1
|
Naghshbandieh A, Naghshbandieh A, Barfi E, Abkhooie L. Assessment of the level of apoptosis in differentiated pseudo-neuronal cells derived from neural stem cells under the influence of various inducers. AMERICAN JOURNAL OF STEM CELLS 2024; 13:250-270. [PMID: 39850017 PMCID: PMC11751472 DOI: 10.62347/bptg6174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/23/2024] [Indexed: 01/25/2025]
Abstract
Development and maintenance of the nervous system are governed by a scheduled cell death mechanism known as apoptosis. Very much how neurons survive and function depends on the degree of death in differentiating pseudo-neuronal cells produced from neural stem cells. Different inducers can affect the degree of death in these cells: hormones, medicines, growth factors, and others. Developing inventive therapies for neurodegenerative illnesses depends on a knowledge of how these inducers impact mortality in differentiated pseudo-neuronal cells. Using flow cytometry, Western blotting, and fluorescence microscopy among other techniques, the degree of death in many pseudo-neuronal cells is evaluated. Flow cytometry generates dead cell counts from measurements of cell size, granularity, and DNA content. Whereas fluorescence microscopy visualizes dead cells using fluorescent dyes or antibodies, Western blotting detects caspases and Bcl-2 family proteins. This review attempts to offer a thorough investigation of present studies on death in differentiated pseudo-neuronal cells produced from neural stem cells under the effect of different inducers. Through investigating how these inducers influence death, the review aims to provide information that might direct the next studies and support treatment plans for neurodegenerative diseases. With an eye toward inducers like retinoic acid, selegiline, cytokines, valproic acid, and small compounds, we examined research to evaluate death rates. The findings offer important new perspectives on the molecular processes guiding death in these cells. There is still a complete lack of understanding of how different factors affect the molecular processes that lead to death, so understanding these processes can contribute to new therapeutic approaches to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Adele Naghshbandieh
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares UniversityTehran, Iran
| | - Atefe Naghshbandieh
- Department of Pharmaceutical Biotechnology and Department of Pharmaceutical and Bimolecular Science, University of MilanMilan, Italy
| | - Elahe Barfi
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
| | - Leila Abkhooie
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical SciencesKhorramabad, Iran
| |
Collapse
|
2
|
Anilkumar S, Wright-Jin E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024; 13:485. [PMID: 38534329 PMCID: PMC10968931 DOI: 10.3390/cells13060485] [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: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
The NF-κB (nuclear factor K-light-chain-enhancer of activated B cells) transcription factor family is critical for modulating the immune proinflammatory response throughout the body. During the resting state, inactive NF-κB is sequestered by IκB in the cytoplasm. The proteasomal degradation of IκB activates NF-κB, mediating its translocation into the nucleus to act as a nuclear transcription factor in the upregulation of proinflammatory genes. Stimuli that initiate NF-κB activation are diverse but are canonically attributed to proinflammatory cytokines and chemokines. Downstream effects of NF-κB are cell type-specific and, in the majority of cases, result in the activation of pro-inflammatory cascades. Acting as the primary immune responders of the central nervous system, microglia exhibit upregulation of NF-κB upon activation in response to pathological conditions. Under such circumstances, microglial crosstalk with other cell types in the central nervous system can induce cell death, further exacerbating the disease pathology. In this review, we will emphasize the role of NF-κB in triggering neuroinflammation mediated by microglia.
Collapse
Affiliation(s)
- Sudha Anilkumar
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA
| | - Elizabeth Wright-Jin
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA
- Division of Neurology, Department of Pediatrics, Nemours Children’s Health, Wilmington, DE 19803, USA
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Xu G, Dong F, Su L, Tan ZX, Lei M, Li L, Wen D, Zhang F. The role and therapeutic potential of nuclear factor κB (NF-κB) in ischemic stroke. Biomed Pharmacother 2024; 171:116140. [PMID: 38211425 DOI: 10.1016/j.biopha.2024.116140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Stroke is a prevalent cerebrovascular condition with a global impact, causing significant rates of illness and death. Despite extensive research, the available treatment options for stroke remain restricted. Hence, it is crucial to gain a deeper understanding of the molecular mechanisms associated with the onset and advancement of stroke in order to establish a theoretical foundation for novel preventive and therapeutic approaches. NF-κB, also known as nuclear factor κB, is a transcription factor responsible for controlling the expression of numerous genes and plays a crucial role in diverse physiological processes. NF-κB is triggered and regulates neuroinflammation and other processes after stroke, promoting the generation of cytokine storms and contributing to the advancement of ischemic stroke (IS). Therefore, NF-κB could potentially play a vital role in stroke by regulating diverse pathophysiological processes. This review provides an overview of the functions of NF-κB in stroke and its governing mechanisms. In addition, our attention is directed towards various potential therapies that aim to inhibit the NF-κB signaling pathway in order to offer valuable insights for the advancement of innovative treatment approaches for stroke.
Collapse
Affiliation(s)
- Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lei Su
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, PR China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
4
|
Zhu Y, Duan S, Wang M, Deng Z, Li J. Neuroimmune Interaction: A Widespread Mutual Regulation and the Weapons for Barrier Organs. Front Cell Dev Biol 2022; 10:906755. [PMID: 35646918 PMCID: PMC9130600 DOI: 10.3389/fcell.2022.906755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Since the embryo, the nervous system and immune system have been interacting to regulate each other’s development and working together to resist harmful stimuli. However, oversensitive neural response and uncontrolled immune attack are major causes of various diseases, especially in barrier organs, while neural-immune interaction makes it worse. As the first defense line, the barrier organs give a guarantee to maintain homeostasis in external environment. And the dense nerve innervation and abundant immune cell population in barrier organs facilitate the neuroimmune interaction, which is the physiological basis of multiple neuroimmune-related diseases. Neuroimmune-related diseases often have complex mechanisms and require a combination of drugs, posing challenges in finding etiology and treatment. Therefore, it is of great significance to illustrate the specific mechanism and exact way of neuro-immune interaction. In this review, we first described the mutual regulation of the two principal systems and then focused on neuro-immune interaction in the barrier organs, including intestinal tract, lungs and skin, to clarify the mechanisms and provide ideas for clinical etiology exploration and treatment.
Collapse
Affiliation(s)
- Yan Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Shixin Duan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Mei Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhili Deng, ; Ji Li,
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhili Deng, ; Ji Li,
| |
Collapse
|
5
|
Zhu M, Li N, Wang Y, Gao S, Wang J, Shen X. Regulation of inflammation by VEGF/BDNF signaling in mouse retinal Müller glial cells exposed to high glucose. Cell Tissue Res 2022; 388:521-533. [PMID: 35394215 DOI: 10.1007/s00441-022-03622-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/25/2022] [Indexed: 11/02/2022]
Abstract
The inflammatory changes seem to play an important role in the development of diabetic retinopathy (DR). Anti-VEGF therapy has been testified to inhibit inflammation in animal models of diabetes, but the detailed mechanisms during this process are not yet clear. Müller glial cells (MGCs) in the mammalian retina are deeply involved in DR, while the BDNF overexpression reduces inflammation in diabetic mice. In this research, we aimed to explore the relationship between VEGF and BDNF in mouse retinal MGCs during inflammation of diabetes. We examined the expression of glutamine-synthetase (GS), glial fibrillary acidic protein (GFAP), vascular-endothelial growth factor (VEGF), interleukin-1beta (IL-1β), and tumor necrosis factor-alpha (TNF-α) at different time points after mouse retinal MGCs exposed to high glucose (25 mM). We also explored changes in the expression of brain-derived neurotrophic factor (BDNF), nuclear factor kappa B (NF-κB), IL-1β, and TNF-α in MGCs after treatments with anti-VEGF, VEGF siRNA, BDNF siRNA, BDNF recombination protein, and NF-κB inhibitor. In mouse retinal MGCs exposed to high glucose, BDNF was increased after treatments with anti-VEGF or VEGF siRNA. BDNF was decreased in MGCs from VEGF overexpressed mice. Moreover, the expressions of NF-κB, IL-1β, and TNF-α changed with BDNF: NF-κB, IL-1β, and TNF-α were increased after treatments with BDNF siRNA; NF-κB, IL-1β, and TNF-α were decreased after treatments with BDNF recombination protein. VEGF may regulate cytokines (IL-1β and TNF-α) by BDNF/NF-κB signaling pathway. The regulation of the VEGF/BDNF/NF-κB signaling pathway may be a significant therapeutic strategy for DR.
Collapse
Affiliation(s)
- Minqi Zhu
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Na Li
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Yanuo Wang
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Shuang Gao
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Jing Wang
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China.
| | - Xi Shen
- Department of Ophthalmology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China.
| |
Collapse
|
6
|
Tkáčová Z, Bhide K, Mochnáčová E, Petroušková P, Hruškovicová J, Kulkarni A, Bhide M. Comprehensive Mapping of the Cell Response to Borrelia bavariensis in the Brain Microvascular Endothelial Cells in vitro Using RNA-Seq. Front Microbiol 2021; 12:760627. [PMID: 34819924 PMCID: PMC8606740 DOI: 10.3389/fmicb.2021.760627] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 12/01/2022] Open
Abstract
Borrelia bavariensis can invade the central nervous system (CNS) by crossing the blood-brain barrier (BBB). It is predicted that B. bavariensis evokes numerous signaling cascades in the human brain microvascular endothelial cells (hBMECs) and exploits them to traverse across the BBB. The complete picture of signaling events in hBMECs induced by B. bavariensis remains uncovered. Using RNA sequencing, we mapped 11,398 genes and identified 295 differentially expressed genes (DEGs, 251 upregulated genes and 44 downregulated genes) in B. bavariensis challenged hBMECs. The results obtained from RNA-seq were validated with qPCR. Gene ontology analysis revealed the participation of DEGs in a number of biological processes like cell communication, organization of the extracellular matrix, vesicle-mediated transport, cell response triggered by pattern recognition receptors, antigen processing via MHC class I, cellular stress, metabolism, signal transduction, etc. The expression of several non-protein coding genes was also evoked. In this manuscript, we discuss in detail the correlation between several signaling cascades elicited and the translocation of BBB by B. bavariensis. The data revealed here may contribute to a better understanding of the mechanisms employed by B. bavariensis to cross the BBB.
Collapse
Affiliation(s)
- Zuzana Tkáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Evelina Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Patrícia Petroušková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Jana Hruškovicová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
7
|
Liu X, Ying J, Wang X, Zheng Q, Zhao T, Yoon S, Yu W, Yang D, Fang Y, Hua F. Astrocytes in Neural Circuits: Key Factors in Synaptic Regulation and Potential Targets for Neurodevelopmental Disorders. Front Mol Neurosci 2021; 14:729273. [PMID: 34658786 PMCID: PMC8515196 DOI: 10.3389/fnmol.2021.729273] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are the major glial cells in the brain, which play a supporting role in the energy and nutritional supply of neurons. They were initially regarded as passive space-filling cells, but the latest progress in the study of the development and function of astrocytes highlights their active roles in regulating synaptic transmission, formation, and plasticity. In the concept of "tripartite synapse," the bidirectional influence between astrocytes and neurons, in addition to their steady-state and supporting function, suggests that any negative changes in the structure or function of astrocytes will affect the activity of neurons, leading to neurodevelopmental disorders. The role of astrocytes in the pathophysiology of various neurological and psychiatric disorders caused by synaptic defects is increasingly appreciated. Understanding the roles of astrocytes in regulating synaptic development and the plasticity of neural circuits could help provide new treatments for these diseases.
Collapse
Affiliation(s)
- Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Tiancheng Zhao
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Sungtae Yoon
- Helping Minds International Charitable Foundation, New York, NY, United States
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
8
|
Han H, Desert R, Das S, Song Z, Athavale D, Ge X, Nieto N. Danger signals in liver injury and restoration of homeostasis. J Hepatol 2020; 73:933-951. [PMID: 32371195 PMCID: PMC7502511 DOI: 10.1016/j.jhep.2020.04.033] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
Damage-associated molecular patterns are signalling molecules involved in inflammatory responses and restoration of homeostasis. Chronic release of these molecules can also promote inflammation in the context of liver disease. Herein, we provide a comprehensive summary of the role of damage-associated molecular patterns as danger signals in liver injury. We consider the role of reactive oxygen species and reactive nitrogen species as inducers of damage-associated molecular patterns, as well as how specific damage-associated molecular patterns participate in the pathogenesis of chronic liver diseases such as alcohol-related liver disease, non-alcoholic steatohepatitis, liver fibrosis and liver cancer. In addition, we discuss the role of damage-associated molecular patterns in ischaemia reperfusion injury and liver transplantation and highlight current studies in which blockade of specific damage-associated molecular patterns has proven beneficial in humans and mice.
Collapse
Affiliation(s)
- Hui Han
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Romain Desert
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Sukanta Das
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Zhuolun Song
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Dipti Athavale
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Xiaodong Ge
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA; Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood St., Suite 1020N, MC 787, Chicago, IL 60612, USA.
| |
Collapse
|
9
|
Sánchez-Garrido AI, Prieto-Vicente V, Blanco-Gozalo V, Arévalo M, Quiros Y, López-Montañés D, López-Hernández FJ, Rodríguez-Pérez A, López-Novoa JM. Preventive Effect of Cardiotrophin-1 Administration before DSS-Induced Ulcerative Colitis in Mice. J Clin Med 2019; 8:jcm8122086. [PMID: 31805674 PMCID: PMC6947259 DOI: 10.3390/jcm8122086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/23/2022] Open
Abstract
Ulcerative colitis is a relatively frequent, chronic disease that impacts significantly the patient's quality of life. Although many therapeutic options are available, additional approaches are needed because many patients either do not respond to current therapies or show significant side effects. Cardiotrophin-1 (CT-1) is a cytokine with potent cytoprotective, anti-inflammatory, and antiapoptotic properties. The purpose of this study was to assess if the administration of CT-1 could reduce colon damage in mice with experimental colitis was induced with 5% dextran sulfate sodium (DSS) in the drinking water. Half of the mice received an i.v. dose of CT-1 (200 µg/kg) 2 h before and 2 and 4 days after DSS administration. Animals were followed during 7 days after DSS administration. The severity of colitis was measured by standard scores. Colon damage was assessed by histology and immunohistochemistry. Inflammatory mediators were measured by Western blot and PCR. CT-1 administration to DSS-treated mice ameliorated both the clinical course (disease activity index), histological damage, inflammation (colon expression of TNF-α, IL-17, IL-10, INF IFN-γ, and iNOS), and apoptosis. Our results suggest that CT-1 administration before induction of colitis improves the clinical course, tissue damage, and inflammation in DSS-induced colitis in mice.
Collapse
Affiliation(s)
- Ana I. Sánchez-Garrido
- Department of Gastroenterology, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.I.S.-G.); (V.P.-V.); (A.R.-P.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
| | - Vanessa Prieto-Vicente
- Department of Gastroenterology, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.I.S.-G.); (V.P.-V.); (A.R.-P.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
| | - Víctor Blanco-Gozalo
- Bio-inRen S.L. Faculty of Medicine, Campus Miguel de Unamuno, 37007 Salamanca, Spain; (V.B.-G.); (Y.Q.)
| | - Miguel Arévalo
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
- Department of Human Anatomy and Histology, University of Salamanca, 37007 Salamanca, Spain
| | - Yaremi Quiros
- Bio-inRen S.L. Faculty of Medicine, Campus Miguel de Unamuno, 37007 Salamanca, Spain; (V.B.-G.); (Y.Q.)
| | - Daniel López-Montañés
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
- Bio-inRen S.L. Faculty of Medicine, Campus Miguel de Unamuno, 37007 Salamanca, Spain; (V.B.-G.); (Y.Q.)
| | - Francisco J. López-Hernández
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
- Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
| | - Antonio Rodríguez-Pérez
- Department of Gastroenterology, University Hospital of Salamanca, 37007 Salamanca, Spain; (A.I.S.-G.); (V.P.-V.); (A.R.-P.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
| | - José M. López-Novoa
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.; (M.A.); (D.L.-M.); (F.J.L.-H.)
- Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-923294500; Fax: +34-923294669
| |
Collapse
|
10
|
Jha NK, Jha SK, Kar R, Nand P, Swati K, Goswami VK. Nuclear factor-kappa β as a therapeutic target for Alzheimer's disease. J Neurochem 2019; 150:113-137. [PMID: 30802950 DOI: 10.1111/jnc.14687] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/16/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a typical progressive, chronic neurodegenerative disorder with worldwide prevalence. Its clinical manifestation involves the presence of extracellular plaques and intracellular neurofibrillary tangles (NFTs). NFTs occur in brain tissues as a result of both Aβ agglomeration and Tau phosphorylation. Although there is no known cure for AD, research into possible cures and treatment options continues using cell-cultures and model animals/organisms. The nuclear factor-kappa β (NF-κβ) plays an active role in the progression of AD. Impairment to this signaling module triggers undesirable phenotypic changes such as neuroinflammation, activation of microglia, oxidative stress related complications, and apoptotic cell death. These imbalances further lead to homeostatic abnormalities in the brain or in initial stages of AD essentially pushing normal neurons toward the degeneration process. Interestingly, the role of NF-κβ signaling associated receptor-interacting protein kinase is currently observed in apoptotic and necrotic cell death, and has been reported in brains. Conversely, the NF-κβ signaling pathway has also been reported to be involved in normal brain functioning. This pathway plays a crucial role in maintaining synaptic plasticity and balancing between learning and memory. Since any impairment in the pathways associated with NF-κβ signaling causes altered neuronal dynamics, neurotherapeutics using compounds including, antioxidants, bioflavonoids, and non-steroidal anti-inflammatory drugs against such abnormalities offer possibilities to rectify aberrant excitatory neuronal activity in AD. In this review, we have provided an extensive overview of the crucial role of NF-κβ signaling in normal brain homeostasis. We have also thoroughly outlined several established pathomechanisms associated with NF-κβ pathways in AD, along with their respective therapeutic approaches.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, Noida Institute of Engineering & Technology (NIET), Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Rohan Kar
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Parma Nand
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Kumari Swati
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Vineet Kumar Goswami
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
11
|
McGregor CE, English AW. The Role of BDNF in Peripheral Nerve Regeneration: Activity-Dependent Treatments and Val66Met. Front Cell Neurosci 2019; 12:522. [PMID: 30687012 PMCID: PMC6336700 DOI: 10.3389/fncel.2018.00522] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/14/2018] [Indexed: 11/29/2022] Open
Abstract
Despite the ability of peripheral nerves to spontaneously regenerate after injury, recovery is generally very poor. The neurotrophins have emerged as an important modulator of axon regeneration, particularly brain derived neurotrophic factor (BDNF). BDNF regulation and signaling, as well as its role in activity-dependent treatments including electrical stimulation, exercise, and optogenetic stimulation are discussed here. The importance of a single nucleotide polymorphism in the BDNF gene, Val66Met, which is present in 30% of the human population and may hinder the efficacy of these treatments in enhancing regeneration after injury is considered. Preliminary data are presented on the effectiveness of one such activity-dependent treatment, electrical stimulation, in enhancing axon regeneration in mice expressing the met allele of the Val66Met polymorphism.
Collapse
Affiliation(s)
- Claire Emma McGregor
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Arthur W English
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
12
|
Cardiotrophin-1 attenuates experimental colitis in mice. Clin Sci (Lond) 2018; 132:985-1001. [PMID: 29572384 DOI: 10.1042/cs20171513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
Cardiotrophin-1 (CT-1) holds potent anti-inflammatory, cytoprotective, and anti-apoptotic effects in the liver, kidneys, and heart. In the present study, the role of endogenous CT-1 and the effect of exogenous CT-1 were evaluated in experimental ulcerative colitis. Colitis was induced in CT-1 knockout and wild-type (WT) mice by administration of dextran sulphate sodium (DSS) in the drinking water during 7 days. CT-1 knockout mice showed higher colon damage and disease severity than WT mice. In addition, CT-1 (200 µg/kg/day, iv) or vehicle (as control) was administered during 3 days to WT, colitic mice, starting on day 4 after initiation of DSS. Disease activity index (DAI), inflammatory markers (tumor necrosis factor α (TNF-α), INFγ, IL-17, IL-10, inducible nitric oxide synthase (iNOS)), colon damage, apoptosis (cleaved caspase 3), nuclear factor κB (NFκB) and STAT-3 activation, and bacterial translocation were measured. Compared with mice treated with DSS, mice also treated with exogenous CT-1 showed lower colon damage, DAI, plasma levels of TNFα, colon expression of TNF-α, INFγ, IL-17, iNOS and cleaved caspase 3, higher NFκB and signal transducer and activator of transcription 3 (STAT3) pathways activation, and absence of bacterial translocation. We conclude that endogenous CT-1 plays a role in the defense and repair response of the colon against ulcerative lesions through an anti-inflammatory and anti-apoptotic effect. Supplementation with exogenous CT-1 ameliorates disease symptoms, which opens a potentially new therapeutic strategy for ulcerative colitis.
Collapse
|
13
|
Tanqueiro SR, Ramalho RM, Rodrigues TM, Lopes LV, Sebastião AM, Diógenes MJ. Inhibition of NMDA Receptors Prevents the Loss of BDNF Function Induced by Amyloid β. Front Pharmacol 2018; 9:237. [PMID: 29695962 PMCID: PMC5904251 DOI: 10.3389/fphar.2018.00237] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/02/2018] [Indexed: 01/18/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays important functions in cell survival and differentiation, neuronal outgrowth and plasticity. In Alzheimer’s disease (AD), BDNF signaling is known to be impaired, partially because amyloid β (Aβ) induces truncation of BDNF main receptor, TrkB-full length (TrkB-FL). We have previously shown that such truncation is mediated by calpains, results in the formation of an intracellular domain (ICD) fragment and causes BDNF loss of function. Since calpains are Ca2+-dependent proteases, we hypothesized that excessive intracellular Ca2+ build-up could be due to dysfunctional N-methyl-d-aspartate receptors (NMDARs) activation. To experimentally address this hypothesis, we investigated whether TrkB-FL truncation by calpains and consequent BDNF loss of function could be prevented by NMDAR blockade. We herein demonstrate that a NMDAR antagonist, memantine, prevented excessive calpain activation and TrkB-FL truncation induced by Aβ25–35. When calpains were inhibited by calpastatin, BDNF was able to increase the dendritic spine density of neurons exposed to Aβ25135. Moreover, NMDAR inhibition by memantine also prevented Aβ-driven deleterious impact of BDNF loss of function on structural (spine density) and functional outcomes (synaptic potentiation). Collectively, these findings support NMDAR/Ca2+/calpains mechanistic involvement in Aβ-triggered BDNF signaling disruption.
Collapse
Affiliation(s)
- Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita M Ramalho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Department of Ophthalmology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Luísa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
14
|
Shaikh SS, Nahorski MS, Woods CG. A third HSAN5 mutation disrupts the nerve growth factor furin cleavage site. Mol Pain 2018; 14:1744806918809223. [PMID: 30296891 PMCID: PMC6207963 DOI: 10.1177/1744806918809223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/20/2018] [Accepted: 09/20/2018] [Indexed: 11/17/2022] Open
Abstract
Bi-allelic dysfunctional mutations in nerve growth factor (NGF) cause the rare human phenotype hereditary sensory and autonomic neuropathy type 5 (HSAN5). We describe a novel NGF mutation in an individual with typical HSAN5 findings. The mutation c.361C>T, p.R121W is at the last residue of the furin cleavage motif Arg-Ser-Lys-Arg in proNGF. We show that the p.R121W mutation completely abolishes the formation of mature NGF-β. Surprisingly, mutant p.R121W cells produced very little proNGF. Instead, the two progressive cleavage products of proNGF were produced, proA-NGF and proB-NGF, with proB-NGF being the predominant NGF-derived peptide and the only peptide secreted by mutant p.R121W cells. We found that the ability of the p.R121W mutation to cause tropomyosin receptor kinase A autophosphorylation and mitogen-activated protein kinase phosphorylation was significantly reduced compared to controls (p < 0.05 and p < 0.01). By studying the PC12 cell line morphology and neurite length over a week, we found the p.R121W mutation had residual, but much reduced, neurotrophic activity when compared to wild-type NGF. Finally, we assessed whether the p.R121W mutation affected apoptosis and found a reduced protective effect compared to wild-type NGF. Our results suggest that the p.R121W NGF mutation causes HSAN5 through negating the ability of furin to cleave proNGF to produce NGF-β.
Collapse
Affiliation(s)
- Samiha S Shaikh
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
| | - Michael S Nahorski
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
| | - C Geoffrey Woods
- Cambridge Institute for Medical Research, Addenbrooke's Biomedical Research Centre, Cambridge, UK
- Department of Clinical Genetics, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
15
|
Peng L, Shu X, Lang C, Yu X. Cardiotrophin-1 stimulates the neural differentiation of human umbilical cord blood-derived mesenchymal stem cells and survival of differentiated cells through PI3K/Akt-dependent signaling pathways. Cytotechnology 2017; 69:933-941. [PMID: 28601931 DOI: 10.1007/s10616-017-0103-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/22/2017] [Indexed: 12/31/2022] Open
Abstract
Cardiotrophin-1 (CT1) plays an important role in the differentiation, development, and survival of neural stem cells. In this study, we analyzed its effects on the stimulation of human umbilical cord blood-derived mesenchymal stem cells in terms of their potential to differentiate into neuron-like cells, their survival characteristics, and the molecular mechanisms involved. The treatment of cells with neural induction medium (NIM) and CT1 generated more cells that were neuron-like and produced stronger expression of neural-lineage markers than cells treated with NIM and without CT1. Bcl-2 and Akt phosphorylation (p-Akt) expression levels increased significantly in cells treated with both NIM and CT1. This treatment also effectively blocked cell death following neural induction and decreased Bax, Bak and cleaved-caspase 3 expression compared with cells treated with NIM without CT1. In addition, the inhibition of phosphatidylinositol 3-kinase (PI3K) abrogated p-Akt and Bcl-2 expression. Thus, PI3K/Akt contribute to CT1-stimulated neural differentiation and to the survival of differentiated cells.
Collapse
Affiliation(s)
- Longying Peng
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaomei Shu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China.
| | - Changhui Lang
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| | - Xiaohua Yu
- Department of Pediatric, First Affiliated Hospital, Zunyi Medical College, Zunyi, 563003, Guizhou Province, China
| |
Collapse
|
16
|
Lee HK, Keum S, Sheng H, Warner DS, Lo DC, Marchuk DA. Natural allelic variation of the IL-21 receptor modulates ischemic stroke infarct volume. J Clin Invest 2016; 126:2827-38. [PMID: 27400126 DOI: 10.1172/jci84491] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 05/12/2016] [Indexed: 02/06/2023] Open
Abstract
Risk for ischemic stroke has a strong genetic basis, but heritable factors also contribute to the extent of damage after a stroke has occurred. We previously identified a locus on distal mouse chromosome 7 that contributes over 50% of the variation in postischemic cerebral infarct volume observed between inbred strains. Here, we used ancestral haplotype analysis to fine-map this locus to 12 candidate genes. The gene encoding the IL-21 receptor (Il21r) showed a marked difference in strain-specific transcription levels and coding variants in neonatal and adult cortical tissue. Collateral vessel connections were moderately reduced in Il21r-deficient mice, and cerebral infarct volume increased 2.3-fold, suggesting that Il21r modulates both collateral vessel anatomy and innate neuroprotection. In brain slice explants, oxygen deprivation (OD) activated apoptotic pathways and increased neuronal cell death in IL-21 receptor-deficient (IL-21R-deficient) mice compared with control animals. We determined that the neuroprotective effects of IL-21R arose from signaling through JAK/STAT pathways and upregulation of caspase 3. Thus, natural genetic variation in murine Il21r influences neuronal cell viability after ischemia by modulating receptor function and downstream signal transduction. The identification of neuroprotective genes based on naturally occurring allelic variations has the potential to inform the development of drug targets for ischemic stroke treatment.
Collapse
|
17
|
Wehner AB, Milen AM, Albin RL, Pierchala BA. The p75 neurotrophin receptor augments survival signaling in the striatum of pre-symptomatic Q175(WT/HD) mice. Neuroscience 2016; 324:297-306. [PMID: 26947127 PMCID: PMC4849895 DOI: 10.1016/j.neuroscience.2016.02.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/26/2016] [Accepted: 02/28/2016] [Indexed: 12/19/2022]
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder characterized by a constellation of motor, cognitive, and psychiatric features. Striatal medium spiny neurons, one of the most affected populations, are dependent on brain-derived neurotrophic factor (BDNF) anterogradely transported from the cortex for proper function and survival. Recent studies suggest both receptors for BDNF, TrkB and p75 neurotrophin receptor (p75), are improperly regulated in the striata of HD patients and mouse models of HD. While BDNF-TrkB signaling almost exclusively promotes survival and metabolic function, p75 signaling is able to induce survival or apoptosis depending on the available ligand and associated co-receptor. We investigated the role of p75 in the Q175 knock-in mouse model of HD by examining the levels and activation of downstream signaling molecules, and subsequently examining Hdh(+/Q175);p75(-/-) mice to determine if p75 represents a promising therapeutic target. In Hdh(+/Q175);p75(+/+) mice, we observed enhanced survival signaling as evidenced by an increase in phosphorylation and activation of Akt and the p65 subunit of NFκB in the striatum at 5 months of age and an increase in XIAP expression compared to Hdh(+/+);p75(+/+) mice; this increase was lost in Hdh(+/Q175);p75(-/-) mice. Hdh(+/Q175);p75(-/-) mice also showed a decrease in Bcl-XL expression by immunoblotting compared to Hdh(+/Q175);p75(+/+) and Hdh(+/+);p75(+/+) littermates. Consistent with diminished survival signaling, DARPP-32 expression decreased both by immunoblotting and by immunohistochemistry in Hdh(+/Q175);p75(-/-) mice compared to Hdh(+/+);p75(+/+), Hdh(+/Q175);p75(+/+), and Hdh(+/+);p75(-/-) littermates. Additionally, striatal volume declined to a greater extent in Hdh(+/Q175);p75(-/-) when compared to Hdh(+/Q175);p75(+/+) littermates at 12 months, indicating a more aggressive onset of degeneration. These data suggest that p75 signaling plays an early role in augmenting pro-survival signaling in the striatum and that disruption of p75 signaling at a pre-symptomatic age may exacerbate pathologic changes in Hdh(+/Q175) mice.
Collapse
Affiliation(s)
- A B Wehner
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - A M Milen
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - R L Albin
- Neuroscience Graduate Program, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - B A Pierchala
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
Quirós Y, Blanco-Gozalo V, Sanchez-Gallego JI, López-Hernandez FJ, Ruiz J, Perez de Obanos MP, López-Novoa JM. Cardiotrophin-1 therapy prevents gentamicin-induced nephrotoxicity in rats. Pharmacol Res 2016; 107:137-146. [PMID: 26996880 DOI: 10.1016/j.phrs.2016.02.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/10/2023]
Abstract
Aminoglycosides are very effective antibiotics for the treatment of severe infections, but they rank among the most frequent causes of drug-induced nephrotoxicity. Thus, prevention of aminoglycoside nephrotoxicity is an unmet therapeutic objective. Cardiotrophin-1 (CT-1), a member of the IL-6 family of cytokines, has been reported to protect the kidney against toxic and ischemic acute kidney injury (AKI). We have assessed the effect of rat CT-1 in the severity of gentamicin (G)-induced AKI. Groups of male Wistar rats received the following for 6 consecutive days: i) isotonic saline solution (group CONT), ii) G, 150mg/kg/day, i.p. (group G), iii) CT-1, 100μg/kg/day i.v. (group CT-1), or iv) G and CT-1 at the doses described above. The G group showed a manifest AKI characterized by low creatinine clearance, high plasma creatinine and urea levels, increased urinary excretion of proteins, glucose and AKI markers such as N-acetyl-glucosaminidase, neutrophil gelatinase-associated lipocalin, kidney-injury molecule-1 and T-gelsolin, increased kidney levels of CD-68, iNOS, IL-1β and TNF-α, and markedly higher histological renal damage and leukocyte infiltration than the CONT and CT-1 groups. Administration of CT-1 together with G reduced almost all of the above-described manifestations of G-induced AKI. The results of this study have potential clinical application, as CT-1 is near to being used as a drug for organ protection.
Collapse
Affiliation(s)
| | | | | | - Francisco J López-Hernandez
- Unidad de Fisiopatología Renal y Cardiovascular, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain; Instituto de Estudios de Ciencias de la Salud de Castilla y León (IESCYL), Salamanca, Spain
| | | | | | - José M López-Novoa
- Unidad de Fisiopatología Renal y Cardiovascular, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
19
|
Karki P, Johnson J, Son DS, Aschner M, Lee E. Transcriptional Regulation of Human Transforming Growth Factor-α in Astrocytes. Mol Neurobiol 2016; 54:964-976. [PMID: 26797516 DOI: 10.1007/s12035-016-9705-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 01/05/2016] [Indexed: 11/25/2022]
Abstract
Transforming growth factor-alpha (TGF-α) is known to play multifunctional roles in the central nervous system (CNS), including the provision of neurotropic properties that protect neurons against various neurotoxic insults. Previously, we reported that TGF-α mediates estrogen-induced enhancement of glutamate transporter GLT-1 function in astrocytes. However, the regulatory mechanism of TGF-α at the transcriptional level remains to be established. Our findings revealed that the human TGF-α promoter contains consensus sites for several transcription factors, such as NF-κB and yin yang 1 (YY1). NF-κB served as a positive regulator of TGF-α promoter activity, corroborated by observations that overexpression of NF-κB p65 increased, while mutation in the NF-κB binding sites in the TGF-α promoter reduced the promoter activity in rat primary astrocytes. Pharmacological inhibition of NF-κB with pyrrolidine dithiocarbamate (PDTC; 50 μM) or quinazoline (QNZ; 10 μM) also abolished TGF-α promoter activity, and NF-κB directly bound to its consensus site in the TGF-α promoter as evidenced by electrophoretic mobility shift assay (EMSA). Dexamethasone (DX) increased TGF-α promoter activity by activation of NF-κB. Treatment of astrocytes with 100 nM of DX for 24 h activated its glucocorticoid receptor and signaling proteins, including MAPK, PI3K/Akt, and PKA, via non-genomic pathways, to enhance TGF-α promoter activity and expression. YY1 served as a critical negative regulator of the TGF-α promoter as overexpression of YY1 decreased, while mutation of YY1 binding site in the promoter increased TGF-α promoter activity. Treatment for 3 h with 250 μM of manganese (Mn), an environmental neurotoxin, decreased astrocytic TGF-α expression by activation of YY1. Taken together, our results suggest that NF-κB is a critical positive regulator, whereas YY1 is a negative regulator of the TGF-α promoter. These findings identify potential molecular targets for neurotherapeutics that may modulate TGF-α regulation and afford neuroprotection.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, 37208, USA
| | - James Johnson
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, 37208, USA
| | - Deok-Soo Son
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, 37208, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eunsook Lee
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN, 37208, USA.
| |
Collapse
|
20
|
Kaltschmidt B, Kaltschmidt C. NF-KappaB in Long-Term Memory and Structural Plasticity in the Adult Mammalian Brain. Front Mol Neurosci 2015; 8:69. [PMID: 26635522 PMCID: PMC4656838 DOI: 10.3389/fnmol.2015.00069] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/30/2015] [Indexed: 11/13/2022] Open
Abstract
The transcription factor nuclear factor kappaB (NF-κB) is a well-known regulator of inflammation, stress, and immune responses as well as cell survival. In the nervous system, NF-κB is one of the crucial components in the molecular switch that converts short- to long-term memory-a process that requires de novo gene expression. Here, the researches published on NF-κB and downstream target genes in mammals will be reviewed, which are necessary for structural plasticity and long-term memory, both under normal and pathological conditions in the brain. Genetic evidence has revealed that NF-κB regulates neuroprotection, neuronal transmission, and long-term memory. In addition, after genetic ablation of all NF-κB subunits, a severe defect in hippocampal adult neurogenesis was observed during aging. Proliferation of neural precursors is increased; however, axon outgrowth, synaptogenesis, and tissue homeostasis of the dentate gyrus are hampered. In this process, the NF-κB target gene PKAcat and other downstream target genes such as Igf2 are critically involved. Therefore, NF-κB activity seems to be crucial in regulating structural plasticity and replenishment of granule cells within the hippocampus throughout the life. In addition to the function of NF-κB in neurons, we will discuss on a neuroinflammatory role of the transcription factor in glia. Finally, a model for NF-κB homeostasis on the molecular level is presented, in order to explain seemingly the contradictory, the friend or foe, role of NF-κB in the nervous system.
Collapse
|
21
|
Vicario A, Kisiswa L, Tann JY, Kelly CE, Ibáñez CF. Neuron-type-specific signaling by the p75NTR death receptor is regulated by differential proteolytic cleavage. J Cell Sci 2015; 128:1507-17. [PMID: 25720379 DOI: 10.1242/jcs.161745] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/16/2015] [Indexed: 11/20/2022] Open
Abstract
Signaling by the p75 neurotrophin receptor (p75(NTR), also known as NGFR) is often referred to as cell-context dependent, but neuron-type-specific signaling by p75(NTR) has not been systematically investigated. Here, we report that p75(NTR) signals very differently in hippocampal neurons (HCNs) and cerebellar granule neurons (CGNs), and we present evidence indicating that this is partly controlled by differential proteolytic cleavage. Nerve growth factor (NGF) induced caspase-3 activity and cell death in HCNs but not in CGNs, whereas it stimulated NFκB activity in CGNs but not in HCNs. HCNs and CGNs displayed different patterns of p75(NTR) proteolytic cleavage. Whereas the p75(NTR) carboxy terminal fragment (CTF) was more abundant than the intracellular domain (ICD) in HCNs, CGNs exhibited fully processed ICD with very little CTF. Pharmacological or genetic blockade of p75(NTR) cleavage by γ-secretase abolished NGF-induced upregulation of NFκB activity and enabled induction of CGN death, phenocopying the functional profile of HCNs. Thus, the activities of multifunctional receptors, such as p75(NTR), can be tuned into narrower activity profiles by cell-type-specific differences in intracellular processes, such as proteolytic cleavage, leading to very different biological outcomes.
Collapse
Affiliation(s)
- Annalisa Vicario
- Department of Neuroscience, Karolinska Institute, Stockholm S-17177, Sweden
| | - Lilian Kisiswa
- Department of Neuroscience, Karolinska Institute, Stockholm S-17177, Sweden
| | - Jason Y Tann
- Life Sciences Institute, Department of Physiology, National University of Singapore, Singapore 117456, Singapore
| | - Claire E Kelly
- Department of Neuroscience, Karolinska Institute, Stockholm S-17177, Sweden
| | - Carlos F Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm S-17177, Sweden Life Sciences Institute, Department of Physiology, National University of Singapore, Singapore 117456, Singapore Department of Physiology, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
22
|
García-Bueno B, Bioque M, Mac-Dowell KS, Barcones MF, Martínez-Cengotitabengoa M, Pina-Camacho L, Rodríguez-Jiménez R, Sáiz PA, Castro C, Lafuente A, Santabárbara J, González-Pinto A, Parellada M, Rubio G, García-Portilla MP, Micó JA, Bernardo M, Leza JC. Pro-/anti-inflammatory dysregulation in patients with first episode of psychosis: toward an integrative inflammatory hypothesis of schizophrenia. Schizophr Bull 2014; 40:376-87. [PMID: 23486748 PMCID: PMC3932081 DOI: 10.1093/schbul/sbt001] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Schizophrenia is a chronic syndrome of unknown etiology, predominantly defined by signs of psychosis. The onset of the disorder occurs typically in late adolescence or early adulthood. Efforts to study pathophysiological mechanisms in early stages of the disease are crucial in order to prompt intervention. METHODS Case-control study of first-episode psychotic (FEP) patients and matched controls. We recruited 117 patients during the first year after their FEP according to the DSM-IV criteria and recruited 106 gender-, race-, and age-matched controls between September 2010 and June 2011. RESULTS Biochemical studies carried out in peripheral mononuclear blood cells (PMBC) and plasma evidence a significant increase in intracellular components of a main proinflammatory pathway, along with a significant decrease in the anti-inflammatory ones. Multivariate logistic regression analyses identified the expression of inducible isoforms of nitric oxide synthase and cyclooxygenase in PMBC and homocysteine plasma levels as the most reliable potential risk factors and the inhibitor of the inflammatory transcription factor NFκB, IκBα, and the anti-inflammatory prostaglandin 15d-PGJ2 as potential protection factors. DISCUSSION Taken as a whole, the results of this study indicate robust phenotypical differences at the cellular machinery level in PMBC of patients with FEP. Although more scientific evidence is needed, the determination of multiple components of pro- and anti-inflammatory cellular pathways including the activity of nuclear receptors has interesting potential as biological markers and potential risk/protective factors for FEP. Due to its soluble nature, a notable finding in this study is that the anti-inflammatory mediator 15d-PGJ2 might be used as plasmatic biomarker for first episodes of psychosis.
Collapse
Affiliation(s)
- Borja García-Bueno
- CIBERSAM and: Department of Pharmacology, Faculty of Medicine, Complutense University and Instituto de Investigación Sanitaria-IIS-Hospital 12 de Octubre (i+12), Madrid, Spain;,These authors contributed equally to this work
| | - Miquel Bioque
- Hospital Clínic, Barcelona, Spain;,These authors contributed equally to this work
| | - Karina S. Mac-Dowell
- CIBERSAM and: Department of Pharmacology, Faculty of Medicine, Complutense University and Instituto de Investigación Sanitaria-IIS-Hospital 12 de Octubre (i+12), Madrid, Spain;,These authors contributed equally to this work
| | - M. Fe Barcones
- Hospital Clínico Universitario, Zaragoza, Spain;,These authors contributed equally to this work
| | | | - Laura Pina-Camacho
- Child and Adolescent Psychiatry Department, IIS Gregorio Marañón, IISGM, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Pilar A. Sáiz
- Department of Psychiatry, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - Carmen Castro
- Department of Physiology, Faculty of Medicine, University of Cádiz, Cádiz, Spain
| | - Amalia Lafuente
- Department of Pharmacology, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Javier Santabárbara
- Hospital Clínico Universitario, Zaragoza, Spain;,Department of Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ana González-Pinto
- Hospital Universitario de Alava (sede Santiago) Universidad Nacional de Educación a Distancia, Vitoria, Spain
| | - Mara Parellada
- Child and Adolescent Psychiatry Department, IIS Gregorio Marañón, IISGM, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | | | - Juan A. Micó
- Department of Pharmacology, Faculty of Medicine, University of Cádiz, Cádiz, Spain
| | - Miguel Bernardo
- Hospital Clínic, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Juan C. Leza
- CIBERSAM and: Department of Pharmacology, Faculty of Medicine, Complutense University and Instituto de Investigación Sanitaria-IIS-Hospital 12 de Octubre (i+12), Madrid, Spain;,These authors contributed equally to this work
| |
Collapse
|
23
|
Lu XM, Shu YH, Qiu CH, Chen KT, Wang YT. Protective effects and anti-apoptotic role of nerve growth factor on spinal cord neurons in sciatic nerve-injured rats. Neurol Res 2014; 36:814-23. [PMID: 24620979 DOI: 10.1179/1743132814y.0000000333] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The purpose of this study is to demonstrate a dependence of spinal cord motoneurons on the communication with their targets, sciatic nerves, and investigate whether the effects of nerve growth factor (NGF) on the spinal cord neuron apoptosis and surviving through the regulation of nuclear factor-kappa B (NF-kappaB) in Schwann cells (SCs) in sciatic nerve injured rats. METHODS Ninety healthy adult Sprague-Dawley rats were divided randomly into normal control group, crushing group, and NGF-intervened group. When sciatic nerve crushed 1, 3, 7, 14, and 21 days, the expression of NF-kappaB in SCs and the apoptosis regulator Bcl-2 and Caspase-3 in spinal cord were examined by immunohistochemistry staining, Western blot analysis, and immunofluorescence double-labeling method, the motor neuron apoptosis were investigated by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL), and the surviving neurons were tested by toluidine blue (Nissl) staining, respectively. All the data were further analyzed with SPSS10·0 application software. RESULTS The level of the expression of NF-kappaB in crushing group enhanced at 1 day after crushing, reached peak at 3 days, and reduced at least until 21 days, which was markedly higher than that in the normal control group. The expression of NF-kappaB in NGF-intervened group showed the same changes, reached peak at 7 days, and reduced until 21 days. However, when compared with crushing group, the expression of NF-kappaB in NGF-intervened group was down-regulated significantly until 3 days after injury, and up-regulated obviously with time going on. The same trend was observed in the time course on motor neuron apoptosis in crushing group and NGF-intervened group after sciatic nerves injury, while the reversing change was found in the surviving neurons. Moreover, the kinetics of Bcl-2 expression in spinal cord was consistent with that of NF-kappaB, while reversing with that of Caspase-3. CONCLUSION The findings revealed that NGF may play a pivotal role of anti-apoptosis in spinal cord neurons through retrograde transport of NF-kappaB in SCs following sciatic nerve injury in rats.
Collapse
|
24
|
Saleh A, Smith DR, Tessler L, Mateo AR, Martens C, Schartner E, Van der Ploeg R, Toth C, Zochodne DW, Fernyhough P. Receptor for advanced glycation end-products (RAGE) activates divergent signaling pathways to augment neurite outgrowth of adult sensory neurons. Exp Neurol 2013; 249:149-59. [DOI: 10.1016/j.expneurol.2013.08.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/27/2013] [Accepted: 08/30/2013] [Indexed: 11/26/2022]
|
25
|
Bottai D, Adami R. Spinal muscular atrophy: new findings for an old pathology. Brain Pathol 2013; 23:613-22. [PMID: 23750936 DOI: 10.1111/bpa.12071] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022] Open
Abstract
Understanding the events that are responsible for a disease is mandatory for setting up a therapeutic strategy. Although spinal muscular atrophy (SMA) is considered a rare neurodegenerative pathology, its impact in our society is really devastating as it strikes young people from birth onward, and it affects their families either emotionally or financially. Moreover, it requires intensive care for the children, and this diverts both parents and relatives from their occupations. Each neuron is very different from one another; therefore, in a neurodegenerative disease, the population of axons, synapses and cell bodies degenerate asynchronously, and subpopulations of neurons have different vulnerabilities. The knowledge of the sequence of events along the lengths of individual neurons is crucial to understand if each synapse degenerates before the corresponding axon, or if each axon degenerates before the corresponding cell body. Early degeneration of one neuronal compartment in disease often reflects molecular defects somewhere else. Up until now, SMA is considered mostly a lower motor neuron disease caused by the loss-of-function mutations in the SMN1 gene; here, we inspect other features that can be altered by this defect, such as the cross talk between muscle and motor neuron and the role of physical inactivity.
Collapse
Affiliation(s)
- Daniele Bottai
- Department of Science Health, University of Milan, Milano, Italy
| | | |
Collapse
|
26
|
Feng X, Yang S, Liu J, Huang J, Peng J, Lin J, Tao J, Chen L. Electroacupuncture ameliorates cognitive impairment through inhibition of NF-κB-mediated neuronal cell apoptosis in cerebral ischemia-reperfusion injured rats. Mol Med Rep 2013; 7:1516-22. [PMID: 23525450 DOI: 10.3892/mmr.2013.1392] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/15/2013] [Indexed: 11/06/2022] Open
Abstract
Cognitive impairment is a serious mental deficit following stroke that severely affects the quality of life of stroke survivors. Nuclear factor‑κB (NF-κB)-mediated neuronal cell apoptosis is involved in the development of post-stroke cognitive impairment; therefore, it has become a promising target for the treatment of impaired cognition. Acupuncture at the Baihui (DU20) and Shenting (DU24) acupoints is commonly used in China to clinically treat post‑stroke cognitive impairment; however, the precise mechanism of its action is largely unknown. In the present study, we evaluated the therapeutic efficacy of electroacupuncture against post-stroke cognitive impairment and investigated the underlying molecular mechanisms using a rat model of focal cerebral ischemia-reperfusion (I/R) injury. Electroacupuncture at Baihui and Shenting was identified to significantly ameliorate neurological deficits and reduce cerebral infarct volume. Additionally, electroacupuncture improved learning and memory ability in cerebral I/R injured rats, demonstrating its therapeutic efficacy against post-stroke cognitive impairment. Furthermore, electroacupuncture significantly suppressed the I/R-induced activation of NF-κB signaling in ischemic cerebral tissues. The inhibitory effect of electroacupuncture on NF-κB activation led to the inhibition of cerebral cell apoptosis. Finally, electroacupuncture markedly downregulated the expression of pro-apoptotic Bax and Fas, two critical downstream target genes of the NF-κB pathway. Collectively, our findings suggest that inhibition of NF-κB‑mediated neuronal cell apoptosis may be one mechanism via which electroacupuncture at Baihui and Shenting exerts a therapeutic effect on post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Xiaodong Feng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Saleh A, Schapansky J, Smith DR, Young N, Odero GL, Aulston B, Fernyhough P, Glazner GW. Normalization of NF-κB activity in dorsal root ganglia neurons cultured from diabetic rats reverses neuropathy-linked markers of cellular pathology. Exp Neurol 2012; 241:169-78. [PMID: 23159890 DOI: 10.1016/j.expneurol.2012.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 10/05/2012] [Accepted: 11/06/2012] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS Dorsal root ganglia (DRG) sensory neurons cultured from 3 to 5 month streptozotocin (STZ)-induced diabetic rats exhibit structural and biochemical changes seen in peripheral nerve fibers in vivo, including axonal swellings, oxidative damage, reduced axonal sprouting, and decreased NF-κB activity. NF-κB is a transcription factor required by DRG neurons for survival and plasticity, and regulates transcription of antioxidant proteins (e.g. MnSOD). We hypothesized that the diabetes-induced decrease in NF-κB activity in DRG contributes to pathological phenomena observed in cultured DRG neurons from diabetic rats. METHODS NF-κB localization was assessed in intact DRG and neuron cultures using immunostaining. NF-κB activity was manipulated in sensory neuron cultures derived from age-matched normal or 3-5 month STZ-diabetic rats using pharmacological means and lentiviral expression of shRNA. The impact of diabetes and altered NF-κB activity on neuronal phenotype involved analysis of neurite outgrowth, neurite morphology, oxidative stress (lipid peroxidation) and expression of MnSOD. RESULTS STZ-induced diabetes caused a significant decrease in nuclear localization of NF-κB subunits p50 and c-rel, but no change in p65 in intact DRG. Inhibition of NF-κB in normal neuron cultures significantly increased axonal swellings and oxidative stress, and reduced both neurite outgrowth and expression of MnSOD. These phenomena mimicked markers of pathology in cultured DRG neurons from diabetic rats. Enhancement of NF-κB activity in cultured diabetic DRG neurons ameliorated the sub-optimal neurite outgrowth and MnSOD levels triggered by diabetes. Exogenous insulin enhanced nuclear localization of p50 and c-rel but not p65 in diabetic neuronal cultures. CONCLUSION/INTERPRETATION The diabetes-induced decrease of nuclear localization of NF-κB subunits p50 and c-rel in DRG contributes to development of in vitro markers of peripheral neuropathy, possibly through impaired mitochondrial ROS scavenging by deficient MnSOD.
Collapse
Affiliation(s)
- A Saleh
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Mahler GJ, Farrar EJ, Butcher JT. Inflammatory cytokines promote mesenchymal transformation in embryonic and adult valve endothelial cells. Arterioscler Thromb Vasc Biol 2012; 33:121-30. [PMID: 23104848 DOI: 10.1161/atvbaha.112.300504] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Inflammatory activation of valve endothelium is an early phase of aortic valve disease pathogenesis, but subsequent mechanisms are poorly understood. Adult valve endothelial cells retain the developmental ability to undergo endothelial-to-mesenchymal transformation (EndMT), but a biological role has not been established. Here, we test whether and how inflammatory cytokines (tumor necrosis factor-α and interleukin-6) regulate EndMT in embryonic and adult valve endothelium. METHODS AND RESULTS Using in vitro 3-dimensional collagen gel culture assays with primary cells, we determined that interleukin-6 and tumor necrosis factor-α induce EndMT and cell invasion in dose-dependent manners. Inflammatory-EndMT occurred through an Akt/nuclear factor-κB-dependent pathway in both adult and embryonic stages. In embryonic valves, inflammatory-EndMT required canonical transforming growth factor-β signaling through activin receptor-like kinases 2 and 5 to drive EndMT. In adult valve endothelium, however, inflammatory-induced EndMT still occurred when activin receptor-like kinases 2 and 5 signaling was blocked. Inflammatory receptor gene expression was significantly upregulated in vivo during embryonic valve maturation. Endothelial-derived mesenchymal cells expressing activated nuclear factor-κB were found distal to calcific lesions in diseased human aortic valves. CONCLUSIONS Inflammatory cytokine-induced EndMT in valve endothelium is present in both embryonic and adult stages, acting through Akt/nuclear factor-κB, but differently using transforming growth factor-β signaling. Molecular signatures of valve EndMT may be important diagnostic and therapeutic targets in early valve disease.
Collapse
Affiliation(s)
- Gretchen J Mahler
- Department of Bioengineering, Binghamton University, Binghamton, NY, USA
| | | | | |
Collapse
|
29
|
Saleh A, Roy Chowdhury SK, Smith DR, Balakrishnan S, Tessler L, Martens C, Morrow D, Schartner E, Frizzi KE, Calcutt NA, Fernyhough P. Ciliary neurotrophic factor activates NF-κB to enhance mitochondrial bioenergetics and prevent neuropathy in sensory neurons of streptozotocin-induced diabetic rodents. Neuropharmacology 2012; 65:65-73. [PMID: 23022047 DOI: 10.1016/j.neuropharm.2012.09.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/12/2012] [Accepted: 09/14/2012] [Indexed: 01/26/2023]
Abstract
Diabetes causes mitochondrial dysfunction in sensory neurons that may contribute to peripheral neuropathy. Ciliary neurotrophic factor (CNTF) promotes sensory neuron survival and axon regeneration and prevents axonal dwindling, nerve conduction deficits and thermal hypoalgesia in diabetic rats. In this study, we tested the hypothesis that CNTF protects sensory neuron function during diabetes through normalization of impaired mitochondrial bioenergetics. In addition, we investigated whether the NF-κB signal transduction pathway was mobilized by CNTF. Neurite outgrowth of sensory neurons derived from streptozotocin (STZ)-induced diabetic rats was reduced compared to neurons from control rats and exposure to CNTF for 24 h enhanced neurite outgrowth. CNTF also activated NF-κB, as assessed by Western blotting for the NF-κB p50 subunit and reporter assays for NF-κB promoter activity. Conversely, blockade of NF-κB signaling using SN50 peptide inhibited CNTF-mediated neurite outgrowth. Studies in mice with STZ-induced diabetes demonstrated that systemic therapy with CNTF prevented functional indices of peripheral neuropathy along with deficiencies in dorsal root ganglion (DRG) NF-κB p50 expression and DNA binding activity. DRG neurons derived from STZ-diabetic mice also exhibited deficiencies in maximal oxygen consumption rate and associated spare respiratory capacity that were corrected by exposure to CNTF for 24 h in an NF-κB-dependent manner. We propose that the ability of CNTF to enhance axon regeneration and protect peripheral nerve from structural and functional indices of diabetic peripheral neuropathy is associated with targeting of mitochondrial function, in part via NF-κB activation, and improvement of cellular bioenergetics.
Collapse
Affiliation(s)
- Ali Saleh
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Mincheva-Tasheva S, Soler RM. NF-κB signaling pathways: role in nervous system physiology and pathology. Neuroscientist 2012; 19:175-94. [PMID: 22785105 DOI: 10.1177/1073858412444007] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracellular pathways related to cell survival regulate neuronal physiology during development and neurodegenerative disorders. One of the pathways that have recently emerged with an important role in these processes is nuclear factor-κB (NF-κB). The activity of this pathway leads to the nuclear translocation of the NF-κB transcription factors and the regulation of anti-apoptotic gene expression. Different stimuli can activate the pathway through different intracellular cascades (canonical, non-canonical, and atypical), contributing to the translocation of specific dimers of the NF-κB transcription factors, and each of these dimers can regulate the transcription of different genes. Recent studies have shown that the activation of this pathway regulates opposite responses such as cell survival or neuronal degeneration. These apparent contradictory effects depend on conditions such as the pathway stimuli, the origin of the cells, or the cellular context. In the present review, the authors summarize these findings and discuss their significance with respect to survival or death in the nervous system.
Collapse
Affiliation(s)
- Stefka Mincheva-Tasheva
- Neuronal Signaling Unit, Dep. Ciencies Mediques Basiques, Facultat de Medicina, Universitat de Lleida-IRBLLEIDA, Lleida, Spain
| | | |
Collapse
|
31
|
Sarnico I, Branca C, Lanzillotta A, Porrini V, Benarese M, Spano PF, Pizzi M. NF-κB and epigenetic mechanisms as integrative regulators of brain resilience to anoxic stress. Brain Res 2012; 1476:203-10. [PMID: 22575713 DOI: 10.1016/j.brainres.2012.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/27/2012] [Accepted: 04/09/2012] [Indexed: 12/20/2022]
Abstract
Brain cells display an amazing ability to respond to several different types of environmental stimuli and integrate this response physiologically. Some of these responses can outlive the original stimulus by days, weeks or even longer. Long-lasting changes in both physiological and pathological conditions occurring in response to external stimuli are almost always mediated by changes in gene expression. To effect these changes, cells have developed an impressive repertoire of signaling systems designed to modulate the activity of numerous transcription factors and epigenetic mechanisms affecting the chromatin structure. Since its initial characterization in the nervous system, NF-κB has shown to respond to multiple signals and elicit pleiotropic activities suggesting that it may play a pivotal role in integration of different types of information within the brain. Ample evidence demonstrates that NF-κB factors are engaged in and necessary for neuronal development and synaptic plasticity, but they also regulate brain response to environmental noxae. By focusing on the complexity of NF-κB transcriptional activity in neuronal cell death, it emerged that the composition of NF-κB active dimers finely tunes the neuronal vulnerability to brain ischemia. Even though we are only beginning to understand the contribution of distinct NF-κB family members to the regulation of gene transcription in the brain, an additional level of regulation of NF-κB activity has emerged as operated by the epigenetic mechanisms modulating histone acetylation. We will discuss NF-κB and epigenetic mechanisms as integrative regulators of brain resilience to anoxic stress and useful drug targets for restoration of brain function. This article is part of a Special Issue entitled: Brain Integration.
Collapse
Affiliation(s)
- Ilenia Sarnico
- Department of Biomedical Sciences & Biotechnologies, University of Brescia and Istituto Nazionale di Neuroscienze, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Wagner-Golbs A, Luhmann HJ. Activity-dependent survival of developing neocortical neurons depends on PI3K signalling. J Neurochem 2011; 120:495-501. [PMID: 22118415 DOI: 10.1111/j.1471-4159.2011.07591.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Spontaneous electrical network activity plays a major role in the control of cell survival in the developing brain. Several intracellular pathways are implicated in transducing electrical activity into gene expression dependent and independent survival signals. These include activation of phosphatidylinositol 3-kinase (PI3K) and its downstream effector Akt, activation of Ras and subsequently MAPK/extracellular signal-regulated kinase (MEK) and extracellular signal-regulated kinase and signalling via calcium/calmodulin-dependent protein kinase (CaMK). In the present study, we analyzed the role of these pathways for the control of neuronal survival in different extracellular potassium concentrations ([K(+) ](ex) ). Organotypic neocortical slice cultures prepared from newborn mice were kept in 5.3, 8.0 and 25.0mM [K(+) ](ex) and treated with specific inhibitors of PI3K, MEK1, CaMKK and a broad spectrum CaMK inhibitor. After 6h of incubation, slices were immunostained for activated caspase 3 (a-caspase 3) and the number of apoptotic cells was quantified by computer based analysis. We found that in 5.3 and 8.0mM [K(+) ](ex) only PI3K was important for neuronal survival. When [K(+) ](ex) was raised to 25.0mM, a concentration above the depolarization block, we found no influence of PI3K on neuronal survival. Our data demonstrate that only the PI3K pathway, and not the MEK1, CaMKK or CaMKs pathway, plays a central role in the regulation of activity-dependent neuronal survival in the developing cerebral cortex.
Collapse
Affiliation(s)
- Antje Wagner-Golbs
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, Mainz, Germany
| | | |
Collapse
|
33
|
Saleh A, Smith DR, Balakrishnan S, Dunn L, Martens C, Tweed CW, Fernyhough P. Tumor necrosis factor-α elevates neurite outgrowth through an NF-κB-dependent pathway in cultured adult sensory neurons: Diminished expression in diabetes may contribute to sensory neuropathy. Brain Res 2011; 1423:87-95. [PMID: 21985959 DOI: 10.1016/j.brainres.2011.09.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 09/14/2011] [Accepted: 09/15/2011] [Indexed: 11/24/2022]
Abstract
The presence of a proinflammatory environment in the sensory neuron axis in diabetes was tested by measuring levels of proinflammatory cytokines in lumbar dorsal root ganglia (DRG) and peripheral nerve from age matched control and streptozotocin (STZ)-induced diabetic rats. The levels of tumor necrosis factor-α (TNFα) and other cytokines were diminished in lumbar DRG from diabetic animals. Consequently, we tested the hypothesis that TNFα modulated axonal plasticity in adult sensory neurons and posited that impairments in this signal transduction pathway may underlie degeneration in diabetic sensory neuropathy. Cultured adult rat sensory neurons were grown under defined conditions and TNFα caused a dose-dependent 2-fold (P<0.05) elevation in neurite outgrowth. Neurons derived from 3 to 5month STZ-induced diabetic rats exhibited significantly reduced levels of neurite outgrowth in response to TNFα. TNFα enhanced NF-κB activity as assessed using Western blotting and plasmid reporter technology. Blockade of TNFα-induction of NF-κB activation caused inhibition of neurite outgrowth in cultured neurons. Immunofluorescent staining for NF-κB subunit p50 within neuronal nuclei revealed that medium to large diameter neurons were most susceptible to NF-κB inhibition and was associated with decreased neurite outgrowth. The results demonstrating reduced cytokine expression in DRG confirm that diabetic sensory neuropathy does not involve a neuroinflammatory component at this stage of the disease in experimental animal models. In addition, it is hypothesized that reduced TNFα expression in the DRG and possibly associated deficits in anterograde transport may contribute to impaired collatoral sprouting and regeneration in target tissue in type 1 diabetes.
Collapse
Affiliation(s)
- Ali Saleh
- Division of Neurodegenerative Disorders, St Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | | | | | | | | | | | | |
Collapse
|
34
|
Burugula B, Ganesh BS, Chintala SK. Curcumin attenuates staurosporine-mediated death of retinal ganglion cells. Invest Ophthalmol Vis Sci 2011; 52:4263-73. [PMID: 21498608 DOI: 10.1167/iovs.10-7103] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Staurosporine (SS) causes retinal ganglion cell (RGC) death in vivo, but the underlying mechanisms have been unclear. Since previous studies on RGC-5 cells indicated that SS induces cell death by elevating proteases, this study was undertaken to investigate whether SS induces RGC loss by elevating proteases in the retina, and curcumin prevents SS-mediated death of RGCs. METHODS Transformed mouse retinal ganglion-like cells (RGC-5) were treated with 2.0 μM SS and various doses of curcumin. Two optimal doses of SS (12.5 and 100 nM) and curcumin (2.5 and 10 μM) were injected into the vitreous of C57BL/6 mice. Matrix metalloproteinase (MMP)-9, tissue plasminogen activator (tPA), and urokinase plasminogen activator (uPA) activities were assessed by zymography assays. Viability of RGC-5 cells was assessed by MTT assays. RGC and amacrine cell loss in vivo was assessed by immunostaining with Brn3a and ChAT antibodies, respectively. Frozen retinal cross sections were immunostained for nuclear factor-κB (NF-κB). RESULTS Staurosporine induced uPA and tPA levels in RGC-5 cells, and MMP-9, uPA, and tPA levels in the retinas and promoted the death of RGC-5 cells in vitro and RGCs and amacrine cells in vivo. In contrast, curcumin attenuated RGC and amacrine cell loss, despite elevated levels of proteases. An NF-κB inhibitory peptide reversed curcumin-mediated protective effect on RGC-5 cells, but did not inhibit protease levels. Curcumin did not inhibit protease levels in vivo, but attenuated RGC and amacrine cell loss by restoring NF-κB expression. CONCLUSIONS The results show that curcumin attenuates RGC and amacrine cell death despite elevated levels of proteases and raises the possibility that it may be used as a plausible adjuvant therapeutic agent to prevent the loss of these cells in retinal degenerative conditions.
Collapse
|
35
|
Garcia N, Tomàs M, Santafe MM, Lanuza MA, Besalduch N, Tomàs J. Blocking p75NTR receptors alters polyinnervationz of neuromuscular synapses during development. J Neurosci Res 2011; 89:1331-41. [DOI: 10.1002/jnr.22620] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 12/20/2010] [Accepted: 01/13/2011] [Indexed: 01/17/2023]
|
36
|
Inhibition of NF-κB signaling commits resveratrol-treated medulloblastoma cells to apoptosis without neuronal differentiation. J Neurooncol 2010; 104:169-77. [PMID: 21161674 DOI: 10.1007/s11060-010-0496-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 12/03/2010] [Indexed: 01/05/2023]
Abstract
Resveratrol promotes differentiation and apoptosis of medulloblastoma cells by suppressing STAT3 signaling and a range of cancer-associated gene expression. However, Bcl-2, a common target of STAT3 and NF-κB signaling, is distinctly up-regulated in resveratrol-treated medulloblastoma cells, indicating potential effects of NF-κB in Bcl-2 expression and anti-medulloblastoma efficiency of resveratrol. To clarify this point, the status of NF-κB signaling and the consequence of NF-κB inhibition in UW228-2 and UW228-3 medulloblastoma cells without and with resveratrol treatment were evaluated by several experimental approaches. The results revealed that resveratrol activated NF-κB signaling in both cell lines at the 4-h treatment point, and the treated cells sequentially exhibited Bcl-2 up-regulation, neuronal-like phenotype with synaptophisin expression, and, eventually, apoptosis. Pyrrolidine dithiocarbamate (PDTC) treatment inhibited NF-κB activation and Bcl-2 expression and committed resveratrol-treated cells to apoptosis at the 8-h time point without the step of neuron-oriented differentiation. On the other hand, a single 50 μg/ml lipopolysaccharide (LPS) treatment activated NF-κB signaling accompanied with sustained proliferation and neuron-like differentiation. Tissue microarray-based immunohistochemical staining showed significantly different (P < 0.001) p65 nuclear translocation between the neurons of tumor-surrounding cerebella (10/10; 100%) and medulloblastoma tissues (20/117; 17.09%). Additionally, synaptophysin production was found in 83.64% of p65-positive and in 40.35% of p65-negative medulloblastoma cases. Our in-vitro and in-vivo results thus demonstrate the dual effects of NF-κB signaling on medulloblastoma cells by delaying resveratrol-induced apoptosis by up-regulating Bcl-2 expression or by involvement in neuronal-like differentiation in the absence of resveratrol. Therefore, appropriate inhibition of NF-κB activation may enhance the anti-medulloblastoma efficacy of resveratrol.
Collapse
|
37
|
Veuger SJ, Durkacz BW. Persistence of unrepaired DNA double strand breaks caused by inhibition of ATM does not lead to radio-sensitisation in the absence of NF-κB activation. DNA Repair (Amst) 2010; 10:235-44. [PMID: 21144805 DOI: 10.1016/j.dnarep.2010.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 11/08/2010] [Accepted: 11/10/2010] [Indexed: 12/21/2022]
Abstract
The stress-inducible transcription complex NF-κB induces the transcription of genes that regulate proliferation and apoptosis. Constitutively activated NF-κB is common in breast cancers, and contributes to malignant progression and therapeutic resistance. Ataxia telangiectasia mutated (ATM) is a key regulator of the cellular response to DNA double strand breaks (DSBs), and recent reports have demonstrated that ATM is required for the activation of NF-κB following DNA damage. We investigated the role of ATM in the NF-κB signalling cascade induced by ionising radiation (IR) in breast cancer cell lines using KU55933, a novel and specific inhibitor of ATM. KU55933 suppressed IR-induced IκBα degradation, p50/p65 nuclear translocation and binding to kB consensus sequences. KU55933 also suppressed transcription of an NF-κB dependent reporter gene and inhibited IR-induced DSB repair as assessed by the neutral Comet assay. KU55933 sensitised cells to IR, with a concurrent increase in caspase 3 activity. Importantly, KU55933 sensitised IKKβ(+/+) and p65(+/+), but not IKKβ(-/-) or p65(-/-), mouse embryonic fibroblasts to IR, despite the equivalent inhibitory effects of KU55933 on DSB repair in both the proficient and the deficient cell lines. P65 siRNA had no effect on DSB repair in either breast cancer cell line. When combined with KU55933, DSB repair was inhibited to the same extent as KU55933 alone in both breast cancer cell lines. P65 siRNA alone sensitised both cell lines to IR. A combination of p65 siRNA and KU55933 resulted in no further sensitisation compared to either one alone. Taken together these data support the hypothesis that KU55933-mediated radio-sensitisation is solely a consequence of its inhibition of NF-κB activation. We conclude that radiotherapy deploying ATM inhibitors may be particularly advantageous in tumours where NF-κB is constitutively activated.
Collapse
Affiliation(s)
- Stephany J Veuger
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| | | |
Collapse
|
38
|
Peng H, Sola A, Moore J, Wen T. Caspase inhibition by cardiotrophin-1 prevents neuronal death in vivo and in vitro. J Neurosci Res 2010; 88:1041-51. [PMID: 19859964 DOI: 10.1002/jnr.22269] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Our previous studies showed that cardiotrophin-1 (CT-1), a cytokine in the interleukin-6 family, protected the developing rat brain against focal cerebral ischemia (FCI) in vivo and prevented cortical neuron death in vitro. However, the mechanisms by which CT-1 prevents neuronal death are not clearly understood. This in vivo study focused on whether CT-1 treatment prevented FCI-induced brain injuries in the postnatal day 7 (P7) rat through modulating activation of the initiator caspase-8 (C-8) and the downstream effector caspase-3 (C-3). FCI caused a significant increase in expressions of cleaved C-8 and C-3 and, meanwhile, a significant decrease in expression of microtubule-associated protein-2 (MAP2) in the left ischemic cortex of the P7 rat brain after FCI. Exogenous treatment of CT-1 significantly reduced the expression of cleaved C-8 or C-3 and attenuated the decline in MAP2 expression in the ischemic cortex from 12 to 24 hr after FCI. Subsequent in vitro experiments demonstrated that CT-1 treatment inhibited sodium nitroprusside (SNP)-induced activation of C-8 and C-3 and loss of MAP2-positive neurons in cortical neuron cultures. More importantly, CT-1 activated several pathways, including Janus kinase 2, signal transducers and activators of transcription 3, nuclear factor kappa B, mitogen-activated protein kinase (MAPK), and MAPK kinase in the cultures exposed to SNP. This is the first suggestion that CT-1 prevents neuronal injury in the developing central nervous system possibly through mediating multiple signal pathways, inhibiting activation of C-8 and C-3.
Collapse
Affiliation(s)
- Hui Peng
- Atlantic Neonatal Research Institute, MidAtlantic Neonatology Associates, and Atlantic Health, Morristown, New Jersey, USA
| | | | | | | |
Collapse
|
39
|
Abstract
The transcription factor NF-kappaB has diverse functions in the nervous system, depending on the cellular context. NF-kappaB is constitutively activated in glutamatergic neurons. Knockout of p65 or inhibition of neuronal NF-kappaB by super-repressor IkappaB resulted in the loss of neuroprotection and defects in learning and memory. Similarly, p50-/- mice have a lower learning ability and are sensitive to neurotoxins. Activated NF-kappaB can be transported retrogradely from activated synapses to the nucleus to translate short-term processes to long-term changes such as axon growth, which is important for long-term memory. In glia, NF-kappaB is inducible and regulates inflammatory processes that exacerbate diseases such as autoimmune encephalomyelitis, ischemia, and Alzheimer's disease. In summary, inhibition of NF-kappaB in glia might ameliorate disease, whereas activation in neurons might enhance memory. This review focuses on results produced by the analysis of genetic models.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, Universitätsstr. 25, D-33501 Bielefeld.
| | | |
Collapse
|
40
|
Kajiya M, Shiba H, Fujita T, Takeda K, Uchida Y, Kawaguchi H, Kitagawa M, Takata T, Kurihara H. Brain-derived neurotrophic factor protects cementoblasts from serum starvation-induced cell death. J Cell Physiol 2009; 221:696-706. [DOI: 10.1002/jcp.21909] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
41
|
Yang RH, Strong JA, Zhang JM. NF-kappaB mediated enhancement of potassium currents by the chemokine CXCL1/growth related oncogene in small diameter rat sensory neurons. Mol Pain 2009; 5:26. [PMID: 19476648 PMCID: PMC2698898 DOI: 10.1186/1744-8069-5-26] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 05/28/2009] [Indexed: 01/24/2023] Open
Abstract
Background Inflammatory processes play important roles in both neuropathic and inflammatory pain states, but the effects of inflammation per se within the sensory ganglia are not well understood. The cytokine growth-related oncogene (GRO/KC; CXCL1) shows strong, rapid upregulation in dorsal root ganglion (DRG) in both nerve injury and inflammatory pain models. We examined the direct effects of GRO/KC on small diameter DRG neurons, which are predominantly nociceptive. Whole cell voltage clamp technique was used to measure voltage-activated potassium (K) currents in acutely cultured adult rat small diameter sensory neurons. Fluorescently labeled isolectin B4 (IB4) was used to classify cells as IB4-positive or IB4-negative. Results In IB4-negative neurons, voltage-activated K current densities of both transient and sustained components were increased after overnight incubation with GRO/KC (1.5 nM), without marked changes in voltage dependence or kinetics. The average values for the slow and fast decay time constants at 20 mV were unchanged by GRO/KC. The amplitude of the fast inactivating component increased significantly with no large shifts in the voltage dependence of inactivation. The increase in K currents was completely blocked by co-incubation with protein synthesis inhibitor cycloheximide (CHX) or NF-κB inhibitors pyrrolidine dithiocarbamate (PDTC) or quinazoline (6-Amino-4-(4-phenoxypheny lethylamino;QNZ). In contrast, the voltage-activated K current of IB4-positive neurons was unchanged by GRO/KC. GRO/KC incubation caused no significant changes in the expression level of eight selected voltage-gated K channel genes in quantitative PCR analysis. Conclusion The results suggest that GRO/KC has important effects in inflammatory processes via its direct actions on sensory neurons, and that activation of NF-κB is involved in the GRO/KC-induced enhancement of K currents.
Collapse
Affiliation(s)
- Rui-Hua Yang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0531, USA.
| | | | | |
Collapse
|
42
|
Pizzi M, Sarnico I, Lanzillotta A, Battistin L, Spano P. Post-ischemic brain damage: NF-kappaB dimer heterogeneity as a molecular determinant of neuron vulnerability. FEBS J 2009; 276:27-35. [PMID: 19087197 DOI: 10.1111/j.1742-4658.2008.06767.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nuclear factor-kappaB (NF-kappaB) has been proposed to serve a dual function as a regulator of neuron survival in pathological conditions associated with neurodegeneration. NF-kappaB is a transcription family of factors comprising five different proteins, namely p50, RelA/p65, c-Rel, RelB and p52, which can combine differently to form active dimers in response to external stimuli. Recent research shows that diverse NF-kappaB dimers lead to cell death or cell survival in neurons exposed to ischemic injury. While the p50/p65 dimer participates in the pathogenesis of post-ischemic injury by inducing pro-apoptotic gene expression, c-Rel-containing dimers increase neuron resistance to ischemia by inducing anti-apoptotic gene transcription. We present, in this report, the latest findings and consider the therapeutic potential of targeting different NF-kappaB dimers to limit ischemia-associated neurodegeneration.
Collapse
Affiliation(s)
- Marina Pizzi
- Division of Pharmacology and Experimental Therapeutics, Department of Biomedical Sciences and Biotechnologies, School of Medicine, University of Brescia, Italy.
| | | | | | | | | |
Collapse
|
43
|
Sarnico I, Lanzillotta A, Benarese M, Alghisi M, Baiguera C, Battistin L, Spano P, Pizzi M. NF-kappaB dimers in the regulation of neuronal survival. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 85:351-62. [PMID: 19607980 DOI: 10.1016/s0074-7742(09)85024-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nuclear factor-kappaB (NF-kappaB) is a dimeric transcription factor composed of five members, p50, RelA/p65, c-Rel, RelB, and p52 that can diversely combine to form the active transcriptional dimer. NF-kappaB controls the expression of genes that regulate a broad range of biological processes in the central nervous system such as synaptic plasticity, neurogenesis, and differentiation. Although NF-kappaB is essential for neuron survival and its activation may protect neurons against oxidative-stresses or ischemia-induced neurodegeneration, NF-kappaB activation can contribute to inflammatory reactions and apoptotic cell death after brain injury and stroke. It was proposed that the death or survival of neurons might depend on the cell type and the timing of NF-kappaB activation. We here discuss recent evidence suggesting that within the same neuronal cell, activation of diverse NF-kappaB dimers drives opposite effects on neuronal survival. Unbalanced activation of NF-kappaB p50/RelA dimer over c-Rel-containing complexes contributes to cell death secondary to the ischemic insult. While p50/RelA acts as transcriptional inducer of Bcl-2 family proapoptotic Bim and Noxa genes, c-Rel dimers specifically promote transcription of antiapototic Bcl-xL gene. Changes in the nuclear content of c-Rel dimers strongly affect the threshold of neuron vulnerability to ischemic insult and agents, likewise leptin, activating a NF-kappaB/c-Rel-dependent transcription elicit neuroprotection in animal models of brain ischemia.
Collapse
Affiliation(s)
- Ilenia Sarnico
- Division of Pharmacology and Experimental Therapeutics, Department of Biomedical Sciences and Biotechnologies, School of Medicine, University of Brescia, Brescia 25123, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Maalouf M, Rho JM, Mattson MP. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. ACTA ACUST UNITED AC 2008; 59:293-315. [PMID: 18845187 DOI: 10.1016/j.brainresrev.2008.09.002] [Citation(s) in RCA: 401] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 09/11/2008] [Accepted: 09/12/2008] [Indexed: 12/18/2022]
Abstract
Both calorie restriction and the ketogenic diet possess broad therapeutic potential in various clinical settings and in various animal models of neurological disease. Following calorie restriction or consumption of a ketogenic diet, there is notable improvement in mitochondrial function, a decrease in the expression of apoptotic and inflammatory mediators and an increase in the activity of neurotrophic factors. However, despite these intriguing observations, it is not yet clear which of these mechanisms account for the observed neuroprotective effects. Furthermore, limited compliance and concern for adverse effects hamper efforts at broader clinical application. Recent research aimed at identifying compounds that can reproduce, at least partially, the neuroprotective effects of the diets with less demanding changes to food intake suggests that ketone bodies might represent an appropriate candidate. Ketone bodies protect neurons against multiple types of neuronal injury and are associated with mitochondrial effects similar to those described during calorie restriction or ketogenic diet treatment. The present review summarizes the neuroprotective effects of calorie restriction, of the ketogenic diet and of ketone bodies, and compares their putative mechanisms of action.
Collapse
Affiliation(s)
- Marwan Maalouf
- Department of Neurobiology, David Geffen School of Medicine, University of California, 63-323 CH5, Box 951763, Los Angeles, CA 90095-1763, USA.
| | | | | |
Collapse
|
45
|
Toma HS, Murina AT, Areaux RG, Neumann DM, Bhattacharjee PS, Foster TP, Kaufman HE, Hill JM. Ocular HSV-1 latency, reactivation and recurrent disease. Semin Ophthalmol 2008; 23:249-73. [PMID: 18584563 DOI: 10.1080/08820530802111085] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ocular infection with HSV-1 continues to be a serious clinical problem despite the availability of effective antivirals. Primary infection with HSV-1 can involve ocular and adenaxial sites and can manifest as blepharitis, conjunctivitis, or corneal epithelial keratitis. After initial ocular infection, HSV-1 can establish latent infection in the trigeminal ganglia for the lifetime of the host. During latency, the viral genome is retained in the neuron without producing viral proteins. However, abundant transcription occurs at the region encoding the latency-associated transcript, which may play significant roles in the maintenance of latency as well as neuronal reactivation. Many host and viral factors are involved in HSV-1 reactivation from latency. HSV-1 DNA is shed into tears and saliva of most adults, but in most cases this does not result in lesions. Recurrent disease occurs as HSV-1 is carried by anterograde transport to the original site of infection, or any other site innervated by the latently infected ganglia, and can reinfect the ocular tissues. Recurrent corneal disease can lead to corneal scarring, thinning, stromal opacity and neovascularization and, eventually, blindness. In spite of intensive antiviral and anti-inflammatory therapy, a significant percentage of patients do not respond to chemotherapy for herpetic necrotizing stromal keratitis. Therefore, the development of therapies that would reduce asymptomatic viral shedding and lower the risks of recurrent disease and transmission of the virus is key to decreasing the morbidity of ocular herpetic disease. This review will highlight basic HSV-1 virology, and will compare the animal models of latency, reactivation, and recurrent ocular disease to the current clinical data.
Collapse
Affiliation(s)
- Hassanain S Toma
- Louisiana State University Health Sciences Center, New Orleans, LA 70112-2234, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
García-Bueno B, Caso JR, Leza JC. Stress as a neuroinflammatory condition in brain: Damaging and protective mechanisms. Neurosci Biobehav Rev 2008; 32:1136-51. [DOI: 10.1016/j.neubiorev.2008.04.001] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 04/02/2008] [Accepted: 04/02/2008] [Indexed: 01/07/2023]
|
47
|
Doherty GH, Oldreive C, Harvey J. Neuroprotective actions of leptin on central and peripheral neurons in vitro. Neuroscience 2008; 154:1297-307. [PMID: 18550292 DOI: 10.1016/j.neuroscience.2008.04.052] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 04/23/2008] [Accepted: 04/25/2008] [Indexed: 11/16/2022]
Abstract
Neuronal cell death and its regulation have been extensively studied as an essential process of both neurodevelopment and neurodegenerative conditions. However it is not clear how circulating hormones influence such processes. Therefore we aimed to determine whether the anti-obesity hormone leptin could promote the survival of murine central and peripheral neurons in vitro. Thus we established primary neuronal cultures of dopaminergic midbrain neurons and trigeminal sensory neurons and induced cell death via either toxic insult or growth factor withdrawal. We demonstrate that leptin promotes the survival of developing peripheral and central neurons via activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3-kinase)/Akt/nuclear factor kappa B (NF-kappaB) -dependent signaling cascades. Specifically, leptin protects dopaminergic midbrain neurons from the apoptotic stimuli, tumor necrosis factor alpha (TNF-alpha) and 6-hydroxydopamine (6-OHDA). In addition, it promotes the survival of postnatal, but not embryonic, trigeminal sensory neurons following neurotrophin withdrawal. Our data reveal a novel neuroprotective role for leptin in the peripheral nervous system while expanding on the known anti-apoptotic role of leptin in the CNS. These findings have important implications for our understanding of neuronal viability.
Collapse
Affiliation(s)
- G H Doherty
- School of Biology, University of St Andrews, West Burn Lane, St Andrews, Fife, UK.
| | | | | |
Collapse
|
48
|
Yang ZF, Lau CK, Ngai P, Lam SP, Ho DW, Poon RTP, Fan ST. Cardiotrophin-1 enhances regeneration of cirrhotic liver remnant after hepatectomy through promotion of angiogenesis and cell proliferation. Liver Int 2008; 28:622-31. [PMID: 18312290 DOI: 10.1111/j.1478-3231.2008.01687.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND/AIM Hepatic resection is not applicable to a certain proportion of hepatocellular carcinoma patients owing to an insufficient liver function reserve. The present study was designed to investigate the effects of cardiotrophin-1 (CT-1) on improving the function of CCl(4)-induced cirrhotic liver remnant after major hepatectomy. METHODS CT-1 was administered to rats after hepatectomy according to different protocols. RESULTS A double-dose CT-1 protocol improved liver function, enlarged the volume of liver remnant, upregulated the expression of von Willebrand factor and increased the number of BrdU(+) or Ki-67(+) hepatocytes. Administration of CT-1 enhanced the expression of nuclear factor-kappaB (P65), vascular endothelial growth factor (VEGF), CyclinD1 and p42/44 in the liver remnant. However, the effects of CT-1 were blocked by a VEGF receptor blocker, PTK787. Although the expression of gp130, a receptor of CT-1, was downregulated in the diseased hepatocytes isolated from the cirrhotic liver, CT-1 could still stimulate the cell proliferation. CT-1 administration enhanced the expression of P65 and VEGF in the diseased hepatocytes, but the augmented P65 and VEGF expression was blocked by PTK787 administration. CONCLUSION Short-term administration of CT-1 could improve the function of cirrhotic liver remnant and stimulate liver regeneration through promotion of angiogenesis and cell proliferation.
Collapse
Affiliation(s)
- Zhen Fan Yang
- Department of Surgery, Centre for Cancer Research, Queen Mary Hospital, University of Hong Kong Medical Centre, Pokfulam, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Siddiq A, Aminova LR, Ratan RR. Prolyl 4-hydroxylase activity-responsive transcription factors: from hydroxylation to gene expression and neuroprotection. FRONTIERS IN BIOSCIENCE : A JOURNAL AND VIRTUAL LIBRARY 2008; 13:2875-87. [PMID: 17981760 PMCID: PMC2570096 DOI: 10.2741/2892] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Most homeostatic processes including gene transcription occur as a result of deviations in physiological tone that threatens the survival of the organism. A prototypical homeostatic stress response includes changes in gene expression following alterations in oxygen, iron or 2-oxoglutarate levels. Each of these cofactors plays an important role in cellular metabolism. Accordingly, a family of enzymes known as the Prolyl 4-hydroxylase (PHD) enzymes are a group of dioxygenases that have evolved to sense changes in 2-oxoglutarate, oxygen and iron via changes in enzyme activity. Indeed, PHDs are a part of an established oxygen sensor system that regulates transcriptional regulation of hypoxia/stress-regulated genes and thus are an important component of events leading to cellular rescue from oxygen, iron or 2-oxoglutarate deprivations. The ability of PHD activity to regulate homeostatic responses to oxygen, iron or 2-oxoglutarate metabolism has led to the development of small molecule inhibitors of the PHDs as a strategy for activating or augmenting cellular stress responses. These small molecules are proving effective in preclinical models of stroke and Parkinson's disease. However the precise protective pathways engaged by PHD inhibition are only beginning to be defined. In the current review, we summarize the role of iron, 2-oxoglutarate and oxygen in the PHD catalyzed hydroxylation reaction and provide a brief discussion of some of the transcription factors that play an effective role in neuroprotection against oxidative stress as a result of changes in PHD activity.
Collapse
Affiliation(s)
- Ambreena Siddiq
- Burke Medical Research Institute, White Plains, New York 10605, USA.
| | | | | |
Collapse
|
50
|
Kairisalo M, Korhonen L, Blomgren K, Lindholm D. X-linked inhibitor of apoptosis protein increases mitochondrial antioxidants through NF-κB activation. Biochem Biophys Res Commun 2007; 364:138-44. [DOI: 10.1016/j.bbrc.2007.09.115] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 09/26/2007] [Indexed: 01/04/2023]
|