1
|
Noel SC, Madranges JF, Gothié JDM, Ewald J, Milnerwood AJ, Kennedy TE, Scott ME. Maternal gastrointestinal nematode infection alters hippocampal neuroimmunity, promotes synaptic plasticity, and improves resistance to direct infection in offspring. Sci Rep 2024; 14:10773. [PMID: 38730262 PMCID: PMC11087533 DOI: 10.1038/s41598-024-60865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.
Collapse
Affiliation(s)
- Sophia C Noel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| | - Jeanne F Madranges
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jean-David M Gothié
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jessica Ewald
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Austen J Milnerwood
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
2
|
von Bernhardi R, Eugenín J. Aging Microglia and Their Impact in the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 37:379-395. [PMID: 39207703 DOI: 10.1007/978-3-031-55529-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Aging is the greatest risk factor for neurodegenerative diseases. Microglia are the resident immune cells in the central nervous system (CNS), playing key roles in its normal functioning, and as mediators for age-dependent changes of the CNS, condition at which they generate a hostile environment for neurons. Transforming Growth Factor β1 (TGFβ1) is a regulatory cytokine involved in immuneregulation and neuroprotection, affecting glial cell inflammatory activation, neuronal survival, and function. TGFβ1 signaling undergoes age-dependent changes affecting the regulation of microglial cells and can contribute to the pathophysiology of neurodegenerative diseases. This chapter focuses on assessing the role of age-related changes on the regulation of microglial cells and their impact on neuroinflammation and neuronal function, for understanding age-dependent changes of the nervous system.
Collapse
Affiliation(s)
- Rommy von Bernhardi
- Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastian, Santiago, Chile.
| | - Jaime Eugenín
- Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
3
|
Sentyabreva AV, Miroshnichenko EA, Melnikova EA, Tsvetkov IS, Kosyreva AM. Morphofunctional Changes in Brain and Peripheral Blood in Adult and Aged Wistar Rats with AlCl 3-Induced Neurodegeneration. Biomedicines 2023; 11:2336. [PMID: 37760778 PMCID: PMC10526012 DOI: 10.3390/biomedicines11092336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/09/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND the general lifespan has been prolonged greatly during the past century, and the incidence of age-associated diseases, including neurodegenerative ones, has increased as well. However, modelling of age-related pathologies is mostly conducted on adult rodents. We studied morphofunctional changes in the brain and peripheral blood of adult Wistar rats in comparison with old Wistar rats to determine age-related physiological changes and differences in adaptive reactions to AlCl3 exposure. METHODS the work was performed on adult and old male Wistar rats. The animals consumed a 100 mg/kg solution of AlCl3 each day for 60 days. Morphological changes of neurons and microglia, mRNA expression levels of pro-inflammatory and anti-inflammatory cytokines, microglia activation markers, amyloid-related proteins, and hallmarks of cellular senescence, monocyte, and lymphocyte subpopulations in the peripheral blood were examined. RESULTS old rats showed increasing hyperchromic neurons in the hippocampus; activation of microglia; upregulation of pro-inflammatory cytokines and cellular senescence markers; downregulation of anti-inflammatory cytokines; and Hif-1a and a decrease in B-cells and monocyte in peripheral blood. CONCLUSION compared to young animals, aged rats respond to aluminum exposure with a severe decline of most cells' function and irreversible neuronal loss. Regarding all reported data, neurodegeneration modelling and investigating of factors capable of accelerating or preventing it should be performed in experimental work on aged animals.
Collapse
Affiliation(s)
- Alexandra Vladislavovna Sentyabreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Alexandrovna Miroshnichenko
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Andreevna Melnikova
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Ivan Sergeevich Tsvetkov
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Anna Mikhailovna Kosyreva
- Avtsyn Research Institute of Human Morphology of “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
4
|
Cheng X, Yan Z, Su Z, Liu J. The transforming growth factor beta ligand TIG-2 modulates the function of neuromuscular junction and muscle energy metabolism in Caenorhabditis elegans. Front Mol Neurosci 2022; 15:962974. [PMID: 36385772 PMCID: PMC9650414 DOI: 10.3389/fnmol.2022.962974] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Deciphering the physiological function of TGF-β (the transforming growth factor beta) family ligands is import for understanding the role of TGF-β in animals' development and aging. Here, we investigate the function of TIG-2, one of the ligands in Caenorhabditis elegans TGF-β family, in animals' behavioral modulation. Our results show that a loss-of-function mutation in tig-2 gene result in slower locomotion speed in the early adulthood and an increased density of cholinergic synapses, but a decreased neurotransmitter release at neuromuscular junctions (NMJs). Further tissue-specific rescue results reveal that neuronal and intestinal TIG-2 are essential for the formation of cholinergic synapses at NMJs. Interestingly, tig-2(ok3416) mutant is characterized with reduced muscle mitochondria content and adenosine triphosphate (ATP) production, although the function of muscle acetylcholine receptors and the morphology muscle fibers in the mutant are comparable to that in wild-type animals. Our result suggests that TIG-2 from different neuron and intestine regulates worm locomotion by modulating synaptogenesis and neurotransmission at NMJs, as well as energy metabolism in postsynaptic muscle cells.
Collapse
Affiliation(s)
- Xinran Cheng
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Zhenzhen Yan
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Zexiong Su
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jie Liu
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Tomiga Y, Higaki Y, Anzai K, Takahashi H. Behavioral defects and downregulation of hippocampal BDNF and nNOS expression in db/db mice did not improved by chronic TGF-β2 treatment. Front Physiol 2022; 13:969480. [PMID: 36091357 PMCID: PMC9452698 DOI: 10.3389/fphys.2022.969480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiological evidence suggests that there is a link between diabetes and mood disorders, such as depression and anxiety. Although peripheral or central inflammation may explain this link, the molecular mechanisms are not fully understood and few effective treatments for diabetes or mood disorders are available. In the present study, we aimed to determine whether transforming growth factor (TGF)-β2, an anti-inflammatory substance, might represent a potential therapeutic agent for diabetes-related mood behaviors. TGF-β2 expression in the hippocampus is affected by anxiolytic drugs and stress exposure, it is able to cross the blood-brain barrier, and it is as an exercise-induced physiological adipokine that regulates glucose homeostasis. Therefore, we hypothesized that a chronic TGF-β2 infusion would ameliorate diabetes-related glucose intolerance and mood dysregulation. To determine the effects of the chronic administration of TGF-β2 on diabetes, we implanted osmotic pumps containing TGF-β2 into type 2 diabetic mice (db/db mice), and age-matched non-diabetic control wild type mice and db/db mice were infused with vehicle (PBS), for 12 consecutive days. To assess anxiety-like behaviors and glucose homeostasis, the mice underwent elevated plus maze testing and intraperitoneal glucose tolerance testing. Hippocampal and perigonadal visceral white adipose tissue perigonadal white adipose tissue samples were obtained 12 days later. Contrary to our hypothesis, TGF-β2 infusion had no effect on diabetes-related glucose intolerance or diabetes-related behavioral defects, such as inactivity. In db/db mice, the expression of inflammatory markers was high in pgWAT, but not in the hippocampus, and the former was ameliorated by TGF-β2 infusion. The expression of brain-derived neurotrophic factor and neuronal nitric oxide synthase, important regulators of anxiety-like behaviors, was low in db/db mice, but TGF-β2 infusion did not affect their expression. We conclude that although TGF-β2 reduces the expression of pro-inflammatory markers in the adipose tissue of diabetic mice, it does not ameliorate their obesity or mood dysregulation.
Collapse
Affiliation(s)
- Yuki Tomiga
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
- *Correspondence: Yuki Tomiga,
| | - Yasuki Higaki
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Keizo Anzai
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| | - Hirokazu Takahashi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
- Liver Center, Saga University Hospital, Saga, Japan
| |
Collapse
|
6
|
Kuznitsov-Yanovsky L, Shapira G, Gildin L, Shomron N, Ben-Yosef D. Transcriptomic Analysis of Human Fragile X Syndrome Neurons Reveals Neurite Outgrowth Modulation by the TGFβ/BMP Pathway. Int J Mol Sci 2022; 23:ijms23169278. [PMID: 36012539 PMCID: PMC9409179 DOI: 10.3390/ijms23169278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/25/2022] Open
Abstract
Fragile X Syndrome (FXS) is the main genetic reason for intellectual disability and is caused by the silencing of fragile X mental retardation protein (FMRP), an RNA-binding protein regulating the translation of many neuronal mRNAs. Neural differentiation of FX human embryonic stem cells (hESC) mimics the neurodevelopment of FXS fetuses and thus serves as a good model to explore the mechanisms underlining the development of FXS. Isogenic hESC clones with and without the FX mutation that share the same genetic background were in vitro differentiated into neurons, and their transcriptome was analyzed by RNA sequencing. FX neurons inactivating FMR1 expression presented delayed neuronal development and maturation, concomitant with dysregulation of the TGFβ/BMP signaling pathway, and genes related to the extracellular matrix. Migration assay showed decreased neurite outgrowth in FX neurons that was rescued by inhibition of the TGFβ/BMP signaling pathway. Our results provide new insights into the molecular pathway by which loss of FMRP affects neuronal network development. In FX neurons, the lack of FMRP dysregulates members of the BMP signaling pathway associated with ECM organization which, in a yet unknown mechanism, reduces the guidance of axonal growth cones, probably leading to the aberrant neuronal network function seen in FXS.
Collapse
Affiliation(s)
- Liron Kuznitsov-Yanovsky
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Guy Shapira
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Lital Gildin
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Dalit Ben-Yosef
- Wolfe PGD Stem Cell Lab, Racine IVF Unit, Lis Maternity Hospital Tel-Aviv Sourasky Medical Center, Tel Aviv 64239, Israel
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
7
|
TGF-β/Smad Signalling in Neurogenesis: Implications for Neuropsychiatric Diseases. Cells 2021; 10:cells10061382. [PMID: 34205102 PMCID: PMC8226492 DOI: 10.3390/cells10061382] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
TGF-β/Smad signalling has been the subject of extensive research due to its role in the cell cycle and carcinogenesis. Modifications to the TGF-β/Smad signalling pathway have been found to produce disparate effects on neurogenesis. We review the current research on canonical and non-canonical TGF-β/Smad signalling pathways and their functions in neurogenesis. We also examine the observed role of neurogenesis in neuropsychiatric disorders and the relationship between TGF-β/Smad signalling and neurogenesis in response to stressors. Overlapping mechanisms of cell proliferation, neurogenesis, and the development of mood disorders in response to stressors suggest that TGF-β/Smad signalling is an important regulator of stress response and is implicated in the behavioural outcomes of mood disorders.
Collapse
|
8
|
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: 20] [Impact Index Per Article: 5.0] [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
|
9
|
Fessel J. If ineffective levels of transforming growth factors and their receptor account for old age being a risk factor for Alzheimer's disease, then increasing TGFBR2 might be therapeutic. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12019. [PMID: 32382652 PMCID: PMC7202202 DOI: 10.1002/trc2.12019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/02/2020] [Accepted: 02/17/2020] [Indexed: 12/19/2022]
Abstract
If it is correct that ineffective levels of transforming growth factors beta and their receptor account for old age being a risk factor for Alzheimer's disease (AD), then increasing TGFBR2 might be therapeutic. Pacltaxel is a direct way to increase TGFBR2 levels. Indirect ways that will increase TGFBR2, include decreasing the levels of c-myc because that will lower the miRNA cluster 17-92, particularly its miR-17 and miR-20a components; and raising EGFR because that also will increase TGFBR2. Metformin and desferrioxamine are drugs that decrease c-myc; and statins increase levels of EGF. Clinical trials using those drugs, would demonstrate whether they decrease the progression from amnestic mild cognitive impairment to AD.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
10
|
TGF-β/Smad3 Signalling Modulates GABA Neurotransmission: Implications in Parkinson's Disease. Int J Mol Sci 2020; 21:ijms21020590. [PMID: 31963327 PMCID: PMC7013528 DOI: 10.3390/ijms21020590] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
γ-Aminobutiryc acid (GABA) is found extensively in different brain nuclei, including parts involved in Parkinson’s disease (PD), such as the basal ganglia and hippocampus. In PD and in different models of the disorder, an increase in GABA neurotransmission is observed and may promote bradykinesia or L-Dopa-induced side-effects. In addition, proteins involved in GABAA receptor (GABAAR) trafficking, such as GABARAP, Trak1 or PAELR, may participate in the aetiology of the disease. TGF-β/Smad3 signalling has been associated with several pathological features of PD, such as dopaminergic neurodegeneration; reduction of dopaminergic axons and dendrites; and α-synuclein aggregation. Moreover, TGF-β/Smad3 intracellular signalling was recently shown to modulate GABA neurotransmission in the context of parkinsonism and cognitive alterations. This review provides a summary of GABA neurotransmission and TGF-β signalling; their implications in PD; and the regulation of GABA neurotransmission by TGF-β/Smad3. There appear to be new possibilities to develop therapeutic approaches for the treatment of PD using GABA modulators.
Collapse
|
11
|
Johnson BV, Kumar R, Oishi S, Alexander S, Kasherman M, Vega MS, Ivancevic A, Gardner A, Domingo D, Corbett M, Parnell E, Yoon S, Oh T, Lines M, Lefroy H, Kini U, Van Allen M, Grønborg S, Mercier S, Küry S, Bézieau S, Pasquier L, Raynaud M, Afenjar A, Billette de Villemeur T, Keren B, Désir J, Van Maldergem L, Marangoni M, Dikow N, Koolen DA, VanHasselt PM, Weiss M, Zwijnenburg P, Sa J, Reis CF, López-Otín C, Santiago-Fernández O, Fernández-Jaén A, Rauch A, Steindl K, Joset P, Goldstein A, Madan-Khetarpal S, Infante E, Zackai E, Mcdougall C, Narayanan V, Ramsey K, Mercimek-Andrews S, Pena L, Shashi V, Schoch K, Sullivan JA, Pinto E Vairo F, Pichurin PN, Ewing SA, Barnett SS, Klee EW, Perry MS, Koenig MK, Keegan CE, Schuette JL, Asher S, Perilla-Young Y, Smith LD, Rosenfeld JA, Bhoj E, Kaplan P, Li D, Oegema R, van Binsbergen E, van der Zwaag B, Smeland MF, Cutcutache I, Page M, Armstrong M, Lin AE, Steeves MA, Hollander ND, Hoffer MJV, Reijnders MRF, Demirdas S, Koboldt DC, Bartholomew D, Mosher TM, Hickey SE, Shieh C, Sanchez-Lara PA, Graham JM, Tezcan K, Schaefer GB, Danylchuk NR, Asamoah A, Jackson KE, Yachelevich N, Au M, Pérez-Jurado LA, Kleefstra T, Penzes P, Wood SA, Burne T, Pierson TM, Piper M, Gécz J, Jolly LA. Partial Loss of USP9X Function Leads to a Male Neurodevelopmental and Behavioral Disorder Converging on Transforming Growth Factor β Signaling. Biol Psychiatry 2020; 87:100-112. [PMID: 31443933 PMCID: PMC6925349 DOI: 10.1016/j.biopsych.2019.05.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND The X-chromosome gene USP9X encodes a deubiquitylating enzyme that has been associated with neurodevelopmental disorders primarily in female subjects. USP9X escapes X inactivation, and in female subjects de novo heterozygous copy number loss or truncating mutations cause haploinsufficiency culminating in a recognizable syndrome with intellectual disability and signature brain and congenital abnormalities. In contrast, the involvement of USP9X in male neurodevelopmental disorders remains tentative. METHODS We used clinically recommended guidelines to collect and interrogate the pathogenicity of 44 USP9X variants associated with neurodevelopmental disorders in males. Functional studies in patient-derived cell lines and mice were used to determine mechanisms of pathology. RESULTS Twelve missense variants showed strong evidence of pathogenicity. We define a characteristic phenotype of the central nervous system (white matter disturbances, thin corpus callosum, and widened ventricles); global delay with significant alteration of speech, language, and behavior; hypotonia; joint hypermobility; visual system defects; and other common congenital and dysmorphic features. Comparison of in silico and phenotypical features align additional variants of unknown significance with likely pathogenicity. In support of partial loss-of-function mechanisms, using patient-derived cell lines, we show loss of only specific USP9X substrates that regulate neurodevelopmental signaling pathways and a united defect in transforming growth factor β signaling. In addition, we find correlates of the male phenotype in Usp9x brain-specific knockout mice, and further resolve loss of hippocampal-dependent learning and memory. CONCLUSIONS Our data demonstrate the involvement of USP9X variants in a distinctive neurodevelopmental and behavioral syndrome in male subjects and identify plausible mechanisms of pathogenesis centered on disrupted transforming growth factor β signaling and hippocampal function.
Collapse
Affiliation(s)
- Brett V Johnson
- University of Adelaide and Robinson Research Institute, Adelaide, Australia
| | - Raman Kumar
- University of Adelaide and Robinson Research Institute, Adelaide, Australia
| | - Sabrina Oishi
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Suzy Alexander
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
| | - Maria Kasherman
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia; Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | | | - Atma Ivancevic
- University of Adelaide and Robinson Research Institute, Adelaide, Australia; BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado
| | - Alison Gardner
- University of Adelaide and Robinson Research Institute, Adelaide, Australia
| | - Deepti Domingo
- University of Adelaide and Robinson Research Institute, Adelaide, Australia
| | - Mark Corbett
- University of Adelaide and Robinson Research Institute, Adelaide, Australia
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tracey Oh
- Department of Medical Genetics, British Columbia Women's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew Lines
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Henrietta Lefroy
- Oxford Centre for Genomic Medicine, Oxford University Hospitals National Health Services Foundation Trust, Oxford, United Kingdom
| | - Usha Kini
- Oxford Centre for Genomic Medicine, Oxford University Hospitals National Health Services Foundation Trust, Oxford, United Kingdom
| | - Margot Van Allen
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sabine Grønborg
- Center for Rare Diseases, Department of Pediatrics and Department of Clinical Genetics, University Hospital Copenhagen, Copenhagen, Denmark
| | - Sandra Mercier
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes and l'Institut du Thorax, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Nantes, Nantes, France
| | - Sébastien Küry
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes and l'Institut du Thorax, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Nantes, Nantes, France
| | - Stéphane Bézieau
- Service de Génétique Médicale, Centre Hospitalier Universitaire Nantes and l'Institut du Thorax, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Université de Nantes, Nantes, France
| | - Laurent Pasquier
- Service de Génétique Clinique, Centre de Référence Déficiences Intellectuelles de Causes Rares, Centre Hospitalier Universitaire Hôpital Sud, Rennes, France
| | - Martine Raynaud
- Centre Hospitalier Régional Universitaire de Tours, Service de Génétique, Unité Nixte de Recherche 1253, iBrain, Université de Tours, Institut National de la Santé et de la Recherche Médicale, Tours, France
| | - Alexandra Afenjar
- Groupe de Recherche Clinique No. 19, ConCer-LD, Département de Génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Armand Trousseau, Centres de Référence Maladies Rares des Déficits Intellectuels de Causes Rares, Paris, France
| | - Thierry Billette de Villemeur
- Sorbonne Université, Groupe de Recherche Clinique No. 19, ConCer-LD, Neuropédiatrie, Centres de Référence Maladies Rares Neurogénétique, Institut National de la Santé et de la Recherche Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Boris Keren
- Hôpital de la Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Julie Désir
- Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Martina Marangoni
- Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicola Dikow
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - David A Koolen
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter M VanHasselt
- Department of Metabolic Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marjan Weiss
- Department of Clinical Genetics, Vrije Universiteit University Medical Center, Amsterdam, The Netherlands
| | - Petra Zwijnenburg
- Medical Genetics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Joaquim Sa
- Medical Genetics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Claudia Falcao Reis
- Medical Genetics Unit, Hospital Pediátrico, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitário de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer, Spain
| | - Olaya Santiago-Fernández
- Departamento de Bioquímica y Biología Molecular, Instituto Universitário de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | | | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Pascal Joset
- Institute of Medical Genetics, University of Zurich, Schlieren, Switzerland
| | - Amy Goldstein
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Elena Infante
- Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elaine Zackai
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Carey Mcdougall
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, Arizona
| | - Keri Ramsey
- Center for Rare Childhood Disorders, Translational Genomics Research Institute, Phoenix, Arizona
| | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Loren Pena
- Division of Human Genetics, Cincinnati Children's Hospital; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Vandana Shashi
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, North Carolina
| | - Kelly Schoch
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, North Carolina
| | - Jennifer A Sullivan
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, Durham, North Carolina
| | - Filippo Pinto E Vairo
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Pavel N Pichurin
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | - Sarah A Ewing
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | - Sarah S Barnett
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Eric W Klee
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | - M Scott Perry
- Jane and John Justin Neuroscience Center, Cook Children's Medical Center, Fort Worth, Texas
| | - Mary Kay Koenig
- Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas
| | - Catherine E Keegan
- Division of Genetics, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Jane L Schuette
- Division of Genetics, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Stephanie Asher
- Translational Medicine & Human Genetics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yezmin Perilla-Young
- Division of Pediatric Genetics and Metabolism, University of North Carolina, Chapel Hill, North Carolina
| | - Laurie D Smith
- Division of Pediatric Genetics and Metabolism, University of North Carolina, Chapel Hill, North Carolina
| | | | - Elizabeth Bhoj
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Paige Kaplan
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Dong Li
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ellen van Binsbergen
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bert van der Zwaag
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Matthew Page
- Translational Medicine, UCB Pharma, Braine-l'Alleud, Belgium
| | | | - Angela E Lin
- Medical Genetics Unit, Mass General Hospital for Children, Boston, Massachusetts
| | - Marcie A Steeves
- Medical Genetics Unit, Mass General Hospital for Children, Boston, Massachusetts
| | | | - Mariëtte J V Hoffer
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Margot R F Reijnders
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Serwet Demirdas
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | - Scott E Hickey
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Christine Shieh
- David Geffen School of Medicine, University of California-Los Angeles, California
| | | | - John M Graham
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kamer Tezcan
- Department of Genetics, Kaiser Permanente, Sacramento, California
| | - G B Schaefer
- Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Noelle R Danylchuk
- Department of Genetic Counseling, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Alexander Asamoah
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kelly E Jackson
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, Kentucky
| | - Naomi Yachelevich
- Clinical Genetics Services, Department of Pediatrics, New York University School of Medicine, New York, New York
| | - Margaret Au
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California
| | - Luis A Pérez-Jurado
- University of Adelaide and Robinson Research Institute, Adelaide, Australia; Women's and Children's Hospital, Adelaide, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia; Hospital del Mar Research Institute, Network Research Centre for Rare Diseases and Universitat Pompeu Fabra, Barcelona, Spain
| | - Tjitske Kleefstra
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stephen A Wood
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Australia
| | - Thomas Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
| | - Tyler Mark Pierson
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California; Department of Neurology and the Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Jozef Gécz
- University of Adelaide and Robinson Research Institute, Adelaide, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia.
| | - Lachlan A Jolly
- University of Adelaide and Robinson Research Institute, Adelaide, Australia.
| |
Collapse
|
12
|
Mikheeva IB, Malkov AE, Pavlik LL, Arkhipov VI, Levin SG. Effect of TGF-beta1 on long-term synaptic plasticity and distribution of AMPA receptors in the CA1 field of the hippocampus. Neurosci Lett 2019; 704:95-99. [PMID: 30953737 DOI: 10.1016/j.neulet.2019.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/06/2019] [Accepted: 04/02/2019] [Indexed: 11/29/2022]
Abstract
Using the methods of electrophysiology and immunohistochemistry, the effect of the transforming factor beta-1(TGF-β1), an anti-inflammatory cytokine, on the long-term post-tetanic potentiation (LTP) in CA1 field hippocampal slices and the distribution of the GluR1 subunit of the AMPA receptor has been studied. It was shown that TGF-β1 at a concentration of 10 ng/ml did not significantly affect the initial stage of LTP and substantially changed the distribution of synaptic AMPA receptors in response to tetanic stimulation. Twenty five minutes after the tetanization, the main pool of AMPA receptors (90%) was due to the postsynaptic density (PSD). By contrast, LTP in the presence of TGF-β1 was accompanied by less pronounced changes in the distribution of AMPA receptors. Their localization in both pre- and postsynaptic regions remained nearly the same as that in the control. It may be suggested that the normal distribution of AMPA receptors in spinous synapses promotes the stabilization of potentiated synapses, thereby retaining LTP for longer terms.
Collapse
Affiliation(s)
- I B Mikheeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - A E Malkov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - L L Pavlik
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - V I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia
| | - S G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia; Pushchino State Institute of Natural Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
13
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
14
|
Frank DF, Brander SM, Hasenbein S, Harvey DJ, Lein PJ, Geist J, Connon RE. Developmental exposure to environmentally relevant concentrations of bifenthrin alters transcription of mTOR and ryanodine receptor-dependent signaling molecules and impairs predator avoidance behavior across early life stages in inland silversides (Menidia beryllina). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 206:1-13. [PMID: 30414561 PMCID: PMC6464817 DOI: 10.1016/j.aquatox.2018.10.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 05/05/2023]
Abstract
Altered transcription of calcium-dependent signaling cascades involving the ryanodine receptor (RyR) and mechanistic target of rapamycin (mTOR) in response to environmental exposures have been described in model vertebrates, including zebrafish, while the relevance for wild fishes remains unknown. To address this knowledge gap, we exposed the euryhaline model species Menidia beryllina (inland silversides) to the insecticide bifenthrin, a known modulator of calcium signaling. The main objectives of this study were to determine: (1) whether exposure of developing silversides to environmentally relevant concentrations of bifenthrin alters their behavior; and (2) whether behavioral changes correlate with altered expression of genes involved in RyR and mTOR-dependent signaling pathways. At six hours post fertilization (hpf), inland silversides were exposed to bifenthrin at 3, 27 and 122 ng/L until 7 days post fertilization (dpf, larvae hatched at 6dpf), followed by a 14-day recovery period in uncontaminated water. Transcriptional responses were measured at 5, 7 and 21 dpf; locomotor behavior following external stimuli and response to an olfactory predator cue were assessed at 7 and 21 dpf. Bifenthrin elicited significant non-monotonic transcriptional responses in the majority of genes examined at 5 dpf and at 21 dpf. Bifenthrin also significantly altered predator avoidance behavior via olfactory mechanisms with main effects identified for animals exposed to 3 and 27 ng/L. Behavioral effects were not detected in response to visual stimuli during acute exposure, but were significant in the predator-cue assessment following the recovery period, suggesting delayed and long-term effects of early developmental exposures to bifenthrin. Our findings demonstrate that at picomolar (pM) concentrations, which are often not represented in ecotoxicological studies, bifenthrin perturbs early development of inland silversides. These developmental impacts are manifested behaviorally at later life stages, specifically as altered patterns of predator avoidance behavior, which have been correlated with population decline. Collectively, these data suggest that bifenthrin may be negatively impacting wild fish populations.
Collapse
Affiliation(s)
- Daniel F Frank
- Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; Aquatic Systems Biology, Department of Ecology and Ecosystem Management, Technical University Munich, Mühlenweg 22, D-85354 Freising, Germany
| | - Susanne M Brander
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA; Department of Biology & Marine Biology, University of North Carolina, Wilmington, NC 28403, USA
| | - Simone Hasenbein
- Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; Aquatic Systems Biology, Department of Ecology and Ecosystem Management, Technical University Munich, Mühlenweg 22, D-85354 Freising, Germany
| | - Danielle J Harvey
- Department of Public Health Sciences, Division of Biostatistics, University of California, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Juergen Geist
- Aquatic Systems Biology, Department of Ecology and Ecosystem Management, Technical University Munich, Mühlenweg 22, D-85354 Freising, Germany
| | - Richard E Connon
- Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| |
Collapse
|
15
|
Astrocytes and the TGF-β1 Pathway in the Healthy and Diseased Brain: a Double-Edged Sword. Mol Neurobiol 2018; 56:4653-4679. [DOI: 10.1007/s12035-018-1396-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/14/2018] [Indexed: 12/14/2022]
|
16
|
Schulte JT, Wierenga CJ, Bruining H. Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions. Neurosci Biobehav Rev 2018; 90:260-271. [PMID: 29729285 DOI: 10.1016/j.neubiorev.2018.05.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/20/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
Neuronal chloride regulation is a determinant factor for the dynamic tuning of GABAergic inhibition during and beyond brain development. This regulation is mainly dependent on the two co-transporters K+/Cl- co-transporter KCC2 and Na+/K+/Cl- co-transporter NKCC1, whose activity can decrease or increase neuronal chloride concentrations respectively. Altered expression and/or activity of either of these co-transporters has been associated with a wide variety of brain disorders including developmental disorders, epilepsy, schizophrenia and stroke. Here, we review current knowledge on chloride transporter expression and activity regulation and highlight the intriguing potential for existing and future interventions to support chloride homeostasis across a wide range of mental disorders and neurological conditions.
Collapse
Affiliation(s)
- Joran T Schulte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands
| | - Corette J Wierenga
- Division of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center, Heidelberglaan 100, 3508 GA Utrecht The Netherlands.
| |
Collapse
|
17
|
Kashima R, Hata A. The role of TGF-β superfamily signaling in neurological disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:106-120. [PMID: 29190314 PMCID: PMC5846707 DOI: 10.1093/abbs/gmx124] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
The TGF-β superfamily signaling is involved in a variety of biological processes during embryogenesis and in adult tissue homeostasis. Faulty regulation of the signaling pathway that transduces the TGF-β superfamily signals accordingly leads to a number of ailments, such as cancer and cardiovascular, metabolic, urinary, intestinal, skeletal, and immune diseases. In recent years, a number of studies have elucidated the essential roles of TGF-βs and BMPs during neuronal development in the maintenance of appropriate innervation and neuronal activity. The new advancement implicates significant roles of the aberrant TGF-β superfamily signaling in the pathogenesis of neurological disorders. In this review, we compile a number of reports implicating the deregulation of TGF-β/BMP signaling pathways in the pathogenesis of cognitive and neurodegenerative disorders in animal models and patients. We apologize in advance that the review falls short of providing details of the role of TGF-β/BMP signaling or mechanisms underlying the pathogenesis of neurological disorders. The goal of this article is to reveal a gap in our knowledge regarding the association between TGF-β/BMP signaling pathways and neuronal tissue homeostasis and development and facilitate the research with a potential to develop new therapies for neurological ailments by modulating the pathways.
Collapse
Affiliation(s)
- Risa Kashima
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
18
|
Meyers EA, Kessler JA. TGF-β Family Signaling in Neural and Neuronal Differentiation, Development, and Function. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022244. [PMID: 28130363 DOI: 10.1101/cshperspect.a022244] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Signaling by the transforming growth factor β (TGF-β) family is necessary for proper neural development and function throughout life. Sequential waves of activation, inhibition, and reactivation of TGF-β family members regulate numerous elements of the nervous system from the earliest stages of embryogenesis through adulthood. This review discusses the expression, regulation, and function of TGF-β family members in the central nervous system at various developmental stages, beginning with induction and patterning of the nervous system to their importance in the adult as modulators of inflammatory response and involvement in degenerative diseases.
Collapse
Affiliation(s)
- Emily A Meyers
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John A Kessler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
19
|
Astrocyte Transforming Growth Factor Beta 1 Protects Synapses against Aβ Oligomers in Alzheimer's Disease Model. J Neurosci 2017; 37:6797-6809. [PMID: 28607171 DOI: 10.1523/jneurosci.3351-16.2017] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive decline, increasingly attributed to neuronal dysfunction induced by amyloid-β oligomers (AβOs). Although the impact of AβOs on neurons has been extensively studied, only recently have the possible effects of AβOs on astrocytes begun to be investigated. Given the key roles of astrocytes in synapse formation, plasticity, and function, we sought to investigate the impact of AβOs on astrocytes, and to determine whether this impact is related to the deleterious actions of AβOs on synapses. We found that AβOs interact with astrocytes, cause astrocyte activation and trigger abnormal generation of reactive oxygen species, which is accompanied by impairment of astrocyte neuroprotective potential in vitro We further show that both murine and human astrocyte conditioned media (CM) increase synapse density, reduce AβOs binding, and prevent AβO-induced synapse loss in cultured hippocampal neurons. Both a neutralizing anti-transforming growth factor-β1 (TGF-β1) antibody and siRNA-mediated knockdown of TGF-β1, previously identified as an important synaptogenic factor secreted by astrocytes, abrogated the protective action of astrocyte CM against AβO-induced synapse loss. Notably, TGF-β1 prevented hippocampal dendritic spine loss and memory impairment in mice that received an intracerebroventricular infusion of AβOs. Results suggest that astrocyte-derived TGF-β1 is part of an endogenous mechanism that protects synapses against AβOs. By demonstrating that AβOs decrease astrocyte ability to protect synapses, our results unravel a new mechanism underlying the synaptotoxic action of AβOs in AD.SIGNIFICANCE STATEMENT Alzheimer's disease is characterized by progressive cognitive decline, mainly attributed to synaptotoxicity of the amyloid-β oligomers (AβOs). Here, we investigated the impact of AβOs in astrocytes, a less known subject. We show that astrocytes prevent synapse loss induced by AβOs, via production of transforming growth factor-β1 (TGF-β1). We found that AβOs trigger morphological and functional alterations in astrocytes, and impair their neuroprotective potential. Notably, TGF-β1 reduced hippocampal dendritic spine loss and memory impairment in mice that received intracerebroventricular infusions of AβOs. Our results describe a new mechanism underlying the toxicity of AβOs and indicate novel therapeutic targets for Alzheimer's disease, mainly focused on TGF-β1 and astrocytes.
Collapse
|
20
|
Qiao J, Rong L, Wang Z, Zhang M. Involvement of Akt/GSK3β/CREB signaling pathway on chronic omethoate induced depressive-like behavior and improvement effects of combined lithium chloride and astaxanthin treatment. Neurosci Lett 2017; 649:55-61. [PMID: 28366776 DOI: 10.1016/j.neulet.2017.03.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 03/26/2017] [Accepted: 03/28/2017] [Indexed: 11/25/2022]
Abstract
Chronic organophosphorus pesticides (OP) exposure is associated with an increased risk of depression, and there is an urgent need to find an effective treatment for the depressive-like symptoms caused by OP. The main purpose of this study was to investigate whether combined lithium chloride (LiCl) and astaxanthin (AST) treatment would manifest synergetic antidepressant effects on mice with chronic OP exposure, and to determine the role of the Akt/GSK3β/CREB signaling pathway. Our results showed that chronic omethoate exposure significantly increased immobility time in behavioral tests and induced neuron damage in HE staining. The expression of p-GSK3β, p-CREB, p-PI3K and p-Akt in hippocampus after OP exposure were significantly down-regulated, while the influences were reversed by LiCl and AST treatment. Moreover, the combined application of AST and LiCl had synergistic therapeutic effects compared to LiCl and AST treatment alone, the expression of p-GSK3β, p-CREB, p-PI3K and p-Akt after combined LiCl-AST treatment were significantly higher than that with single drug application. These results showed that the combination of LiCl and AST could efficiently ameliorate depressive-like behavior induced by omethoate, and Akt/GSK3β/CREB signaling pathway might be responsible for the neuroprotective effect.
Collapse
Affiliation(s)
- Jingwen Qiao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lixia Rong
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhaoyue Wang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Meizeng Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
21
|
Yanguas-Casás N, Barreda-Manso MA, Pérez-Rial S, Nieto-Sampedro M, Romero-Ramírez L. TGFβ Contributes to the Anti-inflammatory Effects of Tauroursodeoxycholic Acid on an Animal Model of Acute Neuroinflammation. Mol Neurobiol 2016; 54:6737-6749. [PMID: 27744574 DOI: 10.1007/s12035-016-0142-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
The bile acid conjugate tauroursodeoxycholic acid (TUDCA) is a neuroprotective agent in various animal models of neuropathologies. We have previously shown the anti-inflammatory properties of TUDCA in an animal model of acute neuroinflammation. Here, we present a new anti-inflammatory mechanism of TUDCA through the regulation of transforming growth factor β (TGFβ) pathway. The bacterial lipopolysaccharide (LPS) was injected intravenously (iv) on TGFβ reporter mice (Smad-binding element (SBE)/Tk-Luc) to study in their brains the real-time activation profile of the TGFβ pathway in a non-invasive way. The activation of the TGFβ pathway in the brain of SBE/Tk-Luc mice increased 24 h after LPS injection, compared to control animals. This activation peak increased further in mice treated with both LPS and TUDCA than in mice treated with LPS only. The enhanced TGFβ activation in mice treated with LPS and TUDCA correlated with both an increase in TGFβ3 transcript in mouse brain and an increase in TGFβ3 immunoreactivity in microglia/macrophages, endothelial cells, and neurons. Inhibition of the TGFβ receptor with SB431542 drug reverted the effect of TUDCA on microglia/macrophages activation and on TGFβ3 immunoreactivity. Under inflammatory conditions, treatment with TUDCA enhanced further the activation of TGFβ pathway in mouse brain and increased the expression of TGFβ3. Therefore, the induction of TGFβ3 by TUDCA might act as a positive feedback, increasing the initial activation of the TGFβ pathway by the inflammatory stimulus. Our findings provide proof-of-concept that TGFβ contributes to the anti-inflammatory effect of TUDCA under neuroinflammatory conditions.
Collapse
Affiliation(s)
- Natalia Yanguas-Casás
- Laboratorio de Plasticidad Neural. Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071, Toledo, Spain.,Laboratorio de Plasticidad Neural, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
| | - M Asunción Barreda-Manso
- Laboratorio de Plasticidad Neural. Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071, Toledo, Spain.,Laboratorio de Plasticidad Neural, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
| | - Sandra Pérez-Rial
- Laboratorio de Neumología, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES, Avenida Reyes Católicos 2, 28040, Madrid, Spain
| | - Manuel Nieto-Sampedro
- Laboratorio de Plasticidad Neural. Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071, Toledo, Spain.,Laboratorio de Plasticidad Neural, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
| | - Lorenzo Romero-Ramírez
- Laboratorio de Plasticidad Neural. Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda s/n, 45071, Toledo, Spain.
| |
Collapse
|
22
|
Roussa E, Speer JM, Chudotvorova I, Khakipoor S, Smirnov S, Rivera C, Krieglstein K. The membrane trafficking and functionality of the K+-Cl- co-transporter KCC2 is regulated by TGF-β2. J Cell Sci 2016; 129:3485-98. [PMID: 27505893 PMCID: PMC5047681 DOI: 10.1242/jcs.189860] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/02/2016] [Indexed: 02/02/2023] Open
Abstract
Functional activation of the neuronal K(+)-Cl(-) co-transporter KCC2 (also known as SLC12A5) is a prerequisite for shifting GABAA responses from depolarizing to hyperpolarizing during development. Here, we introduce transforming growth factor β2 (TGF-β2) as a new regulator of KCC2 membrane trafficking and functional activation. TGF-β2 controls membrane trafficking, surface expression and activity of KCC2 in developing and mature mouse primary hippocampal neurons, as determined by immunoblotting, immunofluorescence, biotinylation of surface proteins and KCC2-mediated Cl(-) extrusion. We also identify the signaling pathway from TGF-β2 to cAMP-response-element-binding protein (CREB) and Ras-associated binding protein 11b (Rab11b) as the underlying mechanism for TGF-β2-mediated KCC2 trafficking and functional activation. TGF-β2 increases colocalization and interaction of KCC2 with Rab11b, as determined by 3D stimulated emission depletion (STED) microscopy and co-immunoprecipitation, respectively, induces CREB phosphorylation, and enhances Rab11b gene expression. Loss of function of either CREB1 or Rab11b suppressed TGF-β2-dependent KCC2 trafficking, surface expression and functionality. Thus, TGF-β2 is a new regulatory factor for KCC2 functional activation and membrane trafficking, and a putative indispensable molecular determinant for the developmental shift of GABAergic transmission.
Collapse
Affiliation(s)
- Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Jan Manuel Speer
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Ilona Chudotvorova
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Shokoufeh Khakipoor
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| | - Sergei Smirnov
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki FIN-00014, Finland
| | - Claudio Rivera
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki FIN-00014, Finland
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, Freiburg D-79104, Germany
| |
Collapse
|
23
|
Scholz B, Doidge AN, Barnes P, Hall J, Wilkinson LS, Thomas KL. The Regulation of Cytokine Networks in Hippocampal CA1 Differentiates Extinction from Those Required for the Maintenance of Contextual Fear Memory after Recall. PLoS One 2016; 11:e0153102. [PMID: 27224427 PMCID: PMC4880201 DOI: 10.1371/journal.pone.0153102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/23/2016] [Indexed: 12/17/2022] Open
Abstract
We investigated the distinctiveness of gene regulatory networks in CA1 associated with the extinction of contextual fear memory (CFM) after recall using Affymetrix GeneChip Rat Genome 230 2.0 Arrays. These data were compared to previously published retrieval and reconsolidation-attributed, and consolidation datasets. A stringent dual normalization and pareto-scaled orthogonal partial least-square discriminant multivariate analysis together with a jack-knifing-based cross-validation approach was used on all datasets to reduce false positives. Consolidation, retrieval and extinction were correlated with distinct patterns of gene expression 2 hours later. Extinction-related gene expression was most distinct from the profile accompanying consolidation. A highly specific feature was the discrete regulation of neuroimmunological gene expression associated with retrieval and extinction. Immunity-associated genes of the tyrosine kinase receptor TGFβ and PDGF, and TNF families' characterized extinction. Cytokines and proinflammatory interleukins of the IL-1 and IL-6 families were enriched with the no-extinction retrieval condition. We used comparative genomics to predict transcription factor binding sites in proximal promoter regions of the retrieval-regulated genes. Retrieval that does not lead to extinction was associated with NF-κB-mediated gene expression. We confirmed differential NF-κBp65 expression, and activity in all of a representative sample of our candidate genes in the no-extinction condition. The differential regulation of cytokine networks after the acquisition and retrieval of CFM identifies the important contribution that neuroimmune signalling plays in normal hippocampal function. Further, targeting cytokine signalling upon retrieval offers a therapeutic strategy to promote extinction mechanisms in human disorders characterised by dysregulation of associative memory.
Collapse
Affiliation(s)
- Birger Scholz
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Amie N. Doidge
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Philip Barnes
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Jeremy Hall
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Lawrence S. Wilkinson
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
- Schools of Psychology and Medicine, Behavioral Genetics Group, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics and Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kerrie L. Thomas
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Neuroscience & Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
24
|
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: 16] [Impact Index Per Article: 2.0] [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
|
25
|
Qu L, Gao Y, Sun H, Wang H, Liu X, Sun D. Role of PTEN-Akt-CREB Signaling Pathway in Nervous System impairment of Rats with Chronic Arsenite Exposure. Biol Trace Elem Res 2016; 170:366-72. [PMID: 26296331 DOI: 10.1007/s12011-015-0478-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/12/2015] [Indexed: 11/25/2022]
Abstract
The nervous system is a target of arsenic toxicity. Phosphatase and tensin homologue deleted on chromosome 10/protein kinase B/cAMP-response element binding protein (PTEN/Akt/CREB) signaling pathway has been reported to be involved in maintaining normal function of the nervous system, modulating growth and proliferation of neurocyte, regulating neuron synaptic plasticity, and long-term memory. And many studies have demonstrated that expressions of PTEN, Akt, and CREB protein were influenced by arsenic, but it is not clear whether this signaling pathway is involved in the nervous system impairment of rats induced by chronic arsenite exposure, and we have addressed this in this study. Eighty male Sprague-Dawley (SD) rats were randomly divided into eight groups (n = 10 each), four groups exposed to NaAsO2 (0, 5, 10, and 50 mg/L NaAsO2 in drinking water) for 3 months, the other four groups exposed to NaAsO2 (0, 5, 10, 50 mg/L NaAsO2 in drinking water) for 6 months. Hematoxylin and eosin (HE) staining showed that chronic arsenite exposure induced varying degrees of damage in cerebral neurons. And arsenite exposure increased arsenic amount in serum and brain samples in a dose- and time-dependent manner. Moreover, the protein levels of PTEN and Akt in brain tissue were not significantly changed compared with the control group, but p-Akt, CREB, and p-CREB were all significantly downregulated in arsenite-exposed groups with a dose-dependent pattern. These results suggested that chronic arsenite exposure negatively regulated the PTEN-Akt-CREB signaling pathway, and dysfunction of the signaling pathway might be one of the mechanisms of nervous system impairment induced by chronic arsenite exposure.
Collapse
Affiliation(s)
- Lisha Qu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China
| | - Hongna Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China
| | - Hui Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China
| | - Dianjun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiologic Epidemiology of Ministry of Health and Education Bureau of Heilongjiang Province(23618504), Harbin Medical University, 157 Baojian Road, Harbin, 150081, China.
| |
Collapse
|
26
|
A key role for TGF-β1 in hippocampal synaptic plasticity and memory. Sci Rep 2015; 5:11252. [PMID: 26059637 PMCID: PMC4462026 DOI: 10.1038/srep11252] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 05/08/2015] [Indexed: 01/06/2023] Open
Abstract
Transforming Growth Factor β1 (TGF-β1) is a well-known neuroprotective and neurotrophic factor demonstrated to play a role in synaptic transmission. However, its involvement in physiological mechanisms underlying synaptic plasticity and memory at hippocampal level has not been thoroughly investigated. Here, we examine the role of TGF-β1 in hippocampal long-term potentiation (LTP) and memory in adult wild type mice. Our data provide evidence that administration of exogenous TGF-β1 is able to convert early-phase-LTP into late-phase-LTP. Furthermore, we show that the block of the endogenous TGF-β1 signaling pathway by the specific TGF-β1 inhibitor SB431542, impairs LTP and object recognition memory. The latter impairment was rescued by administration of exogenous TGF-β1, suggesting that endogenously produced TGF-β1 plays a role in physiological mechanisms underlying LTP and memory. Finally, TGF-β1 functional effect correlates with an increased expression of the phosphorylated transcription factor cAMP-Responsive Element Binding protein.
Collapse
|
27
|
McGehee AM, Moss BJ, Juo P. The DAF-7/TGF-β signaling pathway regulates abundance of the Caenorhabditis elegans glutamate receptor GLR-1. Mol Cell Neurosci 2015; 67:66-74. [PMID: 26054666 DOI: 10.1016/j.mcn.2015.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/03/2015] [Indexed: 11/24/2022] Open
Abstract
Transforming growth factor-β (TGF-β) family signaling pathways have roles in both neuronal development and the regulation of synaptic function. Here we identify a novel role for the Caenorhabditis elegans DAF-7/TGF-β signaling pathway in the regulation of the AMPA-type glutamate receptor GLR-1. We found that the abundance of GLR-1 increases at synapses in the ventral nerve cord (VNC) of animals with loss-of-function mutations in multiple DAF-7/TGF-β pathway components including the TGF-β ligand DAF-7, the type I receptor DAF-1, and the Smads DAF-8 and DAF-14. The GLR-1 defect can be rescued by expression of daf-8 specifically in glr-1-expressing interneurons. The effect on GLR-1 was specific for the DAF-7 pathway because mutations in the DBL-1/TGF-β family pathway did not increase GLR-1 levels in the VNC. Immunoblot analysis indicates that total levels of GLR-1 protein are increased in neurons of DAF-7/TGF-β pathway mutants. The increased abundance of GLR-1 in the VNC of daf-7 pathway mutants is dependent on the transcriptional regulator DAF-3/Smad suggesting that DAF-3-dependent transcription controls GLR-1 levels. Furthermore, we found that glr-1 transcription is increased in daf-7 mutants based on a glr-1 transcriptional reporter. Together these results suggest that the DAF-7/TGF-β signaling pathway functions in neurons and negatively regulates the abundance of GLR-1, in part, by controlling transcription of the receptor itself. Finally, DAF-7/TGF-β pathway mutants exhibit changes in spontaneous locomotion that are dependent on endogenous GLR-1 and consistent with increased glutamatergic signaling. These results reveal a novel mechanism by which TGF-β signaling functions in the nervous system to regulate behavior.
Collapse
Affiliation(s)
- Annette M McGehee
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA; Department of Biology, Suffolk University, Boston, MA 02114, USA.
| | - Benjamin J Moss
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA; Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Peter Juo
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
28
|
Doxorubicin attenuates serotonin-induced long-term synaptic facilitation by phosphorylation of p38 mitogen-activated protein kinase. J Neurosci 2015; 34:13289-300. [PMID: 25274809 DOI: 10.1523/jneurosci.0538-14.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline used widely for cancer chemotherapy. Its primary mode of action appears to be topoisomerase II inhibition, DNA cleavage, and free radical generation. However, in non-neuronal cells, DOX also inhibits the expression of dual-specificity phosphatases (also referred to as MAPK phosphatases) and thereby inhibits the dephosphorylation of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38 MAPK), two MAPK isoforms important for long-term memory (LTM) formation. Activation of these kinases by DOX in neurons, if present, could have secondary effects on cognitive functions, such as learning and memory. The present study used cultures of rat cortical neurons and sensory neurons (SNs) of Aplysia to examine the effects of DOX on levels of phosphorylated ERK (pERK) and phosphorylated p38 (p-p38) MAPK. In addition, Aplysia neurons were used to examine the effects of DOX on long-term enhanced excitability, long-term synaptic facilitation (LTF), and long-term synaptic depression (LTD). DOX treatment led to elevated levels of pERK and p-p38 MAPK in SNs and cortical neurons. In addition, it increased phosphorylation of the downstream transcriptional repressor cAMP response element-binding protein 2 in SNs. DOX treatment blocked serotonin-induced LTF and enhanced LTD induced by the neuropeptide Phe-Met-Arg-Phe-NH2. The block of LTF appeared to be attributable to overriding inhibitory effects of p-p38 MAPK, because LTF was rescued in the presence of an inhibitor (SB203580 [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole]) of p38 MAPK. These results suggest that acute application of DOX might impair the formation of LTM via the p38 MAPK pathway.
Collapse
|
29
|
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: 2.0] [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
|
30
|
Diniz LP, Matias ICP, Garcia MN, Gomes FCA. Astrocytic control of neural circuit formation: highlights on TGF-beta signaling. Neurochem Int 2014; 78:18-27. [PMID: 25125369 DOI: 10.1016/j.neuint.2014.07.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/18/2014] [Accepted: 07/28/2014] [Indexed: 01/23/2023]
Abstract
Brain function depends critically on the coordinated activity of presynaptic and postsynaptic signals derived from both neurons and non-neuronal elements such as glial cells. A key role for astrocytes in neuronal differentiation and circuitry formation has emerged within the last decade. Although the function of glial cells in synapse formation, elimination and efficacy has greatly increased, we are still very far from deeply understanding the molecular and cellular mechanism underlying these events. The present review discusses the mechanisms driving astrocytic control of excitatory and inhibitory synapse formation in the central nervous system, especially the mechanisms mediated by soluble molecules, particularly those from the TGF-β family. Further, we discuss whether and how human astrocytes might contribute to the acquisition of human cognition. We argue that understanding how astrocytic signals regulate synaptic development might offer new insights into human perception, learning, memory, and cognition and, ultimately, provide new targets for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Luan Pereira Diniz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Isadora C Pereira Matias
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Matheus Nunes Garcia
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
31
|
Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol 2014; 141:575-89. [PMID: 25005789 DOI: 10.1007/s00432-014-1772-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/30/2014] [Indexed: 12/13/2022]
Abstract
Malignant gliomas invariably recur after irradiation, showing radioresistance. Meanwhile, cranial irradiation can bring some risk for developing cognitive dysfunction. There is increasing evidence that cytokines play their peculiar roles in these processes. On the one hand, cytokines directly influence the progression of malignant glioma, promoting or suppressing tumor progression. On the other hand, cytokines indirectly contribute to the immunologic response against gliomas, exhibiting pro-inflammatory or immunosuppressive activities. We propose that cytokines are not simply unregulated products from tumor cells or immune cells, but mediators finely adjust the balance between glioma cells and tumor microenvironment after irradiation. The paper, therefore, focuses on the changes of cytokines after irradiation, analyzing how these mediate the response of tumor cells and normal cells to irradiation. In addition, cytokine-based immunotherapeutic strategies, accompanied with irradiation, for the treatment of gliomas are also discussed.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Radiation Oncology, Cancer Centre, Qilu Hospital, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | | | | | | | | |
Collapse
|
32
|
Poon VY, Choi S, Park M. Growth factors in synaptic function. Front Synaptic Neurosci 2013; 5:6. [PMID: 24065916 PMCID: PMC3776238 DOI: 10.3389/fnsyn.2013.00006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/29/2013] [Indexed: 12/15/2022] Open
Abstract
Synapses are increasingly recognized as key structures that malfunction in disorders like schizophrenia, mental retardation, and neurodegenerative diseases. The importance and complexity of the synapse has fuelled research into the molecular mechanisms underlying synaptogenesis, synaptic transmission, and plasticity. In this regard, neurotrophic factors such as netrin, Wnt, transforming growth factor-β (TGF-β), tumor necrosis factor-α (TNF-α), and others have gained prominence for their ability to regulate synaptic function. Several of these factors were first implicated in neuroprotection, neuronal growth, and axon guidance. However, their roles in synaptic development and function have become increasingly clear, and the downstream signaling pathways employed by these factors have begun to be elucidated. In this review, we will address the role of these factors and their downstream effectors in synaptic function in vivo and in cultured neurons.
Collapse
Affiliation(s)
- Vivian Y Poon
- Neuroscience and Behavioral Disorders Program, Duke-NUS Graduate Medical School Singapore, Singapore
| | | | | |
Collapse
|
33
|
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: 166] [Impact Index Per Article: 13.8] [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
|
34
|
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: 3.3] [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
|
35
|
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: 173] [Impact Index Per Article: 14.4] [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
|
36
|
Krieglstein K, Zheng F, Unsicker K, Alzheimer C. More than being protective: functional roles for TGF-β/activin signaling pathways at central synapses. Trends Neurosci 2011; 34:421-9. [PMID: 21742388 DOI: 10.1016/j.tins.2011.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 12/14/2022]
Abstract
It is becoming increasingly clear that members of the transforming growth factor-β (TGF-β) family have roles in the central nervous system that extend beyond their well-established roles as neurotrophic and neuroprotective factors. Recent findings have indicated that the TGF-β signaling pathways are involved in the modulation of both excitatory and inhibitory synaptic transmission in the adult mammalian brain. In this review, we discuss how TGF-β, bone morphogenetic protein and activin signaling at central synapses modulate synaptic plasticity, cognition and affective behavior. We also discuss the implications of these findings for the molecular understanding and potential treatment of neuropsychiatric diseases, such as anxiety, depression and other neurological disorders.
Collapse
Affiliation(s)
- Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | | | | | | |
Collapse
|
37
|
Kim NC, Marqués G. Identification of downstream targets of the bone morphogenetic protein pathway in the Drosophila nervous system. Dev Dyn 2011; 239:2413-25. [PMID: 20652954 DOI: 10.1002/dvdy.22368] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Bone Morphogenetic Protein (BMP) signaling mediated by the receptor Wishful thinking (Wit) is essential for nervous system development in Drosophila. Mutants lacking wit function show defects in neuromuscular junction development and function, specification of neurosecretory phenotypes, and eclosion behavior that result in lethality. The ligand is Glass bottom boat, the Drosophila ortholog of mammalian BMP-7, which acts as a retrograde signal through the Wit receptor. In order to identify transcriptional targets of the BMP pathway in the Drosophila nervous system, we have analyzed the gene expression profile of wit mutant larval central nervous system. Genes differentially expressed identified by microarray analysis have been verified by quantitative PCR and studied by in situ hybridization. Among the genes thus identified, we find solute transporters, neuropeptides, mitochondrial proteins, and novel genes. In addition, several genes are regulated by wit in an isoform-specific manner that suggest regulation of alternative splicing by BMP signaling.
Collapse
Affiliation(s)
- Nam Chul Kim
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
38
|
Abstract
Activins, which are members of the TGF-β superfamily, were initially isolated from gonads and served as modulators of follicle-stimulating hormone secretion. Activins regulate various biological functions, including induction of the dorsal mesoderm, craniofacial development, and differentiation of numerous cell types. Activin receptors are highly expressed in neuronal cells, and activin mRNA expression is upregulated by neuronal activity. Activins also exhibit neuroprotective action during excitotoxic brain injury. However, very little is known about the functional roles of activins in the brain. We recently generated various types of transgenic mice, demonstrating that activins regulate spine formation, behavioral activity, anxiety, adult neurogenesis, late-phase long-term potentiation, and maintenance of long-term memory. The present chapter describes recent progress in the study of the role of activin in the brain.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi, Japan
| | | |
Collapse
|
39
|
Prenatal inflammation impairs adult neurogenesis and memory related behavior through persistent hippocampal TGFβ1 downregulation. Brain Behav Immun 2010; 24:1301-9. [PMID: 20600816 DOI: 10.1016/j.bbi.2010.06.005] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/04/2010] [Accepted: 06/11/2010] [Indexed: 11/22/2022] Open
Abstract
Prenatal exposure to inflammatory stimuli is known to influence adult brain function. In addition, adult hippocampal neurogenesis is impaired by a local pro-inflammatory microenvironment. On this basis, we hypothesized that a pro-inflammatory insult during gestation would have negative effects on adult neurogenesis in the offspring. Pregnant Wistar rats received subcutaneous injections of lipopolysaccharide (LPS; 0.5mg/kg) or saline every other day from gestational day 14 to 20. The adult offspring prenatally treated with LPS showed a decrease in the proliferating cells and the newborn neurons of the dentate gyrus. Furthermore, prenatal LPS treatment impaired performance in the neurogenesis-dependent novel object recognition test. Maternal care was impaired by prenatal LPS administration but did not contribute to the effects of prenatal LPS on adult neurogenesis. Persistent microglial activation and downregulated expression of transforming growth factor beta-1 (TGFβ(1)) occurred specifically in the adult hippocampus of animals treated prenatally with LPS. Importantly, chronic hippocampal TGFβ(1) overexpression restored neurogenesis as well as recognition memory performance to control levels. These findings demonstrate that prenatal inflammation triggered by LPS impairs adult neurogenesis and recognition memory. Furthermore, we provide a model of reduced adult neurogenesis with long-lasting defined alterations in the neurogenic niche. Finally, we show that the expression of a single cytokine (TGFβ(1)) in the hippocampus can restore adult neurogenesis and its related behavior, highlighting the role of TGFβ(1) in these processes.
Collapse
|
40
|
Vincze C, Pál G, Wappler EA, Szabó ER, Nagy ZG, Lovas G, Dobolyi A. Distribution of mRNAs encoding transforming growth factors-beta1, -2, and -3 in the intact rat brain and after experimentally induced focal ischemia. J Comp Neurol 2010; 518:3752-70. [PMID: 20653032 DOI: 10.1002/cne.22422] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transforming growth factors-beta1 (TGF-beta1), -2, and -3 form a small group of related proteins involved in the regulation of proliferation, differentiation, and survival of various cell types. Recently, TGF-betas were also demonstrated to be neuroprotective. In the present study, we investigated their distribution in the rat brain as well as their expression following middle cerebral artery occlusion. Probes were produced for all types of TGF-betas, and in situ hybridization was performed. We demonstrated high TGF-beta1 expression in cerebral cortex, hippocampus, central amygdaloid nucleus, medial preoptic area, hypothalamic paraventricular nucleus, substantia nigra, brainstem reticular formation and motoneurons, and area postrema. In contrast, TGF-beta2 was abundantly expressed in deep cortical layers, dentate gyrus, midline thalamic nuclei, posterior hypothalamic area and mamillary body, superior olive, areas of monoaminergic neurons, spinal trigeminal nucleus, dorsal vagal complex, cerebellum, and choroid plexus, and a high level of TGF-beta3 mRNA was found in cerebral cortex, hippocampus, basal amygdaloid nuclei, lateral septal nucleus, several thalamic nuclei, arcuate and supramamillary nuclei, superior colliculus, superior olive, brainstem reticular formation and motoneurons, area postrema, and inferior olive. Focal brain ischemia induced TGF-betas with markedly different expression patterns. TGF-beta1 was induced in the penumbral region of cortex and striatum, whereas TGF-beta2 and -beta3 were induced in different layers of the ipsilateral cortex. The expression of the subtypes of TGF-betas in different brain regions suggests that they are involved in the regulation of different neurons and bind to different latent TGF-beta binding proteins. Furthermore, they might have subtype-specific functions following ischemic attack.
Collapse
Affiliation(s)
- Csilla Vincze
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary
| | | | | | | | | | | | | |
Collapse
|
41
|
Garay PA, McAllister AK. Novel roles for immune molecules in neural development: implications for neurodevelopmental disorders. Front Synaptic Neurosci 2010; 2:136. [PMID: 21423522 PMCID: PMC3059681 DOI: 10.3389/fnsyn.2010.00136] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 08/03/2010] [Indexed: 11/18/2022] Open
Abstract
Although the brain has classically been considered “immune-privileged”, current research suggests an extensive communication between the immune and nervous systems in both health and disease. Recent studies demonstrate that immune molecules are present at the right place and time to modulate the development and function of the healthy and diseased central nervous system (CNS). Indeed, immune molecules play integral roles in the CNS throughout neural development, including affecting neurogenesis, neuronal migration, axon guidance, synapse formation, activity-dependent refinement of circuits, and synaptic plasticity. Moreover, the roles of individual immune molecules in the nervous system may change over development. This review focuses on the effects of immune molecules on neuronal connections in the mammalian central nervous system – specifically the roles for MHCI and its receptors, complement, and cytokines on the function, refinement, and plasticity of geniculate, cortical and hippocampal synapses, and their relationship to neurodevelopmental disorders. These functions for immune molecules during neural development suggest that they could also mediate pathological responses to chronic elevations of cytokines in neurodevelopmental disorders, including autism spectrum disorders (ASD) and schizophrenia.
Collapse
Affiliation(s)
- Paula A Garay
- Laboratory of Dr. A.K. McAllister, Department of Neurobiology, Physiology, and Behavior, Center for Neuroscience, University of California Davis, CA, USA
| | | |
Collapse
|
42
|
TGF-beta2 alters the characteristics of the neuromuscular junction by regulating presynaptic quantal size. Proc Natl Acad Sci U S A 2010; 107:13515-9. [PMID: 20624974 DOI: 10.1073/pnas.1001695107] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The amount of neurotransmitter released from a presynaptic terminal is the product of the quantal content (number of vesicles) and the presynaptic quantal size (QSpre, amount of transmitter per vesicle). QSpre varies with synaptic use, but its regulation is poorly understood. The motor nerve terminals at the neuromuscular junction (NMJ) contain TGF-beta receptors. We present evidence that TGF-beta2 regulates QSpre at the NMJ. Application of TGF-beta2 to the rat diaphragm NMJ increased the postsynaptic response to both spontaneous and evoked release of acetylcholine, whereas antibodies to TGF-beta2 or its receptor had the converse effect. L-vesamicol and bafilomycin blocked the actions of TGF-beta2, indicating that TGF-beta2 acts by altering the extent of vesicular filling. Recordings of the postsynaptic currents from the diaphragm were consistent with TGF-beta2 having this presynaptic action and a lesser postsynaptic effect on input resistance. TGF-beta2 also decreased quantal content by an atropine-sensitive pathway, indicating that this change is secondary to cholinergic feedback on vesicular release. Consequently, the net actions of TGF-beta2 at the NMJ were to amplify the postsynaptic effects of spontaneous transmission and to diminish the number of vesicles used per evoked stimulus, without diminishing the amount of acetylcholine released.
Collapse
|
43
|
Canonical TGF-beta signaling is required for the balance of excitatory/inhibitory transmission within the hippocampus and prepulse inhibition of acoustic startle. J Neurosci 2010; 30:6025-35. [PMID: 20427661 DOI: 10.1523/jneurosci.0789-10.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Smad4 is a unique nuclear transducer for all TGF-beta signaling pathways and regulates gene transcription during development and tissue homeostasis. To elucidate the postnatal role of TGF-beta signaling in the mammalian brain, we generated forebrain-specific Smad4 knock-out mice. Surprisingly, the mutants showed no alteration in long-term potentiation and water maze, suggesting that Smad4 is not required for spatial learning and memory. However, these mutant mice did show enhancement of paired-pulse facilitation in excitatory synaptic transmission and stronger paired-pulse depression of GABA(A) currents in the hippocampus. The alteration of hippocampal electrophysiology correlated with mouse hyperactivity in homecage and open field tests. Mutant mice also showed overgrooming as well as deficits of prepulse inhibition, a widely used endophenotype of schizophrenia. With a specific real-time PCR array focused on TGF-beta signaling pathway, we identified a novel regulation mechanism of the pathway in the hippocampal neurons, in which Smad4-mediated signaling suppresses the level of extracellular antagonism of TGF-beta ligands through transcriptional regulation of follistatin, a selective inhibitor to activin/TGF-beta signaling in the hippocampus. In summary, we suggest that the canonical TGF-beta signaling pathway is critical for use-dependent modulation of GABA(A) synaptic transmission and dendritic homeostasis; furthermore, a disruption in the balance of the excitatory and inhibitory hippocampal network can result in psychiatric-like behavior.
Collapse
|
44
|
Ageta H, Ikegami S, Miura M, Masuda M, Migishima R, Hino T, Takashima N, Murayama A, Sugino H, Setou M, Kida S, Yokoyama M, Hasegawa Y, Tsuchida K, Aosaki T, Inokuchi K. Activin plays a key role in the maintenance of long-term memory and late-LTP. Learn Mem 2010; 17:176-85. [PMID: 20332189 DOI: 10.1101/lm.16659010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A recent study has revealed that fear memory may be vulnerable following retrieval, and is then reconsolidated in a protein synthesis-dependent manner. However, little is known about the molecular mechanisms of these processes. Activin betaA, a member of the TGF-beta superfamily, is increased in activated neuronal circuits and regulates dendritic spine morphology. To clarify the role of activin in the synaptic plasticity of the adult brain, we examined the effect of inhibiting or enhancing activin function on hippocampal long-term potentiation (LTP). We found that follistatin, a specific inhibitor of activin, blocked the maintenance of late LTP (L-LTP) in the hippocampus. In contrast, administration of activin facilitated the maintenance of early LTP (E-LTP). We generated forebrain-specific activin- or follistatin-transgenic mice in which transgene expression is under the control of the Tet-OFF system. Maintenance of hippocampal L-LTP was blocked in the follistatin-transgenic mice. In the contextual fear-conditioning test, we found that follistatin blocked the formation of long-term memory (LTM) without affecting short-term memory (STM). Furthermore, consolidated memory was selectively weakened by the expression of follistatin during retrieval, but not during the maintenance phase. On the other hand, the maintenance of memory was also influenced by activin overexpression during the retrieval phase. Thus, the level of activin in the brain during the retrieval phase plays a key role in the maintenance of long-term memory.
Collapse
Affiliation(s)
- Hiroshi Ageta
- Mitsubishi Kagaku Institute of Life Sciences, MITILS, Machida, Tokyo 194-8511, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ghose S, Gleason KA, Potts BW, Lewis-Amezcua K, Tamminga CA. Differential expression of metabotropic glutamate receptors 2 and 3 in schizophrenia: a mechanism for antipsychotic drug action? Am J Psychiatry 2009; 166:812-20. [PMID: 19487395 PMCID: PMC2860261 DOI: 10.1176/appi.ajp.2009.08091445] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Preclinical and clinical data implicate the group II metabotropic glutamate receptors mGluR2 and mGluR3 in the pathophysiology of schizophrenia. Moreover, a recent phase II clinical trial demonstrated the antipsychotic efficacy of a mGluR2/mGluR3 agonist. The purpose of the present study was to distinguish the expression of mGluR2 and mGluR3 receptor proteins in schizophrenia and to quantify glutamate carboxypeptidase II (GCP II) in order to explore a role for the metabotropic receptors in schizophrenia therapeutics. GCP II is an enzyme that metabolizes N-acetyl-aspartyl-glutamate (NAAG), which is the only known specific endogenous agonist of mGluR3 in the mammalian brain. METHOD The normal expression levels of mGluR2, mGluR3, and GCP II were determined for 10 regions of the postmortem human brain using specific antibodies. Differences in expression levels of each protein were examined in the dorsolateral prefrontal cortex, temporal cortex, and motor cortex in 15 postmortem schizophrenia subjects and 15 postmortem matched normal comparison subjects. Chronic antipsychotic treatment in rodents was conducted to examine the potential effect of antipsychotic drugs on expression of the three proteins. RESULTS Findings revealed a significant increase in GCP II protein and a reduction in mGluR3 protein in the dorsolateral prefrontal cortex in schizophrenia subjects, with mGluR2 protein levels unchanged. Chronic antipsychotic treatment in rodents did not influence GCP II or mGluR3 levels. CONCLUSIONS Increased GCP II expression and low mGluR3 expression in the dorsolateral prefrontal cortex suggest that NAAG-mediated signaling is impaired in this brain region in schizophrenia. Further, these data implicate the mGluR3 receptor in the antipsychotic action of mGluR2/mGluR3 agonists.
Collapse
Affiliation(s)
- Subroto Ghose
- University of Texas Southwestern Medical Center, NE5.110C, Dallas, TX 75390-9127, USA.
| | | | | | | | | |
Collapse
|
46
|
Polazzi E, Altamira LEP, Eleuteri S, Barbaro R, Casadio C, Contestabile A, Monti B. Neuroprotection of microglial conditioned medium on 6-hydroxydopamine-induced neuronal death: role of transforming growth factor beta-2. J Neurochem 2009; 110:545-56. [PMID: 19457129 DOI: 10.1111/j.1471-4159.2009.06117.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Microglia, the immune cells of the CNS, play essential roles in both physiological and pathological brain states. Here we have used an in vitro model to demonstrate neuroprotection of a 48 h-microglial conditioned medium (MCM) towards cerebellar granule neurons (CGNs) challenged with the neurotoxin 6-hydroxydopamine, which induces a Parkinson-like neurodegeneration, and to identify the protective factor(s). MCM nearly completely protects CGNs from 6-hydroxydopamine neurotoxicity and at least some of the protective factor(s) are peptidic in nature. While the fraction of the medium containing molecules < 30 kDa completely protects CGNs, fractions containing molecules < 10 kDa or > 10 kDa are not neuroprotective. We further demonstrate that microglia release high amounts of transforming growth factor-beta2 (TGF-beta2) and that its exogenous addition to the fraction of the medium not containing it (< 10 kDa) fully restores the neuroprotective action. Moreover, MCM neuroprotection is significantly counteracted by an inhibitor of TGF-beta2 transduction pathway. Our results identify TGF-beta2 as an essential neuroprotective factor released by microglia in its culture medium that requires to be fully effective the concomitant presence of other factor(s) of low molecular weight.
Collapse
|
47
|
Loss of transforming growth factor-beta 2 leads to impairment of central synapse function. Neural Dev 2008; 3:25. [PMID: 18854036 PMCID: PMC2576228 DOI: 10.1186/1749-8104-3-25] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 10/14/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The formation of functional synapses is a crucial event in neuronal network formation, and with regard to regulation of breathing it is essential for life. Members of the transforming growth factor-beta (TGF-beta) superfamily act as intercellular signaling molecules during synaptogenesis of the neuromuscular junction of Drosophila and are involved in synaptic function of sensory neurons of Aplysia. RESULTS Here we show that while TGF-beta2 is not crucial for the morphology and function of the neuromuscular junction of the diaphragm muscle of mice, it is essential for proper synaptic function in the pre-Bötzinger complex, a central rhythm organizer located in the brainstem. Genetic deletion of TGF-beta2 in mice strongly impaired both GABA/glycinergic and glutamatergic synaptic transmission in the pre-Bötzinger complex area, while numbers and morphology of central synapses of knock-out animals were indistinguishable from their wild-type littermates at embryonic day 18.5. CONCLUSION The results demonstrate that TGF-beta2 influences synaptic function, rather than synaptogenesis, specifically at central synapses. The functional alterations in the respiratory center of the brain are probably the underlying cause of the perinatal death of the TGF-beta2 knock-out mice.
Collapse
|
48
|
Shan ZY, Shen JL, Li QM, Wang Y, Huang XY, Guo TY, Liu HW, Lei L, Jin LH. pCREB is involved in neural induction of mouse embryonic stem cells by RA. Anat Rec (Hoboken) 2008; 291:519-26. [PMID: 18383274 DOI: 10.1002/ar.20686] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Mouse embryonic stem (ES) cells can be induced by various chemicals to differentiate into a variety of cell types in vitro. In our study, retinoic acid (RA), one of the most important inducers, used at a concentration of 5 microM, was found to induce the differentiation of ES cells into neural progenitor cells (NPCs). During embryoid body (EB) differentiation, the level of active cyclic AMP response element-binding protein (CREB) was relatively high when 5 microM RA treatment was performed. Inhibition of CREB activity committed EBs to becoming other germ layers, whereas increased expression of CREB enhanced NPC differentiation. Moreover, RA increased the expression of active CREB by enhancing the activity of JNK. Our research suggests that CREB plays a role in RA-induced NPC differentiation by increasing the expression of active JNK.
Collapse
Affiliation(s)
- Zhi-Yan Shan
- Department of Histology and Embryology, Harbin Medical University, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dobolyi A, Palkovits M. Expression of latent transforming growth factor beta binding proteins in the rat brain. J Comp Neurol 2008; 507:1393-408. [PMID: 18196529 DOI: 10.1002/cne.21621] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transforming growth factor-betas are expressed in the brain, have neuroprotective functions, and may be involved in the pathogenesis of neurodegenerative disorders. Their intracellular processing, secretion, and extracellular activation requires latent transforming growth factor-beta binding proteins (LTBPs) as demonstrated in peripheral organs. Here, we first report that the four types of LTBPs are expressed in the rat brain based on reverse-transcriptase polymerase chain reaction (RT-PCR) and that the subtypes have different topographical distributions based on in situ hybridization histochemistry. LTBP-1 has a high expression level in several brain regions including choroid plexus, cerebral cortex, medial amygdaloid nucleus, anteromedial and midline thalamic nuclei, medial preoptic area, arcuate and dorsomedial hypothalamic nuclei, superior olive, and area postrema. LTBP-3 and -4 are the most widely distributed LTBPs. Both are abundant in the cerebral cortex, cerebellum, hypothalamus, amygdala, brainstem motor nuclei, and area postrema. In addition, LTBP-3 mRNA is also abundant in the choroid plexus, globus pallidus, anterior and reticular thalamic nuclei, mamillary body, substantia nigra, red nucleus, pontine nuclei, some brainstem sensory nuclei, and reticular formation, while LTBP-4 is more abundant in the hippocampus and the parabrachial nuclei. In contrast, the expression of LTBP-2 is restricted to cerebral cortex, CA1 neurons of the hippocampus, and perifornical/lateral hypothalamic areas. The hypothalamic cells were identified by double in situ hybridization histochemistry as orexin-synthesizing neurons, demonstrating that LTBP expression can be very specifically regulated. Our data demonstrate that each type of LTBPs have highly distinct distributional patterns suggesting that the expression of LTBPs are specifically regulated in the brain.
Collapse
Affiliation(s)
- Arpád Dobolyi
- Neuromorphological and Neuroendocrine Research Laboratory, Department of Anatomy, Histology and Embryology, Semmelweis University and the Hungarian Academy of Sciences, Budapest, H-1094, Hungary.
| | | |
Collapse
|
50
|
Lacmann A, Hess D, Gohla G, Roussa E, Krieglstein K. Activity-dependent release of transforming growth factor-beta in a neuronal network in vitro. Neuroscience 2007; 150:647-57. [DOI: 10.1016/j.neuroscience.2007.09.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 09/05/2007] [Accepted: 10/11/2007] [Indexed: 01/17/2023]
|