1
|
Zamorano-Cataldo M, Vega-Vásquez I, García-Navarrete C, Toledo J, Bustamante D, Ezquer F, Urra FA, Farfán-Troncoso N, Herrera-Marschitz M, Morales P. Mitochondrial dynamics and sex-specific responses in the developing rat hippocampus: Effect of perinatal asphyxia and mesenchymal stem cell Secretome treatment. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119851. [PMID: 39332539 DOI: 10.1016/j.bbamcr.2024.119851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/27/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024]
Abstract
AIMS Perinatal asphyxia is one of the major causes of neonatal death at birth. Survivors can progress but often suffer from long-term sequelae. We aim to determine the effects of perinatal asphyxia on mitochondrial dynamics and whether mesenchymal stem cell secretome (MSC-S) treatment can alleviate the deleterious effects. MATERIALS AND METHODS Animals were subjected to 21 min of asphyxia at the time of delivery. MSC-S or vehicle was intranasally administered 2 h post-delivery. Mitochondrial mass (D-loop, qPCR), mitochondrial dynamics proteins (Drp1, Fis1 and OPA1, Western blot), mitochondrial dynamics (TOMM20, Immunofluorescence), as well as mitochondrial membrane potential (ΔΨm) (Safranin O) were evaluated at P1 and P7 in the hippocampus. KEY FINDINGS Perinatal asphyxia increased levels of mitochondrial dynamics proteins Drp1 and S-OPA1 at P1 and Fis1 at P7. Mitochondrial density and mass were decreased at P1. Perinatal asphyxia induced sex-specific differences, with increased L-OPA1 in females at P7 and increased mitochondria circularity. In males, asphyxia-exposed animals exhibited a reduced ΔΨm at P7. MSC-S treatment normalised levels of mitochondrial dynamics proteins involved in fission. SIGNIFICANCE This study provides novel insights into the effects of perinatal asphyxia on mitochondrial dynamics in the developing brain and on the therapeutic opportunities provided by mesenchymal stem cell secretome treatment. It also highlights on the relevance of considering sex as a biological variable in perinatal brain injury and therapy development. These findings contribute to the development of targeted, personalised therapies for infants affected by perinatal asphyxia.
Collapse
Affiliation(s)
- M Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile
| | - I Vega-Vásquez
- Advanced Scientific Equipment Network (REDECA), Medical Faculty, Universidad de Chile, Chile
| | - C García-Navarrete
- Advanced Scientific Equipment Network (REDECA), Medical Faculty, Universidad de Chile, Chile
| | - J Toledo
- Advanced Scientific Equipment Network (REDECA), Medical Faculty, Universidad de Chile, Chile
| | - D Bustamante
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile
| | - F Ezquer
- Center for Regenerative Medicine, Medical Faculty, Clínica Alemana, Universidad del Desarrollo, Chile
| | - F A Urra
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile; Laboratory of Metabolic Plasticity and Bioenergetics, Molecular & Clinical Pharmacology Program, Medical Faculty, Universidad de Chile, Chile; Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics (MIBI), Medical Faculty, Universidad de Chile, Chile
| | - N Farfán-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile.
| | - M Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile.
| | - P Morales
- Molecular & Clinical Pharmacology Program, ICBM, Medical Faculty, Universidad de Chile, Chile; Department of Neuroscience, Medical Faculty, Universidad de Chile, Chile.
| |
Collapse
|
2
|
Sasaki H, Nakagawa I, Furuta T, Yokoyama S, Morisaki Y, Saito Y, Nakase H. Mitochondrial Calcium Uniporter (MCU) is Involved in an Ischemic Postconditioning Effect Against Ischemic Reperfusion Brain Injury in Mice. Cell Mol Neurobiol 2024; 44:32. [PMID: 38568450 PMCID: PMC10991049 DOI: 10.1007/s10571-024-01464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
The phenomenon of ischemic postconditioning (PostC) is known to be neuroprotective against ischemic reperfusion (I/R) injury. One of the key processes in PostC is the opening of the mitochondrial ATP-dependent potassium (mito-KATP) channel and depolarization of the mitochondrial membrane, triggering the release of calcium ions from mitochondria through low-conductance opening of the mitochondrial permeability transition pore. Mitochondrial calcium uniporter (MCU) is known as a highly sensitive transporter for the uptake of Ca2+ present on the inner mitochondrial membrane. The MCU has attracted attention as a new target for treatment in diseases, such as neurodegenerative diseases, cancer, and ischemic stroke. We considered that the MCU may be involved in PostC and trigger its mechanisms. This research used the whole-cell patch-clamp technique on hippocampal CA1 pyramidal cells from C57BL mice and measured changes in spontaneous excitatory post-synaptic currents (sEPSCs), intracellular Ca2+ concentration, mitochondrial membrane potential, and N-methyl-D-aspartate receptor (NMDAR) currents under inhibition of MCU by ruthenium red 265 (Ru265) in PostC. Inhibition of MCU increased the occurrence of sEPSCs (p = 0.014), NMDAR currents (p < 0.001), intracellular Ca2+ concentration (p < 0.001), and dead cells (p < 0.001) significantly after reperfusion, reflecting removal of the neuroprotective effects in PostC. Moreover, mitochondrial depolarization in PostC with Ru265 was weakened, compared to PostC (p = 0.004). These results suggest that MCU affects mitochondrial depolarization in PostC to suppress NMDAR over-activation and prevent elevation of intracellular Ca2+ concentrations against I/R injury.
Collapse
Affiliation(s)
- Hiromitsu Sasaki
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan.
| | - Takanori Furuta
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| | - Shohei Yokoyama
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| | - Yudai Morisaki
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| | - Hiroyuki Nakase
- Department of Neurosurgery, Nara Medical University, Shijo-Cho 840, Kashihara City, Nara, 634-8522, Japan
| |
Collapse
|
3
|
Chanana V, Zafer D, Kintner DB, Chandrashekhar JH, Eickhoff J, Ferrazzano PA, Levine JE, Cengiz P. TrkB-mediated neuroprotection in female hippocampal neurons is autonomous, estrogen receptor alpha-dependent, and eliminated by testosterone: a proposed model for sex differences in neonatal hippocampal neuronal injury. Biol Sex Differ 2024; 15:30. [PMID: 38566248 PMCID: PMC10988865 DOI: 10.1186/s13293-024-00596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. METHODS We cultured sexed hippocampal neurons from ERα+/+ and ERα-/- mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. RESULTS Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. CONCLUSIONS OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T.
Collapse
Affiliation(s)
- Vishal Chanana
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Wisconsin, 1500 Highland Ave - T505, Madison, WI, 53705-9345, USA
| | - Dila Zafer
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Wisconsin, 1500 Highland Ave - T505, Madison, WI, 53705-9345, USA
| | - Douglas B Kintner
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Wisconsin, 1500 Highland Ave - T505, Madison, WI, 53705-9345, USA
| | - Jayadevi H Chandrashekhar
- Waisman Center, University of Wisconsin, Madison, WI, USA
- University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jens Eickhoff
- Department of Statistics and Bioinformatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter A Ferrazzano
- Waisman Center, University of Wisconsin, Madison, WI, USA
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Wisconsin, 1500 Highland Ave - T505, Madison, WI, 53705-9345, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin, Madison, WI, USA
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Pelin Cengiz
- Waisman Center, University of Wisconsin, Madison, WI, USA.
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, University of Wisconsin, 1500 Highland Ave - T505, Madison, WI, 53705-9345, USA.
| |
Collapse
|
4
|
Chanana V, Hackett M, Deveci N, Aycan N, Ozaydin B, Cagatay NS, Hanalioglu D, Kintner DB, Corcoran K, Yapici S, Camci F, Eickhoff J, Frick KM, Ferrazzano P, Levine JE, Cengiz P. TrkB-mediated sustained neuroprotection is sex-specific and Erα-dependent in adult mice following neonatal hypoxia ischemia. Biol Sex Differ 2024; 15:1. [PMID: 38178264 PMCID: PMC10765746 DOI: 10.1186/s13293-023-00573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of life-long neurological morbidities that result in learning and memory impairments. Evidence suggests that male neonates are more susceptible to the detrimental effects of HI, yet the mechanisms mediating these sex-specific responses to neural injury in neonates remain poorly understood. We previously tested the effects of treatment with a small molecule agonist of the tyrosine kinase B receptor (TrkB), 7,8-dihydroxyflavone (DHF) following neonatal HI and determined that females, but not males exhibit increased phosphorylation of TrkB and reduced apoptosis in their hippocampi. Moreover, these female-specific effects of the TrkB agonist were found to be dependent upon the expression of Erα. These findings demonstrated that TrkB activation in the presence of Erα comprises one pathway by which neuroprotection may be conferred in a female-specific manner. The goal of this study was to determine the role of Erα-dependent TrkB-mediated neuroprotection in memory and anxiety in young adult mice exposed to HI during the neonatal period. METHODS In this study, we used a unilateral hypoxic ischemic (HI) mouse model. Erα+/+ or Erα-/- mice were subjected to HI on postnatal day (P) 9 and mice were treated with either vehicle control or the TrkB agonist, DHF, for 7 days following HI. When mice reached young adulthood, we used the novel object recognition, novel object location and open field tests to assess long-term memory and anxiety-like behavior. The brains were then assessed for tissue damage using immunohistochemistry. RESULTS Neonatal DHF treatment prevented HI-induced decrements in recognition and location memory in adulthood in females, but not in males. This protective effect was absent in female mice lacking Erα. The female-specific improved recognition and location memory outcomes in adulthood conferred by DHF therapy after neonatal HI tended to be or were Erα-dependent, respectively. Interestingly, DHF triggered anxiety-like behavior in both sexes only in the mice that lacked Erα. When we assessed the severity of injury, we found that DHF therapy did not decrease the percent tissue loss in proportion to functional recovery. We additionally observed that the presence of Erα significantly reduced overall HI-associated mortality in both sexes. CONCLUSIONS These observations provide evidence for a therapeutic role for DHF in which TrkB-mediated sustained recovery of recognition and location memories in females are Erα-associated and dependent, respectively. However, the beneficial effects of DHF therapy did not include reduction of gross tissue loss but may be derived from the enhanced functioning of residual tissues in a cell-specific manner.
Collapse
Affiliation(s)
- Vishal Chanana
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Margaret Hackett
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nazli Deveci
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Aycan
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Burak Ozaydin
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Nur Sena Cagatay
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Damla Hanalioglu
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Douglas B Kintner
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Karson Corcoran
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Sefer Yapici
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Furkan Camci
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
| | - Jens Eickhoff
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Peter Ferrazzano
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison, 1500 Highland Ave-T503, Madison, WI, 53705-9345, USA.
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
5
|
Taparli OE, Shahi PK, Cagatay NS, Aycan N, Ozaydin B, Yapici S, Liu X, Cikla U, Zafer D, Eickhoff JC, Ferrazzano P, Pattnaik BR, Cengiz P. Selectively compromised inner retina function following hypoxic-ischemic encephalopathy in mice: A noninvasive measure of severity of the injury. Neurochem Int 2023; 163:105471. [PMID: 36592700 PMCID: PMC9905320 DOI: 10.1016/j.neuint.2022.105471] [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: 11/03/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
The intricate system of connections between the eye and the brain implies that there are common pathways for the eye and brain that get activated following injury. Hypoxia-ischemia (HI) related encephalopathy is a consequence of brain injury caused by oxygen and blood flow deprivation that may result in visual disturbances and neurodevelopmental disorders in surviving neonates. We have previously shown that the tyrosine receptor kinase B (TrkB) agonist/modulator improves neuronal survival and long-term neuroprotection in a sexually differential way. In this study, we tested the hypotheses that; 1) TrkB agonist therapy improves the visual function in a sexually differential way; 2) Visual function detected by electroretinogram (ERG) correlates with severity of brain injury detected by magnetic resonance (MRI) imaging following neonatal HI in mice. To test our hypotheses, we used C57/BL6 mice at postnatal day (P) 9 and subjected them to either Vannucci's rodent model of neonatal HI or sham surgery. ERG was performed at P 30, 60, and 90. MRI was performed following the completion of the ERG. ERG in these mice showed that the a-wave is normal, but the b-wave amplitude is severely abnormal, reducing the b/a wave amplitude ratio. Inner retina function was found to be perturbed as we detected severely attenuated oscillatory potential after HI. No sex differences were detected in the injury and severity pattern to the retina as well as in response to 7,8-DHF therapy. Strong correlations were detected between the percent change in b/a ratio and percent hemispheric/hippocampal tissue loss obtained by MRI, suggesting that ERG is a valuable noninvasive tool that can predict the long-term severity of brain injury.
Collapse
Affiliation(s)
- Onur E Taparli
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA
| | - Pawan K Shahi
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, WI, USA
| | - Nur Sena Cagatay
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA
| | - Nur Aycan
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA
| | - Burak Ozaydin
- Department of Neurological Surgery, University of Wisconsin-Madison, WI, USA
| | - Sefer Yapici
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA
| | - Xinying Liu
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA
| | - Ulas Cikla
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Neurological Surgery, University of Wisconsin-Madison, WI, USA
| | - Dila Zafer
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA
| | - Jens C Eickhoff
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, WI, USA
| | - Peter Ferrazzano
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA
| | - Bikash R Pattnaik
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, WI, USA.
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison, WI, USA; Department of Waisman Center, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
6
|
Nayak M, Das D, Pradhan J, Ahmed R, Laureano-Melo R, Dandapat J. Epigenetic signature in neural plasticity: the journey so far and journey ahead. Heliyon 2022; 8:e12292. [PMID: 36590572 PMCID: PMC9798197 DOI: 10.1016/j.heliyon.2022.e12292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/31/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Neural plasticity is a remarkable characteristic of the brain which allows neurons to rewire their structure in response to internal and external stimuli. Many external stimuli collectively referred to as 'epigenetic factors' strongly influence structural and functional reorganization of the brain, thereby acting as a potential driver of neural plasticity. DNA methylation and demethylation, histone acetylation, and deacetylation are some of the frontline epigenetic mechanisms behind neural plasticity. Epigenetic signature molecules (mostly proteins) play a pivotal role in epigenetic reprogramming. Though neuro-epigenetics is an incredibly important field of emerging research, the critical role of signature proteins associated with epigenetic alteration and their involvement in neural plasticity needs further attention. This study gives an integrated and systematic overview of the current state of knowledge with a clear idea of types of neural plasticity and the context-dependent role of epigenetic signature molecules and their modulation by some natural bioactive compounds.
Collapse
Affiliation(s)
- Madhusmita Nayak
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India
| | - Diptimayee Das
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Faculty of Allied Health Science, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai India
| | - Jyotsnarani Pradhan
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| | - R.G. Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Roberto Laureano-Melo
- Barra Mansa University Center, R. Ver. Pinho de Carvalho, 267, 27330-550, Barra Mansa, Rio de Janeiro, Brazil
| | - Jagneshwar Dandapat
- Post-Graduate Department of Biotechnology, Utkal University, Bhubaneswar 751004, Odisha, India,Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar 751004, Odisha, India,Corresponding author.
| |
Collapse
|
7
|
Lim SY, Cengiz P. Opioid tolerance and opioid-induced hyperalgesia: Is TrkB modulation a potential pharmacological solution? Neuropharmacology 2022; 220:109260. [PMID: 36165856 DOI: 10.1016/j.neuropharm.2022.109260] [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: 03/14/2022] [Revised: 06/23/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022]
Abstract
Opioids are widely prescribed for moderate to severe pain in patients with acute illness, cancer pain, and chronic noncancer pain. However, long-term opioid use can cause opioid tolerance and opioid-induced hyperalgesia (OIH), contributing to the opioid misuse and addiction crisis. Strategies to mitigate opioid tolerance and OIH are needed to reduce opioid use and its sequelae. Currently, there are few effective pharmacological strategies that reduce opioid tolerance and OIH. The intrinsic tyrosine kinase receptor B (TrkB) ligand, brain-derived neurotrophic factor (BDNF), has been shown to modulate pain. The BDNF-TrkB signaling plays a role in initiating and sustaining elevated pain sensitivity; however, increasing evidence has shown that BDNF and 7,8-dihydroxyflavone (7,8-DHF), a potent blood-brain barrier-permeable ligand to TrkB, exert neuroprotective, anti-inflammatory, and antioxidant effects that may protect against opioid tolerance and OIH. As such, TrkB signaling may be an important therapeutic avenue in opioid tolerance and OIH. Here, we review 1) the mechanisms of pain, opioid analgesia, opioid tolerance, and OIH; 2) the role of BDNF-TrkB in pain modulation; and 3) the neuroprotective effects of 7,8-DHF and their implications for opioid tolerance and OIH.
Collapse
Affiliation(s)
- Sin Yin Lim
- Pharmacy Practice and Translational Research Division, University of Wisconsin-Madison School of Pharmacy, Madison, WI, United States.
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States; Waisman Center, University of Wisconsin-Madison, United States.
| |
Collapse
|
8
|
Zafer D, Adams T, Olson E, Stenman L, Taparli O, Eickhoff J, Cengiz P, Mezu-Ndubuisi OJ. Retinal vascular recovery revealed by retinal imaging following neonatal hypoxia ischemia in mice: Is there a role for tyrosine kinase receptor modulation? Brain Res 2022; 1796:148093. [PMID: 36116486 PMCID: PMC10013450 DOI: 10.1016/j.brainres.2022.148093] [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: 03/12/2022] [Revised: 09/06/2022] [Accepted: 09/11/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Hypoxic ischemic encephalopathy (HIE) secondary to perinatal asphyxia leads to long-term visual disabilities. Dilated retinal exams in human newborns with HIE is an emerging diagnostic tool, but phenotypes of hypoxia ischemia (HI) related retinal vascular injury are unclear. 7,8-Dihydroxyflavone (7,8-DHF) is a TrkB agonist with protective effects on HI-related brain damage. We studied retinal vessels in a mouse model of neonatal HIE and the efficacy of 7,8-DHF in ameliorating HI-related retinal vascular injury. METHODS C57BL6/J mice at post-natal day (P) 9 received unilateral left carotid artery ligation followed by exposure to 10 % oxygen for 50 min. Phosphate buffered saline or 7,8-DHF (5 mg/kg) were administered daily for 7 days intraperitoneally. Control groups of naïve or carotid artery ligation only mice were studied. Fluorescein angiography was performed in acute (two weeks post-exposure) and chronic (four weeks post-exposure) time points. Retinal artery width, retinal vein width, and collateral vessel length were quantified. RESULTS Ligation of the common carotid artery alone caused retinal artery dilation in acute and chronic time points, but had no effect on retinal veins. At acute time point, HI caused increased retinal artery vasodilation, but was reversed by 7,8-DHF. HI caused short collateral vessel formation in ipsilateral eyes, rescued by 7,8-DHF treatment. CONCLUSION Retinal artery vasodilation and collateral vessel formation due to HI were rescued by 7,8-DHF treatment. Retinal and collateral vessel monitoring could be diagnostic biomarkers for HI severity. Studies to elucidate mechanisms of 7,8-DHF action on retinal vessels could aid development of therapies for neonatal HI.
Collapse
Affiliation(s)
- Dila Zafer
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA
| | - Thao Adams
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Ellie Olson
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Lauren Stenman
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| | - Onur Taparli
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA.
| | - Jens Eickhoff
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA.
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin, Madison, WI, USA; Waisman Center, University of Wisconsin, Madison, WI, USA.
| | - Olachi J Mezu-Ndubuisi
- Department of Pediatrics, University of Rochester, Rochester, NY, USA; Department of Ophthalmology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
9
|
Xiong J, Liao J, Liu X, Zhang Z, Adams J, Pacifici R, Ye K. A TrkB agonist prodrug prevents bone loss via inhibiting asparagine endopeptidase and increasing osteoprotegerin. Nat Commun 2022; 13:4820. [PMID: 35973996 PMCID: PMC9381595 DOI: 10.1038/s41467-022-32435-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/26/2022] [Indexed: 11/12/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its tropomyosin-related kinase B receptor (TrkB) are expressed in human osteoblasts and mediate fracture healing. BDNF/TrkB signaling activates Akt that phosphorylates and inhibits asparagine endopeptidase (AEP), which regulates the differentiation fate of human bone marrow stromal cells (hBMSC) and is altered in postmenopausal osteoporosis. Here we show that R13, a small molecular TrkB receptor agonist prodrug, inhibits AEP and promotes bone formation. Though both receptor activator of nuclear factor kappa-Β ligand (RANK-L) and osteoprotegerin (OPG) induced by ovariectomy (OVX) remain comparable between WT and BDNF+/− mice, R13 treatment significantly elevates OPG in both mice without altering RANKL, blocking trabecular bone loss. Strikingly, both R13 and anti-RANK-L exhibit equivalent therapeutic efficacy. Moreover, OVX increases RANK-L and OPG in WT and AEP KO mice with RANK-L/OPG ratio lower in the latter than the former, attenuating bone turnover. 7,8-DHF, released from R13, activates TrkB and its downstream effector CREB, which is critical for OPG augmentation. Consequently, 7,8-DHF represses C/EBPβ/AEP pathway, inhibiting RANK-L-induced RAW264.7 osteoclastogenesis. Therefore, our findings support that R13 exerts its therapeutic efficacy toward osteoporosis via inhibiting AEP and escalating OPG. BDNS and TrkB are involved in bone fracture healing by inhibiting AEP. Here the authors show that a TrkB agonist prodrug can inhibit AEP and promote bone formation in osteoporotic mice.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology (SIAT) Shenzhen, Guangdong, PR China.
| |
Collapse
|
10
|
Qiu H, Qian T, Wu T, Gao T, Xing Q, Wang L. Src Family Kinases Inhibition Ameliorates Hypoxic-Ischemic Brain Injury in Immature Rats. Front Cell Neurosci 2022; 15:746130. [PMID: 34992524 PMCID: PMC8724194 DOI: 10.3389/fncel.2021.746130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/24/2021] [Indexed: 01/06/2023] Open
Abstract
Hypoxic-ischemic (HI) injury is one of the initial factors contributing to neonatal brain injury. Src family kinases (SFKs) are considered to act as molecular hubs for N-methyl-d-aspartate receptor (NMDAR) regulation and participate in the HI injury process. The objectives of this study were to evaluate the levels of phospho-Src (p-Src), the relationship between NMDARs and SFKs, and the effects of SFK inhibition on an immature rat HI brain injury model. The model was induced in 3-day-old Sprague–Dawley rats using the Rice-Vannucci model operation. The level of p-Src was evaluated using Western blotting. The association of NMDARs with SFKs was detected using Western blotting and coimmunoprecipitation. After intraperitoneal injection of 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine (PP2), an SFK-selective inhibitor, neuropathological changes were observed by performing H&E and immunofluorescence staining, and the neurological functions were assessed using the following behavioral tests: modified neurological severity score, open field test, and Morris water maze test. The levels of p-Src first decreased at 0 h after injury, increased at 2 h after injury, and continuously decreased from 6 h to 3 days. Along with the increased p-Src levels observed at 2 h after injury, the phosphorylation of NMDAR subunit NR2B at tyrosine 1472 was increased. Following the administration of PP2, the increased p-Src and NMDAR-2B levels detected at 2 h after injury were decreased, and tissue injury and myelin basic protein expression were improved at 7 days after injury. The PP2 intervention improved the performance of injured rats on behavioral tests. In conclusion, we determined the patterns of p-Src expression after HI brain injury in immature rats and showed a relationship with the activated NMDA receptor. The inhibition of p-Src ameliorates neuropathological changes and damages neurological functions induced by HI injury.
Collapse
Affiliation(s)
- Han Qiu
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Ting Gao
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Qinghe Xing
- Department of Neonatology, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
11
|
Human-Induced Pluripotent Stem Cell-Based Models for Studying Sex-Specific Differences in Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1387:57-88. [PMID: 34921676 DOI: 10.1007/5584_2021_683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The prevalence of neurodegenerative diseases is steadily increasing worldwide, and epidemiological studies strongly suggest that many of the diseases are sex-biased. It has long been suggested that biological sex differences are crucial for neurodegenerative diseases; however, how biological sex affects disease initiation, progression, and severity is not well-understood. Sex is a critical biological variable that should be taken into account in basic research, and this review aims to highlight the utility of human-induced pluripotent stem cells (iPSC)-derived models for studying sex-specific differences in neurodegenerative diseases, with advantages and limitations. In vitro systems utilizing species-specific, renewable, and physiologically relevant cell sources can provide powerful platforms for mechanistic studies, toxicity testings, and drug discovery. Matched healthy, patient-derived, and gene-corrected human iPSCs, from both sexes, can be utilized to generate neuronal and glial cell types affected by specific neurodegenerative diseases to study sex-specific differences in two-dimensional (2D) and three-dimensional (3D) human culture systems. Such relatively simple and well-controlled systems can significantly contribute to the elucidation of molecular mechanisms underlying sex-specific differences, which can yield effective, and potentially sex-based strategies, against neurodegenerative diseases.
Collapse
|
12
|
Li L, Wang T, Chen S, Yue Y, Xu Z, Yuan Y. DNA methylations of brain-derived neurotrophic factor exon VI are associated with major depressive disorder and antidepressant-induced remission in females. J Affect Disord 2021; 295:101-107. [PMID: 34418778 DOI: 10.1016/j.jad.2021.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has been suggested to play important roles in major depressive disorder (MDD) and antidepressant treatment. The main purpose of this study was to evaluate the association of DNA methylation changes in the BDNF gene with MDD and antidepressant treatment. METHODS A total of 291 MDD patients and 100 healthy controls were included and followed up for 6 weeks. The Hamilton Depression Rating Scale-17 (HDRS-17) was used to measure treatment improvement. The life events scales (LES) and childhood trauma questionnaire (CTQ) were used to rate recent and early life stress. DNA methylation levels of CpG sites in the BDNF gene were measured. RESULTS Two CpG sites in BDNF exon VI (BDNF133 and BDNF134) were demonstrated to have significantly higher methylation in MDD patients than in controls (both FDR-adjusted P = 0.001). A logistics regression model indicated that the interaction between the hypermethylation of BDNF133 and negative subscore of LES was associated to MDD (OR=0.0075, P<0.001). Methylation of BDNF140 at baseline was significantly elevated in remitters (FDR-adjusted P = 0.046) at week 6. In subgroup analyses, these findings could be replicated in females, but not in males. LIMITATIONS The methylation status of BDNF after 6 weeks of antidepressant treatment was not measured and the DNA methylation were detected in peripheral blood cells. CONCLUSIONS These findings highlight gender-specific alteration of methylation at several CpG sites in BDNF exon VI as a promising candidate indicator of MDD and antidepressant-induced remission.
Collapse
Affiliation(s)
- Lei Li
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China; Department of Sleep Medicine, The Fourth People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Tianyu Wang
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Suzhen Chen
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Yingying Yue
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Zhi Xu
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China.
| | - Yonggui Yuan
- Institute of Psychosomatics, School of Medicine, Southeast University, Nanjing, 210023, China; Department of Psychosomatics and Psychiatry, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China; Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast university, Nanjing, 210009, China.
| |
Collapse
|
13
|
Zaitoun IS, Sheibani N. Hypoxic-Ischemic Encephalopathy: Impact on Retinal Neurovascular Integrity and Function. J Ophthalmic Vis Res 2021; 16:317-319. [PMID: 34394859 PMCID: PMC8358751 DOI: 10.18502/jovr.v16i3.9427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/01/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Ismail S Zaitoun
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
14
|
Suggesting 7,8-dihydroxyflavone as a promising nutraceutical against CNS disorders. Neurochem Int 2021; 148:105068. [PMID: 34022252 DOI: 10.1016/j.neuint.2021.105068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 11/20/2022]
Abstract
7,8-dihydroxyflavone (DHF), a naturally-occurring plant-based flavone, is a high-affinity tyrosine kinase receptor B (TrkB) agonist and a bioactive molecule of therapeutic interest for neuronal survival, differentiation, synaptic plasticity and neurogenesis. In the family of neurotrophic factors, this small BDNF-mimetic molecule has attracted considerable attention due to its oral bioavailability and ability to cross the blood-brain barrier. Recent evidences have shed light on the neuroprotective role of this pleiotropic flavone against several neurological disorders, including Alzheimer's disease, Parkinson's disease, cerebral ischemia, Huntington's disease, and other CNS disorders. DHF also elicits potent protective actions against toxins-induced insults to brain and neuronal cells. DHF shows promising anti-oxidant and anti-inflammatory properties in ameliorating the neurodegenerative processes affecting the CNS. This review provides an overview of the significant neuroprotective potentials of DHF and discusses how it exerts its multitudinous beneficial effects by modulating different pathways linked with the pathophysiology of CNS disorders, and thus proposes it to be a nutraceutical against a broad spectrum of neurological disorders.
Collapse
|
15
|
Baumgartner NE, Daniel JM. Estrogen receptor α: a critical role in successful female cognitive aging. Climacteric 2021; 24:333-339. [PMID: 33522313 DOI: 10.1080/13697137.2021.1875426] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Due to potential health risks, current recommendations are that individuals who wish to use hormone therapy to treat menopausal symptoms do so for the shortest period of time possible. In our investigation into how short-term use of estrogens in midlife following loss of ovarian function exerts long-term effects on female cognitive aging in rodents, we discovered a link between the ability of previous exposure to estradiol to enhance memory in the long term and its ability to increase estrogen receptor α (ERα) levels in the hippocampus, a brain area important for memory. Follow-up studies in model systems implicate a role for ERα in enhanced cognitive function independent of ovarian or exogenously administered estrogens. Results are consistent with clinical studies in which brain ERα levels in older women and men are related to cognitive functioning and risk of cognitive decline is associated with polymorphisms in the gene that transcribes ERα. Research in preclinical models reveals mechanisms through which ERα can be activated and affect cognition in the absence of ovarian estrogens, including ligand-independent activation via insulin-like growth factor-1 signaling and activation by brain-derived neuroestrogens. This report reviews preclinical and clinical data that collectively point to the importance of ERα in cognition and highlights the need to differentiate the role of estrogen receptors from their classical ligands as we seek approaches to facilitate successful cognitive aging.
Collapse
Affiliation(s)
- N E Baumgartner
- Neuroscience Program, Tulane University, New Orleans, LA, USA.,Brain Institute, Tulane University, New Orleans, LA, USA
| | - J M Daniel
- Neuroscience Program, Tulane University, New Orleans, LA, USA.,Brain Institute, Tulane University, New Orleans, LA, USA.,Department of Psychology, Tulane University, New Orleans, LA, USA
| |
Collapse
|
16
|
Sun Y, Ma L, Jin M, Zheng Y, Wang D, Ni H. Effects of Melatonin on Neurobehavior and Cognition in a Cerebral Palsy Model of plppr5-/- Mice. Front Endocrinol (Lausanne) 2021; 12:598788. [PMID: 33692754 PMCID: PMC7937640 DOI: 10.3389/fendo.2021.598788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cerebral palsy (CP), a group of clinical syndromes caused by non-progressive brain damage in the developing fetus or infant, is one of the most common causes of lifelong physical disability in children in most countries. At present, many researchers believe that perinatal cerebral hypoxic ischemic injury or inflammatory injury are the main causes of cerebral palsy. Previous studies including our works confirmed that melatonin has a protective effect against convulsive brain damage during development and that it affects the expression of various molecules involved in processes such as metabolism, plasticity and signaling in the brain. Integral membrane protein plppr5 is a new member of the plasticity-related protein family, which is specifically expressed in brain and spinal cord, and induces filopodia formation as well as neurite growth. It is highly expressed in the brain, especially in areas of high plasticity, such as the hippocampus. The signals are slightly lower in the cortex, the cerebellum, and in striatum. Noteworthy, during development plppr5 mRNA is expressed in the spinal cord, i.e., in neuron rich regions such as in medial motor nuclei, suggesting that plppr5 plays an important role in the regulation of neurons. However, the existing literature only states that plppr5 is involved in the occurrence and stability of dendritic spines, and research on its possible involvement in neonatal ischemic hypoxic encephalopathy has not been previously reported. We used plppr5 knockout (plppr5-/-) mice and their wild-type littermates to establish a model of hypoxicischemic brain injury (HI) to further explore the effects of melatonin on brain injury and the role of plppr5 in this treatment in an HI model, which mainly focuses on cognition, exercise, learning, and memory. All the tests were performed at 3-4 weeks after HI. As for melatonin treatment, which was performed 5 min after HI injury and followed by every 24h. In these experiments, we found that there was a significant interaction between genotype and treatment in novel object recognition tests, surface righting reflex tests and forelimb suspension reflex tests, which represent learning and memory, motor function and coordination, and the forelimb grip of the mice, respectively. However, a significant main effect of genotype and treatment on performance in all behavioral tests were observed. Specifically, wild-type mice with HI injury performed better than plppr5-/- mice, regardless of treatment with melatonin or vehicle. Moreover, treatment with melatonin could improve behavior in the tests for wild-type mice with HI injury, but not for plppr5-/- mice. This study showed that plppr5 knockout aggravated HI damage and partially weakened the neuroprotection of melatonin in some aspects (such as novel object recognition test and partial nerve reflexes), which deserves further study.
Collapse
|
17
|
Zhao Z, Xue F, Gu Y, Han J, Jia Y, Ye K, Zhang Y. Crosstalk between the muscular estrogen receptor α and BDNF/TrkB signaling alleviates metabolic syndrome via 7,8-dihydroxyflavone in female mice. Mol Metab 2020; 45:101149. [PMID: 33352311 PMCID: PMC7811170 DOI: 10.1016/j.molmet.2020.101149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Objective 7,8-Dihydroxyflavone (7,8-DHF), a small molecular mimetic of brain-derived neurotrophic factor (BDNF), alleviates high-fat diet-induced obesity in female mice in a sex-specific manner by activating muscular tropomyosin-related kinase B (TrkB). However, the underlying molecular mechanism for this sex difference is unknown. Moreover, muscular estrogen receptor α (ERα) plays a critical role in metabolic diseases. Impaired ERα action is often accompanied by metabolic syndrome (MetS) in postmenopausal women. This study investigated whether muscular ERα is involved in the metabolic effects of 7,8-DHF. Methods For the in vivo studies, 72 female C57BL/6J mice were given a low-fat diet or high-fat diet, and both received daily intragastric administration of vehicle or 7,8-DHF for 24 weeks. The hypothalamic-pituitary-ovarian (HPO) axis function was assessed by investigating typical sex-related serum hormones and the ovarian reserve. Indicators of menopausal MetS, including lipid metabolism, insulin sensitivity, bone density, and serum inflammatory cytokines, were also evaluated. The expression levels of ERα and other relevant signaling molecules were also examined. In vitro, the molecular mechanism involved in the interplay of ERα and TrkB receptors was verified in differentiated C2C12 myotubes using several inhibitors and a lentivirus short hairpin RNA-knockdown strategy. Results Long-term oral administration of 7,8-DHF acted as a protective factor for the female HPO axis function, protecting against ovarian failure, earlier menopause, and sex hormone disorders, which was paralleled by the alleviation of MetS coupled with the production of ERα-rich, TrkB-activated, and uncoupling protein 1 (UCP1) high thermogenic skeletal muscle tissues. 7,8-DHF-stimulated transactivation of ERα at serine 118 (S118) and tyrosine 537 (Y537), which was crucial to activate the BDNF/TrkB signaling cascades. In turn, activation of BDNF/TrkB signaling was also required for the ligand-independent activation of ERα, especially at the Y537 phosphorylation site. In addition, Src family kinases played a core role in the interplay of ERα and TrkB, synergistically activating the signaling pathways related to energy metabolism. Conclusions These findings revealed a novel role of 7,8-DHF in protecting the function of the female HPO axis and activating tissue-specific ERα, which improves our understanding of this sex difference in 7,8-DHF-mediated maintenance of metabolic homeostasis and provides new therapeutic strategies for managing MetS in women. 7,8-DHF improves hypothalamic-pituitary-ovarian axis function in mature adult female mice. 7,8-DHF protects against ovarian failure and onset of earlier menopause. 7,8-DHF-induced transactivation of ERα is crucial to activate BDNF/TrkB signaling cascades. 7,8-DHF-induced activations of ERα and BDNF/TrkB signaling are interdependent. Src family kinases play a core role in the crosstalk of ERα and BDNF/TrkB signaling pathways.
Collapse
Affiliation(s)
- Zhenlei Zhao
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Fan Xue
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Yanpei Gu
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Jianxin Han
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Yingxian Jia
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Ying Zhang
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Center for Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
18
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
19
|
Tsuji S, Di Martino E, Mukai T, Tsuji S, Murakami T, Harris RA, Blomgren K, Åden U. Aggravated brain injury after neonatal hypoxic ischemia in microglia-depleted mice. J Neuroinflammation 2020; 17:111. [PMID: 32276642 PMCID: PMC7149909 DOI: 10.1186/s12974-020-01792-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/27/2020] [Indexed: 01/17/2023] Open
Abstract
Background Neuroinflammation plays an important role in neonatal hypoxic-ischemic encephalopathy (HIE). Although microglia are largely responsible for injury-induced inflammatory response, they play beneficial roles in both normal and disease states. However, the effects of microglial depletion on neonatal HIE remain unclear. Methods Tamoxifen was administered to Cx3cr1CreER/+Rosa26DTA/+ (microglia-depleted model) and Cx3cr1CreER/+Rosa26DTA/− (control) mice at P8 and P9 to assess the effect of microglial depletion. The density of microglia was quantified using Iba-1 staining. Moreover, the proportion of resident microglia after the HI insult was analyzed using flow cytometric analysis. At P10, the HI insult was conducted using the Rice-Vannucci procedure at P10. The infarct size and apoptotic cells were analyzed at P13. Cytokine analyses were performed using quantitative polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA) at P13. Results At P10, tamoxifen administration induced > 99% microglial depletion in DTA+ mice. Following HI insult, there was persisted microglial depletion over 97% at P13. Compared to male DTA− mice, male DTA+ mice exhibited significantly larger infarct volumes; however, there were no significant differences among females. Moreover, compared to male DTA− mice, male DTA+ mice had a significantly higher density of TUNEL+ cells in the caudoputamen, cerebral cortex, and thalamus. Moreover, compared to female DTA− mice, female DTA+ mice showed a significantly greater number of TUNEL+ cells in the hippocampus and thalamus. Compared to DTA− mice, ELISA revealed significantly lower IL-10 and TGF-β levels in both male and female DTA+ mice under both normal conditions and after HI (more pronounced). Conclusion We established a microglial depletion model that aggravated neuronal damage and apoptosis after the HI insult, which was predominantly observed in males.
Collapse
Affiliation(s)
- Shunichiro Tsuji
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Elena Di Martino
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Takeo Mukai
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shoko Tsuji
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu City, Shiga, 520-2192, Japan
| | - Robert A Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska Hospital Solna, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Åden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Neonatal Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Cengiz P, Zafer D, Chandrashekhar JH, Chanana V, Bogost J, Waldman A, Novak B, Kintner DB, Ferrazzano PA. Developmental differences in microglia morphology and gene expression during normal brain development and in response to hypoxia-ischemia. Neurochem Int 2019; 127:137-147. [PMID: 30639264 DOI: 10.1016/j.neuint.2018.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/27/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in ischemic brain injury and recovery, however the interplay between brain development and the neuroinflammatory response is poorly understood. We previously described age-dependent differences in the microglial response and the effect of microglial inhibition. Here we investigate whether age-dependent microglial responses may be related to pre-injury developmental differences in microglial phenotype. METHODS Measures of microglia morphology were quantified using semi-automated software analysis of immunostained sections from postnatal day 2 (P2), P9, P30 and P60 mice using IMARIS. Microglia were isolated from P2, P9, P30 and P60 mice, and expression of markers of classical and alternative microglial activation was assessed, as well as transforming growth factor beta (TGF-β) receptor, Serpine1, Mer Tyrosine Kinase (MerTK), and the suppressor of cytokine signaling (SOCS3). Hypoxia-ischemia (HI) was induced in P9 and P30 mice using unilateral carotid artery ligation and exposure to 10% oxygen for 50 min. Microglia morphology and microglial expression of genes in the TGF-β and MerTK pathways were determined in ipsilateral and contralateral hippocampus. RESULTS A progressive and significant increase in microglia branching morphology was seen in all brain regions from P2 to P30. No consistent classical or alternative activation profile was seen in isolated microglia. A clear transition to increased expression of TGF-β and its downstream effector serpine1 was seen between P9 and P30. A similar increase in expression was seen in MerTK and its downstream effector SOCS3. HI resulted in a significant decrease in branching morphology only in the P9 mice, and expression of TGF-β receptor, Serpine1, MerTK, and SOCS3 were elevated in P30 mice compared to P9 post-HI. CONCLUSION Microglia maturation is associated with changes in morphology and gene expression, and microglial responses to ischemia in the developing brain differ based on the age at which injury occurs.
Collapse
Affiliation(s)
- Pelin Cengiz
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Dila Zafer
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jayadevi H Chandrashekhar
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; University of Illinois at Urbana-Champaign, IL, USA
| | - Vishal Chanana
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacob Bogost
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alex Waldman
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Emory University School of Medicine, Atlanta, GA, USA
| | - Becca Novak
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Douglas B Kintner
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter A Ferrazzano
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
21
|
Zafer D, Aycan N, Ozaydin B, Kemanli P, Ferrazzano P, Levine JE, Cengiz P. Sex differences in Hippocampal Memory and Learning following Neonatal Brain Injury: Is There a Role for Estrogen Receptor-α? Neuroendocrinology 2019; 109:249-256. [PMID: 30884486 PMCID: PMC6893032 DOI: 10.1159/000499661] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/17/2019] [Indexed: 01/11/2023]
Abstract
Neonatal encephalopathy due to hypoxia-ischemia (HI) leads to severe, life-long morbidities in thousands of neonates born in the USA and worldwide each year. Varying capacities of long-term episodic memory, verbal working memory, and learning can present without cerebral palsy and have been associated with the severity of neonatal encephalopathy sustained at birth. Among children who sustain a moderate degree of HI at birth, girls have larger hippocampal volumes compared to boys. Clinical studies indicate that female neonatal brains are more resistant to the effects of neonatal HI, resulting in better long-term cognitive outcomes compared to males with comparable brain injury. Our most recent mechanistic studies have addressed the origins and cellular basis of sex differences in hippocampal neuroprotection following neonatal HI-related brain injury and implicate estrogen receptor-α (ERα) in the neurotrophin receptor-mediated hippocampal neuroprotection in female mice. This review summarizes the recent findings on ERα-dependent, neurotrophin-mediated hippocampal neuroprotection and weighs the evidence that this mechanism plays an important role in preservation of long-term memory and learning following HI in females.
Collapse
Affiliation(s)
- Dila Zafer
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Nur Aycan
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Burak Ozaydin
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Pinar Kemanli
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Peter Ferrazzano
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - Jon E Levine
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Pelin Cengiz
- Waisman Center, University of Wisconsin, Madison, Wisconsin, USA,
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA,
| |
Collapse
|
22
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
23
|
Chandran R, Kim T, Mehta SL, Udho E, Chanana V, Cengiz P, Kim H, Kim C, Vemuganti R. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab 2018; 38:1818-1827. [PMID: 29083257 PMCID: PMC6168911 DOI: 10.1177/0271678x17738701] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Uncontrolled oxidative stress contributes to the secondary neuronal death that promotes long-term neurological dysfunction following traumatic brain injury (TBI). Surprisingly, both NADPH oxidase 2 (NOX2) that increases and transcription factor Nrf2 that decreases reactive oxygen species (ROS) are induced after TBI. As the post-injury functional outcome depends on the balance of these opposing molecular pathways, we evaluated the effect of TBI on the motor and cognitive deficits and cortical contusion volume in NOX2 and Nrf2 knockout mice. Genetic deletion of NOX2 improved, while Nrf2 worsened the post-TBI motor function recovery and lesion volume indicating that decreasing ROS levels might be beneficial after TBI. Treatment with either apocynin (NOX2 inhibitor) or TBHQ (Nrf2 activator) alone significantly improved the motor function after TBI, but had no effect on the lesion volume, compared to vehicle control. Whereas, the combo therapy (apocynin + TBHQ) given at either 5 min/24 h or 2 h/24 h improved motor and cognitive function and decreased cortical contusion volume compared to vehicle group. Thus, both the generation and disposal of ROS are important modulators of oxidative stress, and a combo therapy that prevents ROS formation and potentiates ROS disposal concurrently is efficacious after TBI.
Collapse
Affiliation(s)
| | - TaeHee Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Suresh L Mehta
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Eshwar Udho
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Vishal Chanana
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Pelin Cengiz
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - HwuiWon Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Chanul Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,3 William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
24
|
Vijayan M, Kumar S, Yin X, Zafer D, Chanana V, Cengiz P, Reddy PH. Identification of novel circulatory microRNA signatures linked to patients with ischemic stroke. Hum Mol Genet 2018; 27:2318-2329. [PMID: 29701837 PMCID: PMC6005038 DOI: 10.1093/hmg/ddy136] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/01/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are involved in growth, development, and occurrence and progression of many diseases. MiRNA-mediated post-transcriptional regulation is poorly understood in vascular biology and pathology. The purpose of this is to determine circulatory miRNAs as early detectable peripheral biomarkers in patients with ischemic stroke (IS). MiRNAs expression levels were measured in IS serum samples and healthy controls using Illumina deep sequencing analysis and identified differentially expressed miRNAs. Differentially expressed miRNAs were further validated using SYBR-green-based quantitative real-time PCR (qRT-PCR) assay in postmortem IS brains, lymphoblastoid IS cell lines, oxygen and glucose deprivation/reoxygenation -treated human and mouse neuroblastoma cells, and mouse models of hypoxia and ischemia (HI)-induced stroke. A total of 4656 miRNAs were differentially expressed in IS serum samples relative to healthy controls. Out of 4656 miRNAs, 272 were found to be significantly deregulated in IS patients. Interestingly, we found several novel and previously unreported miRNAs in IS patients relative to healthy controls. Further analyses revealed that some candidate miRNAs and its target genes were involved in the regulation of the stroke. To the best of our knowledge, this is the first study identified potential novel candidate miRNAs in IS serum samples from the residents of rural West Texas. MiRNAs identified in this study could potentially be used as a biomarker and the development of novel therapeutic approaches for stroke. Further studies are necessary to better understand miRNAs-regulated stroke cellular changes.
Collapse
Affiliation(s)
- Murali Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Subodh Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Xiangling Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Dila Zafer
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Vishal Chanana
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Pelin Cengiz
- Waisman Center and Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA and
- Department of Public Health, Graduate School of Biomedical Sciences, Lubbock, TX, USA
| |
Collapse
|
25
|
Fang C, Xie L, Liu C, Fu C, Ye W, Liu H, Zhang B. Tanshinone IIA improves hypoxic ischemic encephalopathy through TLR‑4‑mediated NF‑κB signal pathway. Mol Med Rep 2018; 18:1899-1908. [PMID: 29956801 PMCID: PMC6072156 DOI: 10.3892/mmr.2018.9227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/30/2018] [Indexed: 12/23/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is the most common brain injury following hypoxia and/or ischemia caused by various factors during the perinatal period, resulting in detrimental neurological deficits in the nervous system. Tanshinone IIA (Tan‑IIA) is a potential agent for the treatment of cardiovascular and cerebrovascular diseases. In this study, the efficacy of Tan‑IIA was investigated in a newborn mouse model of HIE. The dynamic mechanism of Tan‑IIA was also investigated in the central nervous system of neonate mice. Intravenous injection of Tan‑IIA (5 mg/kg) was administered and changes in oxidative stress, inflammation and apoptosis‑associated proteins in neurons. Histology and immunohistochemistry was used to determine infarct volume and the number of damaged neurons by Fluoro‑Jade C staining. The effects of Tan‑IIA on mice with HIE were evaluated by body weight, brain water content, neurobehavioral tests and blood‑brain barrier permeability. The results demonstrated that the apoptosis rate was decreased following Tan‑IIA administration. Expression levels of pro‑apoptotic proteins, caspase‑3 and caspase‑9 and P53 were downregulated. Expression of Bcl‑2 anti‑apoptotic proteins was upregulated by Tan‑IIA treatment in neuro. Results also found that Tan‑IIA treatment decreased production of inflammatory cytokines such as interleukin‑1, tumor necrosis factor‑α, C‑X‑C motif chemokine 10, and chemokine (C‑C motif) ligand 12. Oxidative stress was also reduced by Tan‑IIA in neurons, as determined by the expression levels of superoxide dismutase, glutathione and catalase, and the production of reactive oxygen species. The results demonstrated that Tan‑IIA treatment reduced the infarct volume and the number of damaged neurons. Furthermore, body weight, brain water content and blood‑brain barrier permeability were markedly improved by Tan‑IIA treatment of newborn mice following HIE. Furthermore, the results indicated that Tan‑IIA decreased Toll‑like receptor‑4 (TLR‑4) and nuclear factor‑κB (NF‑κB) expression in neurons. TLR‑4 treatment of neuronal cell in vitro addition stimulated NF‑κB activity, and further enhanced the production of inflammatory cytokines and oxidative stress levels in neurons. In conclusion, these results suggest that Tan‑IIA treatment is beneficial for improvement of HIE through TLR‑4‑mediated NF‑κB signaling.
Collapse
Affiliation(s)
- Chengzhi Fang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lili Xie
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chunmei Liu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chunhua Fu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Ye
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Liu
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Binghong Zhang
- Department of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
26
|
Zaitoun IS, Cikla U, Zafer D, Udho E, Almomani R, Suscha A, Cengiz P, Sorenson CM, Sheibani N. Attenuation of Retinal Vascular Development in Neonatal Mice Subjected to Hypoxic-Ischemic Encephalopathy. Sci Rep 2018; 8:9166. [PMID: 29907863 PMCID: PMC6003906 DOI: 10.1038/s41598-018-27525-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/05/2018] [Indexed: 11/24/2022] Open
Abstract
A significant proportion of children that survive hypoxic-ischemic encephalopathy (HIE) develop visual impairment. These visual deficits are generally attributed to injuries that occur in the primary visual cortex and other visual processing systems. Recent studies suggested that neuronal damage might also occur in the retina. An important structure affecting the viability of retinal neurons is the vasculature. However, the effects of HIE on the retinal neurovasculature have not been systemically evaluated. Here we investigated whether exposure of postnatal day 9 (P9) neonatal mice to HIE is sufficient to induce neurovascular damage in the retina. We demonstrate that the blood vessels on the surface of the retina, from mice subjected to HIE, were abnormally enlarged with signs of degeneration. The intermediate and deep vascular layers in these retinas failed to form normally, particularly in the periphery. All the vascular damages observed here were irreversible in nature up to 100 days post HIE. We also observed loss of retinal neurons, together with changes in both astrocytes and Müller cells mainly in the inner retina at the periphery. Collectively, our findings suggest that HIE results in profound alterations in the retinal vasculature, indicating the importance of developing therapeutic strategies to protect neurovascular dysfunction not only in the brain but also in the retina for infants exposed to HIE.
Collapse
Affiliation(s)
- Ismail S Zaitoun
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA. .,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| | - Ulas Cikla
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Dila Zafer
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Eshwar Udho
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Reem Almomani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Andrew Suscha
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Christine M Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| |
Collapse
|
27
|
Sukhanova IA, Sebentsova EA, Khukhareva DD, Manchenko DM, Glazova NY, Vishnyakova PA, Inozemtseva LS, Dolotov OV, Vysokikh MY, Levitskaya NG. Gender-dependent changes in physical development, BDNF content and GSH redox system in a model of acute neonatal hypoxia in rats. Behav Brain Res 2018; 350:87-98. [PMID: 29753727 DOI: 10.1016/j.bbr.2018.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/17/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
Perinatal hypoxia-ischaemia is one of the leading factors that negatively influence the development of the central nervous system. Our aim was to investigate the effects of sex on the outcomes of acute neonatal hypoxia (ANH) in rat pups. Male and female Wistar rats were exposed to a hypoxic condition (8% oxygen for 120 min) at postnatal day 2 (P2). Immediately after ANH an increase in HIF1-α gene expression was observed in the rat brains, independently of sex. Brain-derived neurotrophic factor (BDNF) and glutathione peroxidase-4 gene expression was increased in female animals only. Hypoxic pups of both sexes showed a decreased reduced/oxidised glutathione (GSH/GSSG) ratio in the blood and only males had an increased GSH content in the whole brain immediately after hypoxia. Furthermore, an increased BDNF content in the brain was found in both male and female rat pups at 0 h and in serum 4 h after hypoxia, but at 4 h after hypoxia only males had an increased BDNF level in the brain. Only hypoxic males displayed retarded performance in the righting reflex, but in a negative geotaxis test hypoxic pups of both sexes had an increased turnaround time. Moreover, hypoxic female but not male pups demonstrated less weight gain than control littermates for the entire observation period (until P18). These results demonstrate that ANH at P2 leads to both molecular and physiological impairments in a sex-specific manner and the described model could be used to represent mild hypoxic brain damage in very preterm infants.
Collapse
Affiliation(s)
- Iu A Sukhanova
- Lomonosov Moscow State University, Biology Faculty, Moscow, Russia; Federal State Budget Institution 'Research Centre for Obstetrics Gynaecology and Perinatology' Ministry of Healthcare and Social Development of the Russian Federation, Moscow, Russia.
| | - E A Sebentsova
- Institute of Molecular Genetics, Russian Academy of Science, Moscow, Russia
| | - D D Khukhareva
- Lomonosov Moscow State University, Biology Faculty, Moscow, Russia
| | - D M Manchenko
- Lomonosov Moscow State University, Biology Faculty, Moscow, Russia
| | - N Yu Glazova
- Institute of Molecular Genetics, Russian Academy of Science, Moscow, Russia
| | - P A Vishnyakova
- Federal State Budget Institution 'Research Centre for Obstetrics Gynaecology and Perinatology' Ministry of Healthcare and Social Development of the Russian Federation, Moscow, Russia
| | - L S Inozemtseva
- Institute of Molecular Genetics, Russian Academy of Science, Moscow, Russia
| | - O V Dolotov
- Institute of Molecular Genetics, Russian Academy of Science, Moscow, Russia
| | - M Y Vysokikh
- Federal State Budget Institution 'Research Centre for Obstetrics Gynaecology and Perinatology' Ministry of Healthcare and Social Development of the Russian Federation, Moscow, Russia
| | - N G Levitskaya
- Lomonosov Moscow State University, Biology Faculty, Moscow, Russia; Institute of Molecular Genetics, Russian Academy of Science, Moscow, Russia
| |
Collapse
|
28
|
Chan CB, Ye K. Sex differences in brain-derived neurotrophic factor signaling and functions. J Neurosci Res 2017; 95:328-335. [PMID: 27870419 DOI: 10.1002/jnr.23863] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/27/2016] [Accepted: 07/11/2016] [Indexed: 01/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family that plays a critical role in numerous neuronal activities. Recent studies have indicated that some functions or action mechanisms of BDNF vary in a sex-dependent manner. In particular, BDNF content in some brain parts and the tendency to develop BDNF deficiency-related diseases such as depression are greater in female animals. With the support of relevant studies, it has been suggested that sex hormones or steroids can modulate the activities of BDNF, which may account for its functional discrepancy in different sexes. Indeed, the cross-talk between BDNF and sex steroids has been detected for decades, and some sex steroids, such as estrogen, have a positive regulatory effect on BDNF expression and signaling. Thus, the sex of animal models that are used in studying the functions of BDNF is critical. This Mini-Review summarizes our current findings on the differences in expression, signaling, and functions of BDNF between sexes. We also discuss the potential mechanisms for mediating these differential responses, with a specific emphasis on sex steroids. By presenting and discussing these findings, we seek to encourage researchers to take sex influences into consideration when designing experiments, interpreting results, and drawing conclusions. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
29
|
Jover-Mengual T, Castelló-Ruiz M, Burguete MC, Jorques M, López-Morales MA, Aliena-Valero A, Jurado-Rodríguez A, Pérez S, Centeno JM, Miranda FJ, Alborch E, Torregrosa G, Salom JB. Molecular mechanisms mediating the neuroprotective role of the selective estrogen receptor modulator, bazedoxifene, in acute ischemic stroke: A comparative study with 17β-estradiol. J Steroid Biochem Mol Biol 2017; 171:296-304. [PMID: 28479229 DOI: 10.1016/j.jsbmb.2017.05.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022]
Abstract
As the knowledge on the estrogenic system in the brain grows, the possibilities to modulate it in order to afford further neuroprotection in brain damaging disorders so do it. We have previously demonstrated the ability of the selective estrogen receptor modulator, bazedoxifene (BZA), to reduce experimental ischemic brain damage. The present study has been designed to gain insight into the molecular mechanisms involved in such a neuroprotective action by investigating: 1) stroke-induced apoptotic cell death; 2) expression of estrogen receptors (ER) ERα, ERβ and the G-protein coupled estrogen receptor (GPER); and 3) modulation of MAPK/ERK1/2 and PI3K/Akt signaling pathways. For comparison, a parallel study was done with 17β-estradiol (E2)-treated animals. Male Wistar rats subject to transient right middle cerebral artery occlusion (tMCAO, intraluminal thread technique, 60min), were distributed in vehicle-, BZA- (20.7±2.1ng/mL in plasma) and E2- (45.6±7.8pg/mL in plasma) treated groups. At 24h from the onset of tMCAO, RT-PCR, Western blot and histochemical analysis were performed on brain tissue samples. Ischemia-reperfusion per se increased apoptosis as assessed by both caspase-3 activity and TUNEL-positive cell counts, which were reversed by both BZA and E2. ERα and ERβ expression, but not that of GPER, was reduced by the ischemic insult. BZA and E2 had different effects: while BZA increased both ERα and ERβ expression, E2 increased ERα expression but did not change that of ERβ. Both MAPK/ERK1/2 and PI3K/Akt pathways were stimulated under ischemic conditions. While BZA strongly reduced the increased p-ERK1/2 levels, E2 did not. Neither BZA nor E2 modified ischemia-induced increase in p-Akt levels. These results show that modulation of ERα and ERβ expression, as well as of the ERK1/2 signaling pathway accounts, at least in part, for the inhibitory effect of BZA on the stroke-induced apoptotic cell death. This lends mechanistic support to the consideration of BZA as a potential neuroprotective drug in acute ischemic stroke treatment.
Collapse
Affiliation(s)
- Teresa Jover-Mengual
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - María Jorques
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Mikahela A López-Morales
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Andrés Jurado-Rodríguez
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Salvador Pérez
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - José M Centeno
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Francisco J Miranda
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Enrique Alborch
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia Spain.
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Departamento de Fisiología, Universidad de Valencia, Hospital Universitario y Politécnico La Fe, Valencia, Spain; Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Valencia Spain
| |
Collapse
|
30
|
Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res 2016; 95:409-421. [DOI: 10.1002/jnr.23828] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Eduardo Sanches
- Division of Child Development and Growth, Department of Pediatrics; University of Geneva; Geneva Switzerland
| | - Felipe Kawa Odorcyk
- Postgraduate Program of Neurosciences, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Luz Elena Duran-Carabali
- Postgraduate Program of Physiology, Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul; Porto Alegre Rio Grande do Sul Brazil
| | - Simone Nardin Weis
- Department of Cellular Biology; Universidade de Brasília; Brasilia Distrito Federal Brazil
| |
Collapse
|
31
|
Chanana V, Tumturk A, Kintner D, Udho E, Ferrazzano P, Cengiz P. Sex Differences in Mouse Hippocampal Astrocytes after In-Vitro Ischemia. J Vis Exp 2016. [PMID: 27805577 DOI: 10.3791/53695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Astrogliosis following hypoxia/ischemia (HI)-related brain injury plays a role in increased morbidity and mortality in neonates. Recent clinical studies indicate that the severity of brain injury appear to be sex dependent, and that the male neonates are more susceptible to the effects of HI-related brain injury, resulting in more severe neurological outcomes as compared to females with comparable brain injuries. The development of reliable methods to isolate and maintain highly enriched populations of sexed hippocampal astrocytes is essential to understand the cellular basis of sex differences in the pathological consequences of neonatal HI. In this study, we describe a method for creating sex specific hippocampal astrocyte cultures that are subjected to a model of in-vitro ischemia, oxygen-glucose deprivation, followed by reoxygenation. Subsequent reactive astrogliosis was examined by immunostaining for the Glial Fibrillary Acidic Protein (GFAP) and S100B. This method provides a useful tool to study the role of male and female hippocampal astrocytes following neonatal HI, separately.
Collapse
Affiliation(s)
| | | | | | | | - Peter Ferrazzano
- Waisman Center, University of Wisconsin; Department of Pediatrics, University of Wisconsin
| | - Pelin Cengiz
- Waisman Center, University of Wisconsin; Department of Pediatrics, University of Wisconsin;
| |
Collapse
|
32
|
The Potential of Gonadal Hormone Signalling Pathways as Therapeutics for Dementia. J Mol Neurosci 2016; 60:336-348. [PMID: 27525638 DOI: 10.1007/s12031-016-0813-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022]
Abstract
Dementia is an ever-expanding problem facing an ageing society. Currently, there is a sharp paucity of treatment strategies. It has long been known that sex hormones, namely 17β-estradiol and testosterone, possess neuroprotective- and cognitive-enhancing qualities. However, certain lacunae in the knowledge underlying their molecular mechanisms have delayed their use as treatment strategies in dementia. With recent advancements in pharmacology and molecular biology, especially in the development of safer selective oestrogen receptor modulators and the recent discovery of the small-molecule brain-derived neurotrophic factor receptor agonist, 7,8-dihydroxyflavone, the exploitation of these signalling pathways for clinical use has become possible. This review aims to adumbrate the evidence and hurdles underscoring the use of sex hormones in the treatment of dementia as well as discussing some direction that is required to advance the translation of evidence into practise.
Collapse
|