1
|
Zhan Z, Ye M, Jin X. The roles of FLOT1 in human diseases (Review). Mol Med Rep 2023; 28:212. [PMID: 37772385 PMCID: PMC10552069 DOI: 10.3892/mmr.2023.13099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/25/2023] [Indexed: 09/30/2023] Open
Abstract
FLOT1, a scaffold protein of lipid rafts, is involved in several biological processes, including lipid raft protein‑-dependent or clathrin‑independent endocytosis, and the formation of hippocampal synapses, amongst others. Increasing evidence has shown that FLOT1 can function as both a cancer promoter and cancer suppressor dependent on the type of cancer. FLOT1 can affect the occurrence and development of several types of cancer by affecting epithelial‑mesenchymal transition, proliferation of cancer cells, and relevant signaling pathways, and is regulated by long intergenic non‑coding RNAs or microRNAs. In the nervous system, overexpression or abnormally low expression of FLOT1 may lead to the occurrence of neurological diseases, such as Alzheimer's disease, Parkinson's disease, major depressive disorder and other diseases. Additionally, it is also associated with dilated cardiomyopathy, pathogenic microbial infection, diabetes‑related diseases, and gynecological diseases, amongst other diseases. In the present review, the structure and localization of FLOT1, as well as the physiological processes it is involved in are reviewed, and then the upstream and downstream regulation of FLOT1 in human disease, particularly in different types of cancer and neurological diseases are discussed, with a focus on potentially targeting FLOT1 for the clinical treatment of several diseases.
Collapse
Affiliation(s)
- Ziqing Zhan
- Department of Oncology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Science Health Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Meng Ye
- Department of Oncology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Science Health Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiaofeng Jin
- Department of Oncology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315020, P.R. China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Science Health Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
2
|
Bauer KC, York EM, Cirstea MS, Radisavljevic N, Petersen C, Huus KE, Brown EM, Bozorgmehr T, Berdún R, Bernier L, Lee AHY, Woodward SE, Krekhno Z, Han J, Hancock REW, Ayala V, MacVicar BA, Finlay BB. Gut microbes shape microglia and cognitive function during malnutrition. Glia 2022; 70:820-841. [PMID: 35019164 PMCID: PMC9305450 DOI: 10.1002/glia.24139] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022]
Abstract
Fecal-oral contamination promotes malnutrition pathology. Lasting consequences of early life malnutrition include cognitive impairment, but the underlying pathology and influence of gut microbes remain largely unknown. Here, we utilize an established murine model combining malnutrition and iterative exposure to fecal commensals (MAL-BG). The MAL-BG model was analyzed in comparison to malnourished (MAL mice) and healthy (CON mice) controls. Malnourished mice display poor spatial memory and learning plasticity, as well as altered microglia, non-neuronal CNS cells that regulate neuroimmune responses and brain plasticity. Chronic fecal-oral exposures shaped microglial morphology and transcriptional profile, promoting phagocytic features in MAL-BG mice. Unexpectedly, these changes occurred independently from significant cytokine-induced inflammation or blood-brain barrier (BBB) disruption, key gut-brain pathways. Metabolomic profiling of the MAL-BG cortex revealed altered polyunsaturated fatty acid (PUFA) profiles and systemic lipoxidative stress. In contrast, supplementation with an ω3 PUFA/antioxidant-associated diet (PAO) mitigated cognitive deficits within the MAL-BG model. These findings provide valued insight into the malnourished gut microbiota-brain axis, highlighting PUFA metabolism as a potential therapeutic target.
Collapse
Affiliation(s)
- Kylynda C. Bauer
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Elisa M. York
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Mihai S. Cirstea
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Nina Radisavljevic
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Charisse Petersen
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Kelsey E. Huus
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Eric M. Brown
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | | | - Rebeca Berdún
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Louis‐Philippe Bernier
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Amy H. Y. Lee
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Sarah E. Woodward
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Zakhar Krekhno
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
| | - Jun Han
- The Metabolomics Innovation CentreUniversity of VictoriaVictoriaCanada
| | - Robert E. W. Hancock
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB‐Lleida)LleidaSpain
- Department of Experimental MedicineUniversitat de Lleida (UdL)LleidaSpain
| | - Brian A. MacVicar
- Psychiatry Department, Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverCanada
| | - Barton Brett Finlay
- Michael Smith LaboratoriesUniversity of British ColumbiaVancouverCanada
- Microbiology and Immunology DepartmentUniversity of British ColumbiaVancouverCanada
- Biochemistry and Molecular Biology DepartmentUniversity of British ColumbiaVancouverCanada
| |
Collapse
|
3
|
Tiwari RK, Moin A, Rizvi SMD, Shahid SMA, Bajpai P. Modulating neuroinflammation in neurodegeneration-related dementia: can microglial toll-like receptors pull the plug? Metab Brain Dis 2021; 36:829-847. [PMID: 33704660 DOI: 10.1007/s11011-021-00696-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/16/2021] [Indexed: 01/13/2023]
Abstract
Neurodegeneration-associated dementia disorders (NADDs), namely Alzheimer and Parkinson diseases, are developed by a significant portion of the elderly population globally. Extensive research has provided critical insights into the molecular basis of the pathological advancements of these diseases, but an efficient curative therapy seems elusive. A common attribute of NADDs is neuroinflammation due to a chronic inflammatory response within the central nervous system (CNS), which is primarily modulated by microglia. This response within the CNS is positively regulated by cytokines, chemokines, secondary messengers or cyclic nucleotides, and free radicals. Microglia mediated immune activation is regulated by a positive feedback loop in NADDs. The present review focuses on evaluating the crosstalk between inflammatory mediators and microglia, which aggravates both the clinical progression and extent of NADDs by forming a persistent chronic inflammatory milieu within the CNS. We also discuss the role of the human gut microbiota and its effect on NADDs as well as the suitability of targeting toll-like receptors for an immunotherapeutic intervention targeting the deflation of an inflamed milieu within the CNS.
Collapse
Affiliation(s)
- Rohit Kumar Tiwari
- Department of Biosciences, Integral University, Kursi Road, Lucknow, Uttar Pradesh, 226026, India
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Syed Monowar Alam Shahid
- Department of Biochemistry, College of Medicine, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Preeti Bajpai
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| |
Collapse
|
4
|
Microglia in Neurodegenerative Events-An Initiator or a Significant Other? Int J Mol Sci 2021; 22:ijms22115818. [PMID: 34072307 PMCID: PMC8199265 DOI: 10.3390/ijms22115818] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
A change in microglia structure, signaling, or function is commonly associated with neurodegeneration. This is evident in the patient population, animal models, and targeted in vitro assays. While there is a clear association, it is not evident that microglia serve as an initiator of neurodegeneration. Rather, the dynamics imply a close interaction between the various cell types and structures in the brain that orchestrate the injury and repair responses. Communication between microglia and neurons contributes to the physiological phenotype of microglia maintaining cells in a surveillance state and allows the cells to respond to events occurring in their environment. Interactions between microglia and astrocytes is not as well characterized, nor are interactions with other members of the neurovascular unit; however, given the influence of systemic factors on neuroinflammation and disease progression, such interactions likely represent significant contributes to any neurodegenerative process. In addition, they offer multiple target sites/processes by which environmental exposures could contribute to neurodegenerative disease. Thus, microglia at least play a role as a significant other with an equal partnership; however, claiming a role as an initiator of neurodegeneration remains somewhat controversial.
Collapse
|
5
|
TLR-2 neutralization potentiates microglial M1 to M2 switching by the combinatorial treatment of ciprofloxacin and dexamethasone during S. aureus infection. J Neuroimmunol 2020; 344:577262. [DOI: 10.1016/j.jneuroim.2020.577262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
|
6
|
Combination treatment of celecoxib and ciprofloxacin attenuates live S. aureus induced oxidative damage and inflammation in murine microglia via regulation of cytokine balance. J Neuroimmunol 2018; 316:23-39. [DOI: 10.1016/j.jneuroim.2017.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/08/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022]
|
7
|
Shadab M, Jha B, Asad M, Deepthi M, Kamran M, Ali N. Apoptosis-like cell death in Leishmania donovani treated with KalsomeTM10, a new liposomal amphotericin B. PLoS One 2017; 12:e0171306. [PMID: 28170432 PMCID: PMC5295687 DOI: 10.1371/journal.pone.0171306] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/14/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The present study aimed to elucidate the cell death mechanism in Leishmania donovani upon treatment with KalsomeTM10, a new liposomal amphotericin B. METHODOLOGY/PRINCIPAL FINDINGS We studied morphological alterations in promastigotes through phase contrast and scanning electron microscopy. Phosphatidylserine (PS) exposure, loss of mitochondrial membrane potential and disruption of mitochondrial integrity was determined by flow cytometry using annexinV-FITC, JC-1 and mitotraker, respectively. For analysing oxidative stress, generation of H2O2 (bioluminescence kit) and mitochondrial superoxide O2- (mitosox) were measured. DNA fragmentation was evaluated using terminal deoxyribonucleotidyl transferase mediated dUTP nick-end labelling (TUNEL) and DNA laddering assay. We found that KalsomeTM10 is more effective then Ambisome against the promastigote as well as intracellular amastigote forms. The mechanistic study showed that KalsomeTM10 induced several morphological alterations in promastigotes typical of apoptosis. KalsomeTM10 treatment showed a dose- and time-dependent exposure of PS in promastigotes. Further, study on mitochondrial pathway revealed loss of mitochondrial membrane potential as well as disruption in mitochondrial integrity with depletion of intracellular pool of ATP. KalsomeTM10 treated promastigotes showed increased ROS production, diminished GSH levels and increased caspase-like activity. DNA fragmentation and cell cycle arrest was observed in KalsomeTM10 treated promastigotes. Apoptotic DNA fragmentation was also observed in KalsomeTM10 treated intracellular amastigotes. KalsomeTM10 induced generation of ROS and nitric oxide leads to the killing of the intracellular parasites. Moreover, endocytosis is indispensable for KalsomeTM10 mediated anti-leishmanial effect in host macrophage. CONCLUSIONS KalsomeTM10 induces apoptotic-like cell death in L. donovani parasites to exhibit its anti-leishmanial function.
Collapse
Affiliation(s)
- Md. Shadab
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Baijayanti Jha
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Mohammad Asad
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Makaraju Deepthi
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Mohd. Kamran
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Jadavpur, Kolkata, West Bengal, India
- * E-mail:
| |
Collapse
|
8
|
Kumawat KL, Kaushik DK, Goswami P, Basu A. Acute exposure to lead acetate activates microglia and induces subsequent bystander neuronal death via caspase-3 activation. Neurotoxicology 2014; 41:143-53. [DOI: 10.1016/j.neuro.2014.02.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 01/20/2014] [Accepted: 02/04/2014] [Indexed: 01/13/2023]
|
9
|
Račková L. Cholesterol load of microglia: contribution of membrane architecture changes to neurotoxic power? Arch Biochem Biophys 2013; 537:91-103. [PMID: 23831332 DOI: 10.1016/j.abb.2013.06.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 06/21/2013] [Accepted: 06/22/2013] [Indexed: 12/15/2022]
Abstract
Considerable evidence provides a link between hypercholesterolemia and ageing-related neurodegenerative diseases. The present study was aimed to provide a complex view on the effects caused by cholesterol- and cholesterol 5α,6α-epoxide-load in microglia, with particular emphasize put on membrane proteins. Prolonged application of oxysterol significantly enhanced LPS-stimulated association of cytosolic NADPH-oxidase factor p47[phox] with detergent-resistant microdomains (DRMs) in BV-2 cells. Although the treatment with both sterols does not influence the portion of CD36 receptor in DRMs, its apparent surface-cellular expression was altered. Even though sterol-treatment potentiated oxidant production by microglia, as well as their phagocytosis, these effects, however, appeared to be independent of cholesterol profusion in the membrane. In addition, oxysterol-treatment resulted in a loss of DRMs-associated activity of 26S proteasome, the protease critically regulating both protein homeostasis and immune signaling in microglia. Oxysterol relatively ameliorated cytotoxic effects of inflammed microglia on co-cultured PC12 cells. The outcomes of this study suggest that cholesterol and cholesterol oxides can differentially modulate microglia resulting either in impairment of their immune functionalities or enhanced neurotoxic power. Moreover, these findings shed light on possible complexity of this effect, produced by simultaneous affection of the levels, distribution and function of the critical proteins within microglial membrane compartments.
Collapse
Affiliation(s)
- Lucia Račková
- Institute of Experimental Pharmacology & Toxicology Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
10
|
Sierra A, Abiega O, Shahraz A, Neumann H. Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front Cell Neurosci 2013. [PMID: 23386811 DOI: 10.3389/fncel.2013.00006/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Microglia are the resident brain macrophages and they have been traditionally studied as orchestrators of the brain inflammatory response during infections and disease. In addition, microglia has a more benign, less explored role as the brain professional phagocytes. Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and microbes. In addition, microglia phagocytoses brain-specific cargo, such as axonal and myelin debris in spinal cord injury or multiple sclerosis, amyloid-β deposits in Alzheimer's disease, and supernumerary synapses in postnatal development. Common mechanisms of recognition, engulfment, and degradation of the different types of cargo are assumed, but very little is known about the shared and specific molecules involved in the phagocytosis of each target by microglia. More importantly, the functional consequences of microglial phagocytosis remain largely unexplored. Overall, phagocytosis is considered a beneficial phenomenon, since it eliminates dead cells and induces an anti-inflammatory response. However, phagocytosis can also activate the respiratory burst, which produces toxic reactive oxygen species (ROS). Phagocytosis has been traditionally studied in pathological conditions, leading to the assumption that microglia have to be activated in order to become efficient phagocytes. Recent data, however, has shown that unchallenged microglia phagocytose apoptotic cells during development and in adult neurogenic niches, suggesting an overlooked role in brain remodeling throughout the normal lifespan. The present review will summarize the current state of the literature regarding the role of microglial phagocytosis in maintaining tissue homeostasis in health as in disease.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro-Basque Center for Neuroscience Zamudio, Spain ; Department of Neuroscience, University of the Basque Country EHU/UPV Leioa, Spain ; Ikerbasque-Basque Foundation for Science Bilbao, Spain
| | | | | | | |
Collapse
|
11
|
Sierra A, Abiega O, Shahraz A, Neumann H. Janus-faced microglia: beneficial and detrimental consequences of microglial phagocytosis. Front Cell Neurosci 2013; 7:6. [PMID: 23386811 PMCID: PMC3558702 DOI: 10.3389/fncel.2013.00006] [Citation(s) in RCA: 440] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/09/2013] [Indexed: 02/04/2023] Open
Abstract
Microglia are the resident brain macrophages and they have been traditionally studied as orchestrators of the brain inflammatory response during infections and disease. In addition, microglia has a more benign, less explored role as the brain professional phagocytes. Phagocytosis is a term coined from the Greek to describe the receptor-mediated engulfment and degradation of dead cells and microbes. In addition, microglia phagocytoses brain-specific cargo, such as axonal and myelin debris in spinal cord injury or multiple sclerosis, amyloid-β deposits in Alzheimer's disease, and supernumerary synapses in postnatal development. Common mechanisms of recognition, engulfment, and degradation of the different types of cargo are assumed, but very little is known about the shared and specific molecules involved in the phagocytosis of each target by microglia. More importantly, the functional consequences of microglial phagocytosis remain largely unexplored. Overall, phagocytosis is considered a beneficial phenomenon, since it eliminates dead cells and induces an anti-inflammatory response. However, phagocytosis can also activate the respiratory burst, which produces toxic reactive oxygen species (ROS). Phagocytosis has been traditionally studied in pathological conditions, leading to the assumption that microglia have to be activated in order to become efficient phagocytes. Recent data, however, has shown that unchallenged microglia phagocytose apoptotic cells during development and in adult neurogenic niches, suggesting an overlooked role in brain remodeling throughout the normal lifespan. The present review will summarize the current state of the literature regarding the role of microglial phagocytosis in maintaining tissue homeostasis in health as in disease.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro-Basque Center for Neuroscience Zamudio, Spain ; Department of Neuroscience, University of the Basque Country EHU/UPV Leioa, Spain ; Ikerbasque-Basque Foundation for Science Bilbao, Spain
| | | | | | | |
Collapse
|
12
|
Abstract
There is growing evidence that cell membranes can contain domains with different lipid and protein compositions and with different physical properties. Furthermore, it is increasingly appreciated that sphingolipids play a crucial role in the formation and properties of ordered lipid domains (rafts) in cell membranes. This review describes recent advances in our understanding of ordered membrane domains in both cells and model membranes. In addition, how the structure of sphingolipids influences their ability to participate in the formation of ordered domains, as well as how sphingolipid structure alters ordered domain properties, is described. The diversity of sphingolipid structure is likely to play an important role in modulating the biologically relevant properties of "rafts" in cell membranes.
Collapse
|
13
|
The neuroprotective effects of milk fat globule-EGF factor 8 against oligomeric amyloid β toxicity. J Neuroinflammation 2012; 9:148. [PMID: 22742657 PMCID: PMC3418568 DOI: 10.1186/1742-2094-9-148] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/01/2012] [Indexed: 12/25/2022] Open
Abstract
Background Phosphatidylserine receptor is a key molecule that mediates the phagocytosis of apoptotic cells. Milk fat globule-EGF factor 8 (MFG-E8) is a phosphatidylserine receptor that is expressed on various macrophage lineage cells, including microglia in the central nervous system (CNS). Targeted clearance of degenerated neurons by microglia is essential to maintain healthy neural networks. We previously showed that the CX3C chemokine fractalkine is secreted from degenerated neurons and accelerates microglial clearance of neuronal debris via inducing the release of MFG-E8. However, the mechanisms by which microglia produce MFG-E8 and the precise functions of MFG-E8 are unknown. Methods The release of MFG-E8 from microglia treated with conditioned medium from neurons exposed to neurotoxic substances, glutamate or oligomeric amyloid β (oAβ) was measured by ELISA. The neuroprotective effects of MFG-E8 and MFG-E8 − induced microglial phagocytosis of oAβ were assessed by immunocytochemistry. The effects of MFG-E8 on the production of the anti-oxidative enzyme hemeoxygenase-1 (HO-1) were determined by ELISA and immunocytochemisty. Results MFG-E8 was induced in microglia treated with conditioned medium from neurons that had been exposed to neurotoxicants, glutamate or oAβ. MFG-E8 significantly attenuated oAβ-induced neuronal cell death in a primary neuron − microglia coculture system. Microglial phagocytosis of oAβ was accelerated by MFG-E8 treatment due to increased CD47 expression in the absence of neurotoxic molecule production, such as tumor necrosis factor-α, nitric oxide, and glutamate. MFG-E8 − treated microglia induced nuclear factor E(2) − related factor 2 (Nrf2) − mediated HO-1 production, which also contributed to neuroprotection. Conclusions These results suggest that microglia release MFG-E8 in response to signals from degenerated neurons and that MFG-E8 protects oAβ-induced neuronal cell death by promoting microglial phagocytic activity and activating the Nrf2-HO-1 pathway. Thus, MFG-E8 may have novel roles as a neuroprotectant in neurodegenerative conditions.
Collapse
|
14
|
Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer's disease: focus on disease modifying drugs. Br J Clin Pharmacol 2012; 73:504-17. [PMID: 22035455 DOI: 10.1111/j.1365-2125.2011.04134.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Current approved drug treatments for Alzheimer disease (AD) include cholinesterase inhibitors (donepezil, rivastigmine, galantamine) and the NMDA receptor antagonist memantine. These drugs provide symptomatic relief but poorly affect the progression of the disease. Drug discovery has been directed, in the last 10 years, to develop 'disease modifying drugs' hopefully able to counteract the progression of AD. Because in a chronic, slow progressing pathological process, such as AD, an early start of treatment enhances the chance of success, it is crucial to have biomarkers for early detection of AD-related brain dysfunction, usable before clinical onset. Reliable early biomarkers need therefore to be prospectively tested for predictive accuracy, with specific cut off values validated in clinical practice. Disease modifying drugs developed so far include drugs to reduce β amyloid (Aβ) production, drugs to prevent Aβ aggregation, drugs to promote Aβ clearance, drugs targeting tau phosphorylation and assembly and other approaches. Unfortunately none of these drugs has demonstrated efficacy in phase 3 studies. The failure of clinical trials with disease modifying drugs raises a number of questions, spanning from methodological flaws to fundamental understanding of AD pathophysiology and biology. Recently, new diagnostic criteria applicable to presymptomatic stages of AD have been published. These new criteria may impact on drug development, such that future trials on disease modifying drugs will include populations susceptible to AD, before clinical onset. Specific problems with completed trials and hopes with ongoing trials are discussed in this review.
Collapse
Affiliation(s)
- Salvatore Salomone
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry Department of Formative Processes, University of Catania, Viale Andrea Doria 6, Catania, Italy
| | | | | | | | | |
Collapse
|
15
|
Sweepers in the CNS: Microglial Migration and Phagocytosis in the Alzheimer Disease Pathogenesis. Int J Alzheimers Dis 2012; 2012:891087. [PMID: 22666624 PMCID: PMC3359803 DOI: 10.1155/2012/891087] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/02/2012] [Indexed: 01/28/2023] Open
Abstract
Microglia are multifunctional immune cells in the central nervous system (CNS). In the neurodegenerative diseases such as Alzheimer's disease (AD), accumulation of glial cells, gliosis, occurs in the lesions. The role of accumulated microglia in the pathophysiology of AD is still controversial. When neuronal damage occurs, microglia exert diversified functions, including migration, phagocytosis, and production of various cytokines and chemokines. Among these, microglial phagocytosis of unwanted neuronal debris is critical to maintain the healthy neuronal networks. Microglia express many surface receptors implicated in phagocytosis. It has been suggested that the lack of microglial phagocytosis worsens pathology of AD and induces memory impairment. The present paper summarizes recent evidences on implication of microglial chemotaxis and phagocytosis in AD pathology and discusses the mechanisms related to chemotaxis toward injured neurons and phagocytosis of unnecessary debris.
Collapse
|
16
|
Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2011; 8:2980-3018. [PMID: 21845170 PMCID: PMC3155341 DOI: 10.3390/ijerph8072980] [Citation(s) in RCA: 219] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 07/05/2011] [Accepted: 07/13/2011] [Indexed: 02/07/2023]
Abstract
Microglia are resident cells of the brain involved in regulatory processes critical for development, maintenance of the neural environment, injury and repair. They belong to the monocytic-macrophage lineage and serve as brain immune cells to orchestrate innate immune responses; however, they are distinct from other tissue macrophages due to their relatively quiescent phenotype and tight regulation by the CNS microenvironment. Microglia actively survey the surrounding parenchyma and respond rapidly to changes such that any disruption to neural architecture or function can contribute to the loss in regulation of the microglia phenotype. In many models of neurodegeneration and neurotoxicity, early events of synaptic degeneration and neuronal loss are accompanied by an inflammatory response including activation of microglia, perivascular monocytes, and recruitment of leukocytes. In culture, microglia have been shown to be capable of releasing several potentially cytotoxic substances, such as reactive oxygen intermediates, nitric oxide, proteases, arachidonic acid derivatives, excitatory amino acids, and cytokines; however, they also produce various neurotrophic factors and quench damage from free radicals and excitotoxins. As the primary source for pro-inflammatory cytokines, microglia are implicated as pivotal mediators of neuroinflammation and can induce or modulate a broad spectrum of cellular responses. Neuroinflammation should be considered as a balanced network of processes whereby subtle modifications can shift the cells toward disparate outcomes. For any evaluation of neuroinflammation and microglial responses, within the framework of neurotoxicity or degeneration, one key question in determining the consequence of neuroinflammation is whether the response is an initiating event or the consequence of tissue damage. As examples of environmental exposure-related neuroinflammation in the literature, we provide an evaluation of data on manganese and diesel exhaust particles.
Collapse
|
17
|
Miller TW, Isenberg JS, Shih HB, Wang Y, Roberts DD. Amyloid-β inhibits No-cGMP signaling in a CD36- and CD47-dependent manner. PLoS One 2010; 5:e15686. [PMID: 21203512 PMCID: PMC3008726 DOI: 10.1371/journal.pone.0015686] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 11/21/2010] [Indexed: 11/26/2022] Open
Abstract
Amyloid-β interacts with two cell surface receptors, CD36 and CD47, through which the matricellular protein thrombospondin-1 inhibits soluble guanylate cyclase activation. Here we examine whether amyloid-β shares this inhibitory activity. Amyloid-β inhibited both drug and nitric oxide-mediated activation of soluble guanylate cyclase in several cell types. Known cGMP-dependent functional responses to nitric oxide in platelets and vascular smooth muscle cells were correspondingly inhibited by amyloid-β. Functional interaction of amyloid-β with the scavenger receptor CD36 was indicated by inhibition of free fatty acid uptake via this receptor. Both soluble oligomer and fibrillar forms of amyloid-β were active. In contrast, amyloid-β did not compete with the known ligand SIRPα for binding to CD47. However, both receptors were necessary for amyloid-β to inhibit cGMP accumulation. These data suggest that amyloid-β interaction with CD36 induces a CD47-dependent signal that inhibits soluble guanylate cyclase activation. Combined with the pleiotropic effects of inhibiting free fatty acid transport via CD36, these data provides a molecular mechanism through which amyloid-β can contribute to the nitric oxide signaling deficiencies associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Thomas W. Miller
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeff S. Isenberg
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Hubert B. Shih
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Howard Hughes Medical Institute–National Institutes of Health Research Scholars Program, Bethesda, Maryland, United States of America
| | - Yichen Wang
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Non-Steroidal Anti-Inflammatory Drugs and Brain Inflammation: Effects on Microglial Functions. Pharmaceuticals (Basel) 2010; 3:1949-1965. [PMID: 27713336 PMCID: PMC4033961 DOI: 10.3390/ph3061949] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 05/21/2010] [Accepted: 06/11/2010] [Indexed: 01/04/2023] Open
Abstract
The term NSAID refers to structurally diverse chemical compounds that share the ability to inhibit the activity of the prostaglandin (PG) biosynthetic enzymes, the cyclooxygenase (COX) isoforms 1 and 2. The suppression of PG synthesis at sites of inflammation has been regarded as primarily responsible for the beneficial properties of NSAIDs, but several COX-independent effects have been described in recent years. Epidemiological studies indicate that NSAIDs are neuroprotective, although the mechanisms underlying their beneficial effect remain largely unknown. Microglial cells play a major role in brain inflammation and are often viewed as major contributors to the neurodegeneration. Therefore, microglia represent a likely target for NSAIDs within the brain. In the present review, we focused on the direct effects of NSAIDs and selective COX-2 inhibitors on microglial functions and discuss the potential efficacy in controlling brain inflammation.
Collapse
|
19
|
Current Opinion in Clinical Nutrition and Metabolic Care. Current world literature. Curr Opin Clin Nutr Metab Care 2010; 13:215-21. [PMID: 20145440 DOI: 10.1097/mco.0b013e32833643b4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Cimino PJ, Sokal I, Leverenz J, Fukui Y, Montine TJ. DOCK2 is a microglial specific regulator of central nervous system innate immunity found in normal and Alzheimer's disease brain. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1622-30. [PMID: 19729484 DOI: 10.2353/ajpath.2009.090443] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Neuroinflammation is a hallmark of several neurodegenerative diseases, including Alzheimer's disease (AD). Strong epidemiological and experimental evidence supports the use of nonsteroidal anti-inflammatory drugs to reduce AD risk. However, poor outcome in clinical trials and toxicity in a prevention trial have shifted focus away from these cyclooxygenase (COX) inhibitors to seek additional therapeutic targets in the prostaglandin pathway. Previously, the prostaglandin E2 receptor, EP2, was shown to regulate neuroinflammation and reduce Abeta plaque burden in transgenic mice. Unfortunately, widespread EP2 distribution and a direct effect on COX2 induction make EP2 a less desirable target. In this study, we link dedicator of cytokinesis 2 (DOCK2) to the prostaglandin pathway in the brain. Additionally, we show that DOCK2 regulates microglial innate immunity independent of COX2 induction and that DOCK2+ microglia are associated with human AD pathology. Together, these results suggest DOCK2 as a COX2 expression-independent therapeutic target for neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Patrick J Cimino
- Division of Neuropathology, Department of Pathology, the Neurobiology and Behavior Program, University of Washington, Box 359645, Harborview Medical Center, Seattle, WA 98104, USA.
| | | | | | | | | |
Collapse
|
21
|
Persaud-Sawin DA, Lightcap S, Harry GJ. Isolation of rafts from mouse brain tissue by a detergent-free method. J Lipid Res 2008; 50:759-67. [PMID: 19060326 DOI: 10.1194/jlr.d800037-jlr200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Membrane rafts are rich in cholesterol and sphingolipids and have specific proteins associated with them. Due to their small size, their identification and isolation have proved to be problematic. Their insolubility in nonionic detergents, such as Triton-X 100, at 4 degrees C has been the most common means of isolation. However, detergent presence can produce artifacts or interfere with ganglioside distribution. The direction is therefore toward the use of detergent-free protocols. We report an optimized method of raft isolation from lipid-rich brain tissue using a detergent-free method. We compared this to Triton-X 100-based isolation along sucrose or Optiprep gradients using the following endpoints: low protein content, high cholesterol content, presence of Flotillin 1 (Flot1), and absence of transferrin receptor (TfR) proteins. These criteria were met in raft fractions isolated in a detergent-free buffer along a sucrose gradient of 5%/35%/42.5%. The use of optiprep gave less consistent results with respect to protein distribution. We demonstrate that clean raft fractions with minimal myelin contamination can be reproducibly obtained in the top three low-density fractions along a sucrose step gradient.
Collapse
Affiliation(s)
- Dixie-Ann Persaud-Sawin
- Laboratory of Molecular Toxicology/Neurotoxicology Group, National Institute of Environmental Health Sciences, RTP, NC 27709, USA.
| | | | | |
Collapse
|