1
|
Strnad FA, Brown AS, Wieben M, Cortes-Sanchez E, Williams ME, Fung CM. Intrauterine Growth Restriction Alters Postnatal Hippocampal Dentate Gyrus Neuron and Microglia Morphology and Cytokine/Chemokine Milieu in Mice. Life (Basel) 2024; 14:1627. [PMID: 39768335 PMCID: PMC11676380 DOI: 10.3390/life14121627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Infants born with intrauterine growth restriction (IUGR) have up to a five-fold higher risk of learning and memory impairment than those with normal growth. Using a mouse model of hypertensive diseases of pregnancy (HDP) to replicate uteroplacental insufficiency (UPI), we have previously shown that UPI causes premature embryonic hippocampal dentate gyrus (DG) neurogenesis in IUGR offspring. The DG is a brain region that receives the first cortical information for memory formation. In the current study, we examined the postnatal DG neuron morphology one month after delivery (P28) using recombinant adeno-associated viral labeling of neurons. We also examined DG microglia's morphology using immunofluorescent histochemistry and defined the hippocampal cytokine/chemokine milieu using Luminex xMAP technology. We found that IUGR preserved the principal dendrite lengths but decreased the dendritic branching and volume of DG neurons. IUGR augmented DG microglial number and cell size. Lastly, IUGR altered the hippocampal cytokine/chemokine profile in a sex-specific manner. We conclude that the prematurely-generated neuronal progenitors develop abnormal morphologies postnatally in a cell-autonomous manner. Microglia appear to modulate neuronal morphology by interacting with dendrites amidst a complex cytokine/chemokine environment that could ultimately lead to adult learning and memory deficits in our mouse model.
Collapse
Affiliation(s)
- Frank A. Strnad
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; (F.A.S.); (A.S.B.); (M.W.)
| | - Ashley S. Brown
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; (F.A.S.); (A.S.B.); (M.W.)
| | - Matthew Wieben
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; (F.A.S.); (A.S.B.); (M.W.)
| | | | - Megan E. Williams
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA;
| | - Camille M. Fung
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84108, USA; (F.A.S.); (A.S.B.); (M.W.)
| |
Collapse
|
2
|
Casciaro C, Hamada H, Bloise E, Matthews SG. The paternal contribution to shaping the health of future generations. Trends Endocrinol Metab 2024:S1043-2760(24)00275-3. [PMID: 39562264 DOI: 10.1016/j.tem.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024]
Abstract
Paternal health and exposure to adverse environments in the period prior to conception have a profound impact on future generations. Adversities such as stress, diet, and toxicants influence offspring health. Emerging evidence indicates that epigenetic mechanisms including noncoding RNA, DNA methylation, and chromatin remodelling mediate these effects. Preclinical studies have contributed to advancing mechanistic understanding in the field; however, human research is limited and primarily observational. Here, we discuss the evidence linking paternal to offspring health and advocate for further research in this area, which may ultimately inform policy and healthcare guidelines to improve paternal preconception health and offspring outcomes.
Collapse
Affiliation(s)
| | - Hirotaka Hamada
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Enrrico Bloise
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stephen G Matthews
- Department of Physiology, University of Toronto, Toronto, ON, Canada; Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON, Canada.
| |
Collapse
|
3
|
Nayan NM, Husin A, Siran R. The risk of prenatal bisphenol A exposure in early life neurodevelopment: Insights from epigenetic regulation. Early Hum Dev 2024; 198:106120. [PMID: 39293157 DOI: 10.1016/j.earlhumdev.2024.106120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Bisphenols are mainly used as protective coatings for plastics and resin-based materials in various consumer products. Industrial producers have a high demand for bisphenol A (BPA) among all bisphenol substitutes for various consumer products. However, according to reports, prolonged exposure to BPA can cause multiple health issues, including neurodevelopmental disorders in young children. BPA exposure during pregnancy has been considered as the primary cause of increasing the risk of neurological disorders in children as their neural systems are designed to respond to any environmental changes during prenatal life. Recently, there has been an increased focus on the effects of prenatal exposure to BPA, as it has been found to alter gene expression related to epigenetic mechanisms like DNA methylation, histone modification, and microRNA expression. Based on the evidence, frequent interactions can lead to inherited changes in an individual's neural profile. In this review, we delve into the current knowledge regarding the toxicity mechanism of BPA for expecting mothers. Next, we will discuss the possible action of BPA on the epigenetic mechanism during brain development. This is especially important to portray an overview on the role of epigenetic modification caused by prenatal BPA exposure and next, give future directions for improving human health risk assessment caused by BPA exposure.
Collapse
Affiliation(s)
- Norazirah Mat Nayan
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Malaysia; Institute of Medical Molecular and Biotechnology (IMMB) Faculty of Medicine, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Andrean Husin
- Faculty of Dentistry, Universiti Teknologi MARA, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, Malaysia
| | - Rosfaiizah Siran
- Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Malaysia; Neuroscience Research Group (NRG), Faculty of Medicine, Universiti Teknologi MARA, Malaysia.
| |
Collapse
|
4
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
5
|
Sourisse JM, Semmelhack JL, Schunter C. Parental thermal conditions affect the brain activity response to alarm cue in larval zebrafish. PeerJ 2024; 12:e18241. [PMID: 39399440 PMCID: PMC11471146 DOI: 10.7717/peerj.18241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/15/2024] [Indexed: 10/15/2024] Open
Abstract
Temperature is a crucial factor affecting the physiology of ectothermic animals, but exposure to elevated temperature during specific life stages and across generations may confer fish resilience through phenotypic plasticity. In this study, we investigate the effects of developmental and parental temperature on brain activity response to an olfactory cue in the larval zebrafish, Danio rerio. We exposed parents during reproduction and their offspring during development to control (28 °C) or elevated temperature (30 °C) and observed the response of the larval telencephalon to an alarm cue using live calcium imaging. Parental exposure to elevated temperature decreased the time till maximum brain activity response regardless of the offspring's developmental temperature, revealing that parental thermal conditions can affect the excitability of the offspring's neural circuitry. Furthermore, brain activity duration was affected by the interaction between parental and offspring thermal conditions, where longer brain activity duration was seen when either parents or offspring were exposed to elevated temperature. Conversely, we found shorter brain activity duration when the offspring were exposed to the same temperature as their parents, in both control and elevated temperature. This could represent an anticipatory parental effect influencing the offspring's brain response to match the parental environment, or an early developmental effect occurring within a susceptible short time window post-fertilization. Overall, our results suggest that warming can alter processes involved in brain transmission and show that parental conditions could aid in the preparation of their offspring to respond to olfactory stimuli in a warming environment.
Collapse
Affiliation(s)
- Jade M. Sourisse
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
- Marine and Environmental Sciences Centre, Laboratório Marítimo da Guia, Faculdade de Ciências, Universidade de Lisboa, Cascais, Portugal
| | - Julie L. Semmelhack
- The Division of Life Science, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong
| | - Celia Schunter
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Anisman H, Doubad D, Asokumar A, Matheson K. Psychosocial and neurobiological aspects of the worldwide refugee crisis: From vulnerability to resilience. Neurosci Biobehav Rev 2024; 165:105859. [PMID: 39159733 DOI: 10.1016/j.neubiorev.2024.105859] [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: 02/28/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
Anisman, H., Doubad, D., Asokumar, A. & Matheson, K. Psychosocial and neurobiological aspects of the worldwide refugee crisis: From vulnerability to resilience. NEUROSCI BIOBEHAV REV, XXXX. Immigration occurs between countries either to obtain employment, for family reunification or to escape violence and other life-threatening conditions. Refugees and asylum seekers are often obligated to overcome a uniquely challenging set of circumstances prior to and during migration. Settlement following immigration may pose yet another set of stressors related to acculturation to the host country, as well as financial insecurity, discrimination, language barriers, and social isolation. Here we discuss the multiple consequences of immigration experiences, focusing on the health disturbances that frequently develop in adults and children. Aside from the psychosocial influences, immigration-related challenges may cause hormonal, inflammatory immune, and microbiota changes that favor psychological and physical illnesses. Some biological alterations are subject to modification by epigenetic changes, which have implications for intergenerational trauma transmission, as might disruptions in parenting behaviors and family dysfunction. Despite the hardships experienced, many immigrants and their families exhibit positive psychological adjustment after resettlement. We provide information to diminish the impacts associated with immigration and offer strength-based approaches that may foster resilience.
Collapse
Affiliation(s)
- H Anisman
- Carleton University, Department of Neuroscience, Ottawa, Ontario K1S 5B6, Canada.
| | - D Doubad
- Carleton University, Department of Neuroscience, Ottawa, Ontario K1S 5B6, Canada
| | - A Asokumar
- Carleton University, Department of Neuroscience, Ottawa, Ontario K1S 5B6, Canada
| | - K Matheson
- Carleton University, Department of Neuroscience, Ottawa, Ontario K1S 5B6, Canada
| |
Collapse
|
7
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
8
|
Zilkha N, Chuartzman SG, Fishman R, Ben-Dor S, Kimchi T. Maternal high-fat or low-protein diets promote autism-related behavior and altered social behavior within groups in offspring male mice. Sci Rep 2024; 14:19227. [PMID: 39164365 PMCID: PMC11336096 DOI: 10.1038/s41598-024-70062-w] [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: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
Maternal malnutrition has been associated with neurodevelopmental deficits and long-term implications on the offspring's health and behavior. Here, we investigated the effects of maternal low-protein diet (LPD) or obesity-inducing maternal high-fat diet (HFD) on dyadic social interactions, group organization and autism-related behaviors in mice. We found that maternal HFD induced an autism-related behavioral phenotype in the male offspring, including a robust decrease in sociability, increased aggression, cognitive rigidity and repetitive behaviors. Maternal LPD led to a milder yet significant effect on autism-related symptoms, with no effects on olfactory-mediated social behavior. Under naturalistic conditions in a group setting, this manifested in altered behavioral repertoires, increased magnitude in dominance relations, and reduced interactions with novel social stimuli in the HFD male offspring, but not in the LPD offspring. Finally, we found HFD-induced transcriptomic changes in the olfactory bulbs of the male offspring. Together, our findings show that maternal malnutrition induces long-lasting effects on aggression and autism-related behaviors in male offspring, and potential impairments in brain regions processing chemosensory signals.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | | | - Ruth Fishman
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Shifra Ben-Dor
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
9
|
Bellia G, Chang J, Liew Z, Vernetti A, Macari S, Powell K, Chawarska K. Family history of psychiatric conditions and development of siblings of children with autism. Autism Res 2024; 17:1665-1676. [PMID: 38896553 PMCID: PMC11341253 DOI: 10.1002/aur.3175] [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: 03/12/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024]
Abstract
Younger siblings (SIBS) of children with autism exhibit a wide range of clinical and subclinical symptoms including social, cognitive, language, and adaptive functioning delays. Identifying factors linked with this phenotypic heterogeneity is essential for improving understanding of the underlying biology of the heterogenous outcomes and for early identification of the most vulnerable SIBS. Prevalence of neurodevelopmental (NDD) and neuropsychiatric disorders (NPD) is significantly elevated in families of children with autism. It remains unknown, however, if the family history associates with the developmental outcomes among the SIBS. We quantified history of the NDDs and NPDs commonly reported in families of children with autism using a parent interview and assessed autism symptoms, verbal, nonverbal, and adaptive skills in a sample of 229 SIBS. Multiple regression analyses were used to examine links between family history and phenotypic outcomes, whereas controlling for birth year, age, sex, demographics, and parental education. Results suggest that family history of schizophrenia, depression, anxiety, bipolar disorder, and intellectual disability associate robustly with dimensional measures of social affect, verbal and nonverbal IQ, and adaptive functioning in the SIBS. Considering family history of these disorders may improve efforts to predict long-term outcomes in younger siblings of children with autism and inform about familial factors contributing to high phenotypic heterogenetity in this cohort.
Collapse
Affiliation(s)
- Giselle Bellia
- Yale Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Joseph Chang
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut
- Yale Child Study Center, New Haven, Connecticut
| | - Zeyan Liew
- Yale Center for Perinatal, Pediatric and Environmental Epidemiology, Yale School of Public Health, New Haven, Connecticut
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut
| | | | | | | | - Katarzyna Chawarska
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut
- Yale Child Study Center, New Haven, Connecticut
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
10
|
Pilkay SR, Knight AK, Bush NR, LeWinn K, Davis RL, Tylavsky F, Smith AK. Poverty and neighborhood opportunity effects on neonate DNAm developmental age. PLoS One 2024; 19:e0306452. [PMID: 38995877 PMCID: PMC11244791 DOI: 10.1371/journal.pone.0306452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Children from families with low socioeconomic status (SES), as determined by income, experience several negative outcomes, such as higher rates of newborn mortality and behavioral issues. Moreover, associations between DNA methylation and low income or poverty status are evident beginning at birth, suggesting prenatal influences on offspring development. Recent evidence suggests neighborhood opportunities may protect against some of the health consequences of living in low income households. The goal of this study was to assess whether neighborhood opportunities moderate associations between household income (HI) and neonate developmental maturity as measured with DNA methylation. METHODS Umbilical cord blood DNA methylation data was available in 198 mother-neonate pairs from the larger CANDLE cohort. Gestational age acceleration was calculated using an epigenetic clock designed for neonates. Prenatal HI and neighborhood opportunities measured with the Childhood Opportunity Index (COI) were regressed on gestational age acceleration controlling for sex, race, and cellular composition. RESULTS Higher HI was associated with higher gestational age acceleration (B = .145, t = 4.969, p = 1.56x10-6, 95% CI [.087, .202]). Contrary to expectation, an interaction emerged showing higher neighborhood educational opportunity was associated with lower gestational age acceleration at birth for neonates with mothers living in moderate to high HI (B = -.048, t = -2.08, p = .03, 95% CI [-.092, -.002]). Female neonates showed higher gestational age acceleration at birth compared to males. However, within males, being born into neighborhoods with higher social and economic opportunity was associated with higher gestational age acceleration. CONCLUSION Prenatal HI and neighborhood qualities may affect gestational age acceleration at birth. Therefore, policy makers should consider neighborhood qualities as one opportunity to mitigate prenatal developmental effects of HI.
Collapse
Affiliation(s)
- Stefanie R Pilkay
- School of Social Work, Syracuse University, Syracuse, New York, United States of America
| | - Anna K Knight
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta Georgia, United States of America
| | - Nicole R Bush
- Department of Psychiatry, University of California San Francisco School of Medicine, San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, University of California San Francisco, San Francisco, San Francisco, California, United States of America
| | - Kaja LeWinn
- Department of Psychiatry, University of California San Francisco School of Medicine, San Francisco, San Francisco, California, United States of America
| | - Robert L Davis
- Health and Science Center, University of Tennessee, Memphis, Tennessee, United States of America
| | - Frances Tylavsky
- Health and Science Center, University of Tennessee, Memphis, Tennessee, United States of America
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta Georgia, United States of America
| |
Collapse
|
11
|
Vyushina AV, Pritvorova AV, Pivina SG, Ordyan NE. Transgenerational effect of prenatal stress on behavior and lipid peroxidation in brain structures of female rats during the estral cycle. Vavilovskii Zhurnal Genet Selektsii 2024; 28:387-397. [PMID: 39027128 PMCID: PMC11253015 DOI: 10.18699/vjgb-24-44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/19/2023] [Accepted: 02/09/2023] [Indexed: 07/20/2024] Open
Abstract
The effect of stress in pregnant female Wistar rats on the behavior and lipid peroxidation (LP) in the neocortex, hippocampus and hypothalamus in the female F2 generation during the ovarian cycle was investigated. We subjected pregnant females to daily 1-hour immobilization stress from the 15th to the 19th days of pregnancy. Further, family groups were formed from prenatally stressed and control male and female rats of the F1 generation: group 1, the control female and male; group 2, the control female and the prenatally stressed male; group 3, the prenatally stressed female and the control male; group 4, the prenatally stressed female and male. The females of the F2 generation born from these couples were selected into four experimental groups in accordance with the family group. At the age of 3 months, behavior of rats was studied in the "open field" test in two stages of the ovarian cycle - estrus and diestrus. After 7-10 days, the rats were decapitated and the neocortex, hypothalamus and hippocampus were selected to determine the level of diene and triene conjugates, Schiff bases and the degree of lipid oxidation (Klein index). In F2 females with one prenatally stressed parent, there was no interstage difference in locomotor-exploratory activity and anxiety. If both F1 parents were prenatally stressed, female F2 rats retained interstage differences similar to the control pattern, while their locomotor-exploratory activity and time spent in the center of an "open field" decreased in absolute values. In the neocortex of F2 females in groups with prenatally stressed mothers, the level of primary LP products decreased and the level of Schiff bases increased in the estrus stage. In the hippocampus of F2 females in the groups with prenatally stressed fathers, the level of Schiff bases decreased in the estrus stage, and the level of primary LP products increased in group 2 and decreased in group 4. In the hypothalamus of F2 females in the groups with prenatally stressed mothers, the level of Schiff bases increased in the estrus stage and decreased in the diestrus; in addition, in group 3, the level of primary LP products in the estrus stage increased. Thus, we demonstrated the influence of prenatal stress of both F1 mother and F1 father on the behavior and the level of LP in the neocortex, hippocampus and hypothalamus in female rats of the F2 generation in estrus and diestrus.
Collapse
Affiliation(s)
- A V Vyushina
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - A V Pritvorova
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - S G Pivina
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - N E Ordyan
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
12
|
Basak S, Mallick R, Navya Sree B, Duttaroy AK. Placental Epigenome Impacts Fetal Development: Effects of Maternal Nutrients and Gut Microbiota. Nutrients 2024; 16:1860. [PMID: 38931215 PMCID: PMC11206482 DOI: 10.3390/nu16121860] [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: 05/02/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish an exposure threshold due to nutrient deficiencies or other environmental factors that can result in clinically relevant epigenetic alterations that modulate disease risks in the fetus. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development. This review unravels the potential roles of the functional epigenome for targeted intervention to ensure optimal fetal brain development and its performance in later life.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Boga Navya Sree
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
13
|
Cánepa ET, Berardino BG. Epigenetic mechanisms linking early-life adversities and mental health. Biochem J 2024; 481:615-642. [PMID: 38722301 DOI: 10.1042/bcj20230306] [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: 02/19/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/15/2024]
Abstract
Early-life adversities, whether prenatal or postnatal exposure, have been linked to adverse mental health outcomes later in life increasing the risk of several psychiatric disorders. Research on its neurobiological consequences demonstrated an association between exposure to adversities and persistent alterations in the structure, function, and connectivity of the brain. Consistent evidence supports the idea that regulation of gene expression through epigenetic mechanisms are involved in embedding the impact of early-life experiences in the genome and mediate between social environments and later behavioral phenotypes. In addition, studies from rodent models and humans suggest that these experiences and the acquired risk factors can be transmitted through epigenetic mechanisms to offspring and the following generations potentially contributing to a cycle of disease or disease risk. However, one of the important aspects of epigenetic mechanisms, unlike genetic sequences that are fixed and unchangeable, is that although the epigenetic markings are long-lasting, they are nevertheless potentially reversible. In this review, we summarize our current understanding of the epigenetic mechanisms involved in the mental health consequences derived from early-life exposure to malnutrition, maltreatment and poverty, adversities with huge and pervasive impact on mental health. We also discuss the evidence about transgenerational epigenetic inheritance in mammals and experimental data suggesting that suitable social and pharmacological interventions could reverse adverse epigenetic modifications induced by early-life negative social experiences. In this regard, these studies must be accompanied by efforts to determine the causes that promote these adversities and that result in health inequity in the population.
Collapse
Affiliation(s)
- Eduardo T Cánepa
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| | - Bruno G Berardino
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| |
Collapse
|
14
|
Harris JC, Trigg NA, Goshu B, Yokoyama Y, Dohnalová L, White EK, Harman A, Murga-Garrido SM, Ting-Chun Pan J, Bhanap P, Thaiss CA, Grice EA, Conine CC, Kambayashi T. The microbiota and T cells non-genetically modulate inherited phenotypes transgenerationally. Cell Rep 2024; 43:114029. [PMID: 38573852 PMCID: PMC11102039 DOI: 10.1016/j.celrep.2024.114029] [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: 07/12/2023] [Revised: 01/21/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024] Open
Abstract
The host-microbiota relationship has evolved to shape mammalian physiology, including immunity, metabolism, and development. Germ-free models are widely used to study microbial effects on host processes such as immunity. Here, we find that both germ-free and T cell-deficient mice exhibit a robust sebum secretion defect persisting across multiple generations despite microbial colonization and T cell repletion. These phenotypes are inherited by progeny conceived during in vitro fertilization using germ-free sperm and eggs, demonstrating that non-genetic information in the gametes is required for microbial-dependent phenotypic transmission. Accordingly, gene expression in early embryos derived from gametes from germ-free or T cell-deficient mice is strikingly and similarly altered. Our findings demonstrate that microbial- and immune-dependent regulation of non-genetic information in the gametes can transmit inherited phenotypes transgenerationally in mice. This mechanism could rapidly generate phenotypic diversity to enhance host adaptation to environmental perturbations.
Collapse
Affiliation(s)
- Jordan C Harris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Natalie A Trigg
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Departments of Genetics and Pediatrics - Penn Epigenetics Institute, Institute of Regenerative Medicine, and Center for Research on Reproduction and Women's Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bruktawit Goshu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuichi Yokoyama
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lenka Dohnalová
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen K White
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adele Harman
- Transgenic Core, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sofía M Murga-Garrido
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamie Ting-Chun Pan
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Preeti Bhanap
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Colin C Conine
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Departments of Genetics and Pediatrics - Penn Epigenetics Institute, Institute of Regenerative Medicine, and Center for Research on Reproduction and Women's Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Walhovd KB, Krogsrud SK, Amlien IK, Sørensen Ø, Wang Y, Bråthen ACS, Overbye K, Kransberg J, Mowinckel AM, Magnussen F, Herud M, Håberg AK, Fjell AM, Vidal-Pineiro D. Fetal influence on the human brain through the lifespan. eLife 2024; 12:RP86812. [PMID: 38602745 PMCID: PMC11008813 DOI: 10.7554/elife.86812] [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] [Indexed: 04/12/2024] Open
Abstract
Human fetal development has been associated with brain health at later stages. It is unknown whether growth in utero, as indexed by birth weight (BW), relates consistently to lifespan brain characteristics and changes, and to what extent these influences are of a genetic or environmental nature. Here we show remarkably stable and lifelong positive associations between BW and cortical surface area and volume across and within developmental, aging and lifespan longitudinal samples (N = 5794, 4-82 y of age, w/386 monozygotic twins, followed for up to 8.3 y w/12,088 brain MRIs). In contrast, no consistent effect of BW on brain changes was observed. Partly environmental effects were indicated by analysis of twin BW discordance. In conclusion, the influence of prenatal growth on cortical topography is stable and reliable through the lifespan. This early-life factor appears to influence the brain by association of brain reserve, rather than brain maintenance. Thus, fetal influences appear omnipresent in the spacetime of the human brain throughout the human lifespan. Optimizing fetal growth may increase brain reserve for life, also in aging.
Collapse
Affiliation(s)
- Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University HospitalOsloNorway
| | - Stine K Krogsrud
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Inge K Amlien
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Yunpeng Wang
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | | | - Knut Overbye
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Jonas Kransberg
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | | | - Fredrik Magnussen
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Martine Herud
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| | - Asta K Håberg
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and TechnologyOsloNorway
| | - Anders Martin Fjell
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University HospitalOsloNorway
| | - Didac Vidal-Pineiro
- Center for Lifespan Changes in Brain and Cognition, University of OsloOsloNorway
| |
Collapse
|
16
|
Zambon A, Rico LC, Herman M, Gundacker A, Telalovic A, Hartenberger LM, Kuehn R, Romanov RA, Hussaini SA, Harkany T, Pollak DD. Gestational immune activation disrupts hypothalamic neurocircuits of maternal care behavior. Mol Psychiatry 2024; 29:859-873. [PMID: 35581295 PMCID: PMC9112243 DOI: 10.1038/s41380-022-01602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/27/2022]
Abstract
Immune activation is one of the most common complications during pregnancy, predominantly evoked by viral infections. Nevertheless, how immune activation affects mother-offspring relationships postpartum remains unknown. Here, by using the polyinosinic-polycytidylic acid (Poly I:C) model of gestational infection we show that viral-like immune activation at mid-gestation persistently changes hypothalamic neurocircuit parameters in mouse dams and, consequently, is adverse to parenting behavior. Poly I:C-exposed dams favor non-pup-directed exploratory behavior at the expense of pup retrieval. These behavioral deficits are underlain by dendrite pruning and lesser immediate early gene activation in Galanin (Gal)+ neurons with dam-specific transcriptional signatures that reside in the medial preoptic area (mPOA). Reduced activation of an exclusively inhibitory contingent of these distal-projecting Gal+ neurons allows for increased feed-forward inhibition onto putative dopaminergic neurons in the ventral tegmental area (VTA) in Poly I:C-exposed dams. Notably, destabilized VTA output specifically accompanies post-pup retrieval epochs. We suggest that gestational immunogenic insults bias both threat processing and reward perception, manifesting as disfavored infant caregiving.
Collapse
Affiliation(s)
- Alice Zambon
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Mathieu Herman
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amina Telalovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lisa-Marie Hartenberger
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rebekka Kuehn
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - S Abid Hussaini
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
17
|
Gao S, Wang Y, Li X, Liang Y, Jin Z, Yang B, Yuan TF, Tian H, Peng B, Rao Y. Dynamics of N6-methyladenosine modification during Alzheimer's disease development. Heliyon 2024; 10:e26911. [PMID: 38496847 PMCID: PMC10944207 DOI: 10.1016/j.heliyon.2024.e26911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
N6-methyladenosine (m6A) modification is a common RNA modification in the central nervous system and has been linked to various neurological disorders, including Alzheimer's disease (AD). However, the dynamic of mRNA m6A modification and m6A enzymes during the development of AD are not well understood. Therefore, this study examined the expression profiles of m6A and its enzymes in the development of AD. The results showed that changes in the expression levels of m6A regulatory factors occur in the early stages of AD, indicating a potential role for m6A modification in the onset of the disease. Additionally, the analysis of mRNA m6A expression profiles using m6A-seq revealed significant differences in m6A modification between AD and control brains. The genes with differential methylation were found to be enriched in GO and KEGG terms related to processes such as inflammation response, immune system processes. And the differently expressed genes (DEGs) are negatively lryassociated with genes involved in microglia hemostasis, but positively associated with genes related to "disease-associated microglia" (DAM) associated genes. These findings suggest that dysregulation of mRNA m6A modification may contribute to the development of AD by affecting the function and gene expression of microglia.
Collapse
Affiliation(s)
- Shuai Gao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
| | - Yuqing Wang
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
- Department of Medical Science, Medical College of Jinzhou Medical University, Jinzhou, Liaoning, 121010, China
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
| | - Yuqing Liang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhihao Jin
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Baozhi Yang
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China
| | - Hengli Tian
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Fudan University, Shanghai, 200040, China
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
18
|
Sood E, Newburger JW, Anixt JS, Cassidy AR, Jackson JL, Jonas RA, Lisanti AJ, Lopez KN, Peyvandi S, Marino BS. Neurodevelopmental Outcomes for Individuals With Congenital Heart Disease: Updates in Neuroprotection, Risk-Stratification, Evaluation, and Management: A Scientific Statement From the American Heart Association. Circulation 2024; 149:e997-e1022. [PMID: 38385268 DOI: 10.1161/cir.0000000000001211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Over the past decade, new research has advanced scientific knowledge of neurodevelopmental trajectories, factors that increase neurodevelopmental risk, and neuroprotective strategies for individuals with congenital heart disease. In addition, best practices for evaluation and management of developmental delays and disorders in this high-risk patient population have been formulated based on literature review and expert consensus. This American Heart Association scientific statement serves as an update to the 2012 statement on the evaluation and management of neurodevelopmental outcomes in children with congenital heart disease. It includes revised risk categories for developmental delay or disorder and an updated list of factors that increase neurodevelopmental risk in individuals with congenital heart disease according to current evidence, including genetic predisposition, fetal and perinatal factors, surgical and perioperative factors, socioeconomic disadvantage, and parental psychological distress. It also includes an updated algorithm for referral, evaluation, and management of individuals at high risk. Risk stratification of individuals with congenital heart disease with the updated categories and risk factors will identify a large and growing population of survivors at high risk for developmental delay or disorder and associated impacts across the life span. Critical next steps must include efforts to prevent and mitigate developmental delays and disorders. The goal of this scientific statement is to inform health care professionals caring for patients with congenital heart disease and other key stakeholders about the current state of knowledge of neurodevelopmental outcomes for individuals with congenital heart disease and best practices for neuroprotection, risk stratification, evaluation, and management.
Collapse
|
19
|
Huang N, Cui J, Fan G, Pan T, Han K, Xu K, Jiang C, Liu X, Wang F, Ma L, Le Q. Transcriptomic effects of paternal cocaine-seeking on the reward circuitry of male offspring. Transl Psychiatry 2024; 14:120. [PMID: 38409093 PMCID: PMC10897445 DOI: 10.1038/s41398-024-02839-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/10/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
It has been previously established that paternal development of a strong incentive motivation for cocaine can predispose offspring to develop high cocaine-seeking behavior, as opposed to sole exposure to the drug that results in drug resistance in offspring. However, the adaptive changes of the reward circuitry have not been fully elucidated. To infer the key nuclei and possible hub genes that determine susceptibility to addiction in offspring, rats were randomly assigned to three groups, cocaine self-administration (CSA), yoked administration (Yoke), and saline self-administration (SSA), and used to generate F1. We conducted a comprehensive transcriptomic analysis of the male F1 offspring across seven relevant brain regions, both under drug-naïve conditions and after cocaine self-administration. Pairwise differentially expressed gene analysis revealed that the orbitofrontal cortex (OFC) exhibited more pronounced transcriptomic changes in response to cocaine exposure, while the dorsal hippocampus (dHip), dorsal striatum (dStr), and ventral tegmental area (VTA) exhibited changes that were more closely associated with the paternal voluntary cocaine-seeking behavior. Consistently, these nuclei showed decreased dopamine levels, elevated neuronal activation, and elevated between-nuclei correlations, indicating dopamine-centered rewiring of the midbrain circuit in the CSA offspring. To determine if possible regulatory cascades exist that drive the expression changes, we constructed co-expression networks induced by paternal drug addiction and identified three key clusters, primarily driven by transcriptional factors such as MYT1L, POU3F4, and NEUROD6, leading to changes of genes regulating axonogenesis, synapse organization, and membrane potential, respectively. Collectively, our data highlight vulnerable neurocircuitry and novel regulatory candidates with therapeutic potential for disrupting the transgenerational inheritance of vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- Nan Huang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Cui
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyuan Fan
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Tao Pan
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Kunxiu Han
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Kailiang Xu
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, 200438, China
| | - Changyou Jiang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Xing Liu
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Feifei Wang
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China
| | - Lan Ma
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China.
| | - Qiumin Le
- School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Research Unit of Addiction Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai, 200032, China.
| |
Collapse
|
20
|
Sanders AFP, Tirado B, Seider NA, Triplett RL, Lean RE, Neil JJ, Miller JP, Tillman R, Smyser TA, Barch DM, Luby JL, Rogers CE, Smyser CD, Warner BB, Chen E, Miller GE. Prenatal exposure to maternal disadvantage-related inflammatory biomarkers: associations with neonatal white matter microstructure. Transl Psychiatry 2024; 14:72. [PMID: 38307841 PMCID: PMC10837200 DOI: 10.1038/s41398-024-02782-6] [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: 11/17/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
Prenatal exposure to heightened maternal inflammation has been associated with adverse neurodevelopmental outcomes, including atypical brain maturation and psychiatric illness. In mothers experiencing socioeconomic disadvantage, immune activation can be a product of the chronic stress inherent to such environmental hardship. While growing preclinical and clinical evidence has shown links between altered neonatal brain development and increased inflammatory states in utero, the potential mechanism by which socioeconomic disadvantage differentially impacts neural-immune crosstalk remains unclear. In the current study, we investigated associations between socioeconomic disadvantage, gestational inflammation, and neonatal white matter microstructure in 320 mother-infant dyads over-sampled for poverty. We analyzed maternal serum levels of four cytokines (IL-6, IL-8, IL-10, TNF-α) over the course of pregnancy in relation to offspring white matter microstructure and socioeconomic disadvantage. Higher average maternal IL-6 was associated with very low socioeconomic status (SES; INR < 200% poverty line) and lower neonatal corticospinal fractional anisotropy (FA) and lower uncinate axial diffusivity (AD). No other cytokine was associated with SES. Higher average maternal IL-10 was associated with lower FA and higher radial diffusivity (RD) in corpus callosum and corticospinal tracts, higher optic radiation RD, lower uncinate AD, and lower FA in inferior fronto-occipital fasciculus and anterior limb of internal capsule tracts. SES moderated the relationship between average maternal TNF-α levels during gestation and neonatal white matter diffusivity. When these interactions were decomposed, the patterns indicated that this association was significant and positive among very low SES neonates, whereby TNF-α was inversely and significantly associated with inferior cingulum AD. By contrast, among the more advantaged neonates (lower-to-higher SES [INR ≥ 200% poverty line]), TNF-α was positively and significantly associated with superior cingulum AD. Taken together, these findings suggest that the relationship between prenatal cytokine exposure and white matter microstructure differs as a function of SES. These patterns are consistent with a scenario where gestational inflammation's effects on white matter development diverge depending on the availability of foundational resources in utero.
Collapse
Affiliation(s)
- Ashley F P Sanders
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Brian Tirado
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nicole A Seider
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Regina L Triplett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rachel E Lean
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jeffrey J Neil
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - J Philip Miller
- Division of Biostatistics, Institute for Informatics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rebecca Tillman
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tara A Smyser
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Deanna M Barch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Psychological and Brain Sciences, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Joan L Luby
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Cynthia E Rogers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christopher D Smyser
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Newborn Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Edith Chen
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| | - Gregory E Miller
- Institute for Policy Research, Northwestern University, Evanston, IL, 60208, USA
- Department of Psychology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
21
|
Kleeman EA, Reisinger SN, Adithya P, Houston B, Stathatos G, Garnham AL, McLaughlin S, O'Bryan MK, Gubert C, Hannan AJ. Paternal immune activation by Poly I:C modulates sperm noncoding RNA profiles and causes transgenerational changes in offspring behavior. Brain Behav Immun 2024; 115:258-279. [PMID: 37820975 DOI: 10.1016/j.bbi.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 10/02/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023] Open
Abstract
Paternal pre-conceptual environmental experiences, such as stress and diet, can affect offspring brain and behavioral phenotypes via epigenetic modifications in sperm. Furthermore, maternal immune activation due to infection during gestation can reprogram offspring behavior and brain functioning in adulthood. However, the effects of paternal pre-conceptual exposure to immune activation on the behavior and physiology of offspring (F1) and grand-offspring (F2) are not currently known. We explored effects of paternal pre-conceptual exposure to viral-like immune activation on F1 and F2 behavioral and physiological phenotypes using a C57BL/6J mouse model. Males were treated with a single injection (intraperitoneal) of the viral mimetic polyinosinic:polycytidylic acid (Poly I:C: 12 mg/kg) then bred with naïve female mice four weeks after the Poly I:C (or 0.9% saline control) injection. The F1 offspring of Poly I:C treated fathers displayed increased depression-like behavior in the Porsolt swim test, an altered stress response in the novelty-suppressed feeding test, and significant transcriptomic changes in their hippocampus. Additionally, the F1 male offspring of Poly I:C treated F0 males showed significantly increased immune responsivity after a Poly I:C immune challenge (12 mg/kg). Furthermore, the F2 male grand-offspring took longer to enter and travelled significantly shorter distances in the light zone of the light/dark box. An analysis of the small noncoding RNA profiles in sperm from Poly I:C treated males and their male offspring revealed significant effects of Poly I:C on the sperm microRNA content at the time of conception and on the sperm PIWI-interacting RNA content of the male offspring. Notably, eight miRNAs with an FDR < 0.05 (miR-141-3p, miR-126b-5p, miR-669o-5p, miR-10b-3p, miR-471-5p, miR-463-5p, miR-148b-3p, and miR-181c-5p) were found to be significantly downregulated in the sperm of Poly I:C treated males. Collectively, we demonstrate that paternal pre-conceptual exposure to a viral immune challenge results in both intergenerational and transgenerational effects on brain and behavior that may be mediated by alterations in the sperm small noncoding RNA content.
Collapse
Affiliation(s)
- Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Pranav Adithya
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Brendan Houston
- Bio21 Institute, University of Melbourne, Parkville, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Gemma Stathatos
- Bio21 Institute, University of Melbourne, Parkville, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Alexandra L Garnham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Shae McLaughlin
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Moira K O'Bryan
- Bio21 Institute, University of Melbourne, Parkville, Victoria, Australia; School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
22
|
Mallick R, Duttaroy AK. Epigenetic modification impacting brain functions: Effects of physical activity, micronutrients, caffeine, toxins, and addictive substances. Neurochem Int 2023; 171:105627. [PMID: 37827244 DOI: 10.1016/j.neuint.2023.105627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/14/2023]
Abstract
Changes in gene expression are involved in many brain functions. Epigenetic processes modulate gene expression by histone modification and DNA methylation or RNA-mediated processes, which is important for brain function. Consequently, epigenetic changes are also a part of brain diseases such as mental illness and addiction. Understanding the role of different factors on the brain epigenome may help us understand the function of the brain. This review discussed the effects of caffeine, lipids, addictive substances, physical activity, and pollutants on the epigenetic changes in the brain and their modulatory effects on brain function.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1046 Blindern, Oslo, Norway.
| |
Collapse
|
23
|
Tatemoto P, Pértille F, Bernardino T, Zanella R, Guerrero-Bosagna C, Zanella AJ. An enriched maternal environment and stereotypies of sows differentially affect the neuro-epigenome of brain regions related to emotionality in their piglets. Epigenetics 2023; 18:2196656. [PMID: 37192378 DOI: 10.1080/15592294.2023.2196656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 02/15/2023] [Accepted: 03/08/2023] [Indexed: 05/18/2023] Open
Abstract
Epigenetic mechanisms are important modulators of neurodevelopmental outcomes in the offspring of animals challenged during pregnancy. Pregnant sows living in a confined environment are challenged with stress and lack of stimulation which may result in the expression of stereotypies (repetitive behaviours without an apparent function). Little attention has been devoted to the postnatal effects of maternal stereotypies in the offspring. We investigated how the environment and stereotypies of pregnant sows affected the neuro-epigenome of their piglets. We focused on the amygdala, frontal cortex, and hippocampus, brain regions related to emotionality, learning, memory, and stress response. Differentially methylated regions (DMRs) were investigated in these brain regions of male piglets born from sows kept in an enriched vs a barren environment. Within the latter group of piglets, we compared the brain methylomes of piglets born from sows expressing stereotypies vs sows not expressing stereotypies. DMRs emerged in each comparison. While the epigenome of the hippocampus and frontal cortex of piglets is mainly affected by the maternal environment, the epigenome of the amygdala is mainly affected by maternal stereotypies. The molecular pathways and mechanisms triggered in the brains of piglets by maternal environment or stereotypies are different, which is reflected on the differential gene function associated to the DMRs found in each piglets' brain region . The present study is the first to investigate the neuro-epigenomic effects of maternal enrichment in pigs' offspring and the first to investigate the neuro-epigenomic effects of maternal stereotypies in the offspring of a mammal.
Collapse
Affiliation(s)
- Patricia Tatemoto
- Center for Comparative Studies in Sustainability, Health and Welfare, Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, FMVZ, University of São Paulo, Pirassununga, São Paulo, Brazil
| | - Fábio Pértille
- Avian Behavioral Genomics and Physiology Group, IFM Biology, Linköping University, Linköping, Sweden
- Animal Biotechnology Laboratory, Animal Science Department, University of São Paulo - Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, Brazil
- Physiology and Environmental Toxicology Program, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Thiago Bernardino
- Center for Comparative Studies in Sustainability, Health and Welfare, Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, FMVZ, University of São Paulo, Pirassununga, São Paulo, Brazil
- Graduation Program in One Health, University of Santo Amaro, São Paulo Brazil
| | - Ricardo Zanella
- Faculty of Agronomy and Veterinary Medicine, University of Passo Fundo, Passo Fundo, Rio Grande do Sul, Brazil
| | - Carlos Guerrero-Bosagna
- Avian Behavioral Genomics and Physiology Group, IFM Biology, Linköping University, Linköping, Sweden
- Physiology and Environmental Toxicology Program, Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Adroaldo José Zanella
- Center for Comparative Studies in Sustainability, Health and Welfare, Department of Preventive Veterinary Medicine and Animal Health, School of Veterinary Medicine and Animal Science, FMVZ, University of São Paulo, Pirassununga, São Paulo, Brazil
| |
Collapse
|
24
|
Zhang Z, Zhang H, Hu B, Luan Y, Zhu K, Ma B, Zhang Z, Zheng X. R-Loop Defines Neural Stem/Progenitor Cells During Mouse Neurodevelopment. Stem Cells Dev 2023; 32:719-730. [PMID: 37823735 DOI: 10.1089/scd.2023.0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Neural stem/progenitor cells (NSPCs) are present in the mammalian brain throughout life and are involved in neurodevelopment and central nervous system repair. Although typical epigenetic signatures, including DNA methylation, histone modifications, and microRNAs, play a pivotal role in regulation of NSPCs, several of the epigenetic regulatory mechanisms of NSPCs remain unclear. Thus, defining a novel epigenetic feature of NSPCs is crucial for developing stem cell therapy to address neurologic disorders caused by injury. In this study, we aimed to define the R-loop, a three-stranded nucleic acid structure, as an epigenetic characteristic of NSPCs during neurodevelopment. Our results demonstrated that R-loop levels change dynamically throughout neurodevelopment. Cells with high levels of R-loops consistently decreased and were enriched in the area of neurogenesis. Additionally, these cells costained with SOX2 during neurodevelopment. Furthermore, these cells with high R-loop levels expressed Ki-67 and exhibited a high degree of overlap with the transcriptional activation markers, H3K4me3, ser5, and H3K27ac. These findings suggest that R-loops may serve as an epigenetic feature for transcriptional activation in NSPCs, indicating their role in gene expression regulation and neurogenesis.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Stomatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hanyue Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Baoqi Hu
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Luan
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kun Zhu
- Department of Neurology, and The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bo Ma
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhichao Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaoyan Zheng
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
25
|
Kartiganer Z, Rojas G, Riccio M, Tyree A, Noronha K, Wetzel M, Barnett J, McGann J, Garbarino J, Massucci D, Chafi NS, Decker S, McDaniels A, Sabina J, Levchenko D, Perez J, Ng C, Wang K. Improved cell-type identification and comprehensive mapping of regulatory features with spatial epigenomics 96-channel microfluidic platform. GEN BIOTECHNOLOGY 2023; 2:503-514. [PMID: 39380764 PMCID: PMC11460376 DOI: 10.1089/genbio.2023.0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Gene expression is subject to epigenetic regulation and is dependent upon cellular context. Spatial omics tools can provide insight into cellular context; however, development has centered on spatial transcriptomics and proteomics. Deterministic barcoding in tissue for spatial omics sequencing (DBiT-seq) was the first spatial epigenomics platform at the cellular level. Here we present a comparison of spatial epigenomic profiling on both 50-channel and 96-channel platforms. The new 96-channel microfluidics chip design greatly improved precision in cell typing and identification of regulatory elements by spatial-ATAC-seq. Spatial mapping reveals complexity of glial cell and neuronal localization within brain structures as well as cis-regulatory elements controlling cellular function. This technology streamlines spatial analysis of the epigenome and contributes a new layer of spatial omics to uncover the context dependent regulatory mechanisms underpinning development, disease, and normal cellular function.
Collapse
Affiliation(s)
| | | | | | | | - Katelyn Noronha
- Commercial, AtlasXomics, Inc., New Haven, CT
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
| | | | | | - James McGann
- Bioinformatics, AtlasXomics, Inc., New Haven, CT
| | | | | | | | | | | | | | - David Levchenko
- Commercial, AtlasXomics, Inc., New Haven, CT
- Lafayette College, Easton, PA
| | - Jose Perez
- Engineering, AtlasXomics, Inc., New Haven, CT
| | - Colin Ng
- Commercial, AtlasXomics, Inc., New Haven, CT
| | | |
Collapse
|
26
|
Sanz JH, Cox S, Donofrio MT, Ishibashi N, McQuillen P, Peyvandi S, Schlatterer S. [Formula: see text] Trajectories of neurodevelopment and opportunities for intervention across the lifespan in congenital heart disease. Child Neuropsychol 2023; 29:1128-1154. [PMID: 36752083 PMCID: PMC10406974 DOI: 10.1080/09297049.2023.2173162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Children with congenital heart disease (CHD) are at increased risk for neurodevelopmental challenges across the lifespan. These are associated with neurological changes and potential acquired brain injury, which occur across a developmental trajectory and which are influenced by an array of medical, sociodemographic, environmental, and personal factors. These alterations to brain development lead to an array of adverse neurodevelopmental outcomes, which impact a characteristic set of skills over the course of development. The current paper reviews existing knowledge of aberrant brain development and brain injury alongside associated neurodevelopmental challenges across the lifespan. These provide a framework for discussion of emerging and potential interventions to improve neurodevelopmental outcomes at each developmental stage.
Collapse
Affiliation(s)
- Jacqueline H Sanz
- Division of Neuropsychology, Children's National Hospital, Washington, D.C
- Departments of Psychiatry and Behavioral Sciences & Pediatrics at The George Washington University School of Medicine
| | - Stephany Cox
- Department of Pediatrics, Division of Developmental Medicine, Benioff Children's Hospital, University of California, San Francisco, CA
| | - Mary T Donofrio
- Division of Cardiology, Children's National Health System, Washington, D.C
- Department of Pediatrics at The George Washington University School of Medicine
| | - Nobuyuki Ishibashi
- Department of Pediatrics at The George Washington University School of Medicine
- Center for Neuroscience Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington D.C
| | - Patrick McQuillen
- Department of Pediatrics, Division of Developmental Medicine, Benioff Children's Hospital, University of California, San Francisco, CA
| | - Shabnam Peyvandi
- Department of Pediatrics, Division of Developmental Medicine, Benioff Children's Hospital, University of California, San Francisco, CA
| | - Sarah Schlatterer
- Department of Pediatrics at The George Washington University School of Medicine
- Prenatal Pediatrics Institute, Children's National Hospital, Washington, D.C
- Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, D.C
| |
Collapse
|
27
|
Rich MT, Worobey SJ, Mankame S, Pang ZP, Swinford-Jackson SE, Pierce RC. Sex-dependent fear memory impairment in cocaine-sired rat offspring. SCIENCE ADVANCES 2023; 9:eadf6039. [PMID: 37851809 PMCID: PMC10584337 DOI: 10.1126/sciadv.adf6039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 09/14/2023] [Indexed: 10/20/2023]
Abstract
Cocaine self-administration by male rats results in neuronal and behavioral alterations in offspring, including responses to cocaine. Given the high degree of overlap between the brain systems underlying the pathological responses to cocaine and stress, we examined whether sire cocaine taking would influence fear-associated behavioral effects in drug-naïve adult male and female progeny. Sire cocaine exposure had no effect on contextual fear conditioning or its extinction in either male or female offspring. During cued fear conditioning, freezing behavior was enhanced in female, but not male, cocaine-sired progeny. In contrast, male cocaine-sired progeny exhibited enhanced expression of cue-conditioned fear during extinction. Long-term potentiation (LTP) was robust in the basolateral amygdala (BLA), which encodes fear conditioning, of female offspring but was completely absent in male offspring of cocaine-exposed sires. Collectively, these results indicate that cued fear memory is enhanced in the male progeny of cocaine exposed sires, which also have BLA synaptic plasticity deficits.
Collapse
Affiliation(s)
- Matthew T. Rich
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ 08854 USA
| | - Samantha J. Worobey
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ 08854 USA
| | - Sharvari Mankame
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ 08854 USA
| | - Zhiping P. Pang
- Child Health Institute and Department of Neuroscience & Cell Biology, Rutgers University, New Brunswick, NJ 08901, USA
| | | | - R. Christopher Pierce
- Brain Health Institute and Department of Psychiatry, Rutgers University, Piscataway, NJ 08854 USA
| |
Collapse
|
28
|
Ju S, Shin Y, Han S, Kwon J, Choi TG, Kang I, Kim SS. The Gut-Brain Axis in Schizophrenia: The Implications of the Gut Microbiome and SCFA Production. Nutrients 2023; 15:4391. [PMID: 37892465 PMCID: PMC10610543 DOI: 10.3390/nu15204391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Schizophrenia, a severe mental illness affecting about 1% of the population, manifests during young adulthood, leading to abnormal mental function and behavior. Its multifactorial etiology involves genetic factors, experiences of adversity, infection, and gene-environment interactions. Emerging research indicates that maternal infection or stress during pregnancy may also increase schizophrenia risk in offspring. Recent research on the gut-brain axis highlights the gut microbiome's potential influence on central nervous system (CNS) function and mental health, including schizophrenia. The gut microbiota, located in the digestive system, has a significant role to play in human physiology, affecting immune system development, vitamin synthesis, and protection against pathogenic bacteria. Disruptions to the gut microbiota, caused by diet, medication use, environmental pollutants, and stress, may lead to imbalances with far-reaching effects on CNS function and mental health. Of interest are short-chain fatty acids (SCFAs), metabolic byproducts produced by gut microbes during fermentation. SCFAs can cross the blood-brain barrier, influencing CNS activity, including microglia and cytokine modulation. The dysregulation of neurotransmitters produced by gut microbes may contribute to CNS disorders, including schizophrenia. This review explores the potential relationship between SCFAs, the gut microbiome, and schizophrenia. Our aim is to deepen the understanding of the gut-brain axis in schizophrenia and to elucidate its implications for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (Y.S.); (S.H.); (J.K.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
29
|
Abstract
According to the commonly accepted opinion, memory engrams are formed and stored at the level of neural networks due to a change in the strength of synaptic connections between neurons. This hypothesis of synaptic plasticity (HSP), formulated by Donald Hebb in the 1940s, continues to dominate the directions of experimental studies and the interpretations of experimental results in the field. The universal acceptance of the HSP has transformed it from a hypothesis into an incontrovertible theory. In this article, I show that the entire body of experimental and clinical data obtained in studies of long-term memory in mammals and humans is inconsistent with the HSP. Instead, these data suggest that long-term memory is formed and stored at the intracellular level where it is reliably protected from ongoing synaptic activity, including pathological epileptic activity. It seems that the generally accepted HSP became a serious obstacle to understanding the mechanisms of memory and that progress in this field requires rethinking this doctrine and shifting experimental efforts toward exploring the intracellular mechanisms.
Collapse
Affiliation(s)
- Yuri I Arshavsky
- BioCircuits Institute, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
30
|
SHODA A, MURATA M, KIMURA M, HARA Y, YONOICHI S, ISHIDA Y, MANTANI Y, YOKOYAMA T, HIRANO T, IKENAKA Y, HOSHI N. Transgenerational effects of developmental neurotoxicity induced by exposure to a no-observed-adverse-effect level (NOAEL) of neonicotinoid pesticide clothianidin. J Vet Med Sci 2023; 85:1023-1029. [PMID: 37544714 PMCID: PMC10539822 DOI: 10.1292/jvms.23-0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/01/2023] [Indexed: 08/08/2023] Open
Abstract
Neonicotinoid pesticides (NNs) transfer rapidly from mother to offspring, which exhibit neurobehavioral effects. However, no studies have investigated NNs' transgenerational effects. We exposed F0 generation mice (mothers) to a no-observed-adverse-effect level (NOAEL) of clothianidin (CLO) during gestation and lactation, and examined the adult neurobehavioral effects of three generations of offspring (F1, F2, F3). F1 had lower birth weight, decreased locomotor activity, and increased anxiety-like behavior. In F2, body weight was affected, and there was a decreasing trend in locomotor activity and an increasing trend in anxiety-like behavior. In F3, locomotor activity tended to increase. Thus, even when only the mothers were exposed, the effects of CLOs were still observed in F1, F2, and F3 but the effects became smaller.
Collapse
Affiliation(s)
- Asuka SHODA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Midori MURATA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Mako KIMURA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Yukako HARA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Sakura YONOICHI
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Yuya ISHIDA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Youhei MANTANI
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Toshifumi YOKOYAMA
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Tetsushi HIRANO
- Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yoshinori IKENAKA
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Hokkaido, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Nobuhiko HOSHI
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| |
Collapse
|
31
|
Zeid D, Toussaint AB, Dressler CC, Schumacher SP, Do C, Desalvo H, Selamawi D, Bongiovanni AR, Mayberry HL, Carr GV, Wimmer ME. Paternal morphine exposure in rats reduces social play in adolescent male progeny without affecting drug-taking behavior in juvenile males or female offspring. Mol Cell Neurosci 2023; 126:103877. [PMID: 37385516 PMCID: PMC10528482 DOI: 10.1016/j.mcn.2023.103877] [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: 12/15/2022] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 07/01/2023] Open
Abstract
The ongoing opioid addiction crisis necessitates the identification of novel risk factors to improve prevention and treatment of opioid use disorder. Parental opioid exposure has recently emerged as a potential regulator of offspring vulnerability to opioid misuse, in addition to heritable genetic liability. An understudied aspect of this "missing heritability" is the developmental presentation of these cross-generational phenotypes. This is an especially relevant question in the context of inherited addiction-related phenotypes, given the prominent role of developmental processes in the etiology of psychiatric disorders. Paternal morphine self-administration was previously shown to alter the sensitivity to the reinforcing and antinociceptive properties of opioids in the next generation. Here, phenotyping was expanded to include the adolescent period, with a focus on endophenotypes related to opioid use disorders and pain. Paternal morphine exposure did not alter heroin or cocaine self-administration in male and female juvenile progeny. Further, baseline sensory reflexes related to pain were unaltered in morphine-sired adolescent rats of either sex. However, morphine-sired adolescent males exhibited a reduction in social play behavior. Our findings suggest that, in morphine-sired male offspring, paternal opioid exposure does not affect opioid intake during adolescence, suggesting that this phenotype does not emerge until later in life. Altered social behaviors in male morphine-sired adolescents indicate that the changes in drug-taking behavior in adults sired by morphine-exposed sires may be due to more complex factors not yet fully assessed.
Collapse
Affiliation(s)
- Dana Zeid
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Andre B Toussaint
- Zuckerman Mind Brain Behavior Institute, Columbia University, United States of America
| | - Carmen C Dressler
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Samuel P Schumacher
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Chau Do
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Heather Desalvo
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Danait Selamawi
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Angela R Bongiovanni
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Hannah L Mayberry
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America
| | - Gregory V Carr
- Lieber Institute of Brain Development, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, United States of America
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, College of Liberal Arts, Temple University, United States of America.
| |
Collapse
|
32
|
Gundacker A, Cuenca Rico L, Stoehrmann P, Tillmann KE, Weber-Stadlbauer U, Pollak DD. Interaction of the pre- and postnatal environment in the maternal immune activation model. DISCOVER MENTAL HEALTH 2023; 3:15. [PMID: 37622027 PMCID: PMC10444676 DOI: 10.1007/s44192-023-00042-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023]
Abstract
Adverse influences during pregnancy are associated with a range of unfavorable outcomes for the developing offspring. Maternal psychosocial stress, exposure to infections and nutritional imbalances are known risk factors for neurodevelopmental derangements and according psychiatric and neurological manifestations later in offspring life. In this context, the maternal immune activation (MIA) model has been extensively used in preclinical research to study how stimulation of the maternal immune system during gestation derails the tightly coordinated sequence of fetal neurodevelopment. The ensuing consequence of MIA for offspring brain structure and function are majorly manifested in behavioral and cognitive abnormalities, phenotypically presenting during the periods of adolescence and adulthood. These observations have been interpreted within the framework of the "double-hit-hypothesis" suggesting that an elevated risk for neurodevelopmental disorders results from an individual being subjected to two adverse environmental influences at distinct periods of life, jointly leading to the emergence of pathology. The early postnatal period, during which the caregiving parent is the major determinant of the newborn´s environment, constitutes a window of vulnerability to external stimuli. Considering that MIA not only affects the developing fetus, but also impinges on the mother´s brain, which is in a state of heightened malleability during pregnancy, the impact of MIA on maternal brain function and behavior postpartum may importantly contribute to the detrimental consequences for her progeny. Here we review current information on the interaction between the prenatal and postnatal maternal environments in the modulation of offspring development and their relevance for the pathophysiology of the MIA model.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Katharina E. Tillmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse, 17, 1090 Vienna, Austria
| |
Collapse
|
33
|
Martinez ME, Wu Z, Hernandez A. Paternal developmental thyrotoxicosis disrupts neonatal leptin leading to increased adiposity and altered physiology of the melanocortin system. Front Endocrinol (Lausanne) 2023; 14:1210414. [PMID: 37560296 PMCID: PMC10407661 DOI: 10.3389/fendo.2023.1210414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
Background The genetic code does not fully explain individual variability and inheritance of susceptibility to endocrine conditions, suggesting the contribution of epigenetic factors acting across generations. Methods We used a mouse model of developmental thyrotoxicosis (Dio3-/- mouse) to analyze endocrine outcomes in the adult offspring of Dio3-/- males using standard methods for body composition, and baseline and fasting hormonal and gene expression determinations in serum and tissues of relevance to the control of energy balance. Results Compared to controls, adult females with an exposed father (EF females) exhibited higher body weight and fat mass, but not lean mass, a phenotype that was much milder in EF males. After fasting, both EF females and males exhibited a more pronounced decrease in body weight than controls. EF females also showed markedly elevated serum leptin, increased white adipose tissue mRNA expression of leptin and mesoderm-specific transcript but decreased expression of type 2 deiodinase. EF females exhibited decreased serum ghrelin, which showed more pronounced post-fasting changes in EF females than in control females. EF female hypothalami also revealed significant decreases in the expression of pro-opiomelanocortin, agouti-related peptide, neuropeptide Y and melanocortin receptor 4. These markers also showed larger changes in response to fasting in EF females than in control females. Adult EF females showed no abnormalities in serum thyroid hormones, but pituitary expression of thyrotropin-releasing hormone receptor 1 and thyroid gland expression of thyroid-stimulating hormone receptor, thyroid peroxidase and iodotyrosine deiodinase were increased at baseline and showed differential regulation after fasting, with no increase in Trhr1 expression and more pronounced reductions in Tshr, Tpo and Iyd. In EF males, these abnormalities were generally milder. In addition, postnatal day 14 (P14) serum leptin was markedly reduced in EF pups. Discussion A paternal excess of thyroid hormone during development modifies the endocrine programming and energy balance in the offspring in a sexually dimorphic manner, with baseline and dynamic range alterations in the leptin-melanocortin system and thyroid gland, and consequences for adiposity phenotypes. We conclude that thyroid hormone overexposure may have important implications for the non-genetic, inherited etiology of endocrine and metabolic pathologies.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
34
|
Widyarti S, Wibowo S, Sabarudin A, Abhirama I, Sumitro SB. Dysfunctional energy and future perspective of low dose H 2O 2 as protective agent in neurodegenerative disease. Heliyon 2023; 9:e18123. [PMID: 37519743 PMCID: PMC10372669 DOI: 10.1016/j.heliyon.2023.e18123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 05/21/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
The number of people with neurodegenerative disease continues to increase every year. A new perspective is needed to overcome this disease. In this review, researchers collected information about dysfunctional energy in neurodegenerative diseases driven by mitochondria. Mitochondrial dysregulation can cause damage to the neuron system. The increase in the amount and interaction of α-synuclein with SAMM50 and GABARAPL1 in the mitochondria is one of the factors causing neurodegenerative disease. As an energy provider in the body, the existence of harmonization in the regulation of mitochondria, specifically the mitochondrial outer membrane, is important. Low-dose hydrogen peroxide (H2O2) has neuroprotective abilities to overcome the impairment function of mitochondria in neurodegenerative patients. Based on computational simulation of this case, it can be used as a basic concept for the development of the role of H2O2 in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sri Widyarti
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| | - Syahputra Wibowo
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
- Postdoctoral Fellow, Faculty of Biology, Gadjah Mada University, Teknika Selatan Sekip Utara, 55281 Yogyakarta, Indonesia
| | - Akhmad Sabarudin
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| | - Intan Abhirama
- Department of Neurology, Bogor Senior Hospital, Jl.Raya Tajur 16137, West Java, Indonesia
| | - Sutiman Bambang Sumitro
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Malang 65145, East Java, Indonesia
| |
Collapse
|
35
|
Jyothi AK, Thotakura B, Priyadarshini C S, Subramanian M, Rajila HS. Evidence of alterations in the learning and memory in offspring of stress-induced male rats. J Basic Clin Physiol Pharmacol 2023; 34:473-487. [PMID: 34428362 DOI: 10.1515/jbcpp-2020-0183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES There is extensive data pointing to offspring outcomes related to maternal life incidents, but there is less research concerning the association between paternal life events and progeny brain development and behaviour. As male gametogenesis is a continuous process, the incidences happening in life can modify the epigenetic regulation, altering the offspring's development and behaviour. The present study evaluates the effects of paternal stress during different life periods on their offspring's learning ability, memory, morphological and biochemical changes in the prefrontal cortex and hippocampus in the rat model. METHODS Four weeks' old male rats were subjected to five variable stressors at the rate of one per day. Stress received male rats were bred with naive female rats for 1 to 3 nights. The offspring's learning and memory were assessed by the Morris water maze test and automated Y maze. Following behavioural studies, offspring were euthanized to examine global DNA methylation, neurotransmitter levels, namely acetylcholine, glutamate in the hippocampus and frontal cortex. RESULTS The offspring of stress-induced animals exhibited a delay in acquiring learning and defect in memory and altered global DNA methylation in the hippocampus (p=0.000124). There was significant reduction of acetylcholine and glutamate levels in hippocampus (p=0.000018, p=0.00001, respectively) and in prefrontal cortex (p=0.00001, p=0.00001, respectively). HPA axis of offspring was altered considerably (p=0.00001). The histomorphometry of the prefrontal cortex and different hippocampal regions revealed a statistically significant (p<0.05) reduction in neuronal numbers in the offspring of stressed animals compared to that of control. These impacts were markedly high in the offspring of fathers who received stress during both pubertal and adult periods. CONCLUSIONS The findings of this study demonstrate that paternal stress can impact offspring learning and memory.
Collapse
Affiliation(s)
- Ashok Kumar Jyothi
- Department of Anatomy, Basaveshwara Medical College and Hospital, Chitradurga, Karnataka, India
- Department of Anatomy, Tagore Medical College & Hospital, Chennai, Tamil Nadu, India
| | - Balaji Thotakura
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Chennai, Tamil Nadu, India
| | | | - Manickam Subramanian
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | | |
Collapse
|
36
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
37
|
Murashov AK, Pak ES, Mar J, O’Brien K, Fisher-Wellman K, Bhat KM. Paternal Western diet causes transgenerational increase in food consumption in Drosophila with parallel alterations in the offspring brain proteome and microRNAs. FASEB J 2023; 37:e22966. [PMID: 37227156 PMCID: PMC10234493 DOI: 10.1096/fj.202300239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/26/2023]
Abstract
Several lines of evidence indicate that ancestral diet might play an important role in determining offspring's metabolic traits. However, it is not yet clear whether ancestral diet can affect offspring's food choices and feeding behavior. In the current study, taking advantage of Drosophila model system, we demonstrate that paternal Western diet (WD) increases offspring food consumption up to the fourth generation. Paternal WD also induced alterations in F1 offspring brain proteome. Using enrichment analyses of pathways for upregulated and downregulated proteins, we found that upregulated proteins had significant enrichments in terms related to translation and translation factors, whereas downregulated proteins displayed enrichments in small molecule metabolic processes, TCA cycles, and electron transport chain (ETC). Using MIENTURNET miRNA prediction tool, dme-miR-10-3p was identified as the top conserved miRNA predicted to target proteins regulated by ancestral diet. RNAi-based knockdown of miR-10 in the brain significantly increased food consumption, implicating miR-10 as a potential factor in programming feeding behavior. Together, these findings suggest that ancestral nutrition may influence offspring feeding behavior through alterations in miRNAs.
Collapse
Affiliation(s)
- Alexander K. Murashov
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Elena S. Pak
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Jordan Mar
- Department of Molecular Medicine, University of South Florida, Tampa, FL
| | - Kevin O’Brien
- Department of Biostatistics, College of Allied Health Sciences, East Carolina University, Greenville, NC
| | - Kelsey Fisher-Wellman
- Department of Physiology & East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Krishna M. Bhat
- Department of Molecular Medicine, University of South Florida, Tampa, FL
| |
Collapse
|
38
|
Duckworth RA, Chenard KC, Meza L, Beiriz MC. Coping styles vary with species' sociality and life history: A systematic review and meta-regression analysis. Neurosci Biobehav Rev 2023; 151:105241. [PMID: 37216998 DOI: 10.1016/j.neubiorev.2023.105241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/24/2023]
Abstract
Despite a long history of animal studies investigating coping styles, the causal connections between behavior and stress physiology remain unclear. Consistency across taxa in effect sizes would support the idea of a direct causal link maintained by either functional or developmental dependencies. Alternatively, lack of consistency would suggest coping styles are evolutionarily labile. Here, we investigated correlations between personality traits and baseline and stress-induced glucocorticoid levels using a systematic review and meta-analysis. Most personality traits did not consistently vary with either baseline or stress-induced glucocorticoids. Only aggression and sociability showed a consistent negative correlation with baseline glucocorticoids. We found that life history variation affected the relationship between stress-induced glucocorticoid levels and personality traits, especially anxiety and aggression. The relationship between anxiety and baseline glucocorticoids depended on species' sociality with solitary species showing more positive effect sizes. Thus, integration between behavioral and physiological traits depends on species' sociality and life history and suggests high evolutionary lability of coping styles.
Collapse
Affiliation(s)
- Renée A Duckworth
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA.
| | - Kathryn C Chenard
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Lexis Meza
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Maria Carolina Beiriz
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ 85721, USA; Department of Ecology and Natural Resources, Universidade Federal do Ceará, Fortaleza, CE 60440-900, Brazil
| |
Collapse
|
39
|
Cediel-Ulloa A, Lindner S, Rüegg J, Broberg K. Epigenetics of methylmercury. Neurotoxicology 2023; 97:34-46. [PMID: 37164037 DOI: 10.1016/j.neuro.2023.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE OF REVIEW Methylmercury (MeHg) is neurotoxic at high levels and particularly affects the developing brain. One proposed mechanism of MeHg neurotoxicity is alteration of the epigenetic programming. In this review, we summarise the experimental and epidemiological literature on MeHg-associated epigenetic changes. RECENT FINDINGS Experimental and epidemiological studies have identified changes in DNA methylation following in utero exposure to MeHg, and some of the changes appear to be persistent. A few studies have evaluated associations between MeHg-related changes in DNA methylation and neurodevelopmental outcomes. Experimental studies reveal changes in histone modifications after MeHg exposure, but we lack epidemiological studies supporting such changes in humans. Experimental and epidemiological studies have identified microRNA-related changes associated with MeHg; however, more research is needed to conclude if these changes lead to persistent and toxic effects. SUMMARY MeHg appears to interfere with epigenetic processes, potentially leading to persistent changes. However, observed associations of mercury with epigenetic changes are as of yet of unknown relevance to neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Andrea Cediel-Ulloa
- Department of Organism Biology, Uppsala University, Kåbovägen 4, 752 36 Uppsala, Sweden
| | - Sabrina Lindner
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Joëlle Rüegg
- Department of Organism Biology, Uppsala University, Kåbovägen 4, 752 36 Uppsala, Sweden
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
40
|
Lei C, Li N, Chen J, Wang Q. Hypericin Ameliorates Depression-like Behaviors via Neurotrophin Signaling Pathway Mediating m6A Epitranscriptome Modification. Molecules 2023; 28:molecules28093859. [PMID: 37175269 PMCID: PMC10179818 DOI: 10.3390/molecules28093859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Hypericin, one of the major antidepressant constituents of St. John's wort, was shown to exert antidepressant effects by affecting cerebral CYP enzymes, serotonin homeostasis, and neuroinflammatory signaling pathways. However, its exact mechanisms are unknown. Previous clinical studies reported that the mRNA modification N6-methyladenosine (m6A) interferes with the neurobiological mechanism in depressed patients, and it was also found that the antidepressant efficacy of tricyclic antidepressants (TCAs) is related to m6A modifications. Therefore, we hypothesize that the antidepressant effect of hypericin may relate to the m6A modification of epitranscriptomic regulation. We constructed a UCMS mouse depression model and found that hypericin ameliorated depressive-like behavior in UCMS mice. Molecular pharmacology experiments showed that hypericin treatment upregulated the expression of m6A-modifying enzymes METTL3 and WTAP in the hippocampi of UCMS mice. Next, we performed MeRIP-seq and RNA-seq to study m6A modifications and changes in mRNA expression on a genome-wide scale. The genome-wide m6A assay and MeRIP-qPCR results revealed that the m6A modifications of Akt3, Ntrk2, Braf, and Kidins220 mRNA were significantly altered in the hippocampi of UCMS mice after stress stimulation and were reversed by hypericin treatment. Transcriptome assays and qPCR results showed that the Camk4 and Arhgdig genes might be related to the antidepressant efficacy of hypericin. Further gene enrichment results showed that the differential genes were mainly involved in neurotrophic factor signaling pathways. In conclusion, our results show that hypericin upregulates m6A methyltransferase METTL3 and WTAP in the hippocampi of UCMS mice and stabilizes m6A modifications to exert antidepressant effects via the neurotrophin signaling pathway. This suggests that METTL3 and WTAP-mediated changes in m6A modifications may be a potential mechanism for the pathogenesis of depression and the efficacy of antidepressants, and that the neurotrophin signaling pathway plays a key role in this process.
Collapse
Affiliation(s)
- Chunguang Lei
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jianhua Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qingzhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
41
|
Shrivastava K, Swaminathan T, Barlotta A, Athreya V, Choudhry H, Rossi MA. Maternal overnutrition is associated with altered synaptic input to lateral hypothalamic area. Mol Metab 2023; 71:101702. [PMID: 36898526 PMCID: PMC10025284 DOI: 10.1016/j.molmet.2023.101702] [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: 01/04/2023] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
OBJECTIVE Maternal overnutrition is associated with adverse outcomes in offspring, including increased risk for obesity and diabetes. Here, we aim to test the effects of maternal obesity on lateral hypothalamic feeding circuit function and determine the relationship with body weight regulation. METHODS Using a mouse model of maternal obesity, we assessed how perinatal overnutrition affected food intake and body weight regulation in adult offspring. We then used channelrhodopsin-assisted circuit mapping and electrophysiological recordings to assess the synaptic connectivity within an extended amygdala-lateral hypothalamic pathway. RESULTS We show that maternal overnutrition during gestation and throughout lactation produces offspring that are heavier than controls prior to weaning. When weaned onto chow, the body weights of over-nourished offspring normalize to control levels. However, when presented with highly palatable food as adults, both male and female maternally over-nourished offspring are highly susceptible to diet-induced obesity. This is associated with altered synaptic strength in an extended amygdala-lateral hypothalamic pathway, which is predicted by developmental growth rate. Additionally, lateral hypothalamic neurons receiving synaptic input from the bed nucleus of the stria terminalis have enhanced excitatory input following maternal overnutrition which is predicted by early life growth rate. CONCLUSIONS Together, these results demonstrate one way in which maternal obesity rewires hypothalamic feeding circuits to predispose offspring to metabolic dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | - Mark A Rossi
- Child Health Institute of New Jersey, USA; Department of Psychiatry, Robert Wood Johnson Medical School, USA; Brain Health Institute, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
42
|
Risal S, Li C, Luo Q, Fornes R, Lu H, Eriksson G, Manti M, Ohlsson C, Lindgren E, Crisosto N, Maliqueo M, Echiburú B, Recabarren S, Petermann TS, Benrick A, Brusselaers N, Qiao J, Deng Q, Stener-Victorin E. Transgenerational transmission of reproductive and metabolic dysfunction in the male progeny of polycystic ovary syndrome. Cell Rep Med 2023; 4:101035. [PMID: 37148878 DOI: 10.1016/j.xcrm.2023.101035] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/27/2022] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
The transgenerational maternal effects of polycystic ovary syndrome (PCOS) in female progeny are being revealed. As there is evidence that a male equivalent of PCOS may exists, we ask whether sons born to mothers with PCOS (PCOS-sons) transmit reproductive and metabolic phenotypes to their male progeny. Here, in a register-based cohort and a clinical case-control study, we find that PCOS-sons are more often obese and dyslipidemic. Our prenatal androgenized PCOS-like mouse model with or without diet-induced obesity confirmed that reproductive and metabolic dysfunctions in first-generation (F1) male offspring are passed down to F3. Sequencing of F1-F3 sperm reveals distinct differentially expressed (DE) small non-coding RNAs (sncRNAs) across generations in each lineage. Notably, common targets between transgenerational DEsncRNAs in mouse sperm and in PCOS-sons serum indicate similar effects of maternal hyperandrogenism, strengthening the translational relevance and highlighting a previously underappreciated risk of transmission of reproductive and metabolic dysfunction via the male germline.
Collapse
Affiliation(s)
- Sanjiv Risal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Congru Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Qing Luo
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Romina Fornes
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Haojiang Lu
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Gustaw Eriksson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Manti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eva Lindgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nicolas Crisosto
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile, Carlos Schachtebeck 299, Interior Quinta Normal, Santiago, Chile; Endocrinology Unit, Department of Medicine, Clínica Alemana de Santiago, Faculty of Medicine, Clinica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Manuel Maliqueo
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile, Carlos Schachtebeck 299, Interior Quinta Normal, Santiago, Chile
| | - Barbara Echiburú
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile, Carlos Schachtebeck 299, Interior Quinta Normal, Santiago, Chile
| | - Sergio Recabarren
- Laboratory of Animal Physiology and Endocrinology, Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - Teresa Sir Petermann
- Endocrinology and Metabolism Laboratory, West Division, School of Medicine, University of Chile, Carlos Schachtebeck 299, Interior Quinta Normal, Santiago, Chile
| | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Nele Brusselaers
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Global Health Institute, Antwerp University, Antwerp, Belgium
| | - Jie Qiao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | | |
Collapse
|
43
|
van Oers K, van den Heuvel K, Sepers B. The Epigenetics of Animal Personality. Neurosci Biobehav Rev 2023; 150:105194. [PMID: 37094740 DOI: 10.1016/j.neubiorev.2023.105194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/26/2023]
Abstract
Animal personality, consistent individual differences in behaviour, is an important concept for understanding how individuals vary in how they cope with environmental challenges. In order to understand the evolutionary significance of animal personality, it is crucial to understand the underlying regulatory mechanisms. Epigenetic marks such as DNA methylation are hypothesised to play a major role in explaining variation in phenotypic changes in response to environmental alterations. Several characteristics of DNA methylation also align well with the concept of animal personality. In this review paper, we summarise the current literature on the role that molecular epigenetic mechanisms may have in explaining personality variation. We elaborate on the potential for epigenetic mechanisms to explain behavioural variation, behavioural development and temporal consistency in behaviour. We then suggest future routes for this emerging field and point to potential pitfalls that may be encountered. We conclude that a more inclusive approach is needed for studying the epigenetics of animal personality and that epigenetic mechanisms cannot be studied without considering the genetic background.
Collapse
Affiliation(s)
- Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands.
| | - Krista van den Heuvel
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| | - Bernice Sepers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Wageningen, The Netherlands; Behavioural Ecology Group, Wageningen University & Research (WUR), Wageningen, the Netherlands
| |
Collapse
|
44
|
Harris JC, Trigg NA, Goshu B, Yokoyama Y, Dohnalová L, White EK, Harman A, Thaiss CA, Grice EA, Conine CC, Kambayashi T. The microbiota and immune system non-genetically affect offspring phenotypes transgenerationally. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535940. [PMID: 37066207 PMCID: PMC10104111 DOI: 10.1101/2023.04.06.535940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The host-microbiota relationship has evolved to shape mammalian processes, including immunity, metabolism, and development 1-3 . Host phenotypes change in direct response to microbial exposures by the individual. Here we show that the microbiota induces phenotypic change not only in the individual but also in their succeeding generations of progeny. We found that germ-free mice exhibit a robust sebum secretion defect and transcriptional changes in various organs, persisting across multiple generations despite microbial colonization and breeding with conventional mice. Host-microbe interactions could be involved in this process, since T cell-deficient mice, which display defective sebum secretion 4 , also transgenerationally transmit their phenotype to progeny. These phenotypes are inherited by progeny conceived during in vitro fertilization using germ-free sperm and eggs, demonstrating that epigenetic information in the gametes is required for phenotypic transmission. Accordingly, small non-coding RNAs that can regulate embryonic gene expression 5 were strikingly and similarly altered in gametes of germ-free and T cell-deficient mice. Thus, we have uncovered a novel mechanism whereby the microbiota and immune system induce phenotypic changes in successive generations of offspring. This epigenetic form of inheritance could be advantageous for host adaptation to environmental perturbation, where phenotypic diversity can be introduced more rapidly than by genetic mutation.
Collapse
|
45
|
Bucci M, Iozzo P, Merisaari H, Huovinen V, Lipponen H, Räikkönen K, Parkkola R, Salonen M, Sandboge S, Eriksson JG, Nummenmaa L, Nuutila P. Resistance Training Increases White Matter Density in Frail Elderly Women. J Clin Med 2023; 12:jcm12072684. [PMID: 37048767 PMCID: PMC10094827 DOI: 10.3390/jcm12072684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/23/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
We aimed to investigate the effects of maternal obesity on brain structure and metabolism in frail women, and their reversibility in response to exercise. We recruited 37 frail elderly women (20 offspring of lean/normal-weight mothers (OLM) and 17 offspring of obese/overweight mothers (OOM)) and nine non-frail controls to undergo magnetic resonance and diffusion tensor imaging (DTI), positron emission tomography with Fluorine-18-fluorodeoxyglucose (PET), and cognitive function tests (CERAD). Frail women were studied before and after a 4-month resistance training, and controls were studied once. White matter (WM) density (voxel-based morphometry) was higher in OLM than in OOM subjects. Exercise increased WM density in both OLM and OOM in the cerebellum in superior parietal regions in OLM and in cuneal and precuneal regions in OOM. OLM gained more WM density than OOM in response to intervention. No significant results were found from the Freesurfer analysis, nor from PET or DTI images. Exercise has an impact on brain morphology and cognition in elderly frail women.
Collapse
Affiliation(s)
- Marco Bucci
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Theme Inflammation and Aging, Karolinska University Hospital, 141 86 Huddinge, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska University, 171 77 Stockholm, Sweden
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - Harri Merisaari
- Department of Radiology, Turku University Hospital, University of Turku, 20014 Turku, Finland
- Turku Brain and Mind Center, University of Turku, 20014 Turku, Finland
| | - Ville Huovinen
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Department of Radiology, Turku University Hospital, University of Turku, 20014 Turku, Finland
| | - Heta Lipponen
- Turku PET Centre, University of Turku, 20520 Turku, Finland
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, 00014 Helsinki, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, University of Turku, 20014 Turku, Finland
| | | | - Samuel Sandboge
- Finnish Institute for Health and Welfare, 00271 Helsinki, Finland
- Psychology/Welfare Sciences, Faculty of Social Sciences, University of Tampere, 33014 Tampere, Finland
| | - Johan Gunnar Eriksson
- Folkhälsan Research Centre, 00250 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki University Hospital, 00290 Helsinki, Finland
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research, Singapore 138632, Singapore
- Department of Obstetrics & Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | | | - Pirjo Nuutila
- Turku PET Centre, University of Turku, 20520 Turku, Finland
- Department of Endocrinology, Turku University Hospital, 20520 Turku, Finland
| |
Collapse
|
46
|
Humphries A, Simcox K, Howell B. A review of the literature: How does prenatal opioid exposure impact placental health and fetal brain development? Dev Psychobiol 2023; 65:e22378. [PMID: 36946682 DOI: 10.1002/dev.22378] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/07/2022] [Accepted: 01/05/2023] [Indexed: 03/06/2023]
Abstract
In recent years, there has been a sixfold increase in the number of pregnant people with opioid use disorder (OUD). Rates of neonatal opioid withdrawal syndrome (NOWS), previously known as neonatal abstinence syndrome (NAS), have significantly increased in virtually every state and demographic group (Healthcare Cost Utilization Project, HCUP, 2010). NOWS is a condition resulting from chronic exposure to either therapeutic opioid use (e.g., medication for OUD, chronic pain conditions) or nonprescribed opioid use. To date, there is no known prenatal treatment to help decrease the risk of infants developing NOWS and subsequent neurodevelopmental outcomes. Given the increasing support for how placental signaling, or placental programming, may play a role in downstream pathology, prospective research investigating how the placenta is affected by chronic opioid exposure morphologically, histologically, and at the cellular level may open up potential treatment opportunities in this field. In this review, we discuss literature exploring the physiological roles of nitric oxide and dopamine not only in the vascular development of the placenta, but also in fetal cerebral blood flow, neurogenesis, neuronal differentiation, and neuronal activity. We also discuss histological preclinical studies that suggest chronic opioid exposure to induce some combination of placental dysfunction and hypoxia in a manner similar to other well-known placental pathologies, as denoted by the compensatory neovascularization and increased utilization of the placenta's supply of trophoblast cells, which play an essential role in placental angiogenesis. Overall, we found that the current literature, while limited, suggests chronic opioid exposure negatively impacts placental function and fetal brain development on a cellular and histopathological level. We conclude that it is worthwhile to consider the placenta as a therapeutic target with the ultimate goal of decreasing the incidence of NOWS and the long-term impacts of prenatal opioid exposure.
Collapse
Affiliation(s)
- Audrey Humphries
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Kim Simcox
- Obstetrics and Gynecology, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
- Department of Obstetrics and Gynecology, Carilion Clinic, Roanoke, Virginia, USA
| | - Brittany Howell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- Department of Human Development and Family Science, Virginia Tech, Roanoke, Virginia, USA
- Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| |
Collapse
|
47
|
Bregalda A, Carducci C, Viscomi MT, Pierigè F, Biagiotti S, Menotta M, Biancucci F, Pascucci T, Leuzzi V, Magnani M, Rossi L. Myelin basic protein recovery during PKU mice lifespan and the potential role of microRNAs on its regulation. Neurobiol Dis 2023; 180:106093. [PMID: 36948260 DOI: 10.1016/j.nbd.2023.106093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023] Open
Abstract
Untreated phenylketonuria (PKU) patients and PKU animal models show hypomyelination in the central nervous system and white matter damages, which are accompanied by myelin basic protein (MBP) impairment. Despite many assumptions, the primary explanation of the mentioned cerebral outcomes remains elusive. In this study, MBP protein and mRNA expression on brains of wild type (WT) and phenylketonuric (ENU2) mice were analyzed throughout mice lifespan (14-60-180-270-360-540 post-natal days, PND). The results confirmed the low MBP expression at first PND times, while revealed an unprecedented progressive MBP protein expression recovery in aged ENU2 mice. Unexpectedly, unaltered MBP mRNA expression between WT and ENU2 was always observed. Additionally, for the same time intervals, a significant decrease of the phenylalanine concentration in the peripheral blood and brain of ENU2 mice was detected, to date, for the first time. In this scenario, a translational hindrance of MBP during initial and late cerebral development in ENU2 mice was hypothesized, leading to the execution of a microRNA microarray analysis on 60 PND brains, which was followed by a proteomic assay on 60 and 360 PND brains in order to validate in silico miRNA-target predictions. Taken together, miR-218 - 1-3p, miR - 1231-3p and miR-217-5p were considered as the most impactful microRNAs, since a downregulation of their potential targets (MAG, CNTNAP2 and ANLN, respectively) can indirectly lead to a low MBP protein expression. These miRNAs, in addition, follow an opposite expression trend compared to MBP during adulthood, and their target proteins revealed a complete normalization in aged ENU2 mice. In conclusion, these results provide a new perspective on the PKU pathophysiology understanding and on a possible treatment, emphasizing the potential modulating role of differentially expressed microRNAs in MBP expression on PKU brains during PKU mouse lifespan.
Collapse
Affiliation(s)
- Alessandro Bregalda
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy.
| | - Claudia Carducci
- Department of Experimental Medicine, Sapienza University, viale del Policlinico 155, 00161 Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Sciences and Public Health, Sect. Histology and Embryology, Università Cattolica del S. Cuore, Largo F. Vito 1, 00168 Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli", IRCCS, 00168 Rome, Italy
| | - Francesca Pierigè
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy
| | - Sara Biagiotti
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy
| | - Federica Biancucci
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy
| | - Tiziana Pascucci
- Fondazione Santa Lucia IRCCS, via Ardeatina 306, 00142 Rome, Italy; Department of Psychology and Centro "Daniel Bovet", Sapienza University, via dei Marsi 78, 00185 Rome, Italy
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Sapienza University, via dei Sabelli 108, 00185 Rome, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy; EryDel SpA, via Antonio Meucci 3, 20091 Bresso, Milan, Italy
| | - Luigia Rossi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", via Saffi 2, 61029 Urbino, PU, Italy; EryDel SpA, via Antonio Meucci 3, 20091 Bresso, Milan, Italy
| |
Collapse
|
48
|
Gorgij E, Fanaei H, Yaghmaei P, Shahraki MR, Mirahmadi H. Maternal treadmill exercise ameliorates impairment of neurological outcome, caspase-1 and NLRP3 gene expression alteration in neonatal hypoxia-ischemia rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:228-234. [PMID: 36742136 PMCID: PMC9869874 DOI: 10.22038/ijbms.2022.66183.14544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 11/12/2022] [Indexed: 02/07/2023]
Abstract
Objectives Neonatal hypoxia-ischemia (HI) is one of the most important causes of neurological disorders in children. Various studies suggest that maternal exercise during pregnancy has a beneficial impact on the health status of offspring infants. In this study, the effect of maternal treadmill exercise during pregnancy on neurological and molecular changes induced by HI in newborn rats was investigated. Materials and Methods In this experiment, 24 pregnant female rats were divided into two groups; the first group was subjected to treadmill exercise for six weeks. The treadmill exercise program was initiated by running for 17 min at 5-10 m/min at 0 inclination in the first week, followed by running for 21 min at 5-25 m/min at 5° inclination in the second week, running for 25 min at 5-30 m/min at 10° inclination in the third and fourth weeks, running for 25 min at 5-15 m/min at 10° inclination in the fifth and sixth weeks. The second group was left untreated and did not perform the exercise. Newborn rats were assigned to four groups; (1) control, (2) control+exercise, (3) HI, and (4) HI+exercise. HI was developed in the offspring on the 8th postnatal day. One week following the induction of HI, the Garcia test was carried out. The histological morphology of neonates was assessed, and the expression levels of caspase-1 and NLRP3 were evaluated. Results The data showed that maternal exercise during pregnancy significantly improved neural cell death (P<0.001) and the Garcia score (P<0.05), while it attenuated the expression levels of caspase-1 (P<0.001) and NLRP3 (P<0.05) genes in newborn rats induced by HI. Conclusion These results demonstrated that maternal treadmill exercise during pregnancy could reverse the neurological deficits, as well as the expression levels of caspase-1 and NLRP3 genes, which occur in neonatal hypoxia-ischemia.
Collapse
Affiliation(s)
- Elahe Gorgij
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Fanaei
- Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran, Department of Physiology, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Parichehr Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran,Corresponding author: Parichehr Yaghmaei. Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | | | - Hadi Mirahmadi
- Infectious Diseases and Tropical Medicine Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
49
|
Peyvandi S, Rollins C. Fetal Brain Development in Congenital Heart Disease. Can J Cardiol 2023; 39:115-122. [PMID: 36174913 PMCID: PMC9905309 DOI: 10.1016/j.cjca.2022.09.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/08/2022] [Accepted: 09/18/2022] [Indexed: 02/07/2023] Open
Abstract
Neurodevelopmental impairments are the most common extracardiac morbidities among patients with complex congenital heart disease (CHD) across the lifespan. Robust clinical research in this area has revealed several cardiac, medical, and social factors that can contribute to neurodevelopmental outcome in the context of CHD. Studies using brain magnetic resonance imaging (MRI) have been instrumental in identifying quantitative and qualitative difference in brain structure and maturation in this patient population. Full-term newborns with complex CHD are known to have abnormal microstructural and metabolic brain development with patterns similar to those seen in premature infants at approximately 34 to 36 weeks' gestation. With the advent of fetal brain MRI, these brain abnormalities are now documented as they begin in utero, as early as the third trimester. Importantly, disturbed brain development in utero is now known to be independently associated with neurodevelopmental outcome in early childhood, making the prenatal period an important timeframe for potential interventions. Advances in fetal brain MRI provide a robust imaging tool to use in future neuroprotective clinical trials. The causes of abnormal fetal brain development are multifactorial and include cardiovascular physiology, genetic abnormalities, placental impairment, and other environmental and social factors. This review provides an overview of current knowledge of brain development in the context of CHD, common prenatal imaging tools to evaluate the developing fetal brain in CHD, and known risk factors contributing to brain immaturity.
Collapse
Affiliation(s)
- Shabnam Peyvandi
- University of California San Francisco Benioff Children's Hospital, San Francisco, California, USA.
| | - Caitlin Rollins
- Boston Children's Hospital and Harvard University Departments of Neurology, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Barbero G, de Sousa Serro MG, Perez Lujan C, Vitullo AD, González CR, González B. Transcriptome profiling of histone writers/erasers enzymes across spermatogenesis, mature sperm and pre-cleavage embryo: Implications in paternal epigenome transitions and inheritance mechanisms. Front Cell Dev Biol 2023; 11:1086573. [PMID: 36776561 PMCID: PMC9911891 DOI: 10.3389/fcell.2023.1086573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/04/2023] [Indexed: 01/28/2023] Open
Abstract
Accumulating evidence points out that sperm carry epigenetic instructions to embryo in the form of retained histones marks and RNA cargo that can transmit metabolic and behavioral traits to offspring. However, the mechanisms behind epigenetic inheritance of paternal environment are still poorly understood. Here, we curated male germ cells RNA-seq data and analyzed the expression profile of all known histone lysine writers and erasers enzymes across spermatogenesis, unraveling the developmental windows at which they are upregulated, and the specific activity related to canonical and non-canonical histone marks deposition and removal. We also characterized the epigenetic enzymes signature in the mature sperm RNA cargo, showing most of them positive translation at pre-cleavage zygote, suggesting that paternally-derived enzymes mRNA cooperate with maternal factors to embryo chromatin assembly. Our study shows several histone modifying enzymes not described yet in spermatogenesis and even more, important mechanistic aspects behind transgenerational epigenetics. Epigenetic enzymes not only can respond to environmental stressors, but could function as vectors of epigenetic information and participate in chromatin organization during maternal-to-zygote transition.
Collapse
Affiliation(s)
- Gastón Barbero
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Maximiliano G. de Sousa Serro
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires–Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Camila Perez Lujan
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires–Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alfredo D. Vitullo
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Candela R. González
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires–Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina,*Correspondence: Betina González,
| |
Collapse
|