1
|
Sinha S, Wal P, Goudanavar P, Divya S, Kimothi V, Jyothi D, Sharma MC, Wal A. Research on Alzheimer's Disease (AD) Involving the Use of In vivo and In vitro Models and Mechanisms. Cent Nerv Syst Agents Med Chem 2025; 25:123-142. [PMID: 38803173 DOI: 10.2174/0118715249293642240522054929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/11/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by the progressive formation of extracellular amyloid plaques, intracellular neurofibrillary tangles, inflammation, and impaired antioxidant systems. Early detection and intervention are vital for managing AD effectively. OBJECTIVES This review scrutinizes both in-vivo and in-vitro screening models employed in Alzheimer's disease research. in-vivo models, including transgenic mice expressing AD-related mutations, offer profound insights into disease progression and potential therapeutic targets. A thorough understanding of these models and mechanisms will facilitate the development of novel therapies and interventions for Alzheimer's disease. This review aims to provide an overview of the current experimental models in AD research, assess their strengths and weaknesses as model systems, and underscore the future prospects of experimental AD modeling. METHODS We conducted a systematic literature search across multiple databases, such as Pub- Med, Bentham Science, Elsevier, Springer Nature, Wiley, and Research Gate. The search strategy incorporated pertinent keywords related to Alzheimer's disease, in-vivo models, in-vitro models, and screening mechanisms. Inclusion criteria were established to identify studies focused on in-vivo and in-vitro screening models and their mechanisms in Alzheimer's disease research. Studies not meeting the predefined criteria were excluded from the review. RESULTS A well-structured experimental animal model can yield significant insights into the neurobiology of AD, enhancing our comprehension of its pathogenesis and the potential for cutting-edge therapeutic strategies. Given the limited efficacy of current AD medications, there is a pressing need for the development of experimental models that can mimic the disease, particularly in pre-symptomatic stages, to investigate prevention and treatment approaches. To address this requirement, numerous experimental models replicating human AD pathology have been established, serving as invaluable tools for assessing potential treatments. CONCLUSION In summary, this comprehensive review underscores the pivotal role of in-vivo and in-vitro screening models in advancing our understanding of Alzheimer's disease. These models offer invaluable insights into disease progression, pathological mechanisms, and potential therapeutic targets. By conducting a rigorous investigation and evaluation of these models and mechanisms, effective screening and treatment methods for Alzheimer's disease can be devised. The review also outlines future research directions and areas for enhancing AD screening models.
Collapse
Affiliation(s)
- Sweta Sinha
- LCIT School of Pharmacy, Bilaspur, Chattisgarh, India
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy) NH19 Kanpur Agra Highway, Bhaunti Kanpur, India
| | - Prakash Goudanavar
- Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University B.G.Nagara, Karnataka, India
| | | | | | - Divya Jyothi
- NGSM Institute of Pharmaceutical Sciences, Nitte University, Paneer Deralakatte, Mangaluru, 575018, India
| | | | - Ankita Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy) NH19 Kanpur Agra Highway, Bhaunti Kanpur, India
| |
Collapse
|
2
|
Sun W, Chen Y, Yang Y, Wang P, Gong J, Han X, Xu C, Luan H, Li S, Li R, Wen B, Lv S, Wei C. Characteristics and Transcriptomic Analysis of Cholinergic Neurons Derived from Induced Pluripotent Stem Cells with APP Mutation in Alzheimer's Disease. J Alzheimers Dis 2024; 101:637-649. [PMID: 39213067 DOI: 10.3233/jad-240299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background The cholinergic hypothesis is one of the main theories that describe the pathogenesis of Alzheimer's disease (AD). Cholinergic neurons degenerate early and are severely damaged in AD. Despite extensive research, the causes of cholinergic neuron damage and the underlying molecular changes remain unclear. Objective This study aimed to explore the characteristics and transcriptomic changes in cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) with APP mutation. Methods Peripheral blood mononuclear cells from patients with AD and healthy individuals were reprogrammed into iPSCs. The iPSCs were differentiated into cholinergic neurons. Cholinergic neurons were stained, neurotoxically tested, and electrophysiologically and transcriptomically analyzed. Results The iPSCs-derived cholinergic neurons from a patient with AD carrying a mutation in APP displayed enhanced susceptibility to Aβ1-42-induced neurotoxicity, characterized by severe neurotoxic effects, such as cell body coagulation and neurite fragmentation. Cholinergic neurons exhibited electrophysiological impairments and neuronal death after 21 days of culture in the AD group. Transcriptome analysis disclosed 883 differentially expressed genes (DEGs, 420 upregulated and 463 downregulated) participating in several signaling pathways implicated in AD pathogenesis. To assess the reliability of RNA sequencing, the expression of 16 target DEGs was validated using qPCR. Finally, the expression of the 8 core genes in different cell types of brain was analyzed by the AlzData database. Conclusions In this study, iPSCs-derived cholinergic neurons from AD patients with APP mutations exhibit characteristics reminiscent of neurodegenerative disease. Transcriptome analysis revealed the corresponding DEGs and pathways, providing potential biomarkers and therapeutic targets for advancing AD research.
Collapse
Affiliation(s)
- Wenxian Sun
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yufei Chen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yuting Yang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Pin Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jin Gong
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ruina Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Boye Wen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Sirong Lv
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
3
|
Humpel C. Organotypic Brain Slices of ADULT Transgenic Mice: A Tool to Study Alzheimer's Disease. Curr Alzheimer Res 2020; 16:172-181. [PMID: 30543174 DOI: 10.2174/1567205016666181212153138] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/23/2018] [Accepted: 11/29/2018] [Indexed: 01/21/2023]
Abstract
Transgenic mice have been extensively used to study the Alzheimer pathology. In order to reduce, refine and replace (3Rs) the number of animals, ex vivo cultures are used and optimized. Organotypic brain slices are the most potent ex vivo slice culture models, keeping the 3-dimensional structure of the brain and being closest to the in vivo situation. Organotypic brain slice cultures have been used for many decades but were mainly prepared from postnatal (day 8-10) old rats or mice. More recent work (including our lab) now aims to culture organotypic brain slices from adult mice including transgenic mice. Especially in Alzheimer´s disease research, brain slices from adult transgenic mice will be useful to study beta-amyloid plaques, tau pathology and glial activation. This review will summarize the studies using organotypic brain slice cultures from adult mice to mimic Alzheimer's disease and will highlight advantages and also pitfalls using this technique.
Collapse
Affiliation(s)
- Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Humpel C. Organotypic brain slice cultures: A review. Neuroscience 2015; 305:86-98. [PMID: 26254240 PMCID: PMC4699268 DOI: 10.1016/j.neuroscience.2015.07.086] [Citation(s) in RCA: 309] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 12/27/2022]
Abstract
In vitro cell cultures are an important tool for obtaining insights into cellular processes in an isolated system and a supplement to in vivo animal experiments. While primary dissociated cultures permit a single homogeneous cell population to be studied, there is a clear need to explore the function of brain cells in a three-dimensional system where the main architecture of the cells is preserved. Thus, organotypic brain slice cultures have proven to be very useful in investigating cellular and molecular processes of the brain in vitro. This review summarizes (1) the historical development of organotypic brain slices focusing on the membrane technology, (2) methodological aspects regarding culturing procedures, age of donors or media, (3) whether the cholinergic neurons serve as a model of neurodegeneration in Alzheimer’s disease, (4) or the nigrostriatal dopaminergic neurons as a model of Parkinson’s disease and (5) how the vascular network can be studied, especially with regard to a synthetic blood–brain barrier. This review will also highlight some limits of the model and give an outlook on future applications.
Collapse
Affiliation(s)
- C Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria.
| |
Collapse
|
5
|
SMAD Transcription Factor, Sma-9, Attunes TGF-β Signaling Cascade Towards Modulating Amyloid Beta Aggregation and Associated Outcome in Transgenic C. elegans. Mol Neurobiol 2014; 53:109-119. [DOI: 10.1007/s12035-014-8988-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/04/2014] [Indexed: 01/22/2023]
|
6
|
Friedman A, Bar-Klein G, Serlin Y, Parmet Y, Heinemann U, Kaufer D. Should losartan be administered following brain injury? Expert Rev Neurother 2014; 14:1365-75. [PMID: 25346269 DOI: 10.1586/14737175.2014.972945] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Brain injury is a major health concern and associated with delayed neurological complications, including post-injury epilepsy, cognitive and emotional disabilities. Currently, there is no strategy to prevent post-injury delayed complications. We recently showed that dysfunction of the blood-brain barrier, often reported in brain injuries, can lead to epilepsy and neurodegeneration via activation of inflammatory TGF-β signaling in astrocytes. We further showed that the FDA approved angiotensin II type 1 receptor antagonist, losartan, blocks brain TGF-β signaling and prevents epilepsy in the albumin or blood-brain barrier breakdown models of epileptogenesis. Here we discuss the potential of losartan as an anti-epileptogenic and a neuroprotective drug, the rationale of its use following brain injury and the challenges of designing clinical trials. We highlight the urgent need to develop reliable biomarkers for epileptogenesis (and other complications) after brain injury as a pre-requisite to challenge neuroprotective therapies.
Collapse
Affiliation(s)
- Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, PO Box 15000, 5850 College Street, Halifax Nova Scotia B3H 4R2, Canada
| | | | | | | | | | | |
Collapse
|
7
|
Caraci F, Spampinato S, Sortino MA, Bosco P, Battaglia G, Bruno V, Drago F, Nicoletti F, Copani A. Dysfunction of TGF-β1 signaling in Alzheimer's disease: perspectives for neuroprotection. Cell Tissue Res 2011; 347:291-301. [PMID: 21879289 DOI: 10.1007/s00441-011-1230-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 08/07/2011] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects about 35 million people worldwide. Current drugs for AD only treat the symptoms and do not interfere with the underlying pathogenic mechanisms of the disease. AD is characterized by the presence of β-amyloid (Aβ) plaques, neurofibrillary tangles, and neuronal loss. Identification of the molecular determinants underlying Aβ-induced neurodegeneration is an essential step for the development of disease-modifying drugs. Recently, an impairment of the transforming growth factor-β1 (TGF-β1) signaling pathway has been demonstrated to be specific to the AD brain and, particularly, to the early phase of the disease. TGF-β1 is a neurotrophic factor responsible for the initiation and maintenance of neuronal differentiation and synaptic plasticity. The deficiency of TGF-β1 signaling is associated with Aβ pathology and neurofibrillary tangle formation in AD animal models. Reduced TGF-β1 signaling seems to contribute both to microglial activation and to ectopic cell-cycle re-activation in neurons, two events that contribute to neurodegeneration in the AD brain. The neuroprotective features of TGF-β1 indicate the advantage of rescuing TGF-β1 signaling as a means to slow down the neurodegenerative process in AD.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Drug Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Beck K, Schachtrup C. Vascular damage in the central nervous system: a multifaceted role for vascular-derived TGF-β. Cell Tissue Res 2011; 347:187-201. [PMID: 21850492 DOI: 10.1007/s00441-011-1228-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 07/22/2011] [Indexed: 01/16/2023]
Abstract
The brain function depends on a continuous supply of blood. The blood-brain barrier (BBB), which is formed by vascular cells and glia, separates components of the circulating blood from neurons and maintains the precisely regulated brain milieu required for proper neuronal function. A compromised BBB alters the transport of molecules between the blood and brain and has been associated with or shown to precede neurodegenerative disease. Blood components immediately leak into the brain after mechanical damage or as a consequence of a compromised BBB in brain disease changing the extracellular environment at sites of vascular damage. It is intriguing how blood-derived components alter the cellular and molecular constituents of the neurovascular interface after BBB opening. We recently identified an unexpected role for the blood protein fibrinogen, which is deposited in the nervous system promptly after vascular damage, as an initial scar inducer by promoting the availability of active TGF-β. Fibrinogen-bound latent TGF-β interacts with astrocytes, leading to active TGF-β formation and activation of the TGF-β/Smad signaling pathway. Here, we discuss the pleiotropic effects of potentially vascular-derived TGF-β on cells at the neurovascular interface and we speculate how these biological effects might contribute to degeneration and regeneration processes. Summarizing the effects of the components derived from the brain vascular system on nervous system regeneration might support the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Kristina Beck
- Centre of Chronic Immunodeficiency, University Medical Centre Freiburg and University of Freiburg, 79106 Freiburg, Germany
| | | |
Collapse
|
9
|
Mohamed A, Posse de Chaves E. Aβ internalization by neurons and glia. Int J Alzheimers Dis 2011; 2011:127984. [PMID: 21350608 PMCID: PMC3042623 DOI: 10.4061/2011/127984] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 12/23/2010] [Indexed: 11/20/2022] Open
Abstract
In the brain, the amyloid β peptide (Aβ) exists extracellularly and inside neurons. The intracellular accumulation of Aβ in Alzheimer's disease brain has been questioned for a long time. However, there is now sufficient strong evidence indicating that accumulation of Aβ inside neurons plays an important role in the pathogenesis of Alzheimer's disease. Intraneuronal Aβ originates from intracellular cleavage of APP and from Aβ internalization from the extracellular milieu. We discuss here the different molecular mechanisms that are responsible for Aβ internalization in neurons and the links between Aβ internalization and neuronal dysfunction and death. A brief description of Aβ uptake by glia is also presented.
Collapse
Affiliation(s)
- Amany Mohamed
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | | |
Collapse
|
10
|
Grovit-Ferbas K, Harris-White ME. Thinking about HIV: the intersection of virus, neuroinflammation and cognitive dysfunction. Immunol Res 2010; 48:40-58. [DOI: 10.1007/s12026-010-8166-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Caraci F, Battaglia G, Bruno V, Bosco P, Carbonaro V, Giuffrida ML, Drago F, Sortino MA, Nicoletti F, Copani A. TGF-β1 pathway as a new target for neuroprotection in Alzheimer's disease. CNS Neurosci Ther 2009; 17:237-49. [PMID: 19925479 DOI: 10.1111/j.1755-5949.2009.00115.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that affects more than 37 million people worldwide. Current drugs for AD are only symptomatic, but do not interfere with the underlying pathogenic mechanisms of the disease. AD is characterized by the presence of ß-amyloid (Aβ) plaques, neurofibrillary tangles, and neuronal loss. The identification of the molecular determinants underlying AD pathogenesis is a fundamental step to design new disease-modifying drugs. Recently, a specific impairment of transforming-growth-factor-β1 (TGF-β1) signaling pathway has been demonstrated in AD brain. The deficiency of TGF-β1 signaling has been shown to increase both Aβ accumulation and Aβ-induced neurodegeneration in AD models. The loss of function of TGF-ß1 pathway seems also to contribute to tau pathology and neurofibrillary tangle formation. Growing evidence suggests a neuroprotective role for TGF-β1 against Aβ toxicity both in vitro and in vivo models of AD. Different drugs, such as lithium or group II mGlu receptor agonists are able to increase TGF-β1 levels in the central nervous system (CNS), and might be considered as new neuroprotective tools against Aβ-induced neurodegeneration. In the present review, we examine the evidence for a neuroprotective role of TGF-β1 in AD, and discuss the TGF-β1 signaling pathway as a new pharmacological target for the treatment of AD.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Pharmaceutical Sciences, University of Catania, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener 2009; 4:47. [PMID: 19917131 PMCID: PMC2784760 DOI: 10.1186/1750-1326-4-47] [Citation(s) in RCA: 560] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 11/16/2009] [Indexed: 12/13/2022] Open
Abstract
While peripheral immune access to the central nervous system (CNS) is restricted and tightly controlled, the CNS is capable of dynamic immune and inflammatory responses to a variety of insults. Infections, trauma, stroke, toxins and other stimuli are capable of producing an immediate and short lived activation of the innate immune system within the CNS. This acute neuroinflammatory response includes activation of the resident immune cells (microglia) resulting in a phagocytic phenotype and the release of inflammatory mediators such as cytokines and chemokines. While an acute insult may trigger oxidative and nitrosative stress, it is typically short-lived and unlikely to be detrimental to long-term neuronal survival. In contrast, chronic neuroinflammation is a long-standing and often self-perpetuating neuroinflammatory response that persists long after an initial injury or insult. Chronic neuroinflammation includes not only long-standing activation of microglia and subsequent sustained release of inflammatory mediators, but also the resulting increased oxidative and nitrosative stress. The sustained release of inflammatory mediators works to perpetuate the inflammatory cycle, activating additional microglia, promoting their proliferation, and resulting in further release of inflammatory factors. Neurodegenerative CNS disorders, including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), tauopathies, and age-related macular degeneration (ARMD), are associated with chronic neuroinflammation and elevated levels of several cytokines. Here we review the hallmarks of acute and chronic inflammatory responses in the CNS, the reasons why microglial activation represents a convergence point for diverse stimuli that may promote or compromise neuronal survival, and the epidemiologic, pharmacologic and genetic evidence implicating neuroinflammation in the pathophysiology of several neurodegenerative diseases.
Collapse
|
13
|
Cell death and proliferation in acute slices and organotypic cultures of mammalian CNS. Prog Neurobiol 2009; 88:221-45. [DOI: 10.1016/j.pneurobio.2009.01.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 12/09/2008] [Accepted: 01/07/2009] [Indexed: 11/24/2022]
|
14
|
Wieghaus KA, Gianchandani EP, Neal RA, Paige MA, Brown ML, Papin JA, Botchwey EA. Phthalimide neovascular factor 1 (PNF1) modulates MT1-MMP activity in human microvascular endothelial cells. Biotechnol Bioeng 2009; 103:796-807. [PMID: 19326468 PMCID: PMC2711776 DOI: 10.1002/bit.22310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We are creating synthetic pharmaceuticals with angiogenic activity and potential to promote vascular invasion. We previously demonstrated that one of these molecules, phthalimide neovascular factor 1 (PNF1), significantly expands microvascular networks in vivo following sustained release from poly(lactic-co-glycolic acid) (PLAGA) films. In addition, to probe PNF1 mode of action, we recently applied a novel pathway-based compendium analysis to a multi-timepoint, controlled microarray data set of PNF1-treated (vs. control) human microvascular endothelial cells (HMVECs), and we identified induction of tumor necrosis factor-alpha (TNF-alpha) and, subsequently, transforming growth factor-beta (TGF-beta) signaling networks by PNF1. Here we validate this microarray data set with quantitative real-time polymerase chain reaction (RT-PCR) analysis. Subsequently, we probe this data set and identify three specific TGF-beta-induced genes with regulation by PNF1 conserved over multiple timepoints-amyloid beta (A4) precursor protein (APP), early growth response 1 (EGR-1), and matrix metalloproteinase 14 (MMP14 or MT1-MMP)-that are also implicated in angiogenesis. We further focus on MMP14 given its unique role in angiogenesis, and we validate MT1-MMP modulation by PNF1 with an in vitro fluorescence assay that demonstrates the direct effects that PNF1 exerts on functional metalloproteinase activity. We also utilize endothelial cord formation in collagen gels to show that PNF1-induced stimulation of endothelial cord network formation in vitro is in some way MT1-MMP-dependent. Ultimately, this new network analysis of our transcriptional footprint characterizing PNF1 activity 1-48 h post-supplementation in HMVECs coupled with corresponding validating experiments suggests a key set of a few specific targets that are involved in PNF1 mode of action and important for successful promotion of the neovascularization that we have observed by the drug in vivo.
Collapse
Affiliation(s)
- Kristen A. Wieghaus
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, Phone: 434-982-1587, Fax: 434-982-3870
| | - Erwin P. Gianchandani
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, Phone: 434-982-6267, Fax: 434-982-3870
| | - Rebekah A. Neal
- Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, Phone: 434-982-1587, Fax: 434-982-3870
| | - Mikell A. Paige
- Lombardi Comprehensive Cancer Center, Drug Discovery Program, Department of Oncology, Georgetown University Medical Center, New Research Building, EP07, 3970 Reservoir Road, Washington, DC 20057, Phone: 202-687-8605
| | - Milton L. Brown
- Lombardi Comprehensive Cancer Center, Drug Discovery Program, Department of Oncology, Georgetown University Medical Center, New Research Building, EP07, 3970 Reservoir Road, Washington, DC 20057, Phone: 202-687-8605
| | - Jason A. Papin
- Cardiovascular Research Center, Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, Phone: 434-924-8195, Fax: 434-982-3870
| | - Edward A. Botchwey
- Department of Orthopaedic Surgery, Department of Biomedical Engineering, University of Virginia, Box 800759, Health System, Charlottesville, VA 22908, Phone: 434-243-9846, Fax: 434-982-3870
| |
Collapse
|
15
|
Eslami P, Johnson MF, Terzakaryan E, Chew C, Harris-White ME. TGF beta2-induced changes in LRP-1/T beta R-V and the impact on lysosomal A beta uptake and neurotoxicity. Brain Res 2008; 1241:176-87. [PMID: 18804458 PMCID: PMC2651642 DOI: 10.1016/j.brainres.2008.08.086] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 08/21/2008] [Accepted: 08/25/2008] [Indexed: 01/01/2023]
Abstract
Numerous studies suggest a central role for the low-density lipoprotein receptor-related protein/transforming growth factor beta receptor V in Alzheimer's Disease. We continue our investigation of a ligand for this receptor, transforming growth factor beta2, which is also implicated in Alzheimer Disease pathogenesis, but whose mechanism(s) remain elusive. Confocal imaging reveals that transforming growth factor beta2 rapidly targets amyloid beta peptide to the lysosomal compartment in cortical neurons and induces cell death. Low-density lipoprotein receptor-related protein/transforming growth factor beta receptor V is known as an endocytic receptor, delivering proteins to the lysosomal compartment for degradation. Transforming growth factor beta2 may alter this pathway resulting in increased uptake, intracellular accumulation and toxicity of amyloid beta peptide. RT-PCR and Western blot analysis of transforming growth factor beta2-treated cells demonstrate that transforming growth factor beta2 modestly increases the mRNA and protein levels of low-density lipoprotein receptor-related protein/transforming growth factor beta receptor V as well as increases the uptake activity. Furthermore, transforming growth factor beta2 alters the morphology and numbers of lysosomes in neurons. Lucifer Yellow and lysosomal hydrolase analysis show that transforming growth factor beta2 makes lysosomal membranes unstable and leaky and this effect is exacerbated with the addition of amyloid beta protein. Our data support a key role for low-density lipoprotein receptor-related protein/transforming growth factor beta receptor V in mediating transforming growth factor beta2 enhancement of amyloid beta peptide uptake and neurotoxicity.
Collapse
Affiliation(s)
- Pirooz Eslami
- Department of Medicine, University of California, Los Angeles, CA
- Veterans Administration-Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Ming F. Johnson
- Department of Medicine, University of California, Los Angeles, CA
- Veterans Administration-Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Ellen Terzakaryan
- Department of Medicine, University of California, Los Angeles, CA
- Veterans Administration-Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Carolyn Chew
- Department of Medicine, University of California, Los Angeles, CA
- Veterans Administration-Greater Los Angeles Healthcare System, Sepulveda, CA
| | - Marni E. Harris-White
- Department of Medicine, University of California, Los Angeles, CA
- Veterans Administration-Greater Los Angeles Healthcare System, Sepulveda, CA
| |
Collapse
|
16
|
Jiang Q, Heneka M, Landreth GE. The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer's disease: therapeutic implications. CNS Drugs 2008; 22:1-14. [PMID: 18072811 DOI: 10.2165/00023210-200822010-00001] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease is a complex neurodegenerative disorder, with aging, genetic and environmental factors contributing to its development and progression. The complexity of Alzheimer's disease presents substantial challenges for the development of new therapeutic agents. Alzheimer's disease is typified by pathological depositions of beta-amyloid peptides and neurofibrillary tangles within the diseased brain. It has also been demonstrated to be associated with a significant microglia-mediated inflammatory component, dysregulated lipid homeostasis and regional deficits in glucose metabolism within the brain. The peroxisome proliferator-activated receptor-gamma (PPARgamma) is a prototypical ligand-activated nuclear receptor that coordinates lipid, glucose and energy metabolism, and is found in elevated levels in the brains of individuals with Alzheimer's disease. A recently appreciated physiological function of this type of receptor is its ability to modulate inflammatory responses. In animal models of Alzheimer's disease, PPARgamma agonist treatment results in the reduction of amyloid plaque burden, reduced inflammation and reversal of disease-related behavioural impairment. In a recent phase II clinical trial, the use of the PPARgamma agonist rosiglitazone was associated with improved cognition and memory in patients with mild to moderate Alzheimer's disease. Thus, PPARgamma may act to modulate multiple pathophysiological mechanisms that contribute to Alzheimer's disease, and represents an attractive therapeutic target for the treatment of the disease.
Collapse
Affiliation(s)
- Qingguang Jiang
- Department of Neurosciences, Alzheimer Research Laboratory, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
17
|
Caraci F, Battaglia G, Busceti C, Biagioni F, Mastroiacovo F, Bosco P, Drago F, Nicoletti F, Sortino MA, Copani A. TGF-beta 1 protects against Abeta-neurotoxicity via the phosphatidylinositol-3-kinase pathway. Neurobiol Dis 2008; 30:234-42. [PMID: 18356065 DOI: 10.1016/j.nbd.2008.01.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 01/10/2008] [Accepted: 01/26/2008] [Indexed: 11/17/2022] Open
Abstract
beta-Amyloid (A beta) injection into the rat dorsal hippocampus had a small neurotoxic effect that was amplified by i.c.v. injection of SB431542, a selective inhibitor of transforming growth factor-beta (TGF-beta) receptor. This suggested that TGF-beta acts as a factor limiting A beta toxicity. We examined the neuroprotective activity of TGF-beta1 in pure cultures of rat cortical neurons challenged with A beta. Neuronal death triggered by A beta is known to proceed along an aberrant re-activation of the cell cycle, and involves late beta-catenin degradation and tau hyperphosphorylation. TGF-beta1 was equally protective when added either in combination with, or 6 h after A beta. Co-added TGF-beta1 prevented A beta-induced cell cycle reactivation, whereas lately added TGF-beta1 had no effect on the cell cycle, but rescued the late beta-catenin degradation and tau hyperphosphorylation. The phosphatidylinositol-3-kinase (PI-3-K) inhibitor, LY294402, abrogated all effects. Thus, TGF-beta1 blocks the whole cascade of events leading to A beta neurotoxicity by activating the PI-3-K pathway.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Pharmaceutical Sciences, University of Catania, 95125, Catania, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Siegel DA, Davies P, Dobrenis K, Huang M. Tomoregulin-2 is found extensively in plaques in Alzheimer's disease brain. J Neurochem 2006; 98:34-44. [PMID: 16805794 DOI: 10.1111/j.1471-4159.2006.03801.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tomoregulin (TR)2 is a transmembrane protein predominantly expressed in brain. It has a unique extracellular domain, containing epidermal growth factor-like and follistatin-like modules. The ectodomain is released from the cell surface, and thought to function as a neurotrophic factor and dendritogenic agent. During CNS development and in the neuronal storage disease GM2 gangliosidosis, which is characterized by ectopic dendrites, the TR2 ectodomain is present in neuronal nuclei where it may function in dendrite initiation. Data presented here demonstrate that TR2 is found extensively in Alzheimer's disease (AD) plaques. Confocal microscopy shows that TR2 is present throughout plaques. Interestingly, TR2 is absent from plaques in the presenilin-1/amyloid precursor protein mouse model of AD. From these data, and what is known about TR2, it is hypothesized that TR2 may participate in amyloid plaque formation and contribute to the pathogenesis of AD. The human TR2 gene is located on chromosome 2q32.3, near a locus linked to Parkinson's disease. TR2 is reported to be a trophic factor for dopaminergic mesencephalic neurons.
Collapse
Affiliation(s)
- Donald A Siegel
- New York Medical College, Department of Cell Biology and Anatomy, Valhalla, New York, USA.
| | | | | | | |
Collapse
|
19
|
Bravo JA, Parra CS, Arancibia S, Andrés S, Morales P, Herrera-Marschitz M, Herrera L, Lara HE, Fiedler JL. Adrenalectomy promotes a permanent decrease of plasma corticoid levels and a transient increase of apoptosis and the expression of Transforming Growth Factor beta1 (TGF-beta1) in hippocampus: effect of a TGF-beta1 oligo-antisense. BMC Neurosci 2006; 7:40. [PMID: 16712723 PMCID: PMC1481618 DOI: 10.1186/1471-2202-7-40] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Accepted: 05/19/2006] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Corticosterone reduction produced by adrenalectomy (ADX) induces apoptosis in dentate gyrus (DG) of the hippocampus, an effect related to an increase in the expression of the pro-apoptotic gene bax. However it has been reported that there is also an increase of the anti-apoptotic gene bcl-2, suggesting the promotion of a neuroprotective phenomenon, perhaps related to the expression of transforming growth factor beta1 (TGF-beta1). Thus, we have investigated whether TGF-beta1 levels are induced by ADX, and whether apoptosis is increased by blocking the expression of TGF-beta1 with an antisense oligonucleotide (ASO) administered intracerebrally in corticosterone depleted rats. RESULTS It was observed an increase of apoptosis in DG, 2 and 5 days after ADX, in agreement with a reduction of corticosterone levels. However, the effect of ADX on the number of apoptotic positive cells in DG was decreased 5 days after the lesion. In CA1-CA3 regions, the effect was only observed 2 days after ADX. TGF-beta1 mRNA levels were increased 2 days after ADX. The sustained intracerebro-ventricular administration of a TGF-beta1 ASO via an osmotic mini pump increased apoptosis levels in CA and DG regions 5 days after ADX as well as sham-operated control animals. No significant effect was observed following a scrambled-oligodeoxynucleotide treatment. CONCLUSION The changes in both the pattern and the magnitude of apoptotic-cell morphology observed 2 and 5 days after ADX suggest that, as a consequence of the reduction of corticosteroids, some trophic mechanisms restricting cell death to a particular time window are elicited. Sustained intracerebral administration of TGF-beta1 ASO increased the apoptosis promoted by ADX, suggesting that TGF-beta1 plays an anti-apoptotic role in vivo in hippocampus.
Collapse
Affiliation(s)
- Javier A Bravo
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Claudio S Parra
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Sandor Arancibia
- Laboratory of Molecular Mechanisms of Neurodegenerative Diseases, Université de Montpellier, Montpellier, France
| | - Sergio Andrés
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Paola Morales
- Programmes of Molecular & Clinical Pharmacology ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Mario Herrera-Marschitz
- Programmes of Molecular & Clinical Pharmacology ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Luisa Herrera
- Human Genetics, ICBM, Faculty of Medicine, Universidad de Chile, Chile
| | - Hernán E Lara
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| | - Jenny L Fiedler
- Department of Biochemistry and Molecular Biology. Laboratory of Neurobiochemistry. Faculty of Chemical and Pharmaceutical Sciences. Universidad de Chile, Chile
| |
Collapse
|
20
|
Dickson MR, Perry RT, Wiener H, Go RCP. Association studies of transforming growth factor-beta 1 and Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 2005; 139B:38-41. [PMID: 16082716 DOI: 10.1002/ajmg.b.30218] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Substantial laboratory evidence suggests Transforming Growth Factor-beta1 (TGFB1) is linked to Alzheimer's Disease (AD) pathology. The purpose of the study was to estimate the genetic association of TGFB1 with AD while controlling for apolipoprotein E4 allele (APOE4) status, the only well-established genetic risk factor for AD. Two study populations were genotyped for the TGFB1-509 and +869 single nucleotide polymorphisms (SNPs) that have been associated with TGFB1 levels. Constituting these populations were 203 families from the NIMH AD Genetic Initiative with at least two affected siblings and a normal sibling, and a population of 126 African-American (AA) AD cases versus 93 age matched controls. Results from family-based analyses showed a significant association between the TGFB1 -509 SNP and AD for the entire set of 203 families (P = 0.007), and a subset of these families without a homozygous APOE4 family member (P = 0.026). Results from family-based analyses on the TGFB1 +869 SNP were not significant in the 203 families. While associations for the main effects of the TGFB1 +869 and -509 SNP with AD in the AA case-control study were also not significant, results did indicate that TGFB1 may function as an effect modifier of APOE4 risk. Specifically, the odds of AD associated with having at least one APOE4 allele increased in an additive fashion with one or two copies of the higher producer allele when stratified by TGFB1 -509 genotype and by TGFB1 +869 genotype. Results support a role for TGFB1 in AD pathogenesis.
Collapse
Affiliation(s)
- M R Dickson
- Department of Epidemiology and International Health, University of Alabama at Birmingham, Birmingham, AL 35294-0022, USA.
| | | | | | | |
Collapse
|
21
|
Lee EO, Kang JL, Chong YH. The amyloid-beta peptide suppresses transforming growth factor-beta1-induced matrix metalloproteinase-2 production via Smad7 expression in human monocytic THP-1 cells. J Biol Chem 2005; 280:7845-53. [PMID: 15632190 DOI: 10.1074/jbc.m409101200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Accumulation of the amyloid-beta (Abeta) peptide in the brain is a crucial factor in the development of Alzheimer disease. Expression of transforming growth factor-beta1 (TGF-beta1), an immunosuppressive cytokine, has been associated in vivo with Abeta accumulation in transgenic mice and recently with Abeta clearance by activated microglia, suggesting its deleterious and beneficial effects in neuronal cells. In this study, we demonstrated that TGF-beta1 stimulated the production of matrix metalloproteinase-2 (MMP-2) in a time- and dose-dependent manner in a human monocytic THP-1 cell line. Notably, we found that Abeta1-42 consistently inhibited the TGF-beta1-induced production of MMP-2, the endogenous gene containing Smad response elements, whereas the reverse peptide, Abeta42-1, evidenced little effect. Additionally, Abeta1-42 reduced TGF-beta1-induced increase in plasminogen activator inhibitor-1 (PAI-1). This inhibitory effect of Abeta1-42 was also seen in human astroglial T98G cell line. Furthermore, Abeta1-42 significantly induced the expression of Smad7, which appears in turn to mediate the Abeta suppression of the TGF-beta1-induced MMP-2 production. Indeed, Smad7 overexpression mimicked the inhibitory effect of Abeta1-42 on TGF-beta1-induced MMP-2 production. Importantly, Abeta1-42 markedly suppressed the transactivation of the transfected reporter construct, p3TP-Lux, which contains TGF-beta1-inducible Smad response elements. This was concomitant with a decreased MMP-2 production in TGF-beta1-treated cells. Inhibition of cellular Smad7 levels via the small interference RNA method significantly ameliorated the Abeta1-42-mediated suppression of TGF-beta1-inducible transcription reporter activity, thereby restoring MMP-2 induction, whereas Smad7 transfection down-regulated TGF-beta1-inducible transcription reporter activity. Collectively, these data suggest that Abeta1-42 may play an important role in the negative regulation of TGF-beta1-induced MMP-2 production via Smad7 expression.
Collapse
Affiliation(s)
- Eun Ok Lee
- Department of Microbiology, College of Medicine, Division of Molecular Biology and Neuroscience, Ewha Medical Research Institute, Ewha Womans University, 911-1, Mok-6-dong, Yangcheonku, Seoul 158-710, Korea
| | | | | |
Collapse
|
22
|
Harris-White ME, Balverde Z, Lim GP, Kim P, Miller SA, Hammer H, Galasko D, Frautschy SA. Role of LRP in TGF?2-mediated neuronal uptake of A? and effects on memory. J Neurosci Res 2004; 77:217-28. [PMID: 15211588 DOI: 10.1002/jnr.20149] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
There is increasing evidence that soluble amyloid-beta peptide (Abeta) uptake into neurons is an early event in the pathogenesis of Alzheimer's disease (AD). Identification of the early events leading to neuronal dysfunction is key to developing therapeutic strategies, but relative roles of receptors and factors modulating uptake are poorly understood. Studies have shown that transforming growth factor beta (TGFbeta), particularly TGFbeta2, can influence the targeting of Abeta to cells in vitro. TGFbeta2 can target Abeta to neurons in organotypic hippocampal slice cultures (OHSC). We examine a specific mechanism for TGFbeta2-mediated targeting of Abeta to neurons. The receptor-associated protein (RAP), a low-density lipoprotein receptor-related protein (LRP) antagonist, can attenuate the cellular targeting of Abeta both in vitro and in vivo and prevent Abeta/TGFbeta2-induced memory retention deficits. Using both in vitro and in vivo methods, we identify LRP as playing a role in TGFbeta2-mediated Abeta uptake, neurodegeneration, and spatial memory impairment.
Collapse
Affiliation(s)
- Marni E Harris-White
- Department of Medicine, University of California, Los Angeles, California 91343, USA.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Amyloid beta peptide (Abeta) is a major component of senile plaques, one of the principle pathological features in Alzheimer's disease (AD) brains. Fibrillar Abeta has been shown to bind C1 via C1q, the recognition component of the classical complement pathway, resulting in the activation of the complement pathway, thereby initiating an inflammatory cascade in the brain. C1q has also been shown to enhance phagocytic activities of microglia, which could benefit in clearance of apoptotic cells or cellular debris. To begin to define the role of C1q in tissue injury mediated by Abeta, we assessed the appearance of C1q in hippocampal slice cultures treated with freshly solubilized or fibrillar Abeta 1-42. Here we demonstrate a dose- and time-dependent uptake of exogenously applied Abeta by pyramidal neurons in organotypic slice cultures from rat hippocampus. Importantly, when slices were immunostained with antibody against rat C1q, a distinct reactivity for C1q in cells within the neuronal cell layer of cornu ammonis (CA) of hippocampus, primarily the CA1/CA2, was observed in the Abeta-treated slices. No such immunoreactivity was detected in untreated cultures or upon addition of control peptides. ELISA assays also showed an increase in C1q in tissue extracts from slices of the treated group. Similarly, the mRNA level of C1q in slices was increased within 24 h after Abeta treatment. These data demonstrate that upon exposure to Abeta, C1q is expressed in neurons in this organotypic system. The induction of C1q may be an early, perhaps beneficial, tissue or cellular response to injury triggered by particular pathogenic stimuli.
Collapse
Affiliation(s)
- Rong Fan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | | |
Collapse
|
24
|
Zhou YX, Zhao M, Li D, Shimazu K, Sakata K, Deng CX, Lu B. Cerebellar deficits and hyperactivity in mice lacking Smad4. J Biol Chem 2003; 278:42313-20. [PMID: 12896967 DOI: 10.1074/jbc.m308287200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Smad4 is a central mediator of TGF-beta signals, which are known to play essential roles in many biological processes. Using a Cre-loxP approach to overcome early embryonic lethality, we have studied functions of TGF-beta/Smad4 signals in the central nervous system (CNS). No obvious deficits were detected in mice carrying the targeted disruption of Smad4 in the CNS. The overall morphology of the hippocampus appeared normal. There was no change in the proliferation of neuronal precursor cells, nor in several forms of synaptic plasticity. In contrast, deletion of Smad4 resulted in a marked decrease in the number of cerebellar Purkinje cells and parvalbumin-positive interneurons. Accompanied by the abnormality in the cerebellum, mutant mice also exhibited significantly increased vertical activity. Thus, our study reveals an unexpected role for Smad4 in cerebellar development and in the control of motor function.
Collapse
Affiliation(s)
- Yong-Xing Zhou
- Mammalian Genetics Section, National Institute of Diabetes and Digestive and Kidney Diseases/NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Mousseau DD, Chapelsky S, De Crescenzo G, Kirkitadze MD, Magoon J, Inoue S, Teplow DB, O'Connor-McCourt MD. A direct interaction between transforming growth factor (TGF)-betas and amyloid-beta protein affects fibrillogenesis in a TGF-beta receptor-independent manner. J Biol Chem 2003; 278:38715-22. [PMID: 12867422 DOI: 10.1074/jbc.m304080200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) receptor-mediated signaling has been proposed to mediate both the beneficial and deleterious roles for this cytokine in amyloid-beta protein (Abeta) function. In order to assess receptor dependence of these events, we used PC12 cell cultures, which are devoid of TGF-beta receptors. Surprisingly, TGF-beta potentiated the neurotoxic effects of the 40-residue Abeta peptide, Abeta-(1-40), in this model suggesting that there may be a direct, receptor-independent interaction between TGF-beta and Abeta-(1-40). Surface plasmon resonance confirmed that TGF-beta binds with high affinity directly to Abeta-(1-40) and electron microscopy revealed that TGF-beta enhances Abeta-(1-40) oligomerization. Immunohistochemical examination of mouse brain revealed that hippocampal CA1 and dentate gyrus, two regions classically associated with Abeta-mediated pathology, lack TGF-beta Type I receptor immunoreactivity, thus indicating that TGF-beta receptor-mediated signaling would not be favored in these regions. Our observations not only provide for a unique, receptor-independent mechanism of action for TGF-beta, but also help to reconcile the literature interpreting the role of TGF-beta in Abeta function. These data support a critical etiological role for this mechanism in neuropathological amyloidoses.
Collapse
Affiliation(s)
- Darrell D Mousseau
- Cell Signaling Group, Neuropsychiatry Research Unit, Department of Psychiatry, University of Saskatchewan, Saskatoon S7N 5E4, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Mazur-Kolecka B, Frackowiak J, Le Vine H, Haske T, Evans L, Sukontasup T, Golabek A. TGFbeta1 enhances formation of cellular Abeta/apoE deposits in vascular myocytes. Neurobiol Aging 2003; 24:355-64. [PMID: 12498970 DOI: 10.1016/s0197-4580(02)00095-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Brain injury increases the risk of Alzheimer's disease (AD) through unknown mechanisms. We studied deposition of amyloid-beta protein (Abeta) in cells exposed to transforming growth factor beta1 (TGFbeta1), a cytokine that regulates cell metabolism during brain injury, and apolipoproteinE (apoE), the major lipid transporter in the brain. The studies were conducted by using brain vascular smooth muscle cells that are engaged in beta-amyloidosis in vivo and produce Abeta in cell culture. We found that cell treatment with TGFbeta1 together with apoE4 strongly increased the amount of cellular Abeta. The intracellular Abeta co-localized with apoE but not with TGFbeta, similarly as in vascular beta-amyloid. Some cellular Abeta/apoE deposits increased in size and persisted in culture even after the TGFbeta1 and apoE4 were removed. The appearance of cellular deposits of Abeta was associated with increased production of the amyloid-beta precursor protein and cellular retention of its mature form. The results suggest that the concomitant presence of apoE and TGFbeta1 can trigger vascular beta-amyloidosis by inducing intracellular formation of stable Abeta/apoE deposits.
Collapse
Affiliation(s)
- Bozena Mazur-Kolecka
- Department of Pathological Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Bi X, Gall CM, Zhou J, Lynch G. Uptake and pathogenic effects of amyloid beta peptide 1-42 are enhanced by integrin antagonists and blocked by NMDA receptor antagonists. Neuroscience 2002; 112:827-40. [PMID: 12088742 DOI: 10.1016/s0306-4522(02)00132-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Many synapses contain two types of receptors - integrins and N-methyl-D-aspartate (NMDA) receptors - that have been implicated in peptide internalization. The present studies tested if either class is involved in the uptake of the 42-residue form of amyloid beta peptide (Abeta1-42), an event hypothesized to be of importance in the development of Alzheimer's disease. Cultured hippocampal slices were exposed to Abeta1-42 for 6 days in the presence or absence of soluble Gly-Arg-Gly-Asp-Ser-Pro, a peptide antagonist of Arg-Gly-Asp (RGD)-binding integrins, or the disintegrin echistatin. Abeta uptake, as assessed with immunocytochemistry, occurred in 42% of the slices incubated with Abeta peptide alone but in more than 80% of the slices co-treated with integrin antagonists. Uptake was also found in a broader range of hippocampal subfields in RGD-treated slices. Increased sequestration was accompanied by two characteristics of early stage Alzheimer's disease: elevated concentrations of cathepsin D immunoreactivity and activation of microglia. The selective NMDA receptor antagonist D-(-)-2-amino-5-phosphonovalerate completely blocked internalization of Abeta, up-regulation of cathepsin D, and activation of microglia. Our results identify two classes of receptors that cooperatively regulate the internalization of Abeta1-42 and support the hypothesis that characteristic pathologies of Alzheimer's disease occur once critical intraneuronal Abeta concentrations are reached.
Collapse
Affiliation(s)
- X Bi
- Psychiatry and Human Behavior, 101 Theory, Suite 250, University of California at Irvine, 92697, USA.
| | | | | | | |
Collapse
|
28
|
Burton T, Liang B, Dibrov A, Amara F. Transcriptional activation and increase in expression of Alzheimer's beta-amyloid precursor protein gene is mediated by TGF-beta in normal human astrocytes. Biochem Biophys Res Commun 2002; 295:702-12. [PMID: 12099697 DOI: 10.1016/s0006-291x(02)00724-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The overexpression of the Alzheimer amyloid precursor protein (APP) and its subsequent proteolytic processing may be one of several factors contributing to amyloid beta-peptide (Abeta) deposition in plaques and microvasculature in Alzheimer's disease (AD) brain. Cytokines and growth factors can influence the expression of APP in response to brain injury, but the underlying mechanisms are largely unknown. We examined the mechanisms by which transforming growth factor-beta (TGF-beta) affects the expression of APP in normal human astrocytes. We report that, TGF-beta up-regulated the expression of APP at the transcription level as determined by nuclear run-on experiments. Transient transfection of astrocytes with APP gene promoter (-2832 bp) chloramphenicol acetyltransferase (CAT) reporter constructs led to increased reporter activity upon TGF-beta stimulation. This reporter activity was mainly attributed to the APP proximal domain (-488 bp). The increase in APP gene transcription was associated with significant accumulation of intracellular APP, APP carboxyl terminal derived fragments, and total secreted Abeta. In addition, we observed a significant increase in levels of TGF-beta in Abeta plaques and its immediate vicinity in AD-affected brain relative to controls. These results indicate that high levels of TGF-beta in the cortex, may serve to up-regulate APP synthesis in reactive astrocytes and indirectly contributes to Abeta deposition. Closely related processes may induce cerebrovascular pathology in AD brain.
Collapse
Affiliation(s)
- Teralee Burton
- The Dr. John Foerster Centre for Health Research on Aging, St. Boniface General Hospital Research Centre, 351 Taché Avenue, Winnipeg, MB, Canada R3T 3B3
| | | | | | | |
Collapse
|
29
|
Williams S, Souchelnytskyi S, Danik M. TGFbeta2 mediates rapid inhibition of calcium influx in identified cholinergic basal forebrain neurons. Biochem Biophys Res Commun 2002; 290:1321-7. [PMID: 11812008 DOI: 10.1006/bbrc.2002.6351] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factors betas (TGFbetas) are known to have important roles in neuronal survival and can be upregulated in disease. However, unlike many other trophic factors, nothing is known about the rapid neurotransmitter-like actions of TGFbeta in the CNS. We explored this by examining the effects of TGFbeta on calcium influx of large enzymatically dissociated basal forebrain neurons. We show that brief application of TGFbeta2, but not TGFbeta1, to fura-2AM-loaded neurons reversibly and acutely (within seconds) inhibited K(+)-evoked calcium influx. Moreover, using single-cell RT-PCR, we confirmed that the large TGFbeta2-responsive neurons presented a cholinergic phenotype. Investigation of the signaling mechanism underlying TGFbeta2 actions using whole-cell recordings of calcium currents revealed that TGFbeta2-mediated responses were insensitive to the nonhydrolyzable GTP analogue GTPgammaS. However, TGFbeta2-mediated calcium current reductions were prevented by intracellular perfusion of a Smad2/3 peptide antagonist. Together, these results suggest that TGFbeta2 can acutely regulate the excitability of basal forebrain cholinergic neurons through an atypical signaling mechanism.
Collapse
Affiliation(s)
- Sylvain Williams
- Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
30
|
Harris-White ME, Chu T, Miller SA, Simmons M, Teter B, Nash D, Cole GM, Frautschy SA. Estrogen (E2) and glucocorticoid (Gc) effects on microglia and A beta clearance in vitro and in vivo. Neurochem Int 2001; 39:435-48. [PMID: 11578779 DOI: 10.1016/s0197-0186(01)00051-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The accumulation of fibrillar aggregates of beta Amyloid (A beta) in Alzheimer's Disease (AD) brain is associated with chronic brain inflammation. Although activated microglia (mu glia) can potentially clear toxic amyloid, chronic activation may lead to excessive production of neurotoxins. Recent epidemiological and clinical data have raised questions about the use of anti-inflammatory steroids (glucocorticoids, Gcs) and estrogens for treatment or prevention of AD. Since very little is known about steroid effects on mu glial interactions with amyloid, we investigated the effects of the synthetic Gc dexamethasone (DXM) and 17-beta estradiol (E2) in vitro in a murine mu glial-like N9 cell line on toxin production and intracellular A beta accumulation. To determine whether the steroid alterations of A beta uptake in vitro had relevance in vivo, we examined the effects of these steroids on A beta accumulation and mu glial responses to A beta infused into rat brain. Our in vitro data demonstrate for the first time that Gc dose-dependently enhanced mu glial A beta accumulation and support previous work showing that E2 enhances A beta uptake. Despite both steroids enhancing uptake, degradation was impeded, particularly with Gcs. Distinct differences between the two steroids were observed in their effect on toxin production and cell viability. Gc dose-dependently increased toxicity and potentiated A beta induction of nitric oxide, while E2 promoted cell viability and inhibited A beta induction of nitric oxide. The steroid enhancement of mu glial uptake and impedence of degradation observed in vitro were consistent with observations from in vivo studies. In the brains of A beta-infused rats, the mu glial staining in entorhinal cortex layer 3, not associated with A beta deposits was increased in response to A beta infusion and this effect was blocked by feeding rats prednisolone. In contrast, E2 enhanced mu glial staining in A beta-infused rats. A beta-immunoreactive (ir) deposits were quantitatively smaller, appeared denser, and were associated with robust mu glial responses. Despite the fact that steroid produced a smaller more focal deposit, total extracted A beta in cortical homogenate was elevated. Together, the in vivo and in vitro data support a role for steroids in plaque compaction. Our data are also consistent with the hypothesis that although E2 is less potent than Gc in impeding A beta degradation, long term exposure to both steroids could reduce A beta clearance and clinical utility. These data showing Gc potentiation of A beta-induced mu glial toxins may help explain the lack of epidemiological correlation for AD. The failure of both steroids to accelerate A beta degradation may explain their lack of efficacy for treatment of AD.
Collapse
Affiliation(s)
- M E Harris-White
- Department of Medicine, UCLA, C-128 RNRC, Los Angeles, CA 90095-1769, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Masliah E, Ho G, Wyss-Coray T. Functional role of TGF beta in Alzheimer's disease microvascular injury: lessons from transgenic mice. Neurochem Int 2001; 39:393-400. [PMID: 11578774 DOI: 10.1016/s0197-0186(01)00046-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recent studies have implicated pro- and anti-inflammatory cytokines as integral to Alzheimer's disease (AD) pathogenesis. Among them, transforming growth factor-beta (TGF-beta) is emerging as an important factor in regulating inflammatory responses. This multifunctional cytokine might be centrally involved in several aspects of AD pathogenesis by regulating beta-amyloid precursor protein synthesis and processing, plaque formation, astroglial and microglial response and neuronal cell death. Among all of these potential roles, studies in transgenic and infusion animal models have shown that TGF-beta may primarily contribute to AD pathogenesis by influencing A beta production and deposition, which in turn might result in damage to the brain microvasculature. The lessons learned from these models are of great interest not only for understanding of the role of TGF-beta in AD, but also for future treatments where testing of anti-inflammatory agents such as ibuprofen and an amyloid vaccine hold great promise. In this regard, further elucidation of the signal pathways by which TGF-beta exerts its effect in AD might lead to specific targets for further therapeutic intervention.
Collapse
Affiliation(s)
- E Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093-0624, USA.
| | | | | |
Collapse
|
32
|
Morgan TE, Rozovsky I, Sarkar DK, Young-Chan CS, Nichols NR, Laping NJ, Finch CE. Transforming growth factor-beta1 induces transforming growth factor-beta1 and transforming growth factor-beta receptor messenger RNAs and reduces complement C1qB messenger RNA in rat brain microglia. Neuroscience 2001; 101:313-21. [PMID: 11074155 DOI: 10.1016/s0306-4522(00)00387-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Transforming growth factor-beta1 is a multifunctional peptide with increased expression during Alzheimer's disease and other neurodegenerative conditions which involve inflammatory mechanisms. We examined the autoregulation of transforming growth factor-beta1 and transforming growth factor-beta receptors and the effects of transforming growth factor-beta1 on complement C1q in brains of adult Fischer 344 male rats and in primary glial cultures. Perforant path transection by entorhinal cortex lesioning was used as a model for the hippocampal deafferentation of Alzheimer's disease. In the hippocampus ipsilateral to the lesion, transforming growth factor-beta1 peptide was increased >100-fold; the messenger RNAs encoding transforming growth factor-beta1, transforming growth factor-beta type I and type II receptors were also increased, but to a smaller degree. In this acute lesion paradigm, microglia are the main cell type containing transforming growth factor-beta1, transforming growth factor-beta type I and II receptor messenger RNAs, shown by immunocytochemistry in combination with in situ hybridization. Autoregulation of the transforming growth factor-beta1 system was examined by intraventricular infusion of transforming growth factor-beta1 peptide, which increased hippocampal transforming growth factor-beta1 messenger RNA levels in a dose-dependent fashion. Similarly, transforming growth factor-beta1 increased levels of transforming growth factor-beta1 messenger RNA and transforming growth factor-beta type II receptor messenger RNA (IC(50), 5pM) and increased release of transforming growth factor-beta1 peptide from primary microglia cultures. Interactions of transforming growth factor-beta1 with complement system gene expression are also indicated, because transforming growth factor-beta1 decreased C1qB messenger RNA in the cortex and hippocampus, after intraventricular infusion, and in cultured glia. These indications of autocrine regulation of transforming growth factor-beta1 in the rodent brain support a major role of microglia in neural activities of transforming growth factor-beta1 and give a new link between transforming growth factor-beta1 and the complement system. The auto-induction of the transforming growth factor-beta1 system has implications for transgenic mice that overexpress transforming growth factor-beta1 in brain cells and for its potential role in amyloidogenesis.
Collapse
Affiliation(s)
- T E Morgan
- Andrus Gerontology Center and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-0191, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, Tran T, Ubeda O, Ashe KH, Frautschy SA, Cole GM. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer's disease. J Neurosci 2000; 20:5709-14. [PMID: 10908610 PMCID: PMC6772529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2000] [Accepted: 05/05/2000] [Indexed: 02/17/2023] Open
Abstract
The brain in Alzheimer's disease (AD) shows a chronic inflammatory response characterized by activated glial cells and increased expression of cytokines and complement factors surrounding amyloid deposits. Several epidemiological studies have demonstrated a reduced risk for AD in patients using nonsteroidal anti-inflammatory drugs (NSAIDs), prompting further inquiries about how NSAIDs might influence the development of AD pathology and inflammation in the CNS. We tested the impact of chronic orally administered ibuprofen, the most commonly used NSAID, in a transgenic model of AD displaying widespread microglial activation, age-related amyloid deposits, and dystrophic neurites. These mice were created by overexpressing a variant of the amyloid precursor protein found in familial AD. Transgene-positive (Tg+) and negative (Tg-) mice began receiving chow containing 375 ppm ibuprofen at 10 months of age, when amyloid plaques first appear, and were fed continuously for 6 months. This treatment produced significant reductions in final interleukin-1beta and glial fibrillary acidic protein levels, as well as a significant diminution in the ultimate number and total area of beta-amyloid deposits. Reductions in amyloid deposition were supported by ELISA measurements showing significantly decreased SDS-insoluble Abeta. Ibuprofen also decreased the numbers of ubiquitin-labeled dystrophic neurites and the percentage area per plaque of anti-phosphotyrosine-labeled microglia. Thus, the anti-inflammatory drug ibuprofen, which has been associated with reduced AD risk in human epidemiological studies, can significantly delay some forms of AD pathology, including amyloid deposition, when administered early in the disease course of a transgenic mouse model of AD.
Collapse
Affiliation(s)
- G P Lim
- University of California Los Angeles, Departments of Medicine and Neurology, 90095, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Neuroinflammation Working Group, Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin W, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O’Banion MK, Pachter J, Pasinetti G, Plata–Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss–Coray T. Inflammation and Alzheimer's disease. Neurobiol Aging 2000; 21:383-421. [PMID: 10858586 PMCID: PMC3887148 DOI: 10.1016/s0197-4580(00)00124-x] [Citation(s) in RCA: 3336] [Impact Index Per Article: 133.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.
Collapse
Affiliation(s)
| | - Haruhiko Akiyama
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Steven Barger
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Scott Barnum
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Bonnie Bradt
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Joachim Bauer
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Greg M. Cole
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Neil R. Cooper
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Piet Eikelenboom
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Mark Emmerling
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Berndt L. Fiebich
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Caleb E. Finch
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Sally Frautschy
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - W.S.T. Griffin
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Harald Hampel
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Michael Hull
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Gary Landreth
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Lih–Fen Lue
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Robert Mrak
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Ian R. Mackenzie
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Patrick L. McGeer
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - M. Kerry O’Banion
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Joel Pachter
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Guilio Pasinetti
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Carlos Plata–Salaman
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Joseph Rogers
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Russell Rydel
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Yong Shen
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Wolfgang Streit
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Ronald Strohmeyer
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Ikuo Tooyoma
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Freek L. Van Muiswinkel
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Robert Veerhuis
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Douglas Walker
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Scott Webster
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Beatrice Wegrzyniak
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Gary Wenk
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| | - Tony Wyss–Coray
- Sun Health Research Institute, 10515 West Santa Fe Drive, P.O. Box 1278, Sun City, AZ, USA 85372
| |
Collapse
|
35
|
Bocharov EV, Blommers MJ, Kuhla J, Arvinte T, Bürgi R, Arseniev AS. Sequence-specific 1H and 15N assignment and secondary structure of transforming growth factor beta3. JOURNAL OF BIOMOLECULAR NMR 2000; 16:179-180. [PMID: 10723998 DOI: 10.1023/a:1008315600134] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
|
36
|
Wyss-Coray T, Lin C, Sanan DA, Mucke L, Masliah E. Chronic overproduction of transforming growth factor-beta1 by astrocytes promotes Alzheimer's disease-like microvascular degeneration in transgenic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 156:139-50. [PMID: 10623661 PMCID: PMC1868631 DOI: 10.1016/s0002-9440(10)64713-x] [Citation(s) in RCA: 183] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/02/1999] [Indexed: 01/06/2023]
Abstract
Cerebrovascular amyloid deposition and microvascular degeneration are frequently associated with Alzheimer's disease (AD), but the etiology and pathogenetic role of these abnormalities are unknown. Recently, transforming growth factor-beta1 (TGF-beta1) was implicated in cerebrovascular amyloid formation in transgenic mice with astroglial overproduction of TGF-beta1 and in AD. We tested whether TGF-beta1 overproduction induces AD-like cerebrovascular degeneration and analyzed how cerebrovascular abnormalities develop over time in TGF-beta1-transgenic mice. In cerebral microvessels from 3- to 4-month-old TGF-beta1-transgenic mice, which display a prominent perivascular astrocytosis, levels of the basement membrane proteins perlecan and fibronectin were severalfold higher than in vessels from nontransgenic mice. Consistent with this increase, cortical capillary basement membranes of TGF-beta1 mice were significantly thickened. These changes preceded amyloid deposition, which began at around 6 months of age. In 9- and 18-month-old TGF-beta1 mice, various degenerative changes in microvascular cells of the brain were observed. Endothelial cells were thinner and displayed abnormal, microvilli-like protrusions as well as occasional condensation of chromatin, and pericytes occupied smaller areas in capillary profiles than in nontransgenic controls. Similar cerebrovascular abnormalities have been reported in AD. We conclude that chronic overproduction of TGF-beta1 triggers an accumulation of basement membrane proteins and results in AD-like cerebrovascular amyloidosis and microvascular degeneration. Closely related processes may induce cerebrovascular pathology in AD.
Collapse
Affiliation(s)
- T Wyss-Coray
- Gladstone Institute of Neurological Disease, San Francisco, California 94141-9100, USA.
| | | | | | | | | |
Collapse
|