liposomes carrying NGF (Lf/NGF-liposomes) were applied to a monolayer of human brain-microvascular endothelial cells (HBMECs) regulated by human astrocytes (HAs) and to fibrillar Aβ1-42-insulted SK-N-MC cells. An increase in cholesterol mole percentage enhanced the particle size, absolute value of zeta potential, and physical stability, however, reduced the entrapment efficiency and release rate of NGF. In addition, an increase in Lf concentration increased the particle size, surface nitrogen percentage, NGF permeability across the BBB, and viability of HBMECs, HAs, and SK-N-MC cells, however, decreased the absolute value of zeta potential, surface phosphorus percentage, and loading efficiency of Lf. After treating with Lf/NGF-liposomes, a higher Aβ concentration yielded a lower survival of SK-N-MC cells. The current Lf/NGF-liposomes are efficacious drug carriers to target the BBB and inhibit the Aβ-induced neurotoxicity as potential pharmacotherapy for Alzheimer's disease.
Collapse
|
31
|
|
32
|
Harris F, Dennison SR, Phoenix DA. Aberrant action of amyloidogenic host defense peptides: a new paradigm to investigate neurodegenerative disorders? FASEB J 2012; 26:1776-81. [PMID: 22308196 DOI: 10.1096/fj.11-199208] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Host defense peptides (HDPs) are components of the innate immune system with activity against a broad range of microbes. In some cases, it appears that this activity is mediated by the ability of these peptides to permeabilize microbial membranes via the formation of amyloid associated structures. Recent evidence suggests that the naturally occurring function of the Aβ40 and Aβ42 peptides, which are causative agents of Alzheimer's disease, may be to serve as amyloidogenic HDPs. Here, it is hypothesized that the neurotoxicity of these peptides is related to aberrant use of their amyloid-mediated antimicrobial mechanisms, which provides the as yet unexplored paradigm of a relationship among HDPs, neurodegenerative disorders, and other conditions that could contribute to their understanding and remediation.
Collapse
|
33
|
Roberts BR, Ryan TM, Bush AI, Masters CL, Duce JA. The role of metallobiology and amyloid-β peptides in Alzheimer’s disease. J Neurochem 2011; 120 Suppl 1:149-166. [DOI: 10.1111/j.1471-4159.2011.07500.x] [Citation(s) in RCA: 199] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
34
|
Wang L, Yang H, Zhao S, Sato H, Konishi Y, Beach TG, Abdelalim EM, Bisem NJ, Tooyama I. Expression and localization of mitochondrial ferritin mRNA in Alzheimer's disease cerebral cortex. PLoS One 2011; 6:e22325. [PMID: 21799823 PMCID: PMC3140525 DOI: 10.1371/journal.pone.0022325] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 06/24/2011] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial ferritin (MtF) has been identified as a novel ferritin encoded by an intron-lacking gene with specific mitochondrial localization located on chromosome 5q23.1. MtF has been associated with neurodegenerative disorders such as Friedreich ataxia and restless leg syndrome. However, little information is available about MtF in Alzheimer's disease (AD). In this study, therefore, we investigated the expression and localization of MtF messenger RNA (mRNA) in the cerebral cortex of AD and control cases using real-time polymerase chain reaction (PCR) as well as in situ hybridization histochemistry. We also examined protein expression using western-blot assay. In addition, we used in vitro methods to further explore the effect of oxidative stress and β-amyloid peptide (Aβ) on MtF expression. To do this we examined MtF mRNA and protein expression changes in the human neuroblastoma cell line, IMR-32, after treatment with Aβ, H2O2, or both. The neuroprotective effect of MtF on oxidative stress induced by H(2)O(2) was measured by MTT assay. The in situ hybridization studies revealed that MtF mRNA was detected mainly in neurons to a lesser degree in glial cells in the cerebral cortex. The staining intensity and the number of positive cells were increased in the cerebral cortex of AD patients. Real-time PCR and western-blot confirmed that MtF expression levels in the cerebral cortex were significantly higher in AD cases than that in control cases at both the mRNA and the protein level. Cell culture experiments demonstrated that the expression of both MtF mRNA and protein were increased by treatment with H2O2 or a combination of Aβ and H2O2, but not with Aβ alone. Finally, MtF expression showed a significant neuroprotective effect against H2O2-induced oxidative stress (p<0.05). The present study suggests that MtF is involved in the pathology of AD and may play a neuroprotective role against oxidative stress.
Collapse
Affiliation(s)
- Ligang Wang
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|