1
|
Burke MR, Sotiropoulos I, Waites CL. The multiple roles of chronic stress and glucocorticoids in Alzheimer's disease pathogenesis. Trends Neurosci 2024; 47:933-948. [PMID: 39307629 PMCID: PMC11563862 DOI: 10.1016/j.tins.2024.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 11/15/2024]
Abstract
Chronic stress and the accompanying long-term elevation of glucocorticoids (GCs), the stress hormones of the body, increase the risk and accelerate the progression of Alzheimer's disease (AD). Signatures of AD include intracellular tau (MAPT) tangles, extracellular amyloid β (Aβ) plaques, and neuroinflammation. A growing body of work indicates that stress and GCs initiate cellular processes underlying these pathologies through dysregulation of protein homeostasis and trafficking, mitochondrial bioenergetics, and response to damage-associated stimuli. In this review, we integrate findings from mechanistic studies in rodent and cellular models, wherein defined chronic stress protocols or GC administration have been shown to elicit AD-related pathology. We specifically discuss the effects of chronic stress and GCs on tau pathogenesis, including hyperphosphorylation, aggregation, and spreading, amyloid precursor protein (APP) processing and trafficking culminating in Aβ production, immune priming by proinflammatory cytokines and disease-associated molecular patterns, and alterations to glial cell and blood-brain barrier (BBB) function.
Collapse
Affiliation(s)
- Mia R Burke
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Pathobiology and Mechanisms of Disease Graduate Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications, National Centre for Scientific Research (NCSR) Demokritos, Agia Paraskevi, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Department of Neuroscience, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Tohumeken S, Deme P, Yoo SW, Gupta S, Rais R, Slusher BS, Haughey NJ. Neuronal deletion of nSMase2 reduces the production of Aβ and directly protects neurons. Neurobiol Dis 2023; 177:105987. [PMID: 36603748 DOI: 10.1016/j.nbd.2023.105987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023] Open
Abstract
Extracellular vesicles (EVs) have been proposed to regulate the deposition of Aβ. Multiple publications have shown that APP, amyloid processing enzymes and Aβ peptides are associated with EVs. However, very little Aβ is associated with EVs compared with the total amount Aβ present in human plasma, CSF, or supernatants from cultured neurons. The involvement of EVs has largely been inferred by pharmacological inhibition or whole body deletion of the sphingomyelin hydrolase neutral sphingomyelinase-2 (nSMase2) that is a key regulator for the biogenesis of at-least one population of EVs. Here we used a Cre-Lox system to selectively delete nSMase2 from pyramidal neurons in APP/PS1 mice (APP/PS1-SMPD3-Nex1) and found a ∼ 70% reduction in Aβ deposition at 6 months of age and ∼ 35% reduction at 12 months of age in both cortex and hippocampus. Brain ceramides were increased in APP/PS1 compared with Wt mice, but were similar to Wt in APP/PS1-SMPD3-Nex1 mice suggesting that elevated brain ceramides in this model involves neuronally expressed nSMase2. Reduced levels of PSD95 and deficits of long-term potentiation in APP/PS1 mice were normalized in APP/PS1-SMPD3-Nex1 mice. In contrast, elevated levels of IL-1β, IL-8 and TNFα in APP/PS1 mice were not normalized in APP/PS1-SMPD3-Nex1 mice compared with APP/PS1 mice. Mechanistic studies showed that the size of liquid ordered membrane microdomains was increased in APP/PS1 mice, as were the amounts of APP and BACE1 localized to these microdomains. Pharmacological inhibition of nSMase2 activity with PDDC reduced the size of the liquid ordered membrane microdomains, reduced the localization of APP with BACE1 and reduced the production of Aβ1-40 and Aβ1-42. Although inhibition of nSMase2 reduced the release and increased the size of EVs, very little Aβ was associated with EVs in all conditions tested. We also found that nSMase2 directly protected neurons from the toxic effects of oligomerized Aβ and preserved neural network connectivity despite considerable Aβ deposition. These data demonstrate that nSMase2 plays a role in the production of Aβ by stabilizing the interaction of APP with BACE1 in liquid ordered membrane microdomains, and directly protects neurons from the toxic effects of Aβ. The effects of inhibiting nSMase2 on EV biogenesis may be independent from effects on Aβ production and neuronal protection.
Collapse
Affiliation(s)
- Sehmus Tohumeken
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Pragney Deme
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Seung Wan Yoo
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Sujasha Gupta
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Rana Rais
- The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America
| | - Barbara S Slusher
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America; The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America; The Johns Hopkins University School of Medicine, Departments of Pharmacology and Molecular Sciences, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Oncology, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Neuroscience, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Medicine, Baltimore, MD, United States of America
| | - Norman J Haughey
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America.
| |
Collapse
|
3
|
Kaur S, Verma H, Dhiman M, Tell G, Gigli GL, Janes F, Mantha AK. Brain Exosomes: Friend or Foe in Alzheimer's Disease? Mol Neurobiol 2021; 58:6610-6624. [PMID: 34595669 DOI: 10.1007/s12035-021-02547-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/23/2021] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. It is known to be a multifactorial disease and several causes are associated with its occurrence as well as progression. However, the accumulation of amyloid beta (Aβ) is widely considered its major pathogenic hallmark. Additionally, neurofibrillary tangles (NFT), mitochondrial dysfunction, oxidative stress, and aging (cellular senescence) are considered as additional hits affecting the disease pathology. Several studies are now suggesting important role of inflammation in AD, which shifts our thought towards the brain's resident immune cells, microglia, and astrocytes; how they interact with neurons; and how these interactions are affected by intra and extracellular stressful factors. These interactions can be modulated by different mechanisms and pathways, in which exosomes could play an important role. Exosomes are multivesicular bodies secreted by nearly all types of cells. The exosomes secreted by glial cells or neurons affect the interactions and thus the physiology of these cells by transmitting miRNAs, proteins, and lipids. Exosomes can serve as a friend or foe to the neuron function, depending upon the carried signals. Exosomes, from the healthy microenvironment, may assist neuron function and health, whereas, from the stressed microenvironment, they carry oxidative and inflammatory signals to the neurons and thus prove detrimental to the neuronal function. Furthermore, exosomes can cross the blood-brain barrier (BBB), and from the blood plasma they can enter the brain cells and activate microglia and astrocytes. Exosomes can transport Aβ or Tau, cytokines, miRNAs between the cells, and alter the physiology of recipient cells. They can also assist in Aβ clearance and regulation of synaptic activity. The exosomes derived from different cells play different roles, and this field is still in its infancy stage. This review advocates exosomes' role as a friend or foe in neurodegenerative diseases, especially in the case of Alzheimer's disease.
Collapse
Affiliation(s)
- Sharanjot Kaur
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Harkomal Verma
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Biological Sciences , Central University of Punjab, Bathinda, Punjab, India
| | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine, University of Udine, Udine, Italy
- Clinical Neurology, Udine University Hospital, Udine, Italy
| | | | - Anil K Mantha
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Village Ghudda151 401, Punjab, Bathinda, India.
| |
Collapse
|
4
|
Custodia A, Aramburu-Núñez M, Correa-Paz C, Posado-Fernández A, Gómez-Larrauri A, Castillo J, Gómez-Muñoz A, Sobrino T, Ouro A. Ceramide Metabolism and Parkinson's Disease-Therapeutic Targets. Biomolecules 2021; 11:945. [PMID: 34202192 PMCID: PMC8301871 DOI: 10.3390/biom11070945] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Ceramide is a bioactive sphingolipid involved in numerous cellular processes. In addition to being the precursor of complex sphingolipids, ceramides can act as second messengers, especially when they are generated at the plasma membrane of cells. Its metabolic dysfunction may lead to or be a consequence of an underlying disease. Recent reports on transcriptomics and electrospray ionization mass spectrometry analysis have demonstrated the variation of specific levels of sphingolipids and enzymes involved in their metabolism in different neurodegenerative diseases. In the present review, we highlight the most relevant discoveries related to ceramide and neurodegeneration, with a special focus on Parkinson's disease.
Collapse
Affiliation(s)
- Antía Custodia
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Marta Aramburu-Núñez
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Adrián Posado-Fernández
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Ana Gómez-Larrauri
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
- Respiratory Department, Cruces University Hospital, Barakaldo, 48903 Bizkaia, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Antonio Gómez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Alberto Ouro
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| |
Collapse
|
5
|
Canals D, Salamone S, Santacreu BJ, Nemeth E, Aguilar D, Hernandez-Corbacho MJ, Adada M, Staquicini DI, Arap W, Pasqualini R, Haley J, Obeid LM, Hannun YA. Ceramide launches an acute anti-adhesion pro-migration cell signaling program in response to chemotherapy. FASEB J 2020; 34:7610-7630. [PMID: 32307766 PMCID: PMC8265206 DOI: 10.1096/fj.202000205r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/21/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
Chemotherapy has been reported to upregulate sphingomylinases and increase cellular ceramide, often linked to the induction to cell death. In this work, we show that sublethal doses of doxorubicin and vorinostat still increased cellular ceramide, which was located predominantly at the plasma membrane. To interrogate possible functions of this specific pool of ceramide, we used recombinant enzymes to mimic physiological levels of ceramide at the plasma membrane upon chemotherapy treatment. Using mass spectrometry and network analysis, followed by experimental confirmation, the results revealed that this pool of ceramide acutely regulates cell adhesion and cell migration pathways with weak connections to commonly established ceramide functions (eg, cell death). Neutral sphingomyelinase 2 (nSMase2) was identified as responsible for the generation of plasma membrane ceramide upon chemotherapy treatment, and both ceramide at the plasma membrane and nSMase2 were necessary and sufficient to mediate these "side" effects of chemotherapy on cell adhesion and migration. This is the first time a specific pool of ceramide is interrogated for acute signaling functions, and the results define plasma membrane ceramide as an acute signaling effector necessary and sufficient for regulation of cell adhesion and cell migration under chemotherapeutical stress.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Silvia Salamone
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Bruno Jaime Santacreu
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Erika Nemeth
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Daniel Aguilar
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Barcelona, Catalunya, Spain
| | | | - Mohamad Adada
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Daniela I. Staquicini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ, United States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - John Haley
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Northport VA Hospital
- Stony Brook Cancer Center, Stony Brook, NY, United States
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, United States
- Department of Biochemistry, Stony Brook University
- Stony Brook Cancer Center, Stony Brook, NY, United States
| |
Collapse
|
6
|
Interaction of palmitate and LPS regulates cytokine expression and apoptosis through sphingolipids in human retinal microvascular endothelial cells. Exp Eye Res 2018; 178:61-71. [PMID: 30273577 DOI: 10.1016/j.exer.2018.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022]
Abstract
Studies have implicated saturated fatty acid (SFA) and lipopolysaccharide (LPS) in diabetic retinopathy. Since type 2 diabetes is associated with increases in both SFA and LPS in circulation, we investigated how SFA interacts with LPS to regulate proinflammatory cytokine expression and apoptosis in human retinal microvascular endothelial cells (HRMVECs) and the underlying mechanisms. HRMVECs were challenged with palmitate, a major SFA, LPS or palmitate plus LPS and the expression of proinflammatory cytokines were quantified using real-time PCR and enzyme-linked immunosorbent assay. The interaction between palmitate and LPS on inflammatory signaling and sphingolipid metabolism was demonstrated by immunoblotting and lipidomic analysis, respectively. The effect of palmitate and LPS on apoptosis was also studied by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and histone-associated DNA fragment assays. Results showed that palmitate robustly stimulated the expression of proinflammatory cytokines including interleukin (IL)-6 and IL-1β, and the combination of palmitate and LPS further upregulated the proinflammatory cytokines by cooperatively stimulating inflammatory signaling pathways. Results also showed that while palmitate stimulated ceramide (CER) production via CER de novo synthesis and sphingomyelin (SM) hydrolysis, addition of LPS further increased CER de novo synthesis, but not SM hydrolysis. The involvement of sphingolipids in the cooperative stimulation by palmitate and LPS on cytokine expression was indicated by the findings that the inhibitor of CER de novo synthesis or SM hydrolysis attenuated the stimulation of IL-6 expression by palmitate and LPS. In addition, our study showed that fatty acid receptors GPR40 and CD36 were involved in the IL-6 upregulation by palmitate and LPS. Furthermore, palmitate induced apoptosis via CER production, but addition of LPS did not further increase apoptosis. Taken together, this study showed that palmitate interacted with LPS to upregulate cytokine expression via free fatty acid receptor-mediated inflammatory signaling and sphingolipid metabolism in HRMVECs. In contrast, the interaction between palmitate and LPS did not further increase apoptosis.
Collapse
|
7
|
Lu MH, Ji WL, Xu DE, Yao PP, Zhao XY, Wang ZT, Fang LP, Huang R, Lan LJ, Chen JB, Wang TH, Cheng LH, Xu RX, Liu CF, Puglielli L, Ma QH. Inhibition of sphingomyelin synthase 1 ameliorates alzheimer-like pathology in APP/PS1 transgenic mice through promoting lysosomal degradation of BACE1. Exp Neurol 2018; 311:67-79. [PMID: 30243987 DOI: 10.1016/j.expneurol.2018.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/12/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
Sphingolipids emerge as essential modulators in the etiology of Alzheimer's disease (AD) with unclear mechanisms. Elevated levels of SM synthase 1 (SMS1), which catalyzes the synthesis of SM from ceramide and phosphatidylcholine, have been observed in the brains of Alzheimer's disease (AD), where expression of β-site APP cleaving enzyme 1 (BACE1), a rate limiting enzyme in amyloid-β (Aβ) generation, are upregulated. In the present study, we show knockdown of SMS1 via andeno associated virus (serotype 8, AAV8) in the hippocampus of APP/PS1 transgenic mice, attenuates the densities of Aβ plaques, neuroinflammation, synaptic loss and thus rescuing cognitive deficits of these transgenic mice. We further describe that knockdown or inhibition of SMS1 decreases BACE1 stability, which is accompanied with decreased BACE1 levels in the Golgi, whereas enhanced BACE1 levels in the early endosomes and the lysosomes. The reduction of BACE1 levels induced by knockdown or inhibition of SMS1 is prevented by inhibition of lysosomes. Therefore, knockdown or inhibition of SMS1 promotes lysosomal degradation of BACE1 via modulating the intracellular trafficking of BACE1. Knockdown of SMS1 attenuates AD-like pathology through promoting lysosomal degradation of BACE1.
Collapse
Affiliation(s)
- Mei-Hong Lu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Wen-Li Ji
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - De-En Xu
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi 214002, China
| | - Pei-Pei Yao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Xiu-Yun Zhao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhao-Tao Wang
- Affiliated Bayi Brain Hospital, Military General Hospital of Beijing PLA, Southern Medical University, Beijing 100700, China
| | - Li-Pao Fang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Rui Huang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Li-Jun Lan
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ji-Bo Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Ting-Hua Wang
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, China
| | - Li-Hua Cheng
- Affiliated Bayi Brain Hospital, Military General Hospital of Beijing PLA, Southern Medical University, Beijing 100700, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, Military General Hospital of Beijing PLA, Southern Medical University, Beijing 100700, China
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Luigi Puglielli
- Department of Medicine and Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho- Diseases, Institute of Neuroscience, Soochow University, Suzhou 215004, China; Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou 215004, China; Affiliated Bayi Brain Hospital, Military General Hospital of Beijing PLA, Southern Medical University, Beijing 100700, China.
| |
Collapse
|
8
|
Kong JN, Zhu Z, Itokazu Y, Wang G, Dinkins MB, Zhong L, Lin HP, Elsherbini A, Leanhart S, Jiang X, Qin H, Zhi W, Spassieva SD, Bieberich E. Novel function of ceramide for regulation of mitochondrial ATP release in astrocytes. J Lipid Res 2018; 59:488-506. [PMID: 29321137 DOI: 10.1194/jlr.m081877] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/08/2018] [Indexed: 12/14/2022] Open
Abstract
We reported that amyloid β peptide (Aβ42) activated neutral SMase 2 (nSMase2), thereby increasing the concentration of the sphingolipid ceramide in astrocytes. Here, we show that Aβ42 induced mitochondrial fragmentation in wild-type astrocytes, but not in nSMase2-deficient cells or astrocytes treated with fumonisin B1 (FB1), an inhibitor of ceramide synthases. Unexpectedly, ceramide depletion was concurrent with rapid movements of mitochondria, indicating an unknown function of ceramide for mitochondria. Using immunocytochemistry and super-resolution microscopy, we detected ceramide-enriched and mitochondria-associated membranes (CEMAMs) that were codistributed with microtubules. Interaction of ceramide with tubulin was confirmed by cross-linking to N-[9-(3-pent-4-ynyl-3-H-diazirine-3-yl)-nonanoyl]-D-erythro-sphingosine (pacFACer), a bifunctional ceramide analog, and binding of tubulin to ceramide-linked agarose beads. Ceramide-associated tubulin (CAT) translocated from the perinuclear region to peripheral CEMAMs and mitochondria, which was prevented in nSMase2-deficient or FB1-treated astrocytes. Proximity ligation and coimmunoprecipitation assays showed that ceramide depletion reduced association of tubulin with voltage-dependent anion channel 1 (VDAC1), an interaction known to block mitochondrial ADP/ATP transport. Ceramide-depleted astrocytes contained higher levels of ATP, suggesting that ceramide-induced CAT formation leads to VDAC1 closure, thereby reducing mitochondrial ATP release, and potentially motility and resistance to Aβ42 Our data also indicate that inhibiting ceramide generation may protect mitochondria in Alzheimer's disease.
Collapse
Affiliation(s)
- Ji-Na Kong
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Zhihui Zhu
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Physiology, University of Kentucky, Lexington, KY
| | - Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA
| | - Guanghu Wang
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Physiology, University of Kentucky, Lexington, KY
| | - Michael B Dinkins
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA
| | - Liansheng Zhong
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Physiology, University of Kentucky, Lexington, KY.,College of Basic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Hsuan-Pei Lin
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Physiology, University of Kentucky, Lexington, KY
| | - Ahmed Elsherbini
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA.,Department of Physiology, University of Kentucky, Lexington, KY
| | - Silvia Leanhart
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA
| | - Xue Jiang
- Department of Physiology, University of Kentucky, Lexington, KY.,Rehabilitation Center, ShengJing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Haiyan Qin
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Wenbo Zhi
- Center of Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | | | - Erhard Bieberich
- Department of Neuroscience and Regenerative Medicine Augusta University, Augusta, GA .,Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
9
|
Dinkins MB, Dasgupta S, Wang G, Zhu G, He Q, Kong JN, Bieberich E. The 5XFAD Mouse Model of Alzheimer's Disease Exhibits an Age-Dependent Increase in Anti-Ceramide IgG and Exogenous Administration of Ceramide Further Increases Anti-Ceramide Titers and Amyloid Plaque Burden. J Alzheimers Dis 2016; 46:55-61. [PMID: 25720409 DOI: 10.3233/jad-150088] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We present evidence that 5XFAD Alzheimer's disease model mice develop an age-dependent increase in antibodies against ceramide, suggesting involvement of autoimmunity against ceramide in Alzheimer's disease pathology. To test this, we increased serum anti-ceramide IgG (2-fold) by ceramide administration and analyzed amyloid plaque formation in 5XFAD mice. There were no differences in soluble or total amyloid-β levels. However, females receiving ceramide had increased plaque burden (number, area, and size) compared to controls. Ceramide-treated mice showed an increase of serum exosomes (up to 3-fold using Alix as marker), suggesting that systemic anti-ceramide IgG and exosome levels are correlated with enhanced plaque formation.
Collapse
|
10
|
Ong WY, Herr DR, Farooqui T, Ling EA, Farooqui AA. Role of sphingomyelinases in neurological disorders. Expert Opin Ther Targets 2015; 19:1725-42. [DOI: 10.1517/14728222.2015.1071794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Neutral sphingomyelinase-2 is a redox sensitive enzyme: role of catalytic cysteine residues in regulation of enzymatic activity through changes in oligomeric state. Biochem J 2015; 465:371-82. [PMID: 25287744 DOI: 10.1042/bj20140665] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neutral sphingomyelinase-2 (nSMase-2) is the major sphingomyelinase activated in response to pro-inflammatory cytokines and during oxidative stress. It is a membrane-bound 655 amino acid protein containing 22 cysteine residues. In this study, we expressed recombinant mouse nSMase-2 protein in Escherichia coli, and investigated whether nSMase-2 is a redox sensitive enzyme. Our results demonstrate that nSMase-2 exists as both monomers and multimers that are associated with high and low enzymatic activity respectively. Mutational analysis of nSMase-2 identified within its C-terminal catalytic domain several oxidant-sensitive cysteine residues that were shown to be involved in enzyme oligomerization. Changing Cys(617) to Ser for example is a gain-of-function mutation associated with a decreased propensity for oligomerization. Alternatively, nSMase-2 expression in a bacterial strain that lacks endogenous thioredoxin, Rosetta-gami2, results in increased oligomer formation and lower enzyme activity. Phenotypic rescue was accomplished by treating nSMase-2 lysates with recombinant human thioredoxin. This indicates that nSMase-2 may be a novel substrate for thioredoxin. FRET analysis confirmed the presence of nSMase-2 multimers in mammalian HEK cells and their localization to the plasma membrane. In conclusion, our results identify nSMase-2 as a redox-sensitive enzyme, whose basal activity is influenced by thioredoxin-mediated changes in its oligomeric state.
Collapse
|
12
|
Alessenko AV. The potential role for sphingolipids in neuropathogenesis of Alzheimer’s disease. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2013. [DOI: 10.1134/s1990750813020029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Liu L, Martin R, Chan C. Palmitate-activated astrocytes via serine palmitoyltransferase increase BACE1 in primary neurons by sphingomyelinases. Neurobiol Aging 2013; 34:540-50. [PMID: 22727944 PMCID: PMC3459302 DOI: 10.1016/j.neurobiolaging.2012.05.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/27/2012] [Accepted: 05/26/2012] [Indexed: 10/28/2022]
Abstract
Astrocytes play a critical role in neurodegenerative diseases, including Alzheimer's disease (AD). Previously, we showed that saturated free fatty acid, palmitic acid (PA), upregulates β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) level and amyloidogenesis in primary rat neurons mediated by astrocytes. However, the molecular mechanisms by which conditioned media from PA-treated astrocytes upregulates BACE1 level in neurons are unknown. This study demonstrates that serine palmitoyltransferase (SPT) in the astrocytes increases ceramide levels, which enhances the release of cytokines that mediate the activation of neural and acidic sphingomyelinase (SMase) in the neurons, to propagate the deleterious effects of PA (i.e., BACE1 upregulation). In support of the relevance of SPT in AD, our laboratory recently measured and found SPT levels to be significantly upregulated in AD brains as compared with controls. Cytokines, namely tumor necrosis factor-α and interleukin-1β, released into the conditioned media of PA-treated astrocytes activate neural and acidic SMase in the neurons. Neutralizing the cytokines in the PA-treated astrocyte conditioned media reduced BACE1 upregulation. However, inhibiting SPT in the astrocytes decreased the levels of both tumor necrosis factor-α and interleukin-1β in the conditioned media, which in turn reduced the SMase activities and BACE1 level in primary neurons. Thus, our results suggest that the activation of the astrocytes by PA is mediated by SPT, and the activated astrocytes increases BACE1 level in the neurons; the latter is mediate by the SMases.
Collapse
Affiliation(s)
- Li Liu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Rebecca Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
| | - Christina Chan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
14
|
Alessenko A. The potential role for sphingolipids in neuropathogenesis of Alzheimer’s disease. ACTA ACUST UNITED AC 2013. [DOI: 10.18097/pbmc20135901025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review discusses the functional role of sphingolipids in the pathogenesis of Alzheimer's disease. Certain evidence exist that the imbalance of sphingolipids such as sphingomyelin, ceramide, sphingosine, sphingosine-1-phosphate and galactosylceramide in the brain of animals and humans, in the cerebrospinal fluid and blood plasma of patients with Alzheimer's disease play a crucial role in neuronal function by regulating growth, differentiation and cell death in CNS. Activation of sphingomyelinase, which leads to the accumulation of the proapoptotic agent, ceramide, can be considered as a new mechanism for AD and may be a prerequisite for the treatment of this disease by using drugs that inhibit sphingomyelinase activity. The role of sphingolipids as biomarkers for the diagnosis of the early stage of Alzheimer's disease and monitoring the effectiveness of treatment with new drugs is discussed.
Collapse
Affiliation(s)
- A.V. Alessenko
- N.M. Emanuel Institute of Biochemical Physics of the Russian Academy of Sciences
| |
Collapse
|
15
|
Renin inhibitor aliskiren exerts neuroprotection against amyloid beta-peptide toxicity in rat cortical neurons. Neurochem Int 2012; 61:369-77. [DOI: 10.1016/j.neuint.2012.05.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 04/24/2012] [Accepted: 05/09/2012] [Indexed: 12/20/2022]
|
16
|
Chen SD, Yin JH, Hwang CS, Tang CM, Yang DI. Anti-apoptotic and anti-oxidative mechanisms of minocycline against sphingomyelinase/ceramide neurotoxicity: implication in Alzheimer's disease and cerebral ischemia. Free Radic Res 2012; 46:940-50. [PMID: 22583533 DOI: 10.3109/10715762.2012.674640] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sphingolipids represent a major class of lipids in which selected family members act as bioactive molecules that control diverse cellular processes, such as proliferation, differentiation, growth, senescence, migration and apoptosis. Emerging evidence reveals that sphingomyelinase/ceramide pathway plays a pivotal role in neurodegenerative diseases that involve mitochondrial dysfunction, oxidative stress and apoptosis. Minocycline, a semi-synthetic second-generation tetracycline derivative in clinical use for infection control, is also considered an effective protective agent in various neurodegenerative diseases in pre-clinical studies. Acting via multiple mechanisms, including anti-inflammatory, anti-oxidative and anti-apoptotic effects, minocycline is a desirable candidate for clinical trials in both acute brain injury as well as chronic neurodegenerative disorders. This review is focused on the anti-apoptotic and anti-oxidative mechanisms of minocycline against neurotoxicity induced by sphingomyelinase/ceramide in relation to neurodegeneration, particularly Alzheimer's disease and cerebral ischemia.
Collapse
Affiliation(s)
- Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
17
|
The Roles of Neutral Sphingomyelinases in Neurological Pathologies. Neurochem Res 2012; 37:1137-49. [DOI: 10.1007/s11064-011-0692-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 11/25/2011] [Accepted: 12/29/2011] [Indexed: 12/14/2022]
|
18
|
Lin WC, Lin CF, Chen CL, Chen CW, Lin YS. Inhibition of neutrophil apoptosis via sphingolipid signaling in acute lung injury. J Pharmacol Exp Ther 2011; 339:45-53. [PMID: 21724966 DOI: 10.1124/jpet.111.181560] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Acute lung injury (ALI) is characterized by lung inflammation and diffuse infiltration of neutrophils into the alveolar space. The inhibition of alveolar neutrophil apoptosis has been implicated in the pathogenesis of ALI. Although sphingolipids may regulate cell apoptosis, the role of sphingolipids in activated neutrophils during ALI is not clear. In this study, we test the hypothesis that sphingolipids would attenuate neutrophil apoptosis that contributes to the development of ALI. Lipopolysaccharide (LPS)-stimulated human neutrophils, with or without inhibitor treatment, were analyzed for apoptosis. We found that the inhibitory effect of LPS on neutrophil apoptosis was blocked by treatment with the neutral sphingomyelinase (nSMase) inhibitor sphingolactone-24 (Sph-24), sphingosine kinase inhibitor II, and p38 mitogen-activated protein kinase (MAPK) inhibitor 4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine (SB203580) but not by the acidic sphingomyelinase inhibitor chlorpromazine. LPS-activated phosphorylation of p38 MAPK also was attenuated by treatment with Sph-24 and sphingosine kinase inhibitor II. Furthermore, mice with LPS-induced lung injury were treated with the nSMase inhibitor Sph-24 to evaluate its impact on lung injury and survival. The severity of LPS-induced ALI was reduced, and the survival rate was increased in mice treated with Sph-24 compared with that in those given LPS alone. Intracellular levels of sphingolipids in alveolar neutrophils from patients with acute respiratory distress syndrome also were measured. We found that intracellular levels of ceramide and phospho-p38 MAPK were elevated in alveolar neutrophils from acute respiratory distress syndrome patients. Our results demonstrate that activation of the nSMase/sphingosine-1-phosphate pathway to induce p38 MAPK phosphorylation results in inhibition of neutrophil apoptosis, which may contribute to the development of ALI.
Collapse
Affiliation(s)
- Wei-Chieh Lin
- Department of Microbiology and Immunology, Institute of Clinical Medicine, National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
19
|
Chiu WT, Shen SC, Yang LY, Chow JM, Wu CY, Chen YC. Inhibition of HSP90-dependent telomerase activity in amyloid β-induced apoptosis of cerebral endothelial cells. J Cell Physiol 2011; 226:2041-51. [DOI: 10.1002/jcp.22536] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
20
|
Tang CM, Hwang CS, Chen SD, Yang DI. Neuroprotective mechanisms of minocycline against sphingomyelinase/ceramide toxicity: Roles of Bcl-2 and thioredoxin. Free Radic Biol Med 2011; 50:710-21. [PMID: 21184825 DOI: 10.1016/j.freeradbiomed.2010.12.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 12/11/2010] [Accepted: 12/15/2010] [Indexed: 12/17/2022]
Abstract
In this study, we determined whether minocycline may protect rat cortical cultures against neurotoxicity induced by sphingomyelinase/ceramide and explored the underlying mechanisms. We found that minocycline exerted strong neuroprotective effects against toxicity induced by bacterial sphingomyelinase and synthetic C2 ceramide. Minocycline enhanced the production of nitric oxide (NO) with resultant increases in cellular cGMP content. Consistently, minocycline-dependent neuroprotection was abolished by the nitric oxide synthase inhibitor L-N(G)-nitroarginine methyl ester (L-NAME) and the soluble guanylate cyclase (sGC) inhibitor 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ). Western blotting revealed that minocycline restored the expression levels of cGMP-dependent protein kinase (PKG)-1, antioxidative thioredoxin-1, and antiapoptotic Bcl-2 that were down-regulated by bacterial sphingomyelinase. Accordingly, the PKG inhibitor KT5823, the thioredoxin reductase inhibitor 1-chloro-2,4-dinitrobenzene (DNCB), and a Bcl-2 inhibitor significantly abolished the minocycline neuroprotection. The minocycline-dependent restoration of Bcl-2 was abolished by L-NAME, ODQ, and KT5823, but not by DNCB, suggesting the involvement of NO/sGC/PKG but not thioredoxin. Furthermore, minocycline-dependent recovery of thioredoxin-1 was PKG-independent. Taken together, our results indicate that minocycline protects rat cortical neurons against bacterial sphingomyelinase/ceramide toxicity via an NO/cGMP/PKG pathway with induction of Bcl-2 and PKG-independent stimulation of thioredoxin-1.
Collapse
Affiliation(s)
- Ching-Min Tang
- Institute of Brain Science and Brain Research Center, National Yang-Ming University, Taipei City 11221, Taiwan, Republic of China
| | | | | | | |
Collapse
|
21
|
Chen KL, Yuan RY, Hu CJ, Hsu CY. Amyloid-β peptide alteration of tau exon-10 splicing via the GSK3β-SC35 pathway. Neurobiol Dis 2010; 40:378-85. [PMID: 20615469 DOI: 10.1016/j.nbd.2010.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 06/11/2010] [Accepted: 06/26/2010] [Indexed: 10/19/2022] Open
Abstract
Amyloid-beta peptide (Aβ) and Tau protein are the lead constituents in the pathogenesis of Alzheimer's disease (AD). However, their inter-relationship in the disease process remains to be established. Tauopathy refers to a characteristic neurodegenerative process in AD. In tauopathy, Tau accumulates as a consequence of altered pre-mRNA splicing of tau exon 10, resulting in 3R (without exon 10)/4R (with exon 10) imbalance. We studied Aβ effects on tau exon 10 pre-mRNA splicing and relevant signaling events. This is the first demonstration of Aβ alteration of tau exon 10 splicing with an increase in 3R/4R ratio caused by reduced 4R expression. This Aβ action is causally related to its activation of GSK-3β which in turn phosphorylates SC35, an enhancer in tau exon 10 splicing. The establishment of the Aβ-GSK-3β-SC35 cascade broadens insight into development of novel strategies to modulate Aβ action on tau exon 10 splicing for possible prevention of tauopathy.
Collapse
Affiliation(s)
- Kun-Lin Chen
- The Department of Medical Education and Research, Shih Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | |
Collapse
|
22
|
Wu BX, Clarke CJ, Hannun YA. Mammalian neutral sphingomyelinases: regulation and roles in cell signaling responses. Neuromolecular Med 2010; 12:320-30. [PMID: 20552297 DOI: 10.1007/s12017-010-8120-z] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 06/02/2010] [Indexed: 12/22/2022]
Abstract
Ceramide, a bioactive lipid, has been extensively studied and identified as an essential bioactive molecule in mediating cellular signaling pathways. Sphingomyelinase (SMase), (EC 3.1.4.12) catalyzes the cleavage of the phosphodiester bond in sphingomyelin (SM) to form ceramide and phosphocholine. In mammals, three Mg(2+)-dependent neutral SMases termed nSMase1, nSMase2 and nSMase3 have been identified. Among the three enzymes, nSMase2 is the most studied and has been implicated in multiple physiological responses including cell growth arrest, apoptosis, development and inflammation. In this review, we summarize recent findings for the cloned nSMases and discuss the insights for their roles in regulation ceramide metabolism and cellular signaling pathway.
Collapse
Affiliation(s)
- Bill X Wu
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, USA
| | | | | |
Collapse
|
23
|
Hsu MJ, Sheu JR, Lin CH, Shen MY, Hsu CY. Mitochondrial mechanisms in amyloid beta peptide-induced cerebrovascular degeneration. Biochim Biophys Acta Gen Subj 2010; 1800:290-6. [DOI: 10.1016/j.bbagen.2009.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 08/06/2009] [Accepted: 08/11/2009] [Indexed: 01/19/2023]
|
24
|
Manzo F, Tambaro FP, Mai A, Altucci L. Histone acetyltransferase inhibitors and preclinical studies. Expert Opin Ther Pat 2009; 19:761-74. [PMID: 19473103 DOI: 10.1517/13543770902895727] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Drugs able to regulate the histone modifier enzymes are very promising tools for the treatment of several diseases, such as cancer. Histone acetyltransferase (HAT) inhibitors are compounds able to inhibit the catalytic activity of HATs reported to be active in cancer, or in several other diseases, such as Alzheimer (AD), diabetes and hyperlipidaemia. OBJECTIVES Here we review the status and the rationale for the use of HAT inhibitors in the treatment of various diseases. METHODS Patents have been found on the espacenet database; the clinical trials have been reported as in the clinicaltrial.gov website. RESULTS AND CONCLUSION Despite the fact that other drugs able to regulate the histone modifier enzymes (such as histone deacetylase inhibitors) have been already approved for the treatment of cancer, HAT inhibitors seem promising for the treatment of human diseases such as AD and diabetes, although side effects and toxicity need to be investigated.
Collapse
Affiliation(s)
- Fabio Manzo
- Dipartimento di Patologia generale, Seconda Università degli Studi di Napoli, Vico L. De Crecchio 7, 80138, Napoli, IT.
| | | | | | | |
Collapse
|
25
|
Hsu MJ, Hsu CY, Chen BC, Chen MC, Ou G, Lin CH. Apoptosis signal-regulating kinase 1 in amyloid beta peptide-induced cerebral endothelial cell apoptosis. J Neurosci 2007; 27:5719-29. [PMID: 17522316 PMCID: PMC6672775 DOI: 10.1523/jneurosci.1874-06.2007] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A pathological hallmark of Alzheimer's disease is accumulation of amyloid-beta peptide (Abeta) in senile plaques. Abeta has also been implicated in vascular degeneration in cerebral amyloid angiopathy because of its cytotoxic effects on non-neuronal cells, including cerebral endothelial cells (CECs). We explore the role of apoptosis signal-regulating kinase 1 (ASK1) in Abeta-induced death in primary cultures of murine CECs. Abeta induced ASK1 dephosphorylation, which could be prevented by selective inhibition of protein phosphatase 2A (PP2A) but not PP2B. ASK1 dephosphorylation resulted in its dissociation from 14-3-3. ASK1, released from 14-3-3 inhibition, activated p38 mitogen-activated protein kinase (p38MAPK), leading to p53 phosphorylation. p53, a proapoptotic transcription factor, in turn transactivated the expression of Bax, a proapoptotic protein. Transfection with various dominant-negative mutants (DNs), including ASK1 DN and p38MAPK DN, suppressed Abeta-induced p38MAPK activation, p53 phosphorylation, and Bax upregulation and partially prevented CEC death. Bax knockdown using a bax small interfering RNA strategy also reduced Bax expression and subsequent CEC death. These results suggest that Abeta activates the ASK1-p38MAPK-p53-Bax cascade to cause CEC death in a PP2A-dependent manner.
Collapse
Affiliation(s)
- Ming-Jen Hsu
- Graduate Institute of Medical Sciences
- Topnotch Stroke Research Center, Taipei Medical University, Taipei 110, Taiwan, and
| | - Chung Y. Hsu
- Department of Neurology and Chi-Chin Huang Stroke Research Center
- Topnotch Stroke Research Center, Taipei Medical University, Taipei 110, Taiwan, and
| | | | - Mei-Chieh Chen
- Department of Microbiology and Immunology, College of Medicine, and
| | - George Ou
- Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences
- Topnotch Stroke Research Center, Taipei Medical University, Taipei 110, Taiwan, and
| |
Collapse
|
26
|
Gomez-Brouchet A, Pchejetski D, Brizuela L, Garcia V, Altié MF, Maddelein ML, Delisle MB, Cuvillier O. Critical Role for Sphingosine Kinase-1 in Regulating Survival of Neuroblastoma Cells Exposed to Amyloid-β Peptide. Mol Pharmacol 2007; 72:341-9. [PMID: 17522181 DOI: 10.1124/mol.106.033738] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We examined the role of sphingosine kinase-1 (SphK1), a critical regulator of the ceramide/sphingosine 1-phosphate (S1P) biostat, in the regulation of death and survival of SH-SY5Y neuroblastoma cells in response to amyloid beta (Abeta) peptide (25-35). Upon incubation with Abeta, SH-SY5Y cells displayed a marked down-regulation of SphK1 activity coupled with an increase in the ceramide/S1P ratio followed by cell death. This mechanism was redox-sensitive; N-acetylcysteine totally abrogated the down-regulation of SphK1 activity and strongly inhibited Abeta-induced cell death. SphK1 overexpression impaired the cytotoxicity of Abeta, whereas SphK1 silencing by RNA interference mimicked Abeta-induced cell death, thereby establishing a critical role for SphK1. We further demonstrated that SphK1 could mediate the well established cytoprotective action of insulin-like growth factor (IGF-I) against Abeta toxicity. A dominant-negative form of SphK1 or its pharmacological inhibition not only abrogated IGF-I-triggered stimulation of SphK1 but also hampered IGF-I protective effect. Similarly to IGF-I, the neuroprotective action of TGF-beta1 was also dependent on SphK1 activity; activation of SphK1 as well as cell survival were impeded by a dominant-negative form of SphK1. Taken together, these results provide the first illustration of SphK1 role as a critical regulator of death and survival of Abeta-treated cells.
Collapse
Affiliation(s)
- Anne Gomez-Brouchet
- Institut National de la Santé et de la Recherche Médicale U466, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Clarke CJ, Hannun YA. Neutral sphingomyelinases and nSMase2: Bridging the gaps. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1758:1893-901. [PMID: 16938269 DOI: 10.1016/j.bbamem.2006.06.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Revised: 06/09/2006] [Accepted: 06/12/2006] [Indexed: 11/30/2022]
Abstract
There is strong evidence indicating a role for ceramide as a second messenger in processes such as apoptosis, cell growth and differentiation, and cellular responses to stress. Ceramide formation from the hydrolysis of sphingomyelin is considered to be a major pathway of stress-induced ceramide production with magnesium-dependent neutral sphingomyelinase (N-SMase) identified as a prime candidate in this pathway. The recent cloning of a mammalian N-SMase-nSMase2- and generation of nSMase2 knockout/mutant mice have now provided vital tools with which to further study the regulation and roles of this enzyme in both a physiological and pathological context. In the present review, we summarize current knowledge on N-SMase relating this to what is known about nSMase2. We also discuss the future areas of nSMase2 research important for molecular understanding of this enzyme and its physiological roles.
Collapse
Affiliation(s)
- Christopher J Clarke
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | |
Collapse
|
28
|
Posse de Chaves EI. Sphingolipids in apoptosis, survival and regeneration in the nervous system. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1758:1995-2015. [PMID: 17084809 DOI: 10.1016/j.bbamem.2006.09.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Revised: 09/20/2006] [Accepted: 09/21/2006] [Indexed: 12/27/2022]
Abstract
Simple sphingolipids such as ceramide, sphingosine and sphingosine 1-phosphate are key regulators of diverse cellular functions. Their roles in the nervous system are supported by extensive evidence derived primarily from studies in cultured cells. More recently animal studies and studies with human samples have revealed the importance of ceramide and its metabolites in the development and progression of neurodegenerative disorders. The roles of sphingolipids in neurons and glial cells are complex, cell dependent, and many times contradictory. In this review I will summarize the effects elicited by ceramide and ceramide metabolites in cells of the nervous system, in particular those effects related to cell survival and death, emphasizing the molecular mechanisms involved. I also discuss recent evidence for the implication of sphingolipids in the development and progression of certain dementias.
Collapse
Affiliation(s)
- Elena I Posse de Chaves
- Centre for Alzheimer and Neurodegenerative Research, Signal Transduction Research Group and Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada T6G 2H7.
| |
Collapse
|
29
|
Chen SD, Hu CJ, Yang DI, Nassief A, Chen H, Yin K, Xu J, Hsu CY. Pravastatin attenuates ceramide-induced cytotoxicity in mouse cerebral endothelial cells with HIF-1 activation and VEGF upregulation. Ann N Y Acad Sci 2006; 1042:357-64. [PMID: 15965081 DOI: 10.1196/annals.1338.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ceramide is a pro-apoptotic lipid messenger that induces oxidative stress and may mediate apoptosis in cerebral endothelial cells (CECs) induced by TNF-alpha/cycloheximide, lipopolysaccharide, oxidized LDL, IL-1, and amyloid peptide. Exposure of CECs to C2 ceramide for 12 h caused cell death in a concentration-dependent manner, with a LC50 of 30 microM. Statins are inhibitors of 3-hydroxyl-3-methyl coenzyme A reductase which is the rate-limiting enzyme for cholesterol biosynthesis. Pretreatment with pravastatin at 20 microM for 16 h substantially attenuated ceramide cytotoxicity in mouse CECs. Increases in vascular endothelial growth factor (VEGF) expression were detected within 1-3 h after pravastatin treatment. This pravastatin action was accompanied by the activation of hypoxia-inducible factor-1 (HIF-1), a transcription factor known to activate VEGF expression. These results raise the possibility that pravastatin may protect CECs against ceramide-induced death via the HIF-VEGF cascade.
Collapse
Affiliation(s)
- Shang-Der Chen
- Department of Neurology, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Yin KJ, Hsu CY, Hu XY, Chen H, Chen SW, Xu J, Lee JM. Protein phosphatase 2A regulates bim expression via the Akt/FKHRL1 signaling pathway in amyloid-beta peptide-induced cerebrovascular endothelial cell death. J Neurosci 2006; 26:2290-9. [PMID: 16495456 PMCID: PMC6674809 DOI: 10.1523/jneurosci.5103-05.2006] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Amyloid-beta peptide (Abeta)-induced death in cerebral endothelial cells (CECs) is preceded by mitochondrial dysfunction and signaling events characteristic of apoptosis. Mitochondria-dependent apoptosis engages Bcl-2 family proteins, especially the BH3-only homologues, which play a key role in initiating the apoptotic cascade. Here, we report that the expression of bim, but not other BH3-only members, was selectively increased in cerebral microvessels isolated from 18-month-old APPsw (Tg2576) mice, a model of cerebral amyloid angiopathy (CAA), suggesting a pivotal role for Bim in Abeta-induced cerebrovascular degeneration in vivo. A similar expression profile was observed in Abeta-treated CECs. Furthermore, Abeta induction of bim expression involved a pro-apoptotic transcription factor, FKHRL1. FKHRL1 bound to a consensus sequence in the bim promoter region and was activated by Abeta before bim expression. FKHRL1 activity was negatively regulated by phosphorylation catalyzed by Akt, an anti-apoptotic kinase. Akt upregulation by adenoviral gene transfer inhibited Abeta-induced FKHRL1 activation and bim induction. In addition, Abeta increased the activity of protein phosphatase 2A (PP2A), a ceramide-activated protein phosphatase. Suppression of PP2A activity by RNA interference or a specific inhibitor, okadaic acid, effectively suppressed Abeta-induced Akt inactivation and FKHRL1 activation, leading to an attenuation of bim expression and cell death in CECs. Coimmunoprecipitation experiments revealed that Abeta enhanced the binding of the PP2A regulatory subunit PP2ACalphabeta to Akt. These results implicate PP2A as an early regulator of Abeta-induced bim expression and CEC apoptosis via the Akt/FKHRL1 signaling pathway. We raise the possibility that this pathway may play a role in cerebrovascular degeneration in CAA.
Collapse
|
31
|
Ju TC, Chen SD, Liu CC, Yang DI. Protective effects of S-nitrosoglutathione against amyloid beta-peptide neurotoxicity. Free Radic Biol Med 2005; 38:938-49. [PMID: 15749390 DOI: 10.1016/j.freeradbiomed.2004.12.019] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Accepted: 12/14/2004] [Indexed: 01/17/2023]
Abstract
Amyloid beta-peptide (Abeta) is a major constituent of senile plaques in the brains of Alzheimer's disease (AD) patients. We have previously demonstrated ceramide production secondary to Abeta-induced activation of neutral sphingomyelinase (nSMase) in cerebral endothelial cells and oligodendrocytes, which may contribute to cellular injury during progression of AD. In this study, we first established the "Abeta --> nSMase --> ceramide --> free radical --> cell death" pathway in primary cultures of fetal rat cortical neurons. We also provided experimental evidence showing that S-nitrosoglutathione (GSNO), a potent endogenous antioxidant derived from the interaction between nitric oxide (NO) and glutathione, caused dose-dependent protective effects against Abeta/ceramide neurotoxicity via inhibition of caspase activation and production of reactive oxygen species (ROS). This GSNO-mediated neuroprotection appeared to involve activation of cGMP-dependent protein kinase (PKG), phosphatidylinositol 3-kinase (PI3K), and extracellular signal-regulated kinase (ERK). Activation of the cGMP/PKG pathway induced expression of thioredoxin and Bcl-2 that were beneficial to cortical neurons in antagonizing Abeta/ceramide toxicity. Consistently, exogenous application of thioredoxin exerted remarkable neuroprotective efficacy in our experimental paradigm. Results derived from the present study establish a neuroprotective role of GSNO, an endogenous NO carrier, against Abeta toxicity via multiple signaling pathways.
Collapse
Affiliation(s)
- Tzyh-Chwen Ju
- Institute of Neuroscience, Tzu Chi University 701, Section 3, Chung-Yang Road, Hualien 970, Taiwan
| | | | | | | |
Collapse
|
32
|
Beier UH, Görögh T. Implications of galactocerebrosidase and galactosylcerebroside metabolism in cancer cells. Int J Cancer 2005; 115:6-10. [PMID: 15657896 DOI: 10.1002/ijc.20851] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Galactosylcerebroside is known to be overexpressed upon the cellular surface of a variety of cancers. In squamous cell carcinomas of the head and neck, one explanation for galactosylcerebroside accumulation has been identified as a transcriptional repression of the galactocerebrosidase gene. Galactocerebrosidase is the enzyme responsible for degrading galactosylcerebroside to ceramide. Ceramide is an important apoptosis activator, whereas galactosylcerebroside functions as an inhibitor. A shift of the ceramide metabolism balance in favor of glycosylated forms has been identified as a mechanism of drug resistance for several antineoplastic agents. Our review elaborates on possible explanations for galactocerebrosidase suppression and on other explanations for increased glycosphingolipid concentration within cancer cell membranes. Furthermore, conjecturable influences of a repressed galactocerebrosidase expression on tumor biology are to be explained. The inhibiting transcription factors YY1 and AP2 have been identified as potential galactocerebrosidase gene suppressors. The resulting accumulation of galactosylcerebroside promotes a reduction of cellular adhesion and inhibits apoptosis, leading to increased cellular growth, migration and prolonged cell survival contributing to carcinogenesis.
Collapse
Affiliation(s)
- Ulf Henning Beier
- Division of Molecular Oncology, Department of Otorhinolaryngology, Head and Neck Surgery,Christian-Albrechts-University of Kiel, Kiel, Germany
| | | |
Collapse
|