1
|
Fedorova EV, Chernomorets IY, Fedorov DA, Arkhipov VI. Delayed treatment with TGF-β1 associated neuroprotection in trimethyltin-induced hippocampal neurodegeneration. Neurosci Lett 2025; 852:138182. [PMID: 40049360 DOI: 10.1016/j.neulet.2025.138182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/09/2025]
Abstract
In experiments conducted on Wistar rats, the effects of the multifunctional cytokine TGF-β1 were investigated using a neurodegeneration model induced by a single injection of the neurotoxicant trimethyltin chloride (TMT). Animals in the experimental group received intranasal administration of TGF-β1 on days 7 and 9 following TMT injection. Behavioral tests were performed to assess cognitive function, and three weeks after TMT administration, hippocampal morphology was analyzed using Nissl staining. Additionally, the state of microglia was evaluated through immunohistochemical labeling of IBA1. The results revealed that exogenous TGF-β1 significantly modulated the progression of hippocampal neurodegeneration. In the passive avoidance test, TGF-β1 ameliorated TMT-induced long-term memory impairment and promoted neuronal preservation in the CA1 region of the hippocampus, although no such effect was observed in the CA3 and CA4 regions. Furthermore, TGF-β1 treatment reduced microglial activation levels in the hippocampal CA1 region compared to animals treated with TMT alone. These findings suggest that the multifunctional cytokine TGF-β1 exerts a neuroprotective effect in the context of ongoing neurodegeneration when delivered intranasally to the brain. The cytokine's ability to regulate microglial activity appears to contribute, at least in part, to its protective properties.
Collapse
Affiliation(s)
| | | | - Dmitry A Fedorov
- Institute of Theoretical and Experimental Biophysics, Pushchino, Russia
| | | |
Collapse
|
2
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 DOI: 10.1016/j.mcn.2025.104003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
3
|
Gonzalez D, Cuenca X, Allende ML. Knockdown of tgfb1a partially improves ALS phenotype in a transient zebrafish model. Front Cell Neurosci 2024; 18:1384085. [PMID: 38644973 PMCID: PMC11032012 DOI: 10.3389/fncel.2024.1384085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/15/2024] [Indexed: 04/23/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) corresponds to a neurodegenerative disorder marked by the progressive degeneration of both upper and lower motor neurons located in the brain, brainstem, and spinal cord. ALS can be broadly categorized into two main types: sporadic ALS (sALS), which constitutes approximately 90% of all cases, and familial ALS (fALS), which represents the remaining 10% of cases. Transforming growth factor type-β (TGF-β) is a cytokine involved in various cellular processes and pathological contexts, including inflammation and fibrosis. Elevated levels of TGF-β have been observed in the plasma and cerebrospinal fluid (CSF) of both ALS patients and mouse models. In this perspective, we explore the impact of the TGF-β signaling pathway using a transient zebrafish model for ALS. Our findings reveal that the knockdown of tgfb1a lead to a partial prevention of motor axon abnormalities and locomotor deficits in a transient ALS zebrafish model at 48 h post-fertilization (hpf). In this context, we delve into the proposed distinct roles of TGF-β in the progression of ALS. Indeed, some evidence suggests a dual role for TGF-β in ALS progression. Initially, it seems to exert a neuroprotective effect in the early stages, but paradoxically, it may contribute to disease progression in later stages. Consequently, we suggest that the TGF-β signaling pathway emerges as an attractive therapeutic target for treating ALS. Nevertheless, further research is crucial to comprehensively understand the nuanced role of TGF-β in the pathological context.
Collapse
Affiliation(s)
- David Gonzalez
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
- Escuela de Terapia Ocupacional, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Xiomara Cuenca
- Escuela de Terapia Ocupacional, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
| | - Miguel L. Allende
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
4
|
Supriya M, Christopher R, Devi BI, Bhat DI, Shukla D, Kalpana SR. Altered MicroRNA Expression in Intracranial Aneurysmal Tissues: Possible Role in TGF-β Signaling Pathway. Cell Mol Neurobiol 2022; 42:2393-2405. [PMID: 34185228 PMCID: PMC11421602 DOI: 10.1007/s10571-021-01121-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
The molecular mechanisms behind the rupture of intracranial aneurysms remain obscure. MiRNAs are key regulators of a wide array of biological processes altering protein synthesis by binding to target mRNAs. However, variations in miRNA levels in ruptured aneurysmal wall have not been completely examined. We hypothesized that altered miRNA signature in aneurysmal tissues could potentially provide insight into aneurysm pathophysiology. Using a high-throughput miRNA microarray screening approach, we compared the miRNA expression pattern in aneurysm tissues obtained during surgery from patients with aneurysmal subarachnoid hemorrhage (aSAH) with control tissues (GEO accession number GSE161870). We found that the expression of 70 miRNAs was altered. Expressions of the top 10 miRNA were validated, by qRT-PCR and results were correlated with clinical characteristics of aSAH patients. The level of 10 miRNAs (miR-24-3p, miR-26b-5p, miR-27b-3p, miR-125b-5p, miR-143-3p, miR-145-5p, miR-193a-3p, miR-199a-5p, miR-365a-3p/365b-3p, and miR-497-5p) was significantly decreased in patients compared to controls. Expression of miR-125b-5p, miR-143-3p and miR-199a-5p was significantly decreased in patients with poor prognosis and vasospasm. The target genes of few miRNAs were enriched in Transforming growth factor-beta (TGF-β) and Mitogen-activated protein kinases (MAPK) pathways. We found significant negative correlation between the miRNA and mRNA expression (TGF-β1, TGF-β2, SMAD family member 2 (SMAD2), SMAD family member 4 (SMAD4), MAPK1 and MAPK3) in aneurysm tissues. We suggest that miR-26b, miR-199a, miR-497and miR-365, could target multiple genes in TGF-β and MAPK signaling cascades to influence inflammatory processes, extracellular matrix and vascular smooth muscle cell degradation and apoptosis, and ultimately cause vessel wall degradation and rupture.
Collapse
Affiliation(s)
- Manjunath Supriya
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, 560029, India
| | - Rita Christopher
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, Karnataka, 560029, India.
| | - Bhagavatula Indira Devi
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bengaluru, 560029, India
| | - Dhananjaya Ishwar Bhat
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bengaluru, 560029, India
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bengaluru, 560029, India
| | - Saligrama Ramegowda Kalpana
- Department of Pathology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, 560069, India
| |
Collapse
|
5
|
Zhu H, Zhang Y, Zhong Y, Ye Y, Hu X, Gu L, Xiong X. Inflammation-Mediated Angiogenesis in Ischemic Stroke. Front Cell Neurosci 2021; 15:652647. [PMID: 33967696 PMCID: PMC8096981 DOI: 10.3389/fncel.2021.652647] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is the leading cause of disability and mortality in the world, but the pathogenesis of ischemic stroke (IS) is not completely clear and treatments are limited. Mounting evidence indicate that neovascularization is a critical defensive reaction to hypoxia that modulates the process of long-term neurologic recovery after IS. Angiogenesis is a complex process in which the original endothelial cells in blood vessels are differentiated, proliferated, migrated, and finally remolded into new blood vessels. Many immune cells and cytokines, as well as growth factors, are directly or indirectly involved in the regulation of angiogenesis. Inflammatory cells can affect endothelial cell proliferation, migration, and activation by secreting a variety of cytokines via various inflammation-relative signaling pathways and thus participate in the process of angiogenesis. However, the mechanism of inflammation-mediated angiogenesis has not been fully elucidated. Hence, this review aimed to discuss the mechanism of inflammation-mediated angiogenesis in IS and to provide new ideas for clinical treatment of IS.
Collapse
Affiliation(s)
- Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyao Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Transcriptome Sequencing in the Preoptic Region of Rat Dams Reveals a Role of Androgen Receptor in the Control of Maternal Behavior. Int J Mol Sci 2021; 22:ijms22041517. [PMID: 33546359 PMCID: PMC7913516 DOI: 10.3390/ijms22041517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/27/2021] [Indexed: 01/17/2023] Open
Abstract
(1) Background: Preoptic region of hypothalamus is responsible to control maternal behavior, which was hypothesized to be associated with gene expressional changes. (2) Methods: Transcriptome sequencing was first applied in the preoptic region of rat dams in comparison to a control group of mothers whose pups were taken away immediately after parturition and did not exhibit caring behavior 10 days later. (3) Results: Differentially expressed genes were found and validated by quantitative RT-PCR, among them NACHT and WD repeat domain containing 1 (Nwd1) is known to control androgen receptor (AR) protein levels. The distribution of Nwd1 mRNA and AR was similar in the preoptic area. Therefore, we focused on this steroid hormone receptor and found its reduced protein level in rat dams. To establish the function of AR in maternal behavior, its antagonist was administered intracerebroventricularly into mother rats and increased pup-directed behavior of the animals. (4) Conclusions: AR levels are suppressed in the preoptic area of mothers possibly mediated by altered Nwd1 expression in order to allow sustained high-level care for the pups. Thus, our study first implicated the AR in the control of maternal behaviors.
Collapse
|
7
|
Galbiati M, Crippa V, Rusmini P, Cristofani R, Messi E, Piccolella M, Tedesco B, Ferrari V, Casarotto E, Chierichetti M, Poletti A. Multiple Roles of Transforming Growth Factor Beta in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2020; 21:ijms21124291. [PMID: 32560258 PMCID: PMC7352289 DOI: 10.3390/ijms21124291] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor beta (TGFB) is a pleiotropic cytokine known to be dysregulated in many neurodegenerative disorders and particularly in amyotrophic lateral sclerosis (ALS). This motor neuronal disease is non-cell autonomous, as it affects not only motor neurons but also the surrounding glial cells, and the target skeletal muscle fibers. Here, we analyze the multiple roles of TGFB in these cell types, and how TGFB signaling is altered in ALS tissues. Data reported support a crucial involvement of TGFB in the etiology and progression of ALS, leading us to hypothesize that an imbalance of TGFB signaling, diminished at the pre-symptomatic stage and then increased with time, could be linked to ALS progression. A reduced stimulation of the TGFB pathway at the beginning of disease blocks its neuroprotective effects and promotes glutamate excitotoxicity. At later disease stages, the persistent activation of the TGFB pathway promotes an excessive microglial activation and strengthens muscular dysfunction. The therapeutic potential of TGFB is discussed, in order to foster new approaches to treat ALS.
Collapse
|
8
|
Abstract
Astrocytes, initially described as merely support cells, are now known as a heterogeneous population of cells actively involved in a variety of biological functions such as: neuronal migration and differentiation; regulation of cerebral blood flow; metabolic control of extracellular potassium concentration; and modulation of synapse formation and elimination; among others. Cerebellar glial cells have been shown to play a significant role in proliferation, differentiation, migration, and synaptogenesis. However, less evidence is available about the role of neuron-astrocyte interactions during cerebellar development and their impact on diseases of the cerebellum. In this review, we will focus on the mechanisms underlying cellular interactions, specifically neuron-astrocyte interactions, during cerebellar development, function, and disease. We will discuss how cerebellar glia, astrocytes, and Bergmann glia play a fundamental role in several steps of cerebellar development, such as granule cell migration, axonal growth, neuronal differentiation, and synapse formation, and in diseases associated with the cerebellum. We will focus on how astrocytes and thyroid hormones impact cerebellar development. Furthermore, we will provide evidence of how growth factors secreted by glial cells, such as epidermal growth factor and transforming growth factors, control cerebellar organogenesis. Finally, we will argue that glia are a key mediator of cerebellar development and that identification of molecules and pathways involved in neuron-glia interactions may contribute to a better understanding of cerebellar development and associated disorders.
Collapse
|
9
|
Abdel-Rahman RF, Alqasoumi SI, Ogaly HA, Abd-Elsalam RM, El-Banna HA, Soliman GA. Propolis ameliorates cerebral injury in focal cerebral ischemia/reperfusion (I/R) rat model via upregulation of TGF-β1. Saudi Pharm J 2019; 28:116-126. [PMID: 31920438 PMCID: PMC6950965 DOI: 10.1016/j.jsps.2019.11.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
Neuroprotective impact of transforming growth factor β1 (TGF-β1) is increasingly recognized in different brain injuries. Propolis exhibits a broad spectrum of biological and pharmacological properties including neuroprotective action. The objective of the investigation was to explore the involvement of TGF-β1 signaling in the neuroprotective mechanism of propolis in I/R rats. In this study, focal cerebral ischemia model was built by middle cerebral artery occlusion (MCAO) for 2 h followed by reperfusion. The investigation was carried out on 48 rats that were arranged into four groups (n = 12): the sham group, I/R control group, I/R + propolis (50 mg/kg) group and I/R + propolis (100 mg/kg) group. The results revealed that propolis preserved rats against neuronal injury induced by cerebral I/R. It significantly reduced neurological deficit scores and improved motor coordination and locomotor activity in I/R rats. Propolis antagonized the damage induced by cerebral I/R through suppression of malondialdehyde (MDA) and elevation of reduced glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), brain-derived neurotropic factor (BDNF) and dopamine levels in the brain homogenates of I/R rats. Other ameliorations were also observed based on reduction of neurodegeneration and histological alterations in the brain tissues. These results also proposed that the neuroprotective effect of propolis might be related to upregulation of TGF-β1 and suppressed matrix metallopeptidase-9 (MMP9) mRNA expression.
Collapse
Affiliation(s)
| | - Saleh I Alqasoumi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hanan A Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Biochemistry, College of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Reham M Abd-Elsalam
- Department of Pathology, College of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hossny A El-Banna
- Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Gamal A Soliman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.,Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
10
|
Barna J, Dimén D, Puska G, Kovács D, Csikós V, Oláh S, Udvari EB, Pál G, Dobolyi Á. Complement component 1q subcomponent binding protein in the brain of the rat. Sci Rep 2019; 9:4597. [PMID: 30872665 PMCID: PMC6418184 DOI: 10.1038/s41598-019-40788-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/19/2019] [Indexed: 12/17/2022] Open
Abstract
Complement component 1q subcomponent binding protein (C1qbp) is a multifunctional protein involved in immune response, energy homeostasis of cells as a plasma membrane receptor, and a nuclear, cytoplasmic or mitochondrial protein. Recent reports suggested its neuronal function, too, possibly in axon maintenance, synaptic function, and neuroplasticity. Therefore, we addressed to identify C1qbp in the rat brain using in situ hybridization histochemistry and immunolabelling at light and electron microscopic level. C1qbp has a topographical distribution in the brain established by the same pattern of C1qbp mRNA-expressing and protein-containing neurons with the highest abundance in the cerebral cortex, anterodorsal thalamic nucleus, hypothalamic paraventricular (PVN) and arcuate nuclei, spinal trigeminal nucleus. Double labelling of C1qbp with the neuronal marker NeuN, with the astrocyte marker S100, and the microglia marker Iba1 demonstrated the presence of C1qbp in neurons but not in glial cells in the normal brain, while C1qbp appeared in microglia following their activation induced by focal ischemic lesion. Only restricted neurons expressed C1qbp, for example, in the PVN, magnocellular neurons selectively contained C1qbp. Further double labelling by using the mitochondria marker Idh3a antibody suggested the mitochondrial localization of C1qbp in the brain, confirmed by correlated light and electron microscopy at 3 different brain regions. Post-embedding immunoelectron microscopy also suggested uneven C1qbp content of mitochondria in different brain areas but also heterogeneity within single neurons. These data suggest a specific function of C1qbp in the brain related to mitochondria, such as the regulation of local energy supply in neuronal cells.
Collapse
Affiliation(s)
- János Barna
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Diána Dimén
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Gina Puska
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Dávid Kovács
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Vivien Csikós
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Szilvia Oláh
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Edina B Udvari
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
| | - Gabriella Pál
- Hungarian Defence Forces Military Hospital, Budapest, Hungary
| | - Árpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
11
|
Mendes NF, Gaspar JM, Lima-Júnior JC, Donato J, Velloso LA, Araújo EP. TGF-β1 down-regulation in the mediobasal hypothalamus attenuates hypothalamic inflammation and protects against diet-induced obesity. Metabolism 2018; 85:171-182. [PMID: 29660453 DOI: 10.1016/j.metabol.2018.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/01/2018] [Accepted: 04/10/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The consumption of large amounts of dietary fats induces hypothalamic inflammation and impairs the function of the melanocortin system, leading to a defective regulation of caloric intake and whole-body energy expenditure. In mice fed a high-fat diet (HFD), TGF-β1 expression was increased and NF-κB signaling was activated in proopiomelanocortin neurons, which plays an important role in the obesity-associated hypothalamic inflammation scenario. However, whether excessive hypothalamic TGF-β1 impairs energy homeostasis remains unclear. OBJECTIVES We aimed to investigate the role of diet-induced hypothalamic TGF-β1 on inflammation and whole-body energy homeostasis. METHODS A TGF-β1 inhibitory lentiviral shRNA particle was stereotaxically injected bilaterally in the arcuate nucleus (ARC) of C57BL/6 mice fed a HFD. We assessed changes in body mass and adiposity, food intake, inflammatory markers, and the function of energy and glucose metabolism. RESULTS TGF-β1 down-regulation in the ARC-attenuated body-mass gain, reduced fat-mass accumulation, decreased hypothalamic inflammatory markers, and protected against HFD-induced lipohypertrophy of brown adipose tissue. In addition, the inhibition of hypothalamic TGF-β1 increased the locomotor activity and improved whole-body lipid metabolism, which attenuated hepatic fat accumulation and serum triglyceride levels. No changes were observed in food intake and glucose homeostasis. CONCLUSION Hypothalamic TGF-β1 down-regulation attenuates hypothalamic inflammation and improves energy metabolism, resulting in lower body-mass gain and lower fat-mass accumulation, which protects mice from the development of obesity. Our data suggest that modulation of hypothalamic TGF-β1 expression might be an effective strategy to treat obesity.
Collapse
Affiliation(s)
- Natália F Mendes
- School of Nursing, State University of Campinas (UNICAMP), Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil
| | - Joana M Gaspar
- Faculty of Medical Sciences, State University of Campinas (UNICAMP), Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil
| | - José C Lima-Júnior
- Faculty of Medical Sciences, State University of Campinas (UNICAMP), Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Lício A Velloso
- Faculty of Medical Sciences, State University of Campinas (UNICAMP), Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil
| | - Eliana P Araújo
- School of Nursing, State University of Campinas (UNICAMP), Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil.
| |
Collapse
|
12
|
Liu R, Liao X, Li X, Wei H, Liang Q, Zhang Z, Yin M, Zeng X, Liang Z, Hu C. Expression profiles of long noncoding RNAs and mRNAs in post-cardiac arrest rat brains. Mol Med Rep 2018; 17:6413-6424. [PMID: 29512756 PMCID: PMC5928618 DOI: 10.3892/mmr.2018.8703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/01/2018] [Indexed: 01/17/2023] Open
Abstract
To investigate long noncoding (lnc)-RNA and mRNA expression profiles in post-cardiac arrest (CA) brains, an external transthoracic electrical current was applied for 8 min to induce CA (the CA group). A total of 4 rats received sham-operations and served as the blank control (BC) group. Upon return of spontaneous circulation (ROSC), lncRNA and mRNA expression in the rat cerebral cortex was assayed with high-throughput Agilent lncRNA and mRNA microarrays. In total, 37 lncRNAs were upregulated and 21 lncRNAs were downregulated in the CA group, and 258 mRNA transcripts were differentially expressed with 177 mRNAs upregulated and 81 mRNAs downregulated in the CA group. The differentially expressed lncRNAs in the CA group were co-expressed with thousands of mRNAs. The differentially expressed lncRNAs could be clustered into >100 signaling pathways and processes according to Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes analyses. The most common predicted functions involved metabolic pathways, protein synthesis, transport and degradation during CA-ROSC. CA-ROSC led to significant alterations in cerebral lncRNA and mRNA expression profiles. Thus, lncRNA-mRNA network interactions have the potential to regulate vital metabolic pathways and processes involved in CA-ROSC.
Collapse
Affiliation(s)
- Rong Liu
- Department of Emergency, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Xiaoxing Liao
- Department of Emergency, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xin Li
- Department of Emergency, Guangdong Provincial People's Hospital, Guangzhou, Guangdong 510080, P.R. China
| | - Hongyan Wei
- Department of Emergency, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qing Liang
- Department of Emergency, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Zuopeng Zhang
- Department of Emergency, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Meixian Yin
- Department of Emergency, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoyun Zeng
- Department of Emergency, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zijing Liang
- Department of Emergency, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| | - Chunlin Hu
- Department of Emergency, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
13
|
Tapella L, Cerruti M, Biocotino I, Stevano A, Rocchio F, Canonico PL, Grilli M, Genazzani AA, Lim D. TGF-β2 and TGF-β3 from cultured β-amyloid-treated or 3xTg-AD-derived astrocytes may mediate astrocyte-neuron communication. Eur J Neurosci 2018; 47:211-221. [DOI: 10.1111/ejn.13819] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Laura Tapella
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Matteo Cerruti
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Isabella Biocotino
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Alessio Stevano
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Francesca Rocchio
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences; Università degli Studi del Piemonte Orientale “Amedeo Avogadro”; 28100 Novara Italy
| |
Collapse
|
14
|
Cerebrovascular Gene Expression in Spontaneously Hypertensive Rats After Transient Middle Cerebral Artery Occlusion. Neuroscience 2017; 367:219-232. [PMID: 29102661 DOI: 10.1016/j.neuroscience.2017.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/22/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022]
Abstract
Hypertension is a major risk factor for stroke, which is one of the leading global causes of death. In the search for new and effective therapeutic targets in stroke research, we need to understand the influence of hypertension in the vasculature following stroke. We used Affymetrix whole-transcriptome expression profiling as a tool to address gene expression differences between the occluded and non-occluded middle cerebral arteries (MCAs) from spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto (WKY) rats after transient middle cerebral artery occlusion (tMCAO), to provide clues about the pathological mechanisms set in play after stroke. Verified by quantitative PCR, expression of Ccl2, Edn1, Tgfβ2, Olr1 and Serpine1 was significantly increased in the occluded compared to non-occluded MCAs from both SHRs and WKY rats. Additionally, expression of Mmp9, Icam1, Hif1α and Timp1 was increased in the occluded compared to non-occluded MCAs isolated from WKY rats. In comparison between occluded MCAs from SHRs versus occluded MCAs from WKY rats, expression of Ccl2, Olr1 and Serpine1 was significantly increased in SHR MCAs. However, the opposite was observed regarding expression of Edn1. Thus these data suggest that Ccl2, Edn1, Tgfβ2, Olr1 and Serpine1 may be possible mediators of the vascular changes in the occluded MCAs from both SHRs and WKY rats after tMCAO. The aforementioned genes possess biological functions that are consistent with early stroke injuries. In conclusion, these genes may be potential targets in future strategies for acute stroke treatments that can be used in patients with and without hypertension.
Collapse
|
15
|
Nakajima T, Hata R, Kunieda Y, Kondo T. Distribution of Smad mRNA and proteins in the rat brain. J Chem Neuroanat 2017; 90:11-39. [PMID: 29196107 DOI: 10.1016/j.jchemneu.2017.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/27/2017] [Accepted: 11/25/2017] [Indexed: 01/27/2023]
Abstract
Smad proteins are known to transduce the action of TGF-β superfamily proteins including TGF-βs, activins, and bone morphogenetic proteins (BMPs). In this study, we examined the expression of Smad1, -2, -3, -4, -5, and -8 mRNA in the rat brain by means of RT-PCR and in situ hybridization (ISH). In addition, we examined the nuclear accumulation of Smad1, -2, -3, -5, and -8 proteins after intracerebroventricular injection of TGF-β1, activin A, or BMP6 with immunohistochemistry to investigate whether TGF-β, activin, and/or BMP activate Smads in the rat brain. RT-PCR analysis revealed that Smad1, -2, -3, -4, -5, and -8 mRNA was expressed in the brain and that the Smad3 and Smad8 mRNA differed in the expression level between brain regions. For example, there were high levels of expression of Smad3 mRNA in the cerebral cortex, caudate putamen/globus pallidus, and cerebellum, but low levels in the thalamus and midbrain. Expression of Smad8 mRNA was higher in the midbrain, cerebellum, and pons/medulla oblongata in comparison to the olfactory bulb, cerebral cortex, caudate putamen/globus pallidus, hippocampus/dentate gyrus, and thalamus. ISH signals for Smad1 mRNA were widely detected in the brain except for a small number of regions including the olfactory tubercle, posterior region of hypothalamus, and cerebellar nuclei. ISH signals for Smad2 mRNA were abundantly observed in several brain regions including the olfactory bulb, piriform cortex, basal ganglia, cingulate cortex, epithalamus, including the pineal gland and medial habenular nuclei, hypothalamus, inferior colliculi of the midbrain, and some nuclei in the pons, cerebellar cortex, and choroid plexus. ISH signals for Smad3 mRNA were also abundantly observed in several brain regions. Especially strong signals for Smad3 mRNA were observed in the olfactory tubercle, piriform cortex, basal ganglia, dentate gyrus, and cingulate cortex. ISH signals for Smad5 and Smad8 mRNA were restricted to a small number of brain regions, the signal intensity of which was weak. ISH signals for Smad4 mRNA were detected in all regions examined. Intracerebroventricular injection of activin A induced nuclear accumulation of Smad2 and Smad3 immunoreactivity in neurons. In contrast, intracerebroventricular injection of TGF-β1 or BMP6 did not induce nuclear accumulation of the immunoreactivity for any Smad in neurons. These results suggest that activin-Smad signaling plays an important role in brain homeostasis.
Collapse
Affiliation(s)
- Takayuki Nakajima
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan.
| | - Ryusuke Hata
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan
| | - Yuji Kunieda
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan
| | - Tomohiro Kondo
- Department of Integrated Structural Biosciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
16
|
Yang Z, Chen Y, Zhang Y, Li R, Dong C. The role of pro-/anti-inflammation imbalance in Aβ42 accumulation of rat brain co-exposed to fine particle matter and sulfur dioxide. Toxicol Mech Methods 2017; 27:568-574. [DOI: 10.1080/15376516.2017.1337256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zhenhua Yang
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Yunzhu Chen
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Yuexia Zhang
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| |
Collapse
|
17
|
Abstract
While stroke research represents the primary interface between circulation and brain research, the hemostasis system also carries a pivotal role in the mechanism of vascular brain injury. The complex interrelated events triggered by the energy crisis have a specific spatial and temporal pattern arching from the initial damage to the final events of brain repair. The complexity of the pathophysiology make it difficult to model this disease, therefore it is challenging to find appropriate therapeutic targets. The ever-persistent antagonism between the positive results of drug candidates in the experimental stroke models and the failures of the clinical trials prompts changes in the research strategy, especially in the field of potential neuroprotective therapies. System biology approach could initiate new directions in the future for both preclinical and clinical research. Incentive methods aimed at anti-apoptosis mechanisms and the augmentation of post-ischemic brain repair could benefit the facts, that these processes can be targeted much longer following the cell-necrosis in the hyper-acute phase. Sequential monitoring of candidate genes and proteins responsible for stroke progression and post-stroke repair seems to be useful both in therapeutic target-identification, and in clinical testing. Understanding the mechanism behind the effect of selegiline and other drugs capable of activating the anti-apoptotic gene expression could help to find new approaches to enhance the regenerative potential in the remodeling of neuronal and microvascular networks.
Collapse
Affiliation(s)
- Z Nagy
- Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Városmajor Street 68, 1122, Hungary; National Institute of Clinical Neurosciences, Budapest, Amerikai Street 57, 1145, Hungary.
| | - S Nardai
- Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Városmajor Street 68, 1122, Hungary; National Institute of Clinical Neurosciences, Budapest, Amerikai Street 57, 1145, Hungary
| |
Collapse
|
18
|
Lékó AH, Cservenák M, Szabó ÉR, Hanics J, Alpár A, Dobolyi Á. Insulin-like growth factor I and its binding protein-3 are regulators of lactation and maternal responsiveness. Sci Rep 2017; 7:3396. [PMID: 28611445 PMCID: PMC5469809 DOI: 10.1038/s41598-017-03645-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/02/2017] [Indexed: 12/29/2022] Open
Abstract
Adaptation to motherhood includes maternal behaviour and lactation during the postpartum period. The major organizing centres of maternal behaviour and lactation are located in the hypothalamic medial preoptic area (MPOA) and the arcuate nucleus, respectively. Insulin-like growth factor I (IGF-I) is an effector of the growth hormone axis; however, its function in the brain is largely unexplored. We identified increased maternal IGF binding protein-3 (IGFBP-3) expression in preoptic rat microarray data and confirmed it by RT-PCR. In situ hybridization histochemistry showed markedly elevated IGFBP-3 expression in the MPOA and the arcuate nucleus in rat dams. Prolonged intracerebroventricular injection of IGF-I or antagonism of brain IGFBP-3 with an inhibitor (NBI-31772) using osmotic minipumps increased pup retrieval time, suggesting reduced maternal motivation. Suckling-induced prolactin release and pup weight gain were also suppressed by IGF-I, suggesting reduced lactation. In addition, IGF-I-induced tyrosine hydroxylase expression and its specific phosphorylation in tuberoinfundibular dopaminergic neurons suppress prolactin secretion. Thus, IGF-I may inhibit both behavioural and lactational alterations in mothers. Neurons in the MPOA and arcuate nuclei express IGFBP-3 during the postpartum period to neutralize IGF-I effects. IGFBP-3 can prevent the blockade of maternal behaviour and lactation exerted by IGF-I, suggesting a novel modulatory mechanism underlying the behavioural and hormonal effects during central maternal adaptations.
Collapse
Affiliation(s)
- András H Lékó
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary.,MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, 1117, Hungary
| | - Melinda Cservenák
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary.,MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, 1117, Hungary
| | - Éva Rebeka Szabó
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - János Hanics
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, Budapest, Hungary.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Árpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary. .,MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, 1117, Hungary. .,MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, 1117, Hungary.
| |
Collapse
|
19
|
Zhu H, Gui Q, Hui X, Wang X, Jiang J, Ding L, Sun X, Wang Y, Chen H. TGF-β1/Smad3 Signaling Pathway Suppresses Cell Apoptosis in Cerebral Ischemic Stroke Rats. Med Sci Monit 2017; 23:366-376. [PMID: 28110342 PMCID: PMC5282965 DOI: 10.12659/msm.899195] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We desired to observe the changes of transforming growth factor-β1/drosophila mothers against decapentaplegic protein (TGF-β1/Smad3) signaling pathway in the hippocampus region of cerebral ischemic stroke rats so that the effects of this pathway on nerve cells can be investigated. MATERIAL AND METHODS The ischemic stroke models were built by middle cerebral artery occlusion (MCAO) in vivo and oxygen-glucose deprivation (OGD) in vitro. TGF-β1 and TGF-β1 inhibitors were injected into rat models while TGF-β1, TGF-β1 siRNA, Smad3, and Smad3 siRNA were transfected into cells. Infarct sizes were measured using triphenyltetrazolium chloride (TTC) staining, while the apoptosis rate of cells were calculated by Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) staining. Levels of TGF-β1, Smad3, and Bcl-2 were examined by real-time polymerase chain reaction (RT-PCR), immunohistochemical, and Western blot analysis. RESULTS The expressions of TGF-β1/Smad3 signal pathway were significantly increased in both model rats and BV2 cells, whereas the expression of Bcl-2 was down-regulated (P<0.05). The TGF-β1/Smad3 signal pathway exhibited protective effects, including the down-regulation of infarction size in cerebral tissues and the down-regulation of apoptosis rate of BV2 cells by increasing the expression of Bcl-2 (P<0.05). In addition, these effects could be antagonized by the corresponding inhibitors and siRNA (P<0.05). CONCLUSIONS The TGF-β1/Smad3 signaling pathway was up-regulated once cerebral ischemic stroke was simulated. TGF-β1 may activate the expression of Bcl-2 via Smad3 to suppress the apoptosis of neurons.
Collapse
Affiliation(s)
- Haiping Zhu
- Department of Neurosurgery, The First People's Hospital of Changshou City, Changshou, Jiangsu, China (mainland)
| | - Qunfeng Gui
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| | - Xiaobo Hui
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaodong Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Jian Jiang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Xiaoyang Sun
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Yanping Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China (mainland)
| | - Huaqun Chen
- Department of Neurosurgery, Yancheng Third People's Hospital, The affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu, China (mainland)
| |
Collapse
|
20
|
Ruskyte K, Liutkevicienė R, Vilkeviciute A, Vaitkiene P, Valiulytė I, Glebauskiene B, Kriauciuniene L, Zaliuniene D. MMP-14 and TGFβ-1 methylation in pituitary adenomas. Oncol Lett 2016; 12:3013-3017. [PMID: 27698891 DOI: 10.3892/ol.2016.4919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/27/2016] [Indexed: 02/06/2023] Open
Abstract
Pituitary adenoma (PA) is one of the most common abnormalities in the sellar region. Despite the fact that PA is a benign monoclonal neoplasm, it can cause serious complications, including ophthalmological, neurological and endocrinological abnormalities. Currently, the causes that increase the progression of tumors are unknown. Epigenetic silencing of the matrix metalloproteinase-14 (MMP-14) and transforming growth factor beta-1 (TGFβ-1) genes may be associated with the development of PA, since these genes are important in the processes of tumor metastasis and angiogenesis. The purpose of the present study was to determine if the methylation status of the MMP-14 and TGFβ-1 promoters is associated with PA development. In the present study, 120 tissue samples of PA were used. The methylation status of the MMP-14 and TGFβ-1 promoters was investigated by methylation specific-polymerase chain reaction. Statistical analysis was conducted to investigate the associations between the methylation status, age and gender of PA patients, PA tumoral activity, recurrence and invasiveness. The MMP-14 gene was methylated in 30.00% (17/56 functioning and 19/64 non-functioning) of patients with PA, while the TGFβ-1 gene was methylated in 13.33% (9/56 functioning and 7/64 non-functioning) of patients with PA. It was also observed that promoter methylation of MMP-14 correlated with the male gender (58.8 vs. 35.7%, P=0.022), while unmethylated (non-silenced) MMP-14 correlated with the female gender (64.3 vs. 41.7%, P=0.027). Associations between the promoter methylation status of the MMP-14 and TGFβ-1 genes and PA functioning or recurrence were not identified. The present study reveals that silencing of the MMP-14 gene correlates with patients' gender. However, MMP-14 and TGFβ-1 promoter methylation cannot be considered as a prognostic marker in PAs.
Collapse
Affiliation(s)
- Kornelija Ruskyte
- Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Rasa Liutkevicienė
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania; Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Paulina Vaitkiene
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Indre Valiulytė
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Brigita Glebauskiene
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Loresa Kriauciuniene
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania; Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| | - Dalia Zaliuniene
- Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, 50009 Kaunas, Lithuania
| |
Collapse
|
21
|
Araujo APB, Diniz LP, Eller CM, de Matos BG, Martinez R, Gomes FCA. Effects of Transforming Growth Factor Beta 1 in Cerebellar Development: Role in Synapse Formation. Front Cell Neurosci 2016; 10:104. [PMID: 27199658 PMCID: PMC4846658 DOI: 10.3389/fncel.2016.00104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/08/2016] [Indexed: 11/13/2022] Open
Abstract
Granule cells (GC) are the most numerous glutamatergic neurons in the cerebellar cortex and represent almost half of the neurons of the central nervous system. Despite recent advances, the mechanisms of how the glutamatergic synapses are formed in the cerebellum remain unclear. Among the TGF-β family, TGF-beta 1 (TGF-β1) has been described as a synaptogenic molecule in invertebrates and in the vertebrate peripheral nervous system. A recent paper from our group demonstrated that TGF-β1 increases the excitatory synapse formation in cortical neurons. Here, we investigated the role of TGF-β1 in glutamatergic cerebellar neurons. We showed that the expression profile of TGF-β1 and its receptor, TβRII, in the cerebellum is consistent with a role in synapse formation in vitro and in vivo. It is low in the early postnatal days (P1–P9), increases after postnatal day 12 (P12), and remains high until adulthood (P30). We also found that granule neurons express the TGF-β receptor mRNA and protein, suggesting that they may be responsive to the synaptogenic effect of TGF-β1. Treatment of granular cell cultures with TGF-β1 increased the number of glutamatergic excitatory synapses by 100%, as shown by immunocytochemistry assays for presynaptic (synaptophysin) and post-synaptic (PSD-95) proteins. This effect was dependent on TβRI activation because addition of a pharmacological inhibitor of TGF-β, SB-431542, impaired the formation of synapses between granular neurons. Together, these findings suggest that TGF-β1 has a specific key function in the cerebellum through regulation of excitatory synapse formation between granule neurons.
Collapse
Affiliation(s)
- Ana P B Araujo
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Luan P Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Cristiane M Eller
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Beatriz G de Matos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Rodrigo Martinez
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil; Faculdade de Medicina/Departamento de Cirurgia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Flávia C A Gomes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Zepeda R, Contreras V, Pissani C, Stack K, Vargas M, Owen GI, Lazo OM, Bronfman FC. Venlafaxine treatment after endothelin-1-induced cortical stroke modulates growth factor expression and reduces tissue damage in rats. Neuropharmacology 2016; 107:131-145. [PMID: 26965219 DOI: 10.1016/j.neuropharm.2016.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/03/2016] [Accepted: 03/05/2016] [Indexed: 02/02/2023]
Abstract
Neuromodulators, such as antidepressants, may contribute to neuroprotection by modulating growth factor expression to exert anti-inflammatory effects and to support neuronal plasticity after stroke. Our objective was to study whether early treatment with venlafaxine, a serotonin-norepinephrine reuptake inhibitor, modulates growth factor expression and positively contributes to reducing the volume of infarcted brain tissue resulting in increased functional recovery. We studied the expression of BDNF, FGF2 and TGF-β1 by examining their mRNA and protein levels and cellular distribution using quantitative confocal microscopy at 5 days after venlafaxine treatment in control and infarcted brains. Venlafaxine treatment did not change the expression of these growth factors in sham rats. In infarcted rats, BDNF mRNA and protein levels were reduced, while the mRNA and protein levels of FGF2 and TGF-β1 were increased. Venlafaxine treatment potentiated all of the changes that were induced by cortical stroke alone. In particular, increased levels of FGF2 and TGF-β1 were observed in astrocytes at 5 days after stroke induction, and these increases were correlated with decreased astrogliosis (measured by GFAP) and increased synaptophysin immunostaining at twenty-one days after stroke in venlafaxine-treated rats. Finally, we show that venlafaxine reduced infarct volume after stroke resulting in increased functional recovery, which was measured using ladder rung motor tests, at 21 days after stroke. Our results indicate that the early oral administration of venlafaxine positively contributes to neuroprotection during the acute and late events that follow stroke.
Collapse
Affiliation(s)
- Rodrigo Zepeda
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina Contreras
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Pissani
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine Stack
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Vargas
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar M Lazo
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Francisca C Bronfman
- MINREB and Center for Aging and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
23
|
Haas SJP, Zhou X, Machado V, Wree A, Krieglstein K, Spittau B. Expression of Tgfβ1 and Inflammatory Markers in the 6-hydroxydopamine Mouse Model of Parkinson's Disease. Front Mol Neurosci 2016; 9:7. [PMID: 26869879 PMCID: PMC4737885 DOI: 10.3389/fnmol.2016.00007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/14/2016] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by loss of midbrain dopaminergic (mDA) neurons in the substantia nigra (SN). Microglia-mediated neuroinflammation has been described as a common hallmark of PD and is believed to further trigger the progression of neurodegenerative events. Injections of 6-hydroxydopamine (6-OHDA) are widely used to induce degeneration of mDA neurons in rodents as an attempt to mimic PD and to study neurodegeneration, neuroinflammation as well as potential therapeutic approaches. In the present study, we addressed microglia and astroglia reactivity in the SN and the caudatoputamen (CPu) after 6-OHDA injections into the medial forebrain bundle (MFB), and further analyzed the temporal and spatial expression patterns of pro-inflammatory and anti-inflammatory markers in this mouse model of PD. We provide evidence that activated microglia as well as neurons in the lesioned SN and CPu express Transforming growth factor β1 (Tgfβ1), which overlaps with the downregulation of pro-inflammatory markers Tnfα, and iNos, and upregulation of anti-inflammatory markers Ym1 and Arg1. Taken together, the data presented in this study suggest an important role for Tgfβ1 as a lesion-associated factor that might be involved in regulating microglia activation states in the 6-OHDA mouse model of PD in order to prevent degeneration of uninjured neurons by microglia-mediated release of neurotoxic factors such as Tnfα and nitric oxide (NO).
Collapse
Affiliation(s)
| | - Xiaolai Zhou
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell UniversityIthaca, NY, USA
| | - Venissa Machado
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-UniversityFreiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-UniversityFreiburg, Germany; Faculty of Biology, Albert-Ludwigs-UniversityFreiburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center Rostock, Germany
| | - Kerstin Krieglstein
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| | - Björn Spittau
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Germany
| |
Collapse
|
24
|
Liu Z, Chen HQ, Huang Y, Qiu YH, Peng YP. Transforming growth factor-β1 acts via TβR-I on microglia to protect against MPP(+)-induced dopaminergic neuronal loss. Brain Behav Immun 2016; 51:131-143. [PMID: 26254549 DOI: 10.1016/j.bbi.2015.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/07/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is associated with pathogenesis of Parkinson's disease (PD), a neurodegenerative disorder characterized by a progressive loss of dopaminergic (DAergic) neurons within the substantia nigra. Transforming growth factor (TGF)-β1 exerts anti-inflammatory and neuroprotective properties. However, it is unclear if microglia are required for TGF-β1 neuroprotection in PD. Here we used both shRNA and pharmacologic inhibition to determine the role of microglial TGF-β receptor (TβR)-I and its downstream signaling pathways in 1-methyl-4-phenylpyridinium (MPP(+))-induced DAergic neuronal toxicity. As expected, MPP(+) reduced the number of tyrosine hydroxylase (TH)-immunoreactive cells in ventral mesencephalic cell cultures. We found that MPP(+) activated microglia as determined by an upregulation in expression of CD11b and inducible nitric oxide synthase (iNOS), an increase in expression and secretion of tumor necrosis factor (TNF)-α and interleukin (IL)-1β, and a decrease in expression and secretion of the neurotrophic factor, insulin-like growth factor (IGF)-1. Pretreatment with TGF-β1 significantly inhibited all these changes caused by MPP(+). Expression of microglial TβR-I was upregulated by TGF-β1. Silencing of the TβR-I gene in microglia abolished both the neuroprotective and anti-inflammatory properties of TGF-β1. TGF-β1 increased microglial p38 MAPK and Akt phosphorylation, both of which were blocked by the p38 inhibitor SB203580 and the PI3K inhibitor LY294002, respectively. Pretreatment of microglia with either SB203580 or LY294002 impaired the ability of TGF-β1 to inhibit MPP(+)-induced DAergic neuronal loss and microglial activation. These findings establish that TGF-β1 activates TβR-I and its downstream p38 MAPK and PI3K-Akt signaling pathways in microglia to protect against DAergic neuronal loss that characterizes in PD.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Hui-Qiao Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
25
|
Spittau B. Transforming growth factor β1-mediated anti-inflammation slows progression of midbrain dopaminergic neurodegeneration in Parkinson's disease? Neural Regen Res 2015; 10:1578-80. [PMID: 26692847 PMCID: PMC4660743 DOI: 10.4103/1673-5374.165228] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Björn Spittau
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
26
|
Fernández-Klett F, Priller J. The fibrotic scar in neurological disorders. Brain Pathol 2015; 24:404-13. [PMID: 24946078 DOI: 10.1111/bpa.12162] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 05/26/2014] [Indexed: 01/18/2023] Open
Abstract
Tissue fibrosis, or scar formation, is a common response to damage in most organs of the body. The central nervous system (CNS) is special in that fibrogenic cells are restricted to vascular and meningeal niches. However, disruption of the blood-brain barrier and inflammation can unleash stromal cells and trigger scar formation. Astroglia segregate from the inflammatory lesion core, and the so-called "glial scar" composed of hypertrophic astrocytes seals off the intact neural tissue from damage. In the lesion core, a second type of "fibrotic scar" develops, which is sensitive to inflammatory mediators. Genetic fate mapping studies suggest that pericytes and perivascular fibroblasts are activated, but other precursor cells may also be involved in generating a transient fibrous extracellular matrix in the CNS. The stromal cells sense inflammation and attract immune cells, which in turn drive myofibroblast transdifferentiation. We believe that the fibrotic scar represents a major barrier to CNS regeneration. Targeting of fibrosis may therefore prove to be a valuable therapeutic strategy for neurological disorders such as stroke, spinal cord injury and multiple sclerosis.
Collapse
Affiliation(s)
- Francisco Fernández-Klett
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
27
|
Lu F, Xu Y, Wang T. Advance of
transforming growth factor beta
in traumatic brain injury. IBRAIN 2015. [DOI: 10.1002/j.2769-2795.2015.tb00003.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Fan Lu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yang Xu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ting‐Hua Wang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
28
|
Eskandari MR, Abdolmaleky HM, Zhou JR, Thiagalingam S. Reduced Risk of Cancer in Schizophrenia, a Bridge Toward Etio-Pathology and Therapy of Both Diseases. EPIGENETICS TERRITORY AND CANCER 2015:137-166. [DOI: 10.1007/978-94-017-9639-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Shen WX, Chen JH, Lu JH, Peng YP, Qiu YH. TGF-β1 protection against Aβ1-42-induced neuroinflammation and neurodegeneration in rats. Int J Mol Sci 2014; 15:22092-108. [PMID: 25470026 PMCID: PMC4284696 DOI: 10.3390/ijms151222092] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/31/2014] [Accepted: 11/14/2014] [Indexed: 01/08/2023] Open
Abstract
Transforming growth factor (TGF)-β1, a cytokine that can be expressed in the brain, is a key regulator of the brain's responses to injury and inflammation. Alzheimer's disease (AD), the most common neurodegenerative disorder, involves inflammatory processes in the brain in addition to the hallmarks, amyloid-β (Aβ) plaques and neurofibrillary tangles. Recently, we have shown that T-helper (Th) 17 cells, a subpopulation of CD4+ T-cells with high proinflammation, also participate in the brain inflammatory process of AD. However, it is poorly known whether TGF-β1 ameliorates the lymphocyte-mediated neuroinflammation and, thereby, alleviates neurodegeneration in AD. Herein, we administered TGF-β1 via the intracerebroventricle (ICV) in AD model rats, by Aβ1-42 injection in both sides of the hippocampus, to show the neuroprotection of TGF-β1. The TGF-β1 administration after the Aβ1-42 injection ameliorated cognitive deficit and neuronal loss and apoptosis, reduced amyloid precursor protein (APP) expression, elevated protein phosphatase (PP)2A expression, attenuated glial activation and alleviated the imbalance of the pro-inflammatory/anti-inflammatory responses of T-lymphocytes, compared to the Aβ1-42 injection alone. These findings demonstrate that TGF-β1 provides protection against AD neurodegeneration and suggest that the TGF-β1 neuroprotection is implemented by the alleviation of glial and T-cell-mediated neuroinflammation.
Collapse
Affiliation(s)
- Wei-Xing Shen
- School of Biological & Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou 215123, China.
| | - Jia-Hui Chen
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Jian-Hua Lu
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China.
| |
Collapse
|
30
|
Kim MJ, O’Connor MB. Anterograde Activin signaling regulates postsynaptic membrane potential and GluRIIA/B abundance at the Drosophila neuromuscular junction. PLoS One 2014; 9:e107443. [PMID: 25255438 PMCID: PMC4177838 DOI: 10.1371/journal.pone.0107443] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/10/2014] [Indexed: 12/16/2022] Open
Abstract
Members of the TGF-β superfamily play numerous roles in nervous system development and function. In Drosophila, retrograde BMP signaling at the neuromuscular junction (NMJ) is required presynaptically for proper synapse growth and neurotransmitter release. In this study, we analyzed whether the Activin branch of the TGF-β superfamily also contributes to NMJ development and function. We find that elimination of the Activin/TGF-β type I receptor babo, or its downstream signal transducer smox, does not affect presynaptic NMJ growth or evoked excitatory junctional potentials (EJPs), but instead results in a number of postsynaptic defects including depolarized membrane potential, small size and frequency of miniature excitatory junction potentials (mEJPs), and decreased synaptic densities of the glutamate receptors GluRIIA and B. The majority of the defective smox synaptic phenotypes were rescued by muscle-specific expression of a smox transgene. Furthermore, a mutation in actβ, an Activin-like ligand that is strongly expressed in motor neurons, phenocopies babo and smox loss-of-function alleles. Our results demonstrate that anterograde Activin/TGF-β signaling at the Drosophila NMJ is crucial for achieving normal abundance and localization of several important postsynaptic signaling molecules and for regulating postsynaptic membrane physiology. Together with the well-established presynaptic role of the retrograde BMP signaling, our findings indicate that the two branches of the TGF-β superfamily are differentially deployed on each side of the Drosophila NMJ synapse to regulate distinct aspects of its development and function.
Collapse
Affiliation(s)
- Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kandasamy M, Lehner B, Kraus S, Sander PR, Marschallinger J, Rivera FJ, Trümbach D, Ueberham U, Reitsamer HA, Strauss O, Bogdahn U, Couillard-Despres S, Aigner L. TGF-beta signalling in the adult neurogenic niche promotes stem cell quiescence as well as generation of new neurons. J Cell Mol Med 2014; 18:1444-1459. [PMID: 24779367 PMCID: PMC4124027 DOI: 10.1111/jcmm.12298] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/10/2014] [Indexed: 12/16/2022] Open
Abstract
Members of the transforming growth factor (TGF)-β family govern a wide range of mechanisms in brain development and in the adult, in particular neuronal/glial differentiation and survival, but also cell cycle regulation and neural stem cell maintenance. This clearly created some discrepancies in the field with some studies favouring neuronal differentiation/survival of progenitors and others favouring cell cycle exit and neural stem cell quiescence/maintenance. Here, we provide a unifying hypothesis claiming that through its regulation of neural progenitor cell (NPC) proliferation, TGF-β signalling might be responsible for (i) maintaining stem cells in a quiescent stage, and (ii) promoting survival of newly generated neurons and their functional differentiation. Therefore, we performed a detailed histological analysis of TGF-β1 signalling in the hippocampal neural stem cell niche of a transgenic mouse that was previously generated to express TGF-β1 under a tetracycline regulatable Ca-Calmodulin kinase promoter. We also analysed NPC proliferation, quiescence, neuronal survival and differentiation in relation to elevated levels of TGF-β1 in vitro and in vivo conditions. Finally, we performed a gene expression profiling to identify the targets of TGF-β1 signalling in adult NPCs. The results demonstrate that TGF-β1 promotes stem cell quiescence on one side, but also neuronal survival on the other side. Thus, considering the elevated levels of TGF-β1 in ageing and neurodegenerative diseases, TGF-β1 signalling presents a molecular target for future interventions in such conditions.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Bernadette Lehner
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | - Sabrina Kraus
- Department of Experimental Ophthalmology, University of RegensburgRegensburg, Germany
| | - Paul Ramm Sander
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
- Institute of Biophysics and Physical Biochemistry, University of RegensburgRegensburg, Germany
| | - Julia Marschallinger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health (GmbH), Technical University MunichNeuherberg, Germany
| | - Uwe Ueberham
- Paul Flechsig Institute for Brain Research, Department of Neuroanatomy, University of LeipzigLeipzig, Germany
| | - Herbert A Reitsamer
- Department of Ophthalmology, SALK, Paracelsus Medical UniversitySalzburg, Austria
| | - Olaf Strauss
- Department of Experimental Ophthalmology, University of RegensburgRegensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital RegensburgRegensburg, Germany
| | - Sebastien Couillard-Despres
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical UniversitySalzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical UniversitySalzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical UniversitySalzburg, Austria
| |
Collapse
|
32
|
Martínez-Canabal A. Potential neuroprotective role of transforming growth factor β1 (TGFβ1) in the brain. Int J Neurosci 2014; 125:1-9. [PMID: 24628581 DOI: 10.3109/00207454.2014.903947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
TGFβ1 is a growth factor that is known to be expressed in most neurodegenerative diseases and after vascular accidents in the brain. TGFβ1 downregulates the activity of activated microglia and promotes astrogliosis. It also prevents cell death by a known mechanism dependant on astrocytes and the secretion of the plasminogen activator inhibitor 1 (PAI-1). This mechanism can provide light on what is the mechanism of action of TGFβ1 as a protective factor and it can provide the pharmacological principles in which this pathway could be used with therapeutic purposes. TGFβ1 is upregulated in most neurodegenerative diseases, however, its expression appears dramatically blocked in Huntington's disease, the fastest of those diseases in progress after the onset. This fact suggests that TGFβ1 slows down the neurodegenerative process, preventing tissue damage and neural apoptotic death. However, the exact details of TGFβ1 action are still unknown and the physiological roles on the diseases are still mysterious. Interestingly, all the data regarding the roles of TGFβ1 in health and disease have been also confirmed with the use of transgenic knockouts and TGFβ1 overexpressing mice. What possibly came as a surprise from the study of TGFβ1 overexpressing models is that combining its neuroprotective and antiproliferative effects, this cytokine generates a significant disruption in the hippocampal circuitry with its consequent learning and memory deficit.
Collapse
Affiliation(s)
- Alonso Martínez-Canabal
- Department of Molecular Neuropathology, Cell Physiology Institute (IFC), Department of Cell Biology, Faculty of Sciences, National Autonomous University of Mexico (UNAM). Ciudad Universitaria, Circuito exterior S/N, Coyoacan, 04510 Mexico D.F. Mexico
| |
Collapse
|
33
|
Nakajima T, Yanagihara M, Nishii H. Temporal and regional patterns of Smad activation in the rat hippocampus following global ischemia. J Neurol Sci 2013; 337:25-37. [PMID: 24290497 DOI: 10.1016/j.jns.2013.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/25/2013] [Accepted: 11/11/2013] [Indexed: 12/25/2022]
Abstract
In this study, we examined the temporal and regional patterns of Smad activation in the rat hippocampus following global ischemia. We also examined the association between Smad activation and ischemia-induced pathology in the hippocampus. We found that 1) Smad1, -2, -3, and -5 proteins were detected in the rat hippocampus by means of western blot and immunohistochemistry; 2) after 5 min of ischemia, Smad2 and Smad3 proteins accumulated in the nuclei of pyramidal cells in the CA1 region, which is vulnerable to ischemia; 3) after 3 min of ischemia, which was non-lethal, there was no such nuclear accumulation of Smad2 and Smad3 in the CA1 region; 4) following injection of activin A, nuclear accumulation of Smad2 and Smad3 was induced not only in pyramidal cells of the CA1 region, but also in pyramidal cells of the CA3 region as well as in granule cells of the DG region; 5) activin A-induced nuclear accumulation of Smad2 and Smad3 neither caused degeneration of hippocampal neurons nor prevented degeneration induced by ischemia. These results suggest that in the hippocampus, ischemia-induced activation of Smad2 and Smad3 is associated with the response to stress but is not related to neuronal survival or death.
Collapse
Affiliation(s)
- Takayuki Nakajima
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan.
| | - Masafumi Yanagihara
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan
| | - Hideki Nishii
- Department of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ohraikita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
34
|
Trefoil factor 3 as an endocrine neuroprotective factor from the liver in experimental cerebral ischemia/reperfusion injury. PLoS One 2013; 8:e77732. [PMID: 24204940 PMCID: PMC3799633 DOI: 10.1371/journal.pone.0077732] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 09/08/2013] [Indexed: 01/30/2023] Open
Abstract
Cerebral ischemia, while causing neuronal injury, can activate innate neuroprotective mechanisms, minimizing neuronal death. In this report, we demonstrate that experimental cerebral ischemia/reperfusion injury in the mouse causes upregulation of the secretory protein trefoil factor 3 (TFF3) in the hepatocyte in association with an increase in serum TFF3. Partial hepatectomy (~60% liver resection) immediately following cerebral injury significantly lowered the serum level of TFF3, suggesting a contribution of the liver to the elevation of serum TFF3. Compared to wild-type mice, TFF3-/- mice exhibited a significantly higher activity of caspase 3 and level of cell death in the ischemic cerebral lesion, a larger fraction of cerebral infarcts, and a smaller fraction of the injured cerebral hemisphere, accompanied by severer forelimb motor deficits. Intravenous administration of recombinant TFF3 reversed changes in cerebral injury and forelimb motor function due to TFF3 deficiency. These observations suggest an endocrine neuroprotective mechanism involving TFF3 from the liver in experimental cerebral ischemia/reperfusion injury.
Collapse
|
35
|
Swardfager W, Winer DA, Herrmann N, Winer S, Lanctôt KL. Interleukin-17 in post-stroke neurodegeneration. Neurosci Biobehav Rev 2013; 37:436-47. [PMID: 23370232 DOI: 10.1016/j.neubiorev.2013.01.021] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/10/2012] [Accepted: 01/20/2013] [Indexed: 12/15/2022]
Abstract
Stroke is a leading cause of physical disability with neurodegenerative sequelae such as dementia and depression causing significant excess morbidity. Stroke severity can be exacerbated by apoptotic cell death in ischemic tissue, of which inflammatory activity is a key determinant. Studies have identified harmful and beneficial sets of T lymphocytes that infiltrate the brain post-stroke and their activation signals, suggesting that they might be targeted for therapeutic benefit. Animal models and human studies implicate interleukin(IL)-17 and its congeners (e.g. IL-23, IL-21) as mediators of tissue damage in the delayed phase of the inflammatory cascade and the involvement of T lymphocytes in propagating IL-17 release. In this review, we highlight the current understanding of IL-17 secreting cells, including sets of CD4(+) αβ and CD4(-) γδ T lymphocytes, as potentially important mediators of brain pathology post-stroke. Interactions between the IL-17 axis and innate pathways, positive feedback mechanisms that prolong or amplify IL-17, and IL-17 regulatory pathways may offer intervention targets to enhance recovery, prevent long-term decline, and improve quality of life.
Collapse
Affiliation(s)
- Walter Swardfager
- Neuropsychopharmacology Research Group, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | | | | | | | | |
Collapse
|
36
|
Spittau B, Wullkopf L, Zhou X, Rilka J, Pfeifer D, Krieglstein K. Endogenous transforming growth factor-beta promotes quiescence of primary microglia in vitro. Glia 2012; 61:287-300. [PMID: 23065670 DOI: 10.1002/glia.22435] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 09/11/2012] [Indexed: 11/12/2022]
Abstract
Microglia are the immune cells of the central nervous system (CNS) and play important roles under physiological and pathophysiological conditions. Activation of microglia has been reported for a variety of CNS diseases and is believed to be involved in inflammation-mediated neurodegeneration. Loss of TGFβ1 results in increased microgliosis and neurodegeneration in mice which indicates that TGFβ1 is an important regulator of microglial functions in vivo. Here, we addressed the role of endogenous TGFβ signaling for microglia in vitro. We clearly demonstrate active TGFβ signaling in primary microglia and further introduce Klf10 as a new TGFβ target gene in microglia. Moreover, we provide evidence that microglia express and release TGFβ1 that acts in an autocrine manner to activate microglial TGFβ/Smad signaling in vitro. Using microarrays, we identified TGFβ-regulated genes in microglia that are involved in TGFβ1 processing, its extracellular storage as well as activation of latent TGFβ. Finally, we demonstrate that pharmacological inhibition of microglial TGFβ signaling resulted in upregulation of the proinflammatory markers IL6 and iNOS and downregulation of the alternative activation markers Arg1 and Ym1 in vitro. Together, these data clearly show that endogenous TGFβ1 and autocrine TGFβ signaling is important for microglial quiescence in vitro and further suggest the upregulation of TGFβ1 in neurodegenerative diseases as a mechanism to regulate microglia functions and silence neuroinflammation.
Collapse
Affiliation(s)
- Björn Spittau
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-University, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Diniz LP, Almeida JC, Tortelli V, Vargas Lopes C, Setti-Perdigão P, Stipursky J, Kahn SA, Romão LF, de Miranda J, Alves-Leon SV, de Souza JM, Castro NG, Panizzutti R, Gomes FCA. Astrocyte-induced synaptogenesis is mediated by transforming growth factor β signaling through modulation of D-serine levels in cerebral cortex neurons. J Biol Chem 2012; 287:41432-45. [PMID: 23055518 PMCID: PMC3510841 DOI: 10.1074/jbc.m112.380824] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Assembly of synapses requires proper coordination between pre- and postsynaptic elements. Identification of cellular and molecular events in synapse formation and maintenance is a key step to understand human perception, learning, memory, and cognition. A key role for astrocytes in synapse formation and function has been proposed. Here, we show that transforming growth factor β (TGF-β) signaling is a novel synaptogenic pathway for cortical neurons induced by murine and human astrocytes. By combining gain and loss of function approaches, we show that TGF-β1 induces the formation of functional synapses in mice. Further, TGF-β1-induced synaptogenesis involves neuronal activity and secretion of the co-agonist of the NMDA receptor, d-serine. Manipulation of d-serine signaling, by either genetic or pharmacological inhibition, prevented the TGF-β1 synaptogenic effect. Our data show a novel molecular mechanism that might impact synaptic function and emphasize the evolutionary aspect of the synaptogenic property of astrocytes, thus shedding light on new potential therapeutic targets for synaptic deficit diseases.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-590 Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Pál G, Vincze C, Renner É, Wappler EA, Nagy Z, Lovas G, Dobolyi A. Time course, distribution and cell types of induction of transforming growth factor betas following middle cerebral artery occlusion in the rat brain. PLoS One 2012; 7:e46731. [PMID: 23056426 PMCID: PMC3466286 DOI: 10.1371/journal.pone.0046731] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 09/03/2012] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-βs (TGF-β1–3) are cytokines that regulate the proliferation, differentiation, and survival of various cell types. The present study describes the induction of TGF-β1–3 in the rat after focal ischemia at 3 h, 24 h, 72 h and 1 month after transient (1 h) or permanent (24 h) middle cerebral artery occlusion (MCAO) using in situ hybridization histochemistry and quantitative analysis. Double labeling with different markers was used to identify the localization of TGF-β mRNA relative to the penumbra and glial scar, and the types of cells expressing TGF-βs. TGF-β1 expression increased 3 h after MCAO in the penumbra and was further elevated 24 h after MCAO. TGF-β1 was present mostly in microglial cells but also in some astrocytes. By 72 h and 1 month after the occlusion, TGF-β1 mRNA-expressing cells also appeared in microglia within the ischemic core and in the glial scar. In contrast, TGF-β2 mRNA level was increased in neurons but not in astrocytes or microglial cells in layers II, III, and V of the ipsilateral cerebral cortex 24 h after MCAO. TGF-β3 was not induced in cells around the penumbra. Its expression increased in only a few cells in layer II of the cerebral cortex 24 h after MCAO. The levels of TGF-β2 and -β3 decreased at subsequent time points. Permanent MCAO further elevated the levels of all 3 subtypes of TGF-βs suggesting that reperfusion is not a major factor in their induction. TGF-β1 did not co-localize with either Fos or ATF-3, while the co-localization of TGF-β2 with Fos but not with ATF-3 suggests that cortical spreading depolarization, but not damage to neural processes, might be the mechanism of induction for TGF-β2. The results imply that endogenous TGF-βs are induced by different mechanisms following an ischemic attack in the brain suggesting that they are involved in distinct spatially and temporally regulated inflammatory and neuroprotective processes.
Collapse
Affiliation(s)
- Gabriella Pál
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Csilla Vincze
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Éva Renner
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
| | - Edina A. Wappler
- Cardiovascular Center, Department Section of Vascular Neurology, Semmelweis University, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Nagy
- Cardiovascular Center, Department Section of Vascular Neurology, Semmelweis University, Budapest, Hungary
| | - Gábor Lovas
- Department of Neurology, Semmelweis University, Budapest, Hungary
- Department of Neurology, Jahn Ferenc Teaching Hospital, Budapest, Hungary
| | - Arpád Dobolyi
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| |
Collapse
|
39
|
The neuroprotective functions of transforming growth factor beta proteins. Int J Mol Sci 2012; 13:8219-8258. [PMID: 22942700 PMCID: PMC3430231 DOI: 10.3390/ijms13078219] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 05/24/2012] [Accepted: 06/19/2012] [Indexed: 12/26/2022] Open
Abstract
Transforming growth factor beta (TGF-β) proteins are multifunctional cytokines whose neural functions are increasingly recognized. The machinery of TGF-β signaling, including the serine kinase type transmembrane receptors, is present in the central nervous system. However, the 3 mammalian TGF-β subtypes have distinct distributions in the brain suggesting different neural functions. Evidence of their involvement in the development and plasticity of the nervous system as well as their functions in peripheral organs suggested that they also exhibit neuroprotective functions. Indeed, TGF-β expression is induced following a variety of types of brain tissue injury. The neuroprotective function of TGF-βs is most established following brain ischemia. Damage in experimental animal models of global and focal ischemia was shown to be attenuated by TGF-βs. In addition, support for their neuroprotective actions following trauma, sclerosis multiplex, neurodegenerative diseases, infections, and brain tumors is also accumulating. The review will also describe the potential mechanisms of neuroprotection exerted by TGF-βs including anti-inflammatory, -apoptotic, -excitotoxic actions as well as the promotion of scar formation, angiogenesis, and neuroregeneration. The participation of these mechanisms in the neuroprotective effects of TGF-βs during different brain lesions will also be discussed.
Collapse
|
40
|
Neuron-glia signaling: Implications for astrocyte differentiation and synapse formation. Life Sci 2011; 89:524-31. [PMID: 21569780 DOI: 10.1016/j.lfs.2011.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 03/10/2011] [Accepted: 04/09/2011] [Indexed: 11/22/2022]
Abstract
Glial cells are currently viewed as active partners of neurons in synapse formation. The close proximity of astrocytes to the synaptic cleft implicates that they strongly influence synapse function as well as suggests that these cells might be potential targets for neuronal-released molecules. In this review, we discuss the signaling pathways of astrocyte generation and the role of astrocyte-derived molecules in synapse formation in the central nervous system. Further, we discuss the role of the excitatory neurotransmitter, glutamate and transforming growth factor beta 1 (TGF-β1) pathway in astrocyte generation and differentiation. We provide evidence that astrocytes surrounding synapses are target of neuronal activity and shed light into the role of astroglial cells into neurological disorders associated with glutamate neurotoxicity.
Collapse
|