1
|
Camarini R, Marianno P, Hanampa-Maquera M, Oliveira SDS, Câmara NOS. Prenatal Stress and Ethanol Exposure: Microbiota-Induced Immune Dysregulation and Psychiatric Risks. Int J Mol Sci 2024; 25:9776. [PMID: 39337263 PMCID: PMC11431796 DOI: 10.3390/ijms25189776] [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: 06/14/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Changes in maternal gut microbiota due to stress and/or ethanol exposure can have lasting effects on offspring's health, particularly regarding immunity, inflammation response, and susceptibility to psychiatric disorders. The literature search for this review was conducted using PubMed and Scopus, employing keywords and phrases related to maternal stress, ethanol exposure, gut microbiota, microbiome, gut-brain axis, diet, dysbiosis, progesterone, placenta, prenatal development, immunity, inflammation, and depression to identify relevant studies in both preclinical and human research. Only a limited number of reviews were included to support the arguments. The search encompassed studies from the 1990s to the present. This review begins by exploring the role of microbiota in modulating host health and disease. It then examines how disturbances in maternal microbiota can affect the offspring's immune system. The analysis continues by investigating the interplay between stress and dysbiosis, focusing on how prenatal maternal stress influences both maternal and offspring microbiota and its implications for susceptibility to depression. The review also considers the impact of ethanol consumption on gut dysbiosis, with an emphasis on the effects of prenatal ethanol exposure on both maternal and offspring microbiota. Finally, it is suggested that maternal gut microbiota dysbiosis may be significantly exacerbated by the combined effects of stress and ethanol exposure, leading to immune system dysfunction and chronic inflammation, which could increase the risk of depression in the offspring. These interactions underscore the potential for novel mental health interventions that address the gut-brain axis, especially in relation to maternal and offspring health.
Collapse
Affiliation(s)
- Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Priscila Marianno
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Maylin Hanampa-Maquera
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Samuel Dos Santos Oliveira
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
2
|
Reid HMO, Trepanier O, Gross A, Poberezhnyk P, Snowden T, Conway K, Breit KR, Rodriguez C, Thomas JD, Christie BR. Prenatal ethanol and cannabis exposure have sex- and region-specific effects on somatostatin and neuropeptide Y interneurons in the rat hippocampus. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1289-1301. [PMID: 38789401 PMCID: PMC11236510 DOI: 10.1111/acer.15350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Cannabis is increasingly being legalized and socially accepted around the world and is often used with alcohol in social settings. We recently showed that in utero exposure to both substances can alter the density of parvalbumin-expressing interneurons in the hippocampus. Here we investigate the effects of in utero alcohol and cannabis exposure, alone or in combination, on somatostatin- and neuropeptide Y-positive (NPY) interneurons. These are separate classes of interneurons important for network synchrony and inhibition in the hippocampus. METHODS A 2 (Ethanol, Air) × 2 (tetrahydrocannabinol [THC], Vehicle) design was used to expose pregnant Sprague-Dawley rats to either ethanol or air, in addition to either THC or the inhalant vehicle solution, during gestational days 5-20. Immunohistochemistry for somatostatin- and NPY-positive interneurons was performed in 50 μm tissue sections obtained at postnatal day 70. RESULTS Exposure to THC in utero had region-specific and sex-specific effects on the density of somatostatin-positive interneurons in the adult rat hippocampus. A female-specific decrease in NPY interneuron cell density was observed in the CA1 region following THC exposure. Combined exposure to alcohol and THC reduced NPY neurons selectively in the ventral dentate gyrus hippocampal subfield. However, overall, co-exposure to alcohol and cannabis had neither additive nor synergistic effects on interneuron populations in other areas of the hippocampus. CONCLUSIONS These results illustrate how alcohol and cannabis exposure in utero may affect hippocampal function by altering inhibitory processes in a sex-specific manner.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Owen Trepanier
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Allyson Gross
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Polina Poberezhnyk
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Taylor Snowden
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Kate Conway
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Kristen R Breit
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
- Department of Psychology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| | - Cristina Rodriguez
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| |
Collapse
|
3
|
Bakhireva LN, Ma X, Wiesel A, Wohrer FE, DiDomenico J, Jacobson SW, Roberts MH. Dose-response effect of prenatal alcohol exposure on perinatal outcomes. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:703-714. [PMID: 38554141 DOI: 10.1111/acer.15284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/12/2024] [Accepted: 02/05/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND A better understanding of the effects of lower levels of prenatal alcohol exposure (PAE), as a common exposure, is needed. The goal of this study was to examine the effects of mild-moderate PAE and episodic binge drinking on perinatal outcomes. METHODS The data were obtained from three prospective cohorts with a combined sample of 281 participants: 125 with PAE and 156 without PAE. Alcohol-related measures included the Alcohol Use Disorders Identification Test, timeline follow-back questionnaires (covering the periconceptional period, mid-gestation, and late gestation), and biomarkers. Absolute alcohol per day (AAD) and per drinking day (AADD), number of binge episodes, and maximum number of drinks in a 24-h period were estimated. Perinatal outcomes included gestational age and anthropometric measures. Data were analyzed using correlation and multivariable regression analysis. RESULTS Among women with PAE, average alcohol consumption across the periconceptional period and pregnancy was 0.37 oz ± 0.74 AA/day (~5 drinks/week). After adjusting for tobacco co-exposure and sociodemographic characteristics, significant associations between all alcohol measures and gestational age at delivery were observed, including cumulative measures of AAD (β = -0.58; 95% CI: -0.98; -0.17) and AADD (β = -0.58; 95% CI: -0.90; -0.26) during pregnancy and the periconceptional period. A significant association between the maximum number of drinks in a 24-h period and birth length percentile (β = -0.70; 95% CI: -1.36; -0.04) was observed in the final model. PAE was associated with lower birth weight percentile in univariate analyses only. CONCLUSIONS Results of this study demonstrate a negative association between mild-moderate PAE and episodic binge drinking with gestational age at delivery and birth length percentile after controlling for other factors. Robust negative effects of PAE, including in the periconceptional period before pregnancy recognition, on duration of gestation highlight the need for primary prevention efforts aimed at PAE in persons of reproductive age.
Collapse
Affiliation(s)
- Ludmila N Bakhireva
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Xingya Ma
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Alexandria Wiesel
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Fiona E Wohrer
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Jared DiDomenico
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Sandra W Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Melissa H Roberts
- College of Pharmacy Substance Use Research and Education Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
4
|
Dunn BR, Olguin SL, Davies S, Pavlik NG, Brigman JL, Hamilton D, Savage DD, Maxwell JR. Sex-specific alterations in cognitive control following moderate prenatal alcohol exposure and transient systemic hypoxia ischemia in the rat. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:640-652. [PMID: 38302722 PMCID: PMC11015983 DOI: 10.1111/acer.15276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) continues to be a worldwide problem. Affected offspring display impaired neurodevelopment, including difficulties with executive control. Although PAE has also been associated with decreased blood flow to fetuses, the relationship between PAE and altered blood flow is not well understood. METHODS We used preclinical models of PAE, transient systemic hypoxia ischemia (TSHI), and PAE + TSHI combined to assess the effects on neurodevelopmental outcomes using translationally relevant touchscreen operant platform testing. Twenty-eight Long-Evans (Blue Spruce, Strain HsdBlu:LE) dams were randomly assigned to one of four experimental groups: Saccharin Control (Sham), 5% Ethanol (PAE), TSHI, or 5% Ethanol and TSHI (PAE + TSHI). Dams consumed either saccharin or 5% ethanol during gestation. TSHI was induced on Embryonic Day 19 (E19) during an open laparotomy where the uterine arteries were transiently occluded for 1 h. Pups were born normally and, after weaning, were separated by sex. A total of 80 offspring, 40 males and 40 females, were tested on the 5-Choice Continuous Performance paradigm (5C-CPT). RESULTS Female offspring were significantly impacted by TSHI, but not PAE, with an increase in false alarms and a decrease in hit rates, omissions, accuracy, and correct choice latencies. In contrast, male offspring were mildly affected by PAE, but not TSHI, showing decreases in premature responses and increases in accuracy. No significant interactions between PAE and TSHI were detected on any measure. CONCLUSION Transient systemic hypoxia ischemia impaired performance on the 5C-CPT in females, leading to a bias toward stimulus responsivity regardless of stimulus type. In contrast, TSHI did not affect male offspring, and only slight effects of PAE were seen. Together, these data suggest that TSHI in females may cause alterations in cortical structures that override alterations caused by moderate PAE.
Collapse
Affiliation(s)
- Brooke R. Dunn
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Sarah L. Olguin
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Suzy Davies
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Nathaniel G. Pavlik
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA
| | - Derek Hamilton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA
| | - Daniel D. Savage
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Psychology, University of New Mexico, Albuquerque, NM, USA
| | - Jessie R. Maxwell
- Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, USA
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
5
|
Maxwell JR, DiDomenico J, Roberts MH, Marquez LE, Rai R, Weinberg J, Jacobson SW, Stephen J, Bakhireva LN. Impact of low-level prenatal alcohol exposure and maternal stress on autonomic regulation. Pediatr Res 2024; 95:350-358. [PMID: 37674025 PMCID: PMC11089775 DOI: 10.1038/s41390-023-02799-5] [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: 02/28/2023] [Revised: 06/07/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) impacts the neurodevelopment of the fetus, including the infant's ability to self-regulate. Heart rate variability (HRV), that is, the beat-to-beat variability in heart rate, is a non-invasive measurement that can indicate autonomic nervous system (ANS) function/dysfunction. METHODS The study consisted of a subset of our ENRICH-2 cohort: 80 participants (32 PAE and 48 Controls) who had completed three visits during pregnancy. The participants completed a comprehensive assessment of PAE and other substances throughout pregnancy and assessments for stress, anxiety, and depression in the third trimester. At 24 h of age, infant HRV was assessed in the hospital during the clinically indicated heel lance; 3- to 5-min HRV epochs were obtained during baseline, heel lancing, and recovery episodes. RESULTS Parameters of HRV differed in infants with PAE compared to Controls during the recovery phase of the heel lance (respiratory sinus arrhythmia (RSA) and high-frequency (HF), p < 0.05). Increased maternal stress was also strongly associated with abnormalities in RSA, HF, and low-frequency / high-frequency (LF/HF, p's < 0.05). CONCLUSIONS Alterations in ANS regulation associated with PAE and maternal stress may reflect abnormal development of the hypothalamic-pituitary-adrenal axis and have long term implications for infant responsiveness and self-regulation. IMPACT Previous studies have focused on effects of moderate to heavy prenatal alcohol exposure (PAE) on autonomic dysregulation, but little is known about the effects of lower levels of PAE on infant self-regulation and heart rate variability (HRV). Prenatal stress is another risk factor for autonomic dysregulation. Mild PAE impacts infant self-regulation, which can be assessed using HRV. However, the effect of prenatal stress is stronger than that of mild PAE or other mental health variables on autonomic dysregulation.
Collapse
Affiliation(s)
- Jessie R Maxwell
- Department of Pediatrics, University of New Mexico, Albuquerque, NM, Mexico.
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, Mexico.
| | - Jared DiDomenico
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, Mexico
| | - Melissa H Roberts
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, Mexico
| | - Lidia Enriquez Marquez
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, Mexico
| | - Rajani Rai
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, Mexico
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Sandra W Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julia Stephen
- The Mind Research Network, a Division of Lovelace Biomedical Research Institute, University of New Mexico, Albuquerque, NM, Mexico
| | - Ludmila N Bakhireva
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, Mexico
| |
Collapse
|
6
|
Noor S, Sun MS, Pasmay AA, Pritha AN, Ruffaner-Hanson CD, Nysus MV, Jimenez DC, Murphy M, Savage DD, Valenzuela CF, Milligan ED. Prenatal alcohol exposure promotes NLRP3 inflammasome-dependent immune actions following morphine treatment and paradoxically prolongs nerve injury-induced pathological pain in female mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:2262-2277. [PMID: 38151779 PMCID: PMC10764094 DOI: 10.1111/acer.15214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/23/2023] [Accepted: 10/18/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuroimmune dysregulation from prenatal alcohol exposure (PAE) may contribute to neurological deficits associated with fetal alcohol spectrum disorders (FASD). PAE is a risk factor for developing peripheral immune and spinal glial sensitization and release of the proinflammatory cytokine IL-1β, which lead to neuropathic pain (allodynia) from minor nerve injury. Although morphine acts on μ-opioid receptors, it also activates immune receptors, TLR4, and the NLRP3 inflammasome that induces IL-1β. We hypothesized that PAE induces NLRP3 sensitization by morphine following nerve injury in adult mice. METHODS We used an established moderate PAE paradigm, in which adult PAE and non-PAE control female mice were exposed to a minor sciatic nerve injury, and subsequent allodynia was measured using the von Frey fiber test. In control mice with standard sciatic damage or PAE mice with minor sciatic damage, the effects of the NLRP3 inhibitor, MCC950, were examined during chronic allodynia. Additionally, minor nerve-injured mice were treated with morphine, with or without MCC950. In vitro studies examined the TLR4-NLRP3-dependent proinflammatory response of peripheral macrophages to morphine and/or lipopolysaccharide, with or without MCC950. RESULTS Mice with standard sciatic damage or PAE mice with minor sciatic damage developed robust allodynia. Blocking NLRP3 activation fully reversed allodynia in both control and PAE mice. Morphine paradoxically prolonged allodynia in PAE mice, while control mice with minor nerve injury remained stably non-allodynic. Allodynia resolved sooner in nerve-injured PAE mice without morphine treatment than in morphine-treated mice. MCC950 treatment significantly shortened allodynia in morphine-treated PAE mice. Morphine potentiated IL-1β release from TLR4-activated PAE immune cells, while MCC950 treatment greatly reduced it. CONCLUSIONS In female mice, PAE prolongs allodynia following morphine treatment through NLRP3 activation. TLR4-activated PAE immune cells showed enhanced IL-1β release with morphine via NLRP3 actions. Similar studies are needed to examine the adverse impact of morphine in males with PAE. These results are predictive of adverse responses to opioid pain therapeutics in individuals with FASD.
Collapse
Affiliation(s)
- Shahani Noor
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Melody S Sun
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Andrea A Pasmay
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Ariana N Pritha
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Monique V Nysus
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Diane C Jimenez
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Minerva Murphy
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Daniel D Savage
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - C Fernando Valenzuela
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Erin D Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
7
|
Pinson MR, Bake S, Hurst DA, Samiya NT, Sohrabji F, Miranda RC. Prenatal alcohol alters inflammatory signatures in enteric portal tissues following adult-onset cerebrovascular ischemic stroke. iScience 2023; 26:107920. [PMID: 37810225 PMCID: PMC10550726 DOI: 10.1016/j.isci.2023.107920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Prenatal alcohol exposure (PAE) impairs recovery from cerebrovascular ischemic stroke in adult rodents. Since the gut becomes dysbiotic following stroke, we assessed links between PAE and enteric portal inflammation. Adult control and PAE rat offspring received a unilateral endothelin-1-induced occlusion of the middle cerebral artery. Post-stroke behavioral disabilities and brain cytokines were assessed. Mesenteric adipose and liver transcriptomes were assessed from stroke-exposed and stroke-naive offspring. We identified, in the liver of stroke-naive animals, a moderate correlation between PAE and a gene network for inflammatory necroptosis. PAE inhibited the acute-phase brain inflammatory cytokine response to stroke. Post-stroke neurological function was correlated with an adipose gene network associated with B-lymphocyte differentiation and nuclear factor κB (NF-κB) signaling and with a liver pro-inflammatory gene network. Collectively, PAE inhibits brain inflammation but results in an inflammatory signature in enteric portal tissues after stroke, suggesting that PAE persistently and adversely impacts the gut-brain axis following adult-onset disease.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - David A Hurst
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Nadia T Samiya
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| |
Collapse
|
8
|
Gui L, Luo X, Zhou L, Wei Q, Gu J. Peripheral CD4 + /CD8 + T cell composition distinct from healthy individuals is shared by ankylosing spondylitis and rheumatoid arthritis. Int J Rheum Dis 2023; 26:2014-2023. [PMID: 37635355 DOI: 10.1111/1756-185x.14860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/04/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) and rheumatoid arthritis (RA) are chronic inflammatory joint diseases, linking to the alterations of immune cells. We attempted to assess whether the alterations in the composition of CD4+ /CD8+ T cells are different between AS and RA and identify the characteristic cells between male and female patients. METHODS The proportions of CD3+ or double positive T cells, 6 CD4+ T subsets and 9 CD8+ T cell subsets were detected by flow cytometry and compared in 30 healthy individuals, 42 AS patients and 45 RA patients. The differentially altered cells were individually analyzed for associations with disease activity parameters. In addition, their proportions were compared between different genders in the 3 groups. RESULTS The proportions of CD4+ T cells, naive CD4+ T cells and central memory CD4+ T cells were lower in AS patients (P = 0.001, P = 0.002 and P = 0.007, respectively) and RA patients (P = 0.032, P < 0.001 and P = 0.016, respectively), but the proportion of effector memory ones was higher when compared with healthy populations (both P < 0.001), as were the decrease of naive/central memory CD8+ T cells in AS (P = 0.003 and P = 0.016, respectively) and RA (P < 0.001 and P = 0.006, respectively), and the increased tendency of terminally differentiated CD8+ T cells. However, these above-mentioned cells, regulatory T (Treg) cells and CD8+ T cells with different CD127 expressions between AS and RA were similar in proportion. Furthermore, naive CD4+ T cells were positively associated with C-reactive protein (CRP) in AS, whereas CD4+ T cells and terminally differentiated CD8+ T of RA patients were associated with CRP in RA. The gender-related alterations predominantly displayed the overexpressions of Treg cells and naive CD8+ T cells in female patients with AS and RA, respectively. CONCLUSIONS AS patients and RA patients have some similar peripheral CD4+ /CD8+ T cell subsets but are distinct from healthy individuals, which may contribute to disease severity. Females are respectively characterized by the up-regulation of Treg cells and naive CD8+ T cells in AS patients and RA patients. The study offers an in-depth understanding of the role of T cell subsets in the similarities of the disorders and helps us to monitor disease changes and may offer a theoretical basis of developing novel therapies against common targets.
Collapse
Affiliation(s)
- Lian Gui
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiqing Luo
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liuzhong Zhou
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiujing Wei
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Bake S, Rouzer SK, Mavuri S, Miranda RC, Mahnke AH. The interaction of genetic sex and prenatal alcohol exposure on health across the lifespan. Front Neuroendocrinol 2023; 71:101103. [PMID: 37802472 PMCID: PMC10922031 DOI: 10.1016/j.yfrne.2023.101103] [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: 04/09/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Prenatal alcohol exposure (PAE) can reprogram the development of cells and tissues, resulting in a spectrum of physical and neurobehavioral teratology. PAE immediately impacts fetal growth, but its effects carry forward post-parturition, into adolescence and adulthood, and can result in a cluster of disabilities, collectively termed Fetal Alcohol Spectrum Disorders. Emerging preclinical and clinical research investigating neurological and behavioral outcomes in exposed offspring point to genetic sex as an important modifier of the effects of PAE. In this review, we discuss the literature on sex differences following PAE, with studies spanning the fetal period through adulthood, and highlight gaps in research where sex differences are likely, but currently under-investigated. Understanding how sex and PAE interact to affect offspring health outcomes across the lifespan is critical for identifying the full complement of PAE-associated secondary conditions, and for refining targeted interventions to improve the quality of life for individuals with PAE.
Collapse
Affiliation(s)
- Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Siara K Rouzer
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Shruti Mavuri
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Medical Research and Education Building I, 8447 Riverside Parkway, Bryan, TX 77807-3620, United States.
| |
Collapse
|
10
|
Chaudoin TR, Bonasera SJ, Dunaevsky A, Padmashri R. Exploring behavioral phenotypes in a mouse model of fetal alcohol spectrum disorders. Dev Neurobiol 2023; 83:184-204. [PMID: 37433012 PMCID: PMC10546278 DOI: 10.1002/dneu.22922] [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: 12/12/2022] [Revised: 04/18/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Fetal alcohol spectrum disorders are one of the leading causes of developmental abnormalities worldwide. Maternal consumption of alcohol during pregnancy leads to a diverse range of cognitive and neurobehavioral deficits. Although moderate-to-heavy levels of prenatal alcohol exposure (PAE) have been associated with adverse offspring outcomes, there is limited data on the consequences of chronic low-level PAE. Here, we use a model of maternal voluntary alcohol consumption throughout gestation in a mouse model to investigate the effects of PAE on behavioral phenotypes during late adolescence and early adulthood in male and female offspring. Body composition was measured by dual-energy X-ray absorptiometry. Baseline behaviors, including feeding, drinking, and movement, were examined by performing home cage monitoring studies. The impact of PAE on motor function, motor skill learning, hyperactivity, acoustic reactivity, and sensorimotor gating was investigated by performing a battery of behavioral tests. PAE was found to be associated with altered body composition. No differences in overall movement, food, or water consumption were observed between control and PAE mice. Although PAE offspring of both sexes exhibited deficits in motor skill learning, no differences were observed in basic motor skills such as grip strength and motor coordination. PAE females exhibited a hyperactive phenotype in a novel environment. PAE mice exhibited increased reactivity to acoustic stimuli, and PAE females showed disrupted short-term habituation. Sensorimotor gating was not altered in PAE mice. Collectively, our data show that chronic low-level exposure to alcohol in utero results in behavioral impairments.
Collapse
Affiliation(s)
- Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
11
|
Upreti D, Rouzer SK, Bowring A, Labbe E, Kumar R, Miranda RC, Mahnke AH. Microbiota and nutrition as risk and resiliency factors following prenatal alcohol exposure. Front Neurosci 2023; 17:1182635. [PMID: 37397440 PMCID: PMC10308314 DOI: 10.3389/fnins.2023.1182635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Alcohol exposure in adulthood can result in inflammation, malnutrition, and altered gastroenteric microbiota, which may disrupt efficient nutrient extraction. Clinical and preclinical studies have documented convincingly that prenatal alcohol exposure (PAE) also results in persistent inflammation and nutrition deficiencies, though research on the impact of PAE on the enteric microbiota is in its infancy. Importantly, other neurodevelopmental disorders, including autism spectrum and attention deficit/hyperactivity disorders, have been linked to gut microbiota dysbiosis. The combined evidence from alcohol exposure in adulthood and from other neurodevelopmental disorders supports the hypothesis that gut microbiota dysbiosis is likely an etiological feature that contributes to negative developmental, including neurodevelopmental, consequences of PAE and results in fetal alcohol spectrum disorders. Here, we highlight published data that support a role for gut microbiota in healthy development and explore the implication of these studies for the role of altered microbiota in the lifelong health consequences of PAE.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Amanda H. Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, United States
| |
Collapse
|
12
|
Lei A, Breit KR, Thomas JD. Prenatal alcohol and tetrahydrocannabinol exposure: Effects on spatial and working memory. Front Neurosci 2023; 17:1192786. [PMID: 37383100 PMCID: PMC10293645 DOI: 10.3389/fnins.2023.1192786] [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: 03/23/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction Alcohol and cannabis are widely used recreational drugs that can negatively impact fetal development, leading to cognitive impairments. However, these drugs may be used simultaneously and the effects of combined exposure during the prenatal period are not well understood. Thus, this study used an animal model to investigate the effects of prenatal exposure to ethanol (EtOH), Δ-9-tetrahydrocannabinol (THC), or the combination on spatial and working memory. Methods Pregnant Sprague-Dawley rats were exposed to vaporized ethanol (EtOH; 68 ml/h), THC (100 mg/ml), the combination, or vehicle control during gestational days 5-20. Adolescent male and female offspring were evaluated using the Morris water maze task to assess spatial and working memory. Results Prenatal THC exposure impaired spatial learning and memory in female offspring, whereas prenatal EtOH exposure impaired working memory. The combination of THC and EtOH did not exacerbate the effects of either EtOH or THC, although subjects exposed to the combination were less thigmotaxic, which might represent an increase in risk-taking behavior. Discussion Our results highlight the differential effects of prenatal exposure to THC and EtOH on cognitive and emotional development, with substance- and sex-specific patterns. These findings highlight the potential harm of THC and EtOH on fetal development and support public health policies aimed at reducing cannabis and alcohol use during pregnancy.
Collapse
Affiliation(s)
- Annie Lei
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA, United States
| | - Kristen R. Breit
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA, United States
- Department of Psychology, West Chester University of Pennsylvania, West Chester, PA, United States
| | - Jennifer D. Thomas
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA, United States
| |
Collapse
|
13
|
Noor S, Pritha AN, Pasmay AA, Sanchez JE, Sanchez JJ, Fernandez-Oropeza AK, Sun MS, Dell’Orco M, Davies S, Savage DD, Mellios N, Milligan ED. Prenatal alcohol exposure dysregulates spinal and circulating immune cell circular RNA expression in adult female rats with chronic sciatic neuropathy. Front Neurosci 2023; 17:1180308. [PMID: 37360167 PMCID: PMC10288115 DOI: 10.3389/fnins.2023.1180308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Alcohol consumption during pregnancy is associated with Fetal Alcohol Spectrum Disorders (FASD) that results in a continuum of central nervous system (CNS) deficits. Emerging evidence from both preclinical and clinical studies indicate that the biological vulnerability to chronic CNS disease in FASD populations is driven by aberrant neuroimmune actions. Our prior studies suggest that, following minor nerve injury, prenatal alcohol exposure (PAE) is a risk factor for developing adult-onset chronic pathological touch sensitivity or allodynia. Allodynia in PAE rats occurs concurrently with heightened proinflammatory peripheral and spinal glial-immune activation. However, minor nerve-injured control rats remain non-allodynic, and corresponding proinflammatory factors are unaltered. A comprehensive molecular understanding of the mechanism(s) that underlie PAE-induced proinflammatory bias during adulthood remains elusive. Non-coding circular RNAs (circRNAs) are emerging as novel modulators of gene expression. Here, we hypothesized that PAE induces dysregulation of circRNAs that are linked to immune function under basal and nerve-injured conditions during adulthood. Utilizing a microarray platform, we carried out the first systematic profiling of circRNAs in adult PAE rats, prior to and after minor nerve injury. The results demonstrate a unique circRNA profile in adult PAE rats without injury; 18 circRNAs in blood and 32 spinal circRNAs were differentially regulated. Following minor nerve injury, more than 100 differentially regulated spinal circRNAs were observed in allodynic PAE rats. Bioinformatic analysis identified that the parental genes of these circRNAs are linked to the NF-κB complex, a central transcription factor for pain-relevant proinflammatory cytokines. Quantitative real-time PCR was employed to measure levels of selected circRNAs and linear mRNA isoforms. We have validated that circVopp1 was significantly downregulated in blood leukocytes in PAE rats, concurrent with downregulation of Vopp1 mRNA levels. Spinal circVopp1 levels were upregulated in PAE rats, regardless of nerve injury. Additionally, PAE downregulated levels of circItch and circRps6ka3, which are linked to immune regulation. These results demonstrate that PAE exerts long-lasting dysregulation of circRNA expression in blood leukocytes and the spinal cord. Moreover, the spinal circRNA expression profile following peripheral nerve injury is differentially modulated by PAE, potentially contributing to PAE-induced neuroimmune dysregulation.
Collapse
|
14
|
Jiao Y, Zhao Z, Li X, Li L, Xiao D, Wan S, Wu T, Li T, Li P, Zhao R. Salidroside ameliorates memory impairment following long-term ethanol intake in rats by modulating the altered intestinal microbiota content and hippocampal gene expression. Front Microbiol 2023; 14:1172936. [PMID: 37362918 PMCID: PMC10288325 DOI: 10.3389/fmicb.2023.1172936] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Background Salidroside (Sal), the main component of a famous herb Rhodiola rosea L, enhances memory performance and reduces fatigue. Therefore, this study assessed the effect of Sal on memory impairment induced by a long-term intake of ethanol (EtOH) in rats and investigated its relevant mechanisms using gut microbiota metagenomic analysis and hippocampal transcriptomic analysis. Methods Eighteen male SD rats were divided into the normal control group (CON group), EtOH model group (Model group), and Sal treatment group (Sal group). The rats in the Model and Sal groups intragastrically (i.g.) received 2 g/kg EtOH for 30 consecutive days, whereas the CON group was given an equal volume of distilled water. Meanwhile, the rats in the Sal group were administered i.g. 30 mg/kg Sal 60 min after EtOH intake. All rats were tested in the eight-arm maze for their memory function every 3 days. On the 30th day, metagenomic analyses of gut microbiota and transcriptomic analyses of the hippocampus were performed. Results Compared with the Model group, Sal treatment reduced the total time to complete the eight-arm maze task, decreased the number of arm entries, and abated the working memory error that was significant from the 9th day. Additionally, Sal intervention improved the gut microbiota composition, such as the increased abundance of Actinobacteria and Bifidobacterium, which was related to the metabolism of amino acids and terpenoid carbohydrate, endocrine function, and signal transduction by neurotransmitters. In the hippocampus, the EtOH intake differentially expressed 68 genes (54 genes increased, whereas 14 genes decreased), compared with the CON group, whereas Sal intervention affected these changes: 15 genes increased whereas 11 genes decreased. And, enrichment analyses revealed these genes were related to the structural components of the ribosome, mRNA splicing process, protein translation, mitochondria function, and immunological reaction. Finally, a correlation analysis found the memory impairment was positively correlated with the abnormal upregulation of Tomm7 but negatively correlated with decreased abundance of gut Alistipes_indistinctus, Lactobacillus_taiwanensis, Lactobacillus_paragasseri, and Lactobacillus johnsonii. Conclusion Sal improved memory impairment caused by long-term EtOH intake in rats, which may be related to its regulation of gut dysbiosis and hippocampal dysfunction.
Collapse
Affiliation(s)
- Yu Jiao
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Zhenglin Zhao
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Xin Li
- Department of Psychiatry, The Fourth Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Lulu Li
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Dan Xiao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, Heilongjiang, China
- Department of Medicine and Health, Zhengzhou Research Institute of Harbin Institute of Technology, Zhengzhou, Henan, China
| | - Siyuan Wan
- Department of Preventive Medicine, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Tong Wu
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Tong Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Rongjie Zhao
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| |
Collapse
|
15
|
Papageorgiou G, Amoah SK, Pierotti C, Otero M, Eckel S, Coffey K, Allan AM, Caldwell KK, Mellios N. Prenatal alcohol exposure results in brain region- and sex-specific changes in circHomer1 expression in adult mouse brain. Front Neurosci 2023; 17:1087950. [PMID: 36875647 PMCID: PMC9983553 DOI: 10.3389/fnins.2023.1087950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/16/2023] [Indexed: 02/19/2023] Open
Abstract
Circular RNAs (circRNAs) are a novel category of covalently-closed non-coding RNAs mainly derived from the back-splicing of exons or introns of protein-coding genes. In addition to their inherent high overall stability, circRNAs, have been shown to have strong functional effects on gene expression via a multitude of transcriptional and post-transcriptional mechanisms. Furthermore, circRNAs, appear to be particularly enriched in the brain and able to influence both prenatal development and postnatal brain function. However, little is known about the potential involvement of circRNAs in the long term influence of prenatal alcohol exposure (PAE) in the brain and their relevance for Fetal Alcohol Spectrum Disorders (FASD). Using circRNA-specific quantification, we have found that circHomer1, an activity-dependent circRNA derived from Homer protein homolog 1 (Homer1) and enriched in postnatal brain, is significantly down-regulated in the male frontal cortex and hippocampus of mice subjected to modest PAE. Our data further suggest that the expression of H19, an imprinted embryonic brain-enriched long non-coding RNA (lncRNA), is significantly up-regulated in the frontal cortex of male PAE mice. Furthermore, we show opposing changes in the developmental- and brain region specific- expression of circHomer1 and H19. Lastly, we show that knockdown of H19 results in robust increases in circHomer1 but not linear HOMER1 mRNA expression in human glioblastoma cell lines. Taken together, our work uncovers notable sex- and brain region-specific alterations in circRNA and lncRNA expression following PAE and introduces novel mechanistic insights with potential relevance to FASD.
Collapse
Affiliation(s)
- Grigorios Papageorgiou
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Stephen K. Amoah
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Caroline Pierotti
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Madison Otero
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Sophie Eckel
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kacie Coffey
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Andrea M. Allan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Kevin K. Caldwell
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Nikolaos Mellios
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
- Autophagy, Inflammation, and Metabolism (AIM) Center, Albuquerque, NM, United States
| |
Collapse
|
16
|
Bake S, Hurst DA, Miranda RC, Sohrabji F. Prenatal alcohol exposure exacerbates acute sensorimotor deficits and impedes long-term behavioral recovery from the effects of an adult-onset cerebrovascular ischemic stroke. Alcohol Clin Exp Res 2022; 46:2267-2279. [PMID: 36203340 PMCID: PMC10100487 DOI: 10.1111/acer.14952] [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: 06/21/2022] [Revised: 08/18/2022] [Accepted: 09/24/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is a significant risk factor for developmental disability, although its health consequences across the lifespan are poorly understood. Here, we hypothesized that latent brain and systemic consequences of PAE influence resiliency to adult-onset neurological disease, specifically, cerebrovascular ischemic stroke. METHODS Pregnant Sprague-Dawley rats were exposed episodically to ethanol during the fetal neurogenic period. Adult (5 months) male and female PAE and control offspring were subjected to endothelin-1-induced unilateral middle cerebral artery occlusion. In the acute injury phase outcomes including stroke volume and neurological, endocrine, and gut permeability markers were assessed. Because the effects of stroke in human populations evolve over months to years, we also assessed hippocampal- and amygdala-dependent memory function and social interaction preference up to 6 months following a stroke, in middle-aged offspring. RESULTS Prenatal alcohol exposure did not alter infarct volume, but significantly increased neurological deficits in both sexes, and impaired interhemispheric sensorimotor integration in PAE females. The IGF-1/IGFBP3 ratio, a measure of bioavailable IGF-1, was significantly reduced, while circulating levels of bacterial lipopolysaccharide, an inflammagen, were significantly increased in PAE males. In PAE females, the circulating IGF-1/IGFBP3 ratio was significantly increased and estradiol-17b levels were significantly reduced. The intestinal fatty acid binding protein, a surrogate marker of gut permeability was also significantly increased in PAE females. Longer-term deficits in hippocampal-associated memory and social interactions were observed in PAE males, while deficits in amygdala-dependent memory were observed in PAE females. CONCLUSIONS PAE contributes to adverse effects on brain health and decreased resiliency in response to a common adult-onset neurovascular disease, cerebrovascular ischemic stroke.
Collapse
Affiliation(s)
- Shameena Bake
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - David A Hurst
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - Rajesh C Miranda
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, Texas, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, Texas, USA
| |
Collapse
|
17
|
Risbud R, Breit KR, Thomas JD. Early developmental alcohol exposure alters behavioral outcomes following adolescent re-exposure in a rat model. Alcohol Clin Exp Res 2022; 46:1993-2009. [PMID: 36117379 PMCID: PMC9722643 DOI: 10.1111/acer.14950] [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: 06/20/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Prenatal alcohol exposure alters brain development, affecting cognitive, motor, and emotional domains, and potentially leading to greater alcohol intake during adolescence. The present study investigated whether early alcohol exposure modifies vulnerability to behavioral alterations associated with adolescent alcohol exposure in a rodent model. METHODS Sprague-Dawley rats received ethanol or sham intubations during two developmental periods: (1) the third trimester equivalent of brain development in humans (postnatal days [PD] 4-9) and (2) adolescence (PD 28-42). Both exposures resulted in blood alcohol concentrations around 200 mg/dl. Subjects were tested in the open field (PD 45-48) and on hippocampal and prefrontal cortical (PFC) dependent tasks: the Morris water maze (PD 52-58) and trace fear conditioning (PD 63-64). RESULTS Neonatal alcohol exposure reduced forebrain and cerebellar weight, increased open-field activity, and slowed acquisition of trace fear conditioning. Adolescent alcohol exposure did not disrupt learning or significantly induce gross brain pathology, suggesting that 200 mg/dl/day of ethanol disrupts cognitive development during the 3rd trimester equivalent, but not during adolescence. Interestingly, females exposed to alcohol only during adolescence exhibited an increased conditioned fear response and more rapid habituation of locomotor activity in the open field, suggesting alterations in emotional responding. Moreover, subjects exposed to a combination of neonatal and adolescent alcohol exposure spent significantly more time in the center of the open field chamber than other groups. Similarly, males exposed to the combination exhibited less thigmotaxis in the Morris water maze. CONCLUSIONS These results indicate that combined exposure to alcohol during these two critical periods reduces anxiety-related behaviors and/or increases risk taking in a sex-dependent manner, suggesting that prenatal alcohol exposure may affect risk for emotional consequences of adolescent alcohol exposure.
Collapse
Affiliation(s)
- R.D. Risbud
- Center for Behavioral Teratology, San Diego State University
| | - K. R. Breit
- Center for Behavioral Teratology, San Diego State University
| | - J. D. Thomas
- Center for Behavioral Teratology, San Diego State University
| |
Collapse
|
18
|
Choline Supplementation Modifies the Effects of Developmental Alcohol Exposure on Immune Responses in Adult Rats. Nutrients 2022; 14:nu14142868. [PMID: 35889826 PMCID: PMC9316525 DOI: 10.3390/nu14142868] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Prenatal alcohol exposure can disrupt the development of numerous systems, including the immune system. Indeed, alterations in cytokine levels may contribute to the neuropathological, behavioral, and cognitive problems, and other adverse outcomes observed in individuals with fetal alcohol spectrum disorders. Importantly, supplementation with the essential nutrient choline can improve performance in hippocampal-dependent behaviors; thus, the present study examined the effects of choline on plasma and hippocampal cytokines in adult rats exposed to ethanol in early development. From postnatal day (PD) 4–9 (third trimester equivalent), pups received ethanol (5.25 g/kg/day) or Sham intubations. Subjects were treated with choline chloride (100 mg/kg/day) or saline from PD10–30. On PD60, plasma and hippocampal tissue was collected before and after an immune challenge (lipopolysaccharide (LPS); 50 ug/kg). Prior to the immune challenge, ethanol-exposed subjects showed an overall increase in hippocampal pro-inflammatory cytokines, an effect mitigated by choline supplementation. In contrast, in the plasma, choline reduced LPS-related increases in pro-inflammatory markers, particularly in ethanol-exposed subjects. Thus, early choline supplementation may modify both brain and peripheral inflammation. These results suggest that early choline can mitigate some long-term effects of ethanol exposure on hippocampal inflammation, which may contribute to improved hippocampal function, and could also influence peripheral immune responses that may impact overall health.
Collapse
|
19
|
Carugati M, Goodlett CR, Cudd TA, Washburn SE. The effects of gestational choline supplementation on cerebellar Purkinje cell number in the sheep model of binge alcohol exposure during the first trimester-equivalent. Alcohol 2022; 100:11-21. [PMID: 35114358 PMCID: PMC8983574 DOI: 10.1016/j.alcohol.2022.01.002] [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: 09/15/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 11/01/2022]
Abstract
Individuals with fetal alcohol spectrum disorders (FASD) incur enduring brain damage and neurodevelopmental impairments from prenatal alcohol exposure (PAE). Preclinical rodent models have demonstrated that choline supplementation during development can reduce the severity of adverse neurodevelopmental consequences of PAE. This study used the sheep model to evaluate dietary choline supplementation during pregnancy as a therapeutic intervention, testing the hypothesis that choline can ameliorate alcohol-induced cerebellar Purkinje cell loss. Pregnant ewes were randomly assigned either to a normal control [NC] group (n = 8), or to groups given intravenous infusions of alcohol (or saline) from gestational days 4-41 (the first trimester-equivalent). A weekly binge-drinking pattern was modeled, with three consecutive days of infusions of saline [SAL], 1.75 g/kg/day alcohol [1.75ALC], or 2.5 g/kg/day alcohol [2.5ALC] followed by four days off. Infused ewes were randomly assigned to receive dietary supplements throughout pregnancy of choline (10 mg/kg/day) or placebo (n = 8 per group). Mean blood alcohol concentrations (BAC) were significantly higher in the 2.5ALC groups (287 mg/dL) than the 1.75ALC groups (197 mg/dL). Lamb cerebella were harvested on postnatal day 180 and processed for stereological counts of Purkinje cells. Both alcohol doses caused significant reductions in Purkinje number relative to NC and SAL-Placebo groups, confirming previous findings. Effects of choline supplementation depended on infusion group: it significantly protected against Purkinje cell loss in the 2.5ALC group, had no effect in the 1.75ALC group, and significantly reduced numbers in the SAL-Choline group (though neither the SAL-Choline nor the SAL-Placebo group differed from the NC group). The protection by choline evident only in the 2.5ALC group suggests that multiple, BAC-dependent mechanisms of cerebellar damage may be activated with alcohol exposure in the first trimester, and that choline may protect against pathogenic mechanisms that emerge at higher BACs. These outcomes extend the evidence that early choline supplementation can mitigate some neurodevelopmental defects resulting from binge-like PAE.
Collapse
Affiliation(s)
- Megan Carugati
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States
| | - Charles R Goodlett
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN, 46202, United States
| | - Timothy A Cudd
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States
| | - Shannon E Washburn
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States.
| |
Collapse
|
20
|
Osterlund Oltmanns JR, Schaeffer EA, Goncalves Garcia M, Donaldson TN, Acosta G, Sanchez LM, Davies S, Savage DD, Wallace DG, Clark BJ. Sexually dimorphic organization of open field behavior following moderate prenatal alcohol exposure. Alcohol Clin Exp Res 2022; 46:861-875. [PMID: 35315075 PMCID: PMC9117438 DOI: 10.1111/acer.14813] [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: 12/13/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) can produce deficits in a wide range of cognitive functions but is especially detrimental to behaviors requiring accurate spatial information processing. In open field environments, spatial behavior is organized such that animals establish "home bases" marked by long stops focused around one location. Progressions away from the home base are circuitous and slow, while progressions directed toward the home base are non-circuitous and fast. The impact of PAE on the organization of open field behavior has not been experimentally investigated. METHODS In the present study, adult female and male rats with moderate PAE or saccharin exposure locomoted a circular high walled open field for 30 minutes under lighted conditions. RESULTS The findings indicate that PAE and sex influence the organization of open field behavior. Consistent with previous literature, PAE rats exhibited greater locomotion in the open field. Novel findings from the current study indicate that PAE and sex also impact open field measures specific to spatial orientation. While all rats established a home base on the periphery of the open field, PAE rats, particularly males, exhibited significantly less clustered home base stopping with smaller changes in heading between stops. PAE also impaired progression measures specific to distance estimation, while sex alone impacted progression measures specific to direction estimation. CONCLUSIONS These findings support the conclusion that adult male rats have an increased susceptibility to the effects of PAE on the organization of open field behavior.
Collapse
Affiliation(s)
| | - Ericka A Schaeffer
- Department of Psychology, Northern Illinois University, Dekalb, Illinois, USA
| | | | - Tia N Donaldson
- Department of Psychology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Gabriela Acosta
- Department of Psychology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Lilliana M Sanchez
- Department of Psychology, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Suzy Davies
- Department of Neurosciences, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Daniel D Savage
- Department of Psychology, The University of New Mexico, Albuquerque, New Mexico, USA.,Department of Neurosciences, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Douglas G Wallace
- Department of Psychology, Northern Illinois University, Dekalb, Illinois, USA
| | - Benjamin J Clark
- Department of Psychology, The University of New Mexico, Albuquerque, New Mexico, USA.,Department of Neurosciences, The University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
21
|
Prenatal and adolescent alcohol exposure programs immunity across the lifespan: CNS-mediated regulation. Pharmacol Biochem Behav 2022; 216:173390. [PMID: 35447157 DOI: 10.1016/j.pbb.2022.173390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/28/2022] [Accepted: 04/11/2022] [Indexed: 12/31/2022]
Abstract
For many individuals, first exposure to alcohol occurs either prenatally due to maternal drinking, or during adolescence, when alcohol consumption is most likely to be initiated. Prenatal Alcohol Exposure (PAE) and its associated Fetal Alcohol Spectrum Disorders (FASD) in humans is associated with earlier initiation of alcohol use and increased rates of Alcohol Use Disorders (AUD). Initiation of alcohol use and misuse in early adolescence correlates highly with later AUD diagnosis as well. Thus, PAE and adolescent binge drinking set the stage for long-term health consequences due to adverse effects of alcohol on subsequent immune function, effects that may persist across the lifespan. The overarching goal of this review, therefore, is to determine the extent to which early developmental exposure to alcohol produces long-lasting, and potentially life-long, changes in immunological function. Alcohol affects the whole body, yet most studies are narrowly focused on individual features of immune function, largely ignoring the systems-level interactions required for effective host defense. We therefore emphasize the crucial role of the Central Nervous System (CNS) in orchestrating host defense processes. We argue that alcohol-mediated disruption of host immunity can occur through both (a) direct action of ethanol on neuroimmune processes, that subsequently disrupt peripheral immune function (top down); and (b) indirect action of ethanol on peripheral immune organs/cells, which in turn elicit consequent changes in CNS neuroimmune function (bottom up). Recognizing that alcohol consumption across the entire body, we argue in favor of integrative, whole-organism approaches toward understanding alcohol effects on immune function, and highlight the need for more work specifically examining long-lasting effects of early developmental exposure to alcohol (prenatal and adolescent periods) on host immunity.
Collapse
|
22
|
Macht VA, Vetreno RP, Crews FT. Cholinergic and Neuroimmune Signaling Interact to Impact Adult Hippocampal Neurogenesis and Alcohol Pathology Across Development. Front Pharmacol 2022; 13:849997. [PMID: 35308225 PMCID: PMC8926387 DOI: 10.3389/fphar.2022.849997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 01/21/2023] Open
Abstract
Alcohol (ethanol) use and misuse is a costly societal issue that can affect an individual across the lifespan. Alcohol use and misuse typically initiates during adolescence and generally continues into adulthood. Not only is alcohol the most widely abused drug by adolescents, but it is also one of the most widely abused drugs in the world. In fact, high rates of maternal drinking make developmental ethanol exposure the most preventable cause of neurological deficits in the Western world. Preclinical studies have determined that one of the most consistent effects of ethanol is its disruption of hippocampal neurogenesis. However, the severity, persistence, and reversibility of ethanol’s effects on hippocampal neurogenesis are dependent on developmental stage of exposure and age at assessment. Complicating the neurodevelopmental effects of ethanol is the concurrent development and maturation of neuromodulatory systems which regulate neurogenesis, particularly the cholinergic system. Cholinergic signaling in the hippocampus directly regulates hippocampal neurogenesis through muscarinic and nicotinic receptor actions and indirectly regulates neurogenesis by providing anti-inflammatory regulatory control over the hippocampal environmental milieu. Therefore, this review aims to evaluate how shifting maturational patterns of the cholinergic system and its regulation of neuroimmune signaling impact ethanol’s effects on adult neurogenesis. For example, perinatal ethanol exposure decreases basal forebrain cholinergic neuron populations, resulting in long-term developmental disruptions to the hippocampus that persist into adulthood. Exaggerated neuroimmune responses and disruptions in adult hippocampal neurogenesis are evident after environmental, developmental, and pharmacological challenges, suggesting that perinatal ethanol exposure induces neurogenic deficits in adulthood that can be unmasked under conditions that strain neural and immune function. Similarly, adolescent ethanol exposure persistently decreases basal forebrain cholinergic neuron populations, increases hippocampal neuroimmune gene expression, and decreases hippocampal neurogenesis in adulthood. The effects of neither perinatal nor adolescent ethanol are mitigated by abstinence whereas adult ethanol exposure-induced reductions in hippocampal neurogenesis are restored following abstinence, suggesting that ethanol-induced alterations in neurogenesis and reversibility are dependent upon the developmental period. Thus, the focus of this review is an examination of how ethanol exposure across critical developmental periods disrupts maturation of cholinergic and neuroinflammatory systems to differentially affect hippocampal neurogenesis in adulthood.
Collapse
Affiliation(s)
- Victoria A Macht
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|