1
|
Sullivan EL, Bogdan R, Bakhireva L, Levitt P, Jones J, Sheldon M, Croff JM, Thomason M, Lo JO, MacIntyre L, Shrivastava S, Cioffredi LA, Edlow AG, Howell BR, Chaiyachati BH, Lashley-Simms N, Molloy K, Lam C, Stoermann AM, Trinh T, Ambalavanan N, Neiderhiser JM. Biospecimens in the HEALthy Brain and Child Development (HBCD) Study: Rationale and protocol. Dev Cogn Neurosci 2024; 70:101451. [PMID: 39326174 PMCID: PMC11460495 DOI: 10.1016/j.dcn.2024.101451] [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: 03/06/2024] [Revised: 07/17/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The HEALthy Brain and Child Development (HBCD) Study, a multi-site prospective longitudinal cohort study, will examine human brain, cognitive, behavioral, social, and emotional development beginning prenatally and planned through early childhood. The longitudinal collection of biological samples from over 7000 birthing parents and their children within the HBCD study enables research on pre- and postnatal exposures (e.g., substance use, toxicants, nutrition), and biological processes (e.g., genetics, epigenetic signatures, proteins, metabolites) on neurobehavioral developmental outcomes. The following biosamples are collected from the birthing parent: 1) blood (i.e., whole blood, serum, plasma, buffy coat, and dried blood spots) during pregnancy, 2) nail clippings during pregnancy and one month postpartum, 3) urine during pregnancy, and 4) saliva during pregnancy and at in-person postnatal assessments. The following samples are collected from the child at in-person study assessments: 1) saliva, 2) stool, and 3) urine. Additionally, placenta tissue, cord blood, and cord tissue are collected by a subset of HBCD sites. Here, we describe the rationale for the collection of these biospecimens, their current and potential future uses, the collection protocol, and collection success rates during piloting. This information will assist research teams in the planning of future studies utilizing this collection of biological samples.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Departments of Psychiatry and Behavioral Neuroscience, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA.
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, USA.
| | - Ludmila Bakhireva
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| | - Pat Levitt
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA; Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Joseph Jones
- United States Drug Testing Laboratories, Des Plaines, IL, USA
| | | | - Julie M Croff
- Department of Rural Health, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Moriah Thomason
- Department of Child and Adolescent Psychiatry & Department of Population Health, New York University Langone Health, New York City, NY, USA
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leigh MacIntyre
- McGill University, Montreal, QC, Canada; Lasso Informatics, Montreal, QC, Canada
| | | | - Leigh-Anne Cioffredi
- Dept of Pediatrics, Larner College of Medicine at the University of Vermont, Burlington, VT, USA; Vermont Children's Hospital, Burlington, VT, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brittany R Howell
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Human Development and Family Science, Virginia Tech, Blacksburg, VA, USA
| | - Barbara H Chaiyachati
- Dept of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; PolicyLab & Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nicole Lashley-Simms
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kelly Molloy
- Departments of Psychiatry and Behavioral Neuroscience, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
| | - Cris Lam
- University of California, San Diego, San Diego, CA, USA
| | | | - Thanh Trinh
- University of California, San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
2
|
Camacho-Morales A, Cárdenas-Tueme M. Prenatal Programming of Monocyte Chemotactic Protein-1 Signaling in Autism Susceptibility. Mol Neurobiol 2024; 61:6119-6134. [PMID: 38277116 DOI: 10.1007/s12035-024-03940-z] [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: 09/12/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that involves functional and structural defects in selective central nervous system (CNS) regions, harming the individual capability to process and respond to external stimuli, including impaired verbal and non-verbal communications. Etiological causes of ASD have not been fully clarified; however, prenatal activation of the innate immune system by external stimuli might infiltrate peripheral immune cells into the fetal CNS and activate cytokine secretion by microglia and astrocytes. For instance, genomic and postmortem histological analysis has identified proinflammatory gene signatures, microglia-related expressed genes, and neuroinflammatory markers in the brain during ASD diagnosis. Active neuroinflammation might also occur during the developmental stage, promoting the establishment of a defective brain connectome and increasing susceptibility to ASD after birth. While still under investigation, we tested the hypothesis whether the monocyte chemoattractant protein-1 (MCP-1) signaling is prenatally programmed to favor peripheral immune cell infiltration and activate microglia into the fetal CNS, setting susceptibility to autism-like behavior. In this review, we will comprehensively provide the current understanding of the prenatal activation of MCP-1 signaling by external stimuli during the developmental stage as a new selective node to promote neuroinflammation, brain structural alterations, and behavioral defects associated to ASD diagnosis.
Collapse
Affiliation(s)
- Alberto Camacho-Morales
- College of Medicine, Department of Biochemistry, Universidad Autónoma de Nuevo Leon, Monterrey, NL, Mexico.
- Center for Research and Development in Health Sciences, Neurometabolism Unit, Universidad Autónoma de Nuevo Leon, San Nicolás de los Garza, Monterrey, NL, Mexico.
| | - Marcela Cárdenas-Tueme
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de La Salud and The Institute for Obesity Research, 64710, Monterrey, Mexico
- Nutrition Unit, Center for Research and Development in Health Sciences, Universidad Autonoma de Nuevo Leon, 64460, Monterrey, Mexico
| |
Collapse
|
3
|
Abuiessa SA, Helmy MM, El-Gowelli HM, El-Gowilly SM, El-Mas MM. Gestationally administered RAS modulators reprogram endotoxic cardiovascular and inflammatory profiles in adult male offspring of preeclamptic rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03305-2. [PMID: 39046530 DOI: 10.1007/s00210-024-03305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Previous studies showed that preeclampsia (PE) amplifies cardiovascular dysfunction induced by endotoxemia in adult male, but not female, offspring. Here, we asked if such aggravated endotoxic insult could be nullified by modulators of the renin-angiotensin system (RAS). PE was induced by gestational administration of Nω-nitro-L-arginine methyl ester(L-NAME, a nitric oxide synthase inhibitor). Adult male offspring of PE mothers treated gestationally with angiotensin 1-7 (Ang1-7, angiotensin II-derived vasodilator), losartan (AT1 receptor antagonist), pioglitazone (peroxisome proliferator-activated receptor gamma, PPARγ, agonist), or combined losartan/pioglitazone were instrumented with femoral indwelling catheters and challenged intravenously with a 5-mg/kg dose of lipopolysaccharides (LPS, 5 mg/kg). LPS caused significant decreases in blood pressure (BP) and spectral index of overall heart rate variability and increases in heart rate and left ventricular contractility (dP/dtmax). These effects were mostly reduced to similar magnitudes by individual drug therapies. In offspring born to Ang1-7-treated dams, the spectral index of cardiac sympathovagal balance showed elevated sympathetic dominance in response to LPS. Immunohistochemistry revealed that Ang1-7, but not losartan/pioglitazone, abolished the exaggerated increases in toll-like receptor 4 (TLR-4) expression caused by PE/LPS in heart tissues and neuronal circuits of brainstem rostral ventrolateral medulla (RVLM). By contrast, the losartan/pioglitazone regimen, but not Ang1-7, decreased and increased angiotensin converting enzyme (ACE) and ACE2 expression, respectively. Together, gestational fetal reprogramming of Ang II (depression) and Ang1-7 (activation) arms of RAS effectively counterbalance worsened endotoxic cardiovascular and inflammatory profiles in adult male offspring of PE rats.
Collapse
Affiliation(s)
- Salwa A Abuiessa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mai M Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Hanan M El-Gowelli
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Sahar M El-Gowilly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mahmoud M El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Al-Jabriyah Block 4, Hawally, Jabriya, Kuwait.
| |
Collapse
|
4
|
Le Belle JE, Condro M, Cepeda C, Oikonomou KD, Tessema K, Dudley L, Schoenfield J, Kawaguchi R, Geschwind D, Silva AJ, Zhang Z, Shokat K, Harris NG, Kornblum HI. Acute rapamycin treatment reveals novel mechanisms of behavioral, physiological, and functional dysfunction in a maternal inflammation mouse model of autism and sensory over-responsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602602. [PMID: 39026891 PMCID: PMC11257517 DOI: 10.1101/2024.07.08.602602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Maternal inflammatory response (MIR) during early gestation in mice induces a cascade of physiological and behavioral changes that have been associated with autism spectrum disorder (ASD). In a prior study and the current one, we find that mild MIR results in chronic systemic and neuro-inflammation, mTOR pathway activation, mild brain overgrowth followed by regionally specific volumetric changes, sensory processing dysregulation, and social and repetitive behavior abnormalities. Prior studies of rapamycin treatment in autism models have focused on chronic treatments that might be expected to alter or prevent physical brain changes. Here, we have focused on the acute effects of rapamycin to uncover novel mechanisms of dysfunction and related to mTOR pathway signaling. We find that within 2 hours, rapamycin treatment could rapidly rescue neuronal hyper-excitability, seizure susceptibility, functional network connectivity and brain community structure, and repetitive behaviors and sensory over-responsivity in adult offspring with persistent brain overgrowth. These CNS-mediated effects are also associated with alteration of the expression of several ASD-,ion channel-, and epilepsy-associated genes, in the same time frame. Our findings suggest that mTOR dysregulation in MIR offspring is a key contributor to various levels of brain dysfunction, including neuronal excitability, altered gene expression in multiple cell types, sensory functional network connectivity, and modulation of information flow. However, we demonstrate that the adult MIR brain is also amenable to rapid normalization of these functional changes which results in the rescue of both core and comorbid ASD behaviors in adult animals without requiring long-term physical alterations to the brain. Thus, restoring excitatory/inhibitory imbalance and sensory functional network modularity may be important targets for therapeutically addressing both primary sensory and social behavior phenotypes, and compensatory repetitive behavior phenotypes.
Collapse
|
5
|
Salari Z, Moslemizadeh A, Tezerji SS, Sabet N, Parizi AS, Khaksari M, Sheibani V, Jafari E, Shafieipour S, Bashiri H. Sex-dependent alterations of inflammatory factors, oxidative stress, and histopathology of the brain-gut axis in a VPA-induced autistic-like model of rats. Birth Defects Res 2024; 116:e2310. [PMID: 38563145 DOI: 10.1002/bdr2.2310] [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: 08/03/2023] [Revised: 12/23/2023] [Accepted: 01/21/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION In this study, we aimed to investigate the inflammatory factors, oxidative stress, and histopathological consequences of the brain-gut axis in male and female rats prenatally exposed to VPA. METHODS Pregnant Wistar rats were randomly divided into two groups. The animals received saline, and valproic acid (VPA) (600 mg/kg, i.p.) on embryonic day 12.5 (E12.5). All offspring were weaned on postnatal day 21, and the experiments were done in male and female rats on day 60. The brain and intestine tissues were extracted to assess histopathology, inflammation, and oxidative stress. RESULTS An increase of interleukin-1β (IL-1β) and interleukin-6 (IL-6) and a decrease of interleukin-10 (IL-10) were observed in the two sexes and two tissues of the autistic rats. In the VPA-exposed animals, malondialdehyde (MDA) and protein carbonyl (PC) increased in the brain of both sexes and the intestines of only the males. The total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT) significantly decreased in both tissues of male and female autistic groups. Histopathological evaluation showed that the %apoptosis of the cortex in the autistic male and female groups was more than in controls whereas this parameter in the CA1 and CA3 was significant only in the male rats. In the intestine, histopathologic changes were seen only in the male autistic animals. CONCLUSION The inflammatory and antioxidant factors were in line in the brain-gut axis in male and female rats prenatally exposed to VPA. Histopathological consequences were more significant in the VPA-exposed male animals.
Collapse
Affiliation(s)
- Zahra Salari
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Sheibani Tezerji
- Department of Behavioural and Molecular Neurobiology, Regensburg Center for Neuroscience, University of Regensburg, Regensburg, Germany
| | - Nazanin Sabet
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Saeidpour Parizi
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Jafari
- Pathology and Stem Cells Research Center, Department of Pathology, Afzalipour School of Medicine, Kerman University of Medical Science, Kerman, Iran
| | - Sara Shafieipour
- Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Liu X, Zhu Y, Seamans M, Nianogo R, Janzen C, Fei Z, Chen L. Gestational diabetes mellitus and risk of neurodevelopmental disorders in young offspring: does the risk differ by race and ethnicity? Am J Obstet Gynecol MFM 2024; 6:101217. [PMID: 37940104 DOI: 10.1016/j.ajogmf.2023.101217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Previous studies examined the associations of gestational diabetes mellitus with autism spectrum disorder and attention deficit hyperactivity disorder. However, the associations between gestational diabetes mellitus and other neurodevelopmental disorders, such as the common speech/language disorder and developmental coordination disorder, are rarely studied, and whether the associations vary by race/ethnicity remains unknown. OBJECTIVE This study aimed to examine the associations of gestational diabetes mellitus with individual neurodevelopmental disorders in young offspring, and to investigate whether the associations vary by race/ethnicity. STUDY DESIGN This retrospective cohort study (Glucose in Relation to Women and Babies' Health [GrownB]) included 14,480 mother-offspring pairs in a large medical center in the United States from March 1, 2013 to August 31, 2021. We ascertained gestational diabetes mellitus using the validated ICD (International Classification of Diseases) codes (ICD-9: 648.8x; ICD-10: O24.4x), and identified neurodevelopmental disorders (speech/language disorder, developmental coordination disorder, autism spectrum disorder, and other neurodevelopmental disorders [attention deficit hyperactivity disorder, behavioral disorder, intellectual disability, and learning difficulty]) and their combinations using validated algorithms. We compared the hazard of neurodevelopmental disorders during the entire follow-up period between offspring born to mothers with and without gestational diabetes mellitus using multivariable Cox regression models. RESULTS Among all mothers, 19.9% were Asian, 21.8% were Hispanic, 41.0% were non-Hispanic White, and 17.3% were of other/unknown race/ethnicity. During the median follow-up of 3.5 years (range, 1.0-6.3 years) after birth, 8.7% of offspring developed at least 1 neurodevelopmental disorder. Gestational diabetes mellitus was associated with a higher risk of speech/language disorder (adjusted hazard ratio, 1.59 [95% confidence interval, 1.07-2.35]), developmental coordination disorder (2.36 [1.37-4.04]), autism spectrum disorder (3.16 [1.36-7.37]), other neurodevelopmental disorders (3.12 [1.51-6.47]), any neurodevelopmental disorder (1.86 [1.36-2.53]), the combination of speech/language disorder and autism spectrum disorder (3.79 [1.35-10.61]), and the combination of speech/language disorder and developmental coordination disorder (4.22 [1.69-10.51]) among offspring born to non-Hispanic White mothers. No associations between gestational diabetes mellitus and any neurodevelopmental disorders or their combinations were observed among offspring born to mothers of other racial/ethnic groups. CONCLUSION We observed an elevated risk of neurodevelopmental disorders among young offspring born to non-Hispanic White mothers with gestational diabetes mellitus, but not among other racial/ethnic groups.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA (Drs Liu, Seamans, Nianogo, and Chen)
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, Oakland, CA. (Dr Zhu); Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA (Dr Zhu)
| | - Marissa Seamans
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA (Drs Liu, Seamans, Nianogo, and Chen)
| | - Roch Nianogo
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA (Drs Liu, Seamans, Nianogo, and Chen); California Center for Population Research, University of California Los Angeles, Los Angeles, CA (Dr Nianogo)
| | - Carla Janzen
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA (Dr Janzen)
| | - Zhe Fei
- Department of Statistics, University of California Riverside, Riverside, CA (Dr Fei)
| | - Liwei Chen
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA (Drs Liu, Seamans, Nianogo, and Chen).
| |
Collapse
|
7
|
Luo Y, Lv K, Du Z, Zhang D, Chen M, Luo J, Wang L, Liu T, Gong H, Fan X. Minocycline improves autism-related behaviors by modulating microglia polarization in a mouse model of autism. Int Immunopharmacol 2023; 122:110594. [PMID: 37441807 DOI: 10.1016/j.intimp.2023.110594] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/13/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with few pharmacological treatments. Minocycline, a tetracycline derivative that inhibits microglial activation, has been well-identified with anti-inflammatory properties and neuroprotective effects. A growing body of research suggests that ASD is associated with neuroinflammation, abnormal neurotransmitter levels, and neurogenesis. Thus, we hypothesized that minocycline could improve autism-related behaviors by inhibiting microglia activation and altering neuroinflammation. To verify our hypothesis, we used a mouse model of autism, BTBR T + Itpr3tf/J (BTBR). As expected, minocycline administration rescued the sociability and repetitive, stereotyped behaviors of BTBR mice while having no effect in C57BL/6J mice. We also found that minocycline improved neurogenesis and inhibited microglia activation in the hippocampus of BTBR mice. In addition, minocycline treatment inhibited Erk1/2 phosphorylation in the hippocampus of BTBR mice. Our findings show that minocycline administration alleviates ASD-like behaviors in BTBR mice and improves neurogenesis, suggesting that minocycline supplementation might be a potential strategy for improving ASD symptoms.
Collapse
Affiliation(s)
- Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Zhulin Du
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China; School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dandan Zhang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Mei Chen
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Jing Luo
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
8
|
Mitchell AJ, Dunn GA, Sullivan EL. The Influence of Maternal Metabolic State and Nutrition on Offspring Neurobehavioral Development: A Focus on Preclinical Models. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:450-460. [PMID: 34915175 PMCID: PMC9086110 DOI: 10.1016/j.bpsc.2021.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022]
Abstract
The prevalence of both obesity and neurodevelopmental disorders has increased substantially over the last several decades. Early environmental factors, including maternal nutrition and metabolic state during gestation, influence offspring neurodevelopment. Both human and preclinical models demonstrate a link between poor maternal nutrition, altered metabolic state, and risk of behavioral abnormalities in offspring. This review aims to highlight evidence from the current literature connecting maternal nutrition and the associated metabolic changes with neural and behavioral outcomes in the offspring, as well as identify possible mechanisms underlying these neurodevelopmental outcomes. Owing to the highly correlated nature of poor nutrition and obesity in humans, preclinical animal models are important in distinguishing the unique effects of maternal nutrition and metabolic state on offspring brain development. We use a translational lens to highlight results from preclinical animal models of maternal obesogenic diet related to alterations in behavioral and neurodevelopmental outcomes in offspring. Specifically, we aim to highlight results that resemble behavioral phenotypes described in the diagnostic criteria of neurodevelopmental conditions in humans. Finally, we examine the proinflammatory nature of maternal obesity and consumption of a high-fat diet as a mechanism for neurodevelopmental alterations that may alter offspring behavior later in life. It is important that future studies examine potential therapeutic interventions and prevention strategies to interrupt the transgenerational transmission of the disease. Given the tremendous risk to the next generation, changes need to be made to ensure that all pregnant people have access to nutritious food and are informed about the optimal diet for their developing child.
Collapse
Affiliation(s)
- A J Mitchell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon
| | - Geoffrey A Dunn
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Elinor L Sullivan
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Department of Psychiatry, Oregon Health & Science University, Portland, Oregon; Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon; Department of Human Physiology, University of Oregon, Eugene, Oregon.
| |
Collapse
|
9
|
Maternal P2X7 receptor inhibition prevents autism-like phenotype in male mouse offspring through the NLRP3-IL-1β pathway. Brain Behav Immun 2022; 101:318-332. [PMID: 35065198 DOI: 10.1016/j.bbi.2022.01.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/29/2021] [Accepted: 01/16/2022] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition caused by interactions of environmental and genetic factors. Recently we showed that activation of the purinergic P2X7 receptors is necessary and sufficient to convert maternal immune activation (MIA) to ASD-like features in male offspring mice. Our aim was to further substantiate these findings and identify downstream signaling pathways coupled to P2X7 upon MIA. Maternal treatment with the NLRP3 antagonist MCC950 and a neutralising IL-1β antibody during pregnancy counteracted the development of autistic characteristics in offspring mice. We also explored time-dependent changes of a widespread cytokine and chemokine profile in maternal blood and fetal brain samples of poly(I:C)/saline-treated dams. MIA-induced increases in plasma IL-1β, RANTES, MCP-1, and fetal brain IL-1β, IL-2, IL-6, MCP-1 concentrations are regulated by the P2X7/NLRP3 pathway. Offspring treatment with the selective P2X7 receptor antagonist JNJ47965567 was effective in the prevention of autism-like behavior in mice using a repeated dosing protocol. Our results highlight that in addition to P2X7, NLRP3, as well as inflammatory cytokines, may also be potential biomarkers and therapeutic targets of social deficits and repetitive behaviors observed in autism spectrum disorder.
Collapse
|
10
|
Carthy E, Ellender T. Histamine, Neuroinflammation and Neurodevelopment: A Review. Front Neurosci 2021; 15:680214. [PMID: 34335160 PMCID: PMC8317266 DOI: 10.3389/fnins.2021.680214] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
The biogenic amine, histamine, has been shown to critically modulate inflammatory processes as well as the properties of neurons and synapses in the brain, and is also implicated in the emergence of neurodevelopmental disorders. Indeed, a reduction in the synthesis of this neuromodulator has been associated with the disorders Tourette's syndrome and obsessive-compulsive disorder, with evidence that this may be through the disruption of the corticostriatal circuitry during development. Furthermore, neuroinflammation has been associated with alterations in brain development, e.g., impacting synaptic plasticity and synaptogenesis, and there are suggestions that histamine deficiency may leave the developing brain more vulnerable to proinflammatory insults. While most studies have focused on neuronal sources of histamine it remains unclear to what extent other (non-neuronal) sources of histamine, e.g., from mast cells and other sources, can impact brain development. The few studies that have started exploring this in vitro, and more limited in vivo, would indicate that non-neuronal released histamine and other preformed mediators can influence microglial-mediated neuroinflammation which can impact brain development. In this Review we will summarize the state of the field with regard to non-neuronal sources of histamine and its impact on both neuroinflammation and brain development in key neural circuits that underpin neurodevelopmental disorders. We will also discuss whether histamine receptor modulators have been efficacious in the treatment of neurodevelopmental disorders in both preclinical and clinical studies. This could represent an important area of future research as early modulation of histamine from neuronal as well as non-neuronal sources may provide novel therapeutic targets in these disorders.
Collapse
Affiliation(s)
- Elliott Carthy
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Tommas Ellender
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
The association between gestational diabetes and ASD and ADHD: a systematic review and meta-analysis. Sci Rep 2021; 11:5136. [PMID: 33664319 PMCID: PMC7933135 DOI: 10.1038/s41598-021-84573-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
There is growing evidence for a role of maternal diabetes in the pathogenesis of neurodevelopmental disorders. However, the specific association between gestational diabetes (GDM), as opposed to pre-gestational diabetes, has been poorly isolated. Thus the aim was to systematically review and meta-analyse literature pertaining to prevalence and risk for two neurodevelopmental disorders: autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD), when exposed to GDM. PubMed, Cochrane Library, EMBASE, PsycINFO and CINAHL were systematically searched for eligible literature, with forward and backward citation tracking. Screening for eligibility, risk of bias assessment and data extraction were performed by two independent reviewers. 18 studies measuring ASD and 15 measuring ADHD met inclusion criteria. On meta-analysis there was an increased risk of ASD (OR 1.42; 95% CI 1.22, 1.65) but not ADHD (OR 1.01; 95% CI 0.79, 1.28). We discuss potential mechanisms for these differing risks. Greater understanding of risk factors, including GDM, for these neurodevelopmental disorders and potential mechanisms may help inform strategies aimed at prevention of exposure to these adversities during pregnancy.
Collapse
|
12
|
Emerging mechanisms of valproic acid-induced neurotoxic events in autism and its implications for pharmacological treatment. Biomed Pharmacother 2021; 137:111322. [PMID: 33761592 DOI: 10.1016/j.biopha.2021.111322] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a sort of mental disorder marked by deficits in cognitive and communication abilities. To date no effective cure for this pernicious disease has been available. Valproic acid (VPA) is a broad-spectrum, antiepileptic drug, and it is also a potent teratogen. Epidemiological studies have shown that children exposed to VPA are at higher risk for ASD during the first trimester of their gestational development. Several animal and human studies have demonstrated important behavioral impairments and morphological changes in the brain following VPA treatment. However, the mechanism of VPA exposure-induced ASD remains unclear. Several factors are involved in the pathological phase of ASD, including aberrant excitation/inhibition of synaptic transmission, neuroinflammation, diminished neurogenesis, oxidative stress, etc. In this review, we aim to outline the current knowledge of the critical pathophysiological mechanisms underlying VPA exposure-induced ASD. This review will give insight toward understanding the complex nature of VPA-induced neuronal toxicity and exploring a new path toward the development of novel pharmacological treatment against ASD.
Collapse
|
13
|
Santos ALG, Leão ERLP, Almeida Miranda D, Souza DNC, Picanço Diniz CW, Diniz DG. BALB/c female subjected to valproic acid during gestational period exhibited greater microglial and behavioral changes than male mice: A significant contra intuitive result. Int J Dev Neurosci 2020; 81:37-50. [DOI: 10.1002/jdn.10072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 01/25/2023] Open
Affiliation(s)
- Alinne Lorrany Gomes Santos
- Núcleo de Pesquisas em Oncologia Programa de Pós‐Graduação em Oncologia e Ciências Médicas Hospital Universitário João de Barros BarretoUniversidade Federal do Pará Belém Brasil
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
| | - Ellen Rose Leandro Ponce Leão
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
| | - Diego Almeida Miranda
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
| | - Dilza Nazaré Colares Souza
- Núcleo de Pesquisas em Oncologia Programa de Pós‐Graduação em Oncologia e Ciências Médicas Hospital Universitário João de Barros BarretoUniversidade Federal do Pará Belém Brasil
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
| | - Cristovam Wanderley Picanço Diniz
- Núcleo de Pesquisas em Oncologia Programa de Pós‐Graduação em Oncologia e Ciências Médicas Hospital Universitário João de Barros BarretoUniversidade Federal do Pará Belém Brasil
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
| | - Daniel Guerreiro Diniz
- Núcleo de Pesquisas em Oncologia Programa de Pós‐Graduação em Oncologia e Ciências Médicas Hospital Universitário João de Barros BarretoUniversidade Federal do Pará Belém Brasil
- Laboratório de Investigações em Neurodegeneração e Infecção Hospital Universitário João de Barros BarretoInstituto de Ciências BiológicasUniversidade Federal do Pará Belém Brasil
- Laboratório de Microscopia Eletrônica Instituto Evandro Chagas Belém Brasil
| |
Collapse
|
14
|
de Leão ERLP, de Souza DNC, de Moura LVB, da Silveira Júnior AM, Dos Santos ALG, Diniz DG, Diniz CWP, Sosthenes MCK. Lateral septum microglial changes and behavioral abnormalities of mice exposed to valproic acid during the prenatal period. J Chem Neuroanat 2020; 111:101875. [PMID: 33127448 DOI: 10.1016/j.jchemneu.2020.101875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Most animal model studies of autism spectrum disorder (ASD) have been performed in males, which may be a reflex of the 3-times higher prevalence in boys than in girls. For this reason, little is known about the mechanisms underlying disease progression in females, and nothing is known about potential associations between microglial changes in the lateral septum (LS) and adult female cognition. Prenatal exposure to valproic acid (VPA) in mice has been widely used as an experimental model of autism-like behaviors associated with cellular changes. However, no study has reported the influence of VPA exposure in utero and its consequences on limbic system-dependent tasks or the microglial response in the LS in adult female mice. We compared the exploratory activity and risk assessment in novel environments of BALB/c control mice to mice exposed in utero to VPA and estimated the total number of microglia in the LS using an optical fractionator. On day 12.5 of pregnancy, females received diluted VPA or saline by gavage. After weaning, VPA exposed or control pups were separately housed in standard laboratory cages. At 5 months of age, all mice underwent behavioral testing and their brain sections were immunolabelled using IBA-1 antibody. In the open field test, VPA group showed a greater distance traveled, which was accompanied by less immobility, less time spent on the periphery and a greater number, crossed lines. Similar findings were found in the elevated plus maze test, where VPA mice traveled greater distances, immobility was significantly higher than that of control and VPA group spent less time on the closed arms of apparatus. Stereological analysis demonstrated higher microglial total number and density in the LS of VPA mice, as the cell count was greater, but the volume was similar. Therefore, we suggest that an increase in microglia in the LS may be part of the cellular changes associated with behavioral dysfunction in the VPA model of ASD.
Collapse
Affiliation(s)
- Ellen Rose Leandro Ponce de Leão
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Dilza Nazaré Colares de Souza
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Larissa Victória Barra de Moura
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Antonio Morais da Silveira Júnior
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Alinne Lorrany Gomes Dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil; Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário "João de Barros Barreto", Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil.
| |
Collapse
|
15
|
Placental programming of neuropsychiatric disease. Pediatr Res 2019; 86:157-164. [PMID: 31003234 DOI: 10.1038/s41390-019-0405-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022]
Abstract
The placenta is vital for fetal growth, and compromised function is associated with abnormal development, especially of the brain. Linking placental function to brain development is a new field we have dubbed neuroplacentology. Approximately 380,000 infants in the United States each year abruptly lose placental support upon premature birth, and more than 10% of pregnancies are affected by more insidious placental dysfunction such as preeclampsia or infection. Abnormal fetal brain development or injury can lead to life-long neurological impairments, including psychiatric disorders. The majority of research connecting placental compromise to fetal brain injury has focused on gas exchange or nutritional programming, neglecting the placenta's essential neuroendocrine role. We will review the current evidence that placental dysfunction, particularly endocrine dysfunction, secretion of pro-inflammatory cytokines, or barrier breakdown may place many thousands of fetuses at risk for life-long neurodevelopmental impairments each year. Understanding how specific placental factors shape brain development and increase the risk for later psychiatric disorders, including autism, attention deficit disorder, and schizophrenia, paves the way for novel treatment strategies to maintain the normal developmental milieu and protect from further injury.
Collapse
|
16
|
Lei XY, Li YJ, Ou JJ, Li YM. Association between parental body mass index and autism spectrum disorder: a systematic review and meta-analysis. Eur Child Adolesc Psychiatry 2019; 28:933-947. [PMID: 30470952 DOI: 10.1007/s00787-018-1259-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Studies have examined the association between parental body mass index (BMI) and autism spectrum disorder (ASD) in offspring, with inconsistent results, especially regarding maternal obesity, overweight and underweight. Cochrane Library, EMBASE, PubMed and PsycINFO databases were searched up to March 2018 for relevant observational studies with no language restriction. Our literature search identified 13 eligible studies for meta-analysis (involving 943,293 children and 30,337 cases). For maternal BMI (13 studies), both maternal obesity [OR 1.41 (95% CI 1.19-1.67)] and maternal overweight [OR 1.16 (95% CI 1.05-1.27)] were significantly associated with ASD, while maternal underweight was not associated with ASD [OR 1.08 (95% CI 0.98-1.20)]. For paternal BMI (three studies), no association was found (paternal obesity: OR 1.28, 95% CI 0.94-1.74; overweight: OR 1.07, 95% CI 0.99-1.15; underweight: OR 1.12, 95% CI 0.87-1.44). Pooled estimates were robust in sensitivity analysis and subgroup analyses. Publication bias may exist for studies assessing maternal BMI and ASD risk, but the filled estimates were not altered. Relative to normal weight, maternal obesity and overweight were significantly associated with increased ASD risk, while maternal underweight was not associated with ASD. Although no association between paternal BMI and ASD was found, current evidence is limited (three studies). Future studies are warranted to address more confounding factors and to identify potential mediators of the association, but pre-pregnancy weight control is suggested.
Collapse
Affiliation(s)
- Xian-Yang Lei
- Office of the President, Central South University, Changsha, 410083, Hunan, China
| | - Yong-Jiang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jian-Jun Ou
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ya-Min Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
17
|
Lawford HLS, Lee AC, Kumar S, Liley HG, Bora S. Establishing a conceptual framework of the impact of placental malaria on infant neurodevelopment. Int J Infect Dis 2019; 84:54-65. [PMID: 31028878 DOI: 10.1016/j.ijid.2019.04.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 01/11/2023] Open
Abstract
A novel conceptual framework to describe the relationship between placental malaria and adverse infant neurodevelopmental outcomes is proposed. This conceptual framework includes three distinct stages: (1) maternal and environmental risk factors for the development of placental malaria; (2) placental pathology and inflammation associated with placental malaria infection; and (3) postnatal impacts of placental malaria. The direct, indirect, and bidirectional effects of these risk factors on infant neurodevelopment across the three stages were critically examined. These factors ultimately culminate in an infant phenotype that not only leads to adverse birth outcomes, but also to increased risks of neurological, cognitive, and behavioural deficits that may impact the quality of life in this high-risk population. Multiple risk factors were identified in this conceptual framework; nonetheless, based on current evidence, a key knowledge gap is the uncertainty regarding which are the most important and how exactly they interact.
Collapse
Affiliation(s)
- Harriet L S Lawford
- Mothers, Babies and Women's Health Program, Mater Research Institute, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
| | - Anne Cc Lee
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sailesh Kumar
- Mothers, Babies and Women's Health Program, Mater Research Institute, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
| | - Helen G Liley
- Mothers, Babies and Women's Health Program, Mater Research Institute, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia
| | - Samudragupta Bora
- Mothers, Babies and Women's Health Program, Mater Research Institute, Faculty of Medicine, The University of Queensland, South Brisbane, QLD, Australia.
| |
Collapse
|
18
|
Maldonado-Ruiz R, Garza-Ocañas L, Camacho A. Inflammatory domains modulate autism spectrum disorder susceptibility during maternal nutritional programming. Neurochem Int 2019; 126:109-117. [PMID: 30880046 DOI: 10.1016/j.neuint.2019.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/30/2019] [Accepted: 03/11/2019] [Indexed: 12/21/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disease which involves functional and structural defects in selective central nervous system (CNS) regions harming capability to process and respond to external stimuli. In addition to genetic background, etiological causes of ASD have not been fully clarified. Maternal immune activation (MIA) during pregnancy have been proposed as a potential etiological cause leading to aberrant synaptic pruning and microglia-mediated neurogenesis impairment. Several clinical studies suggest that pro-inflammatory profile during maternal obesity associates with a higher risk of having a child with autism. In this context, the effect of maternal programing by high fat diet overconsumption during pregnancy sets a pro-inflammatory profile partly dependent on an epigenetic program of immunity which promotes brain micro and macrostructural abnormalities in the offspring that might last through adulthood accompanied by phenotypic changes in ASD subjects. Of note, maternal programming of inflammation during development seems to integrate the CNS and peripheral immune system cross-talk which arrays central inflammatory domains coordinating ASD behavior. In this review, we discuss basic and clinical studies regarding the effects of obesity-induced MIA on peripheral immune cells and microglia priming and their relationship with brain structural alterations in ASD models. Also, we show supportive evidence stating the role of maternal programming on epigenetic gene activation in immune cells of ASD subjects. We suggest that maternal programming by hypercaloric diets during development sets a central and peripheral immune cross-talk which potentially might modulate brain macro and microstructural defects leading to autism susceptibility.
Collapse
Affiliation(s)
- Roger Maldonado-Ruiz
- Universidad Autónoma de Nuevo Leon, Facultad de Medicina, Biochemistry Department, Mexico; Universidad Autónoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Neurometabolism Unit, Mexico
| | - Lourdes Garza-Ocañas
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina, Department of Pharmacology, Mexico
| | - Alberto Camacho
- Universidad Autónoma de Nuevo Leon, Facultad de Medicina, Biochemistry Department, Mexico; Universidad Autónoma de Nuevo Leon, Centro de Investigación y Desarrollo en Ciencias de la Salud, Neurometabolism Unit, Mexico.
| |
Collapse
|
19
|
Maternal Overnutrition Programs Central Inflammation and Addiction-Like Behavior in Offspring. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8061389. [PMID: 30027100 PMCID: PMC6031166 DOI: 10.1155/2018/8061389] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/18/2018] [Accepted: 05/27/2018] [Indexed: 12/21/2022]
Abstract
Obesity or maternal overnutrition during pregnancy and lactation might have long-term consequences in offspring health. Fetal programming is characterized by adaptive responses to specific environmental conditions during early life stages. Programming alters gene expression through epigenetic modifications leading to a transgenerational effect of behavioral phenotypes in the offspring. Maternal intake of hypercaloric diets during fetal development programs aberrant behaviors resembling addiction in offspring. Programming by hypercaloric surplus sets a gene expression pattern modulating axonal pruning, synaptic signaling, and synaptic plasticity in selective regions of the reward system. Likewise, fetal programming can promote an inflammatory phenotype in peripheral and central sites through different cell types such as microglia and T and B cells, which contribute to disrupted energy sensing and behavioral pathways. The molecular mechanism that regulates the central and peripheral immune cross-talk during fetal programming and its relevance on offspring's addictive behavior susceptibility is still unclear. Here, we review the most relevant scientific reports about the impact of hypercaloric nutritional fetal programming on central and peripheral inflammation and its effects on addictive behavior of the offspring.
Collapse
|
20
|
Peretti S, Mariano M, Mazzocchetti C, Mazza M, Pino MC, Verrotti Di Pianella A, Valenti M. Diet: the keystone of autism spectrum disorder? Nutr Neurosci 2018; 22:825-839. [PMID: 29669486 DOI: 10.1080/1028415x.2018.1464819] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Children with autism are characterized by an impairment of social interaction and repetitive patterns of behaviour. Autism is a heterogeneous span of disorders with unknown aetiology. Research has grown significantly and has suggested that environmental risk factors acting during the prenatal period could influence the neurodevelopment of offspring. The literature suggests that the maternal diet during pregnancy has a fundamental role in the etiopathogenesis of autism. Indeed, a maternal diet that is high in some nutrients has been associated with an increase or reduction in the risk of develop Autism Spectrum Disorders (ASD). The diet of ASD children is also a key factor for the worsening of ASD symptoms. Children with autism have food selectivity and limited diets due to smell, taste, or other characteristics of foods. This determines eating routines and food intake patterns, with consequent deficiency or excess of some aliments. Several studies have tried to show a possible relationship between nutritional status and autism. In this review we describe, emphasizing the limits and benefits, the main current empirical studies that have examined the role of maternal diet during gestation and diet of ASD children as modifiable risk factors at the base of development or worsening of symptoms of autism.
Collapse
Affiliation(s)
- S Peretti
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - M Mariano
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - C Mazzocchetti
- Department of Pediatrics, San Salvatore Hospital, University of L'Aquila, Località Coppito, L'Aquila 67100, Italy
| | - M Mazza
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - M C Pino
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| | - A Verrotti Di Pianella
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Department of Pediatrics, San Salvatore Hospital, University of L'Aquila, Località Coppito, L'Aquila 67100, Italy
| | - M Valenti
- Department of Applied Clinical Sciences and Biotechnology, Section of Clinical Epidemiology and Environmental Medicine, University of L'Aquila, Via Vetoio, Località Coppito, 67100 L'Aquila, Italy.,Abruzzo Region Health System, Reference Regional Centre for Autism, L'Aquila, Italy
| |
Collapse
|
21
|
Andersen CH, Thomsen PH, Nohr EA, Lemcke S. Maternal body mass index before pregnancy as a risk factor for ADHD and autism in children. Eur Child Adolesc Psychiatry 2018; 27:139-148. [PMID: 28712019 DOI: 10.1007/s00787-017-1027-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/10/2017] [Indexed: 12/15/2022]
Abstract
The risk of attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorders (ASD) may be influenced by environmental factors such as maternal obesity before pregnancy. Previous studies investigating those associations have found divergent results. We aim to investigate in a large birth cohort this association further in children with ADHD, ASD and comorbid ADHD and ASD. Our study population consisted of 81,892 mother-child pairs participating in the Danish National Birth Cohort (DNBC). Information about pre-pregnancy weight and height was collected in week 16 of pregnancy; the analysis was divided into groups based on BMI. Children with a clinical diagnosis of ADHD and/or ASD were identified in the Danish health registries at an average age of 13.3 years. Hazard ratios (HRs) were estimated using time-to-event analysis. Compared to normal weight mothers, the risk of having a child with ADHD was significantly increased if the mother was overweight (HR = 1.28 [95% CI 1.15;1.48]), obese (HR = 1.47 [95% CI 1.26;1.71]) or severely obese (HR = 1.95 [95% CI 1.58;2.40]). The same pattern was seen for the combined ADHD and ASD group. Regarding ASD, an increased risk was observed in underweight (HR = 1.30 [95% CI 1.01;1.69]) and obese (HR = 1.39 [95% CI 1.11;1.75]) mothers. Subgroup analysis revealed that the association in the ADHD group could mostly be attributable to the hyperactive group. Maternal obesity before pregnancy is a risk factor for ADHD in children. Maternal obesity as well as underweight may also be associated with an increased risk for ASD.
Collapse
Affiliation(s)
| | - Per Hove Thomsen
- Center for Child and Adolescent Psychiatry, Aarhus University Hospital, Risskov, Denmark
| | - Ellen Aagaard Nohr
- Research Unit for Gynaecology and Obstetrics, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sanne Lemcke
- Center for Child and Adolescent Psychiatry, Aarhus University Hospital, Risskov, Denmark.
| |
Collapse
|
22
|
Goldschmidt J. What Happened to Paul? Manifestation of Abnormal Pain Response for Individuals With Autism Spectrum Disorder. QUALITATIVE HEALTH RESEARCH 2017; 27:1133-1145. [PMID: 27117957 DOI: 10.1177/1049732316644415] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
During the progression of a pilot nutrition intervention designed to teach cooking skills to young adults with autism spectrum disorder (ASD), one participant-Paul-fell in the parking lot. Prior to the accident, Paul had been making significant gains in the program and had communicated in a number of ways his enthusiasm. After his accident, which resulted in broken and dislocated bones in his ankle, his demeanor was dramatically altered, program gains were lost, and staff noted the appearance of many new challenging behaviors. This article analyzes Paul's behavior in reference to the pain response in autism. For some time, it was believed that many individuals with ASD did not experience pain based on anecdotal reports of how individuals responded to injury with seeming indifference. This view has given way of late to a more nuanced understanding of how atypical sensory processing and stimulus over-selectivity spill over into pain pathways and pain amplification mechanisms. The consequence is not a reduction in pain sensation, but a different expression of pain, determined by that individual's particular communicative, cognitive, or physiological challenges. From this perspective, many of the disruptive and harmful behaviors that emerged after Paul's accident can be seen as a delayed response to the incident. This article concludes by arguing that professionals across all domains of health care need to begin to see behavior as communicative for those with ASD. This is particularly true of changes in behavior, which can be significant indicators of health care problems rather than something to be dismissed as another manifestation of the condition.
Collapse
|
23
|
Tohidpour A, Morgun AV, Boitsova EB, Malinovskaya NA, Martynova GP, Khilazheva ED, Kopylevich NV, Gertsog GE, Salmina AB. Neuroinflammation and Infection: Molecular Mechanisms Associated with Dysfunction of Neurovascular Unit. Front Cell Infect Microbiol 2017; 7:276. [PMID: 28676848 PMCID: PMC5476750 DOI: 10.3389/fcimb.2017.00276] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is a complex inflammatory process in the central nervous system, which is sought to play an important defensive role against various pathogens, toxins or factors that induce neurodegeneration. The onset of neurodegenerative diseases and various microbial infections are counted as stimuli that can challenge the host immune system and trigger the development of neuroinflammation. The homeostatic nature of neuroinflammation is essential to maintain the neuroplasticity. Neuroinflammation is regulated by the activity of neuronal, glial, and endothelial cells within the neurovascular unit, which serves as a “platform” for the coordinated action of pro- and anti-inflammatory mechanisms. Production of inflammatory mediators (cytokines, chemokines, reactive oxygen species) by brain resident cells or cells migrating from the peripheral blood, results in the impairment of blood-brain barrier integrity, thereby further affecting the course of local inflammation. In this review, we analyzed the most recent data on the central nervous system inflammation and focused on major mechanisms of neurovascular unit dysfunction caused by neuroinflammation and infections.
Collapse
Affiliation(s)
- Abolghasem Tohidpour
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Andrey V Morgun
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia.,Department of Paediatrics, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Elizaveta B Boitsova
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia.,Department of Children Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Natalia A Malinovskaya
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Galina P Martynova
- Department of Children Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Elena D Khilazheva
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Natalia V Kopylevich
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Galina E Gertsog
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| | - Alla B Salmina
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Prof. V.F. Voino-YasenetskyKrasnoyarsk, Russia
| |
Collapse
|
24
|
Wright C, Shin JH, Rajpurohit A, Deep-Soboslay A, Collado-Torres L, Brandon NJ, Hyde TM, Kleinman JE, Jaffe AE, Cross AJ, Weinberger DR. Altered expression of histamine signaling genes in autism spectrum disorder. Transl Psychiatry 2017; 7:e1126. [PMID: 28485729 PMCID: PMC5534955 DOI: 10.1038/tp.2017.87] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
The histaminergic system (HS) has a critical role in cognition, sleep and other behaviors. Although not well studied in autism spectrum disorder (ASD), the HS is implicated in many neurological disorders, some of which share comorbidity with ASD, including Tourette syndrome (TS). Preliminary studies suggest that antagonism of histamine receptors 1-3 reduces symptoms and specific behaviors in ASD patients and relevant animal models. In addition, the HS mediates neuroinflammation, which may be heightened in ASD. Together, this suggests that the HS may also be altered in ASD. Using RNA sequencing (RNA-seq), we investigated genome-wide expression, as well as a focused gene set analysis of key HS genes (HDC, HNMT, HRH1, HRH2, HRH3 and HRH4) in postmortem dorsolateral prefrontal cortex (DLPFC) initially in 13 subjects with ASD and 39 matched controls. At the genome level, eight transcripts were differentially expressed (false discovery rate <0.05), six of which were small nucleolar RNAs (snoRNAs). There was no significant diagnosis effect on any of the individual HS genes but expression of the gene set of HNMT, HRH1, HRH2 and HRH3 was significantly altered. Curated HS gene sets were also significantly differentially expressed. Differential expression analysis of these gene sets in an independent RNA-seq ASD data set from DLPFC of 47 additional subjects confirmed these findings. Understanding the physiological relevance of an altered HS may suggest new therapeutic options for the treatment of ASD.
Collapse
Affiliation(s)
- C Wright
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,AstraZeneca Postdoc Program, Innovative Medicines and Early Development, Waltham, MA, USA
| | - J H Shin
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - A Rajpurohit
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - A Deep-Soboslay
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - L Collado-Torres
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - N J Brandon
- AstraZeneca Neuroscience, Innovative Medicines and Early Development, Waltham, MA, USA
| | - T M Hyde
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J E Kleinman
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - A E Jaffe
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - A J Cross
- AstraZeneca Neuroscience, Innovative Medicines and Early Development, Waltham, MA, USA
| | - D R Weinberger
- Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Lieber Institute for Brain Development, Clinical Sciences, Johns Hopkins School of Medicine, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, 3rd Floor, Baltimore, MD 21205, USA. E-mail:
| |
Collapse
|
25
|
Fortunato JJ, da Rosa N, Martins Laurentino AO, Goulart M, Michalak C, Borges LP, da Cruz Cittadin Soares E, Reis PA, de Castro Faria Neto HC, Petronilho F. Effects of ω-3 fatty acids on stereotypical behavior and social interactions in Wistar rats prenatally exposed to lipopolysaccarides. Nutrition 2017; 35:119-127. [DOI: 10.1016/j.nut.2016.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/21/2016] [Accepted: 10/29/2016] [Indexed: 02/07/2023]
|
26
|
Sivanesan S, Tan A, Jeyaraj R, Lam J, Gole M, Hardan A, Ashkan K, Rajadas J. Pharmaceuticals and Stem Cells in Autism Spectrum Disorders: Wishful Thinking? World Neurosurg 2017; 98:659-672. [DOI: 10.1016/j.wneu.2016.09.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/24/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022]
|
27
|
Getz KD, Anderka MT, Werler MM, Jick SS. Maternal Pre-pregnancy Body Mass Index and Autism Spectrum Disorder among Offspring: A Population-Based Case-Control Study. Paediatr Perinat Epidemiol 2016; 30:479-87. [PMID: 27239935 PMCID: PMC5849232 DOI: 10.1111/ppe.12306] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have attributed high maternal weight gain during pregnancy and pre-pregnancy obesity to a higher risk for autism spectrum disorder (ASD). Maternal underweight was not previously explored with respect to ASD risk. METHODS We evaluated the association between maternal pre-pregnancy body mass index (BMI) and ASD occurrence among singletons born into the General Practice Research Database from 1993 to 2008. Case subjects were children with a diagnosis of ASD from birth to 2010. Up to four control subjects were matched to each case subject on birth year, sex, and general practice. Restricted cubic splines were used to assess the non-linearity of the association between maternal BMI and ASD. All study subjects were classified as underweight, normal weight, overweight, or obese based on maternal pre-pregnancy BMI using the WHO Classification Standard. Conditional logistic regression was used to calculate unadjusted and multivariable adjusted odds ratios for the association between categorical BMI (reference=normal weight) and the occurrence of ASD. RESULTS The association between maternal BMI and ASD occurrence was non-linear and J-shaped. The adjusted ORs for maternal underweight and obesity were 1.43 (95% CI 1.01, 2.04) and 1.54 (95% CI 1.26, 1.89) respectively. CONCLUSIONS Results suggest that extremes in maternal BMI may be associated with modest increases in the risk for ASD among offspring.
Collapse
Affiliation(s)
- Kelly D. Getz
- Boston University School of Public Health, Boston, MA
| | | | | | - Susan S. Jick
- Boston University School of Public Health, Boston, MA,Boston Collaborative Drug Surveillance Program, Lexington, MA
| |
Collapse
|
28
|
Young AMH, Chakrabarti B, Roberts D, Lai MC, Suckling J, Baron-Cohen S. From molecules to neural morphology: understanding neuroinflammation in autism spectrum condition. Mol Autism 2016; 7:9. [PMID: 26793298 PMCID: PMC4719563 DOI: 10.1186/s13229-016-0068-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Growing evidence points toward a critical role for early (prenatal) atypical neurodevelopmental processes in the aetiology of autism spectrum condition (ASC). One such process that could impact early neural development is inflammation. We review the evidence for atypical expression of molecular markers in the amniotic fluid, serum, cerebrospinal fluid (CSF), and the brain parenchyma that suggest a role for inflammation in the emergence of ASC. This is complemented with a number of neuroimaging and neuropathological studies describing microglial activation. Implications for treatment are discussed.
Collapse
Affiliation(s)
- Adam M H Young
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Bhismadev Chakrabarti
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Science, University of Reading, Reading, UK
| | - David Roberts
- School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, Canada ; Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - John Suckling
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK ; Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; CLASS Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
29
|
Sullivan EL, Riper KM, Lockard R, Valleau JC. Maternal high-fat diet programming of the neuroendocrine system and behavior. Horm Behav 2015; 76:153-61. [PMID: 25913366 PMCID: PMC4619177 DOI: 10.1016/j.yhbeh.2015.04.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/20/2015] [Accepted: 04/06/2015] [Indexed: 01/05/2023]
Abstract
This article is part of a Special Issue "SBN 2014". Maternal obesity, metabolic state, and diet during gestation have profound effects on offspring development. The prevalence of neurodevelopmental and mental health disorders has risen rapidly in the last several decades in parallel with the rise in obesity rates. Evidence from epidemiological studies indicates that maternal obesity and metabolic complications increase the risk of offspring developing behavioral disorders such as attention deficit hyperactivity disorder (ADHD), autism spectrum disorders (ASD), and schizophrenia. Animal models show that a maternal diet high in fat similarly disrupts behavioral programming of offspring, with animals showing social impairments, increased anxiety and depressive behaviors, reduced cognitive development, and hyperactivity. Maternal obesity, metabolic conditions, and high fat diet consumption increase maternal leptin, insulin, glucose, triglycerides, and inflammatory cytokines. This leads to increased risk of placental dysfunction, and altered fetal neuroendocrine development. Changes in brain development that likely contribute to the increased risk of behavioral and mental health disorders include increased inflammation in the brain, as well as alterations in the serotonergic system, dopaminergic system and hypothalamic-pituitary-adrenal (HPA) axis.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Biology, University of Portland, Portland, OR, USA; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA.
| | - Kellie M Riper
- Department of Biology, University of Portland, Portland, OR, USA
| | - Rachel Lockard
- Department of Biology, University of Portland, Portland, OR, USA
| | - Jeanette C Valleau
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| |
Collapse
|
30
|
Poddighe D, Castelli L, Marseglia GL, Bruni P. A sudden onset of a pseudo-neurological syndrome after HPV-16/18 AS04-adjuvated vaccine: might it be an autoimmune/inflammatory syndrome induced by adjuvants (ASIA) presenting as a somatoform disorder? Immunol Res 2015; 60:236-46. [PMID: 25388965 DOI: 10.1007/s12026-014-8575-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In last centuries, vaccines reduced the incidence of several infectious diseases. In last decades, some vaccines aimed at preventing also some cancers, where viruses play a causative role. However, several adverse events have been described after vaccines, but a causal relationship has been established only in a minority of cases. Here, we describe a pseudo-neurological syndrome occurred shortly after the administration of the bivalent HPV vaccine. Some autoimmune disorders, including neurological demyelinating diseases, have been reported after HPV vaccines, but the patient showed no organic lesions. The patient was diagnosed as having a functional somatoform syndrome, which was supposed to be autoimmune/inflammatory syndrome induced by adjuvants (ASIA), seen the temporal link with vaccination and the presence of anti-phospholipid autoantibodies. Immunological mechanisms of vaccines-and of adjuvants-have not been completely elucidated yet, and although there is no evidence of statistical association with many post-vaccination events, a causal link with vaccine cannot be excluded in some individuals.
Collapse
Affiliation(s)
- Dimitri Poddighe
- Department of Pediatrics, Azienda Ospedaliera di Melegnano, Milan, Italy,
| | | | | | | |
Collapse
|
31
|
El Gohary TM, El Aziz NA, Darweesh M, Sadaa ES. Plasma level of transforming growth factor β 1 in children with autism spectrum disorder. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.ejenta.2014.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
Abstract
Growing evidence suggests that maternal immune activation has a significant impact on the immuno-competence of the offspring. The present study aimed to characterize region-specific effects of maternal immune activation on the offspring’s neuroimmune function. The offspring born to dams treated with saline or lipopolysaccharide (LPS) at gestational day 18 was stimulated with saline or LPS at postnatal day 21, and the mRNA expression of various inflammatory genes in different brain regions of the offspring was analyzed. The offspring born to saline-treated dams exhibited a typical neuroimmune response with elevated levels of cytokines and chemokines following LPS stimulation in all four brain regions examined. In contrast, the offspring born to LPS-treated dams exhibited significantly reduced mRNA induction of cytokines and chemokines following LPS stimulation in the prefrontal cortex but not in the brainstem when compared with pups born to saline-treated dams. Furthermore, the mRNA expression of LPS-induced I-κBζ was significantly attenuated in the prefrontal cortex when compared with pups born to saline-treated dams. These results suggest that maternal LPS may have differential effects on the neuroimmune function in different regions of the offspring brain, and highlight the importance of maternal milieu in the development of neuroimmune function in the offspring.
Collapse
Affiliation(s)
- Heping Zhou
- Department of Biological Sciences, Seton Hall University, New Jersey, USA
| |
Collapse
|
33
|
Gesundheit B, Ashwood P, Keating A, Naor D, Melamed M, Rosenzweig JP. Therapeutic properties of mesenchymal stem cells for autism spectrum disorders. Med Hypotheses 2014; 84:169-77. [PMID: 25592283 DOI: 10.1016/j.mehy.2014.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
Recent studies of autism spectrum disorders (ASD) highlight hyperactivity of the immune system, irregular neuronal growth and increased size and number of microglia. Though the small sample size in many of these studies limits extrapolation to all individuals with ASD, there is mounting evidence of both immune and nervous system related pathogenesis in at least a subset of patients with ASD. Given the disturbing rise in incidence rates for ASD, and the fact that no pharmacological therapy for ASD has been approved by the Food and Drug Administration (FDA), there is an urgent need for new therapeutic options. Research in the therapeutic effects of mesenchymal stem cells (MSC) for other immunological and neurological conditions has shown promising results in preclinical and even clinical studies. MSC have demonstrated the ability to suppress the immune system and to promote neurogenesis with a promising safety profile. The working hypothesis of this paper is that the potentially synergistic ability of MSC to modulate a hyperactive immune system and its ability to promote neurogenesis make it an attractive potential therapeutic option specifically for ASD. Theoretical mechanisms of action will be suggested, but further research is necessary to support these hypothetical pathways. The choice of tissue source, type of cell, and most appropriate ages for therapeutic intervention remain open questions for further consideration. Concern over poor regulatory control of stem cell studies or treatment, and the unique ethical challenges that each child with ASD presents, demands that future research be conducted with particular caution before widespread use of the proposed therapeutic intervention is implemented.
Collapse
Affiliation(s)
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; Department of Medical Microbiology and Immunology, and the MIND Institute, University of California Davis, USA.
| | - Armand Keating
- Division of Hematology, University of Toronto, Cell Therapy Program, Princess Margaret Hospital, Toronto, Canada.
| | - David Naor
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | - Michal Melamed
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | | |
Collapse
|
34
|
Haldipur P, Gillies GS, Janson OK, Chizhikov VV, Mithal DS, Miller RJ, Millen KJ. Foxc1 dependent mesenchymal signalling drives embryonic cerebellar growth. eLife 2014; 3. [PMID: 25513817 PMCID: PMC4281880 DOI: 10.7554/elife.03962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/16/2014] [Indexed: 01/02/2023] Open
Abstract
Loss of Foxc1 is associated with Dandy-Walker malformation, the most common human cerebellar malformation characterized by cerebellar hypoplasia and an enlarged posterior fossa and fourth ventricle. Although expressed in the mouse posterior fossa mesenchyme, loss of Foxc1 non-autonomously induces a rapid and devastating decrease in embryonic cerebellar ventricular zone radial glial proliferation and concurrent increase in cerebellar neuronal differentiation. Subsequent migration of cerebellar neurons is disrupted, associated with disordered radial glial morphology. In vitro, SDF1α, a direct Foxc1 target also expressed in the head mesenchyme, acts as a cerebellar radial glial mitogen and a chemoattractant for nascent Purkinje cells. Its receptor, Cxcr4, is expressed in cerebellar radial glial cells and conditional Cxcr4 ablation with Nes-Cre mimics the Foxc1-/- cerebellar phenotype. SDF1α also rescues the Foxc1-/- phenotype. Our data emphasizes that the head mesenchyme exerts a considerable influence on early embryonic brain development and its disruption contributes to neurodevelopmental disorders in humans.
Collapse
Affiliation(s)
- Parthiv Haldipur
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Gwendolyn S Gillies
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Olivia K Janson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Memphis, United States
| | - Divakar S Mithal
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Chicago, United States
| | - Richard J Miller
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Chicago, United States
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States
| |
Collapse
|
35
|
Schwartz CE. Aberrant tryptophan metabolism: the unifying biochemical basis for autism spectrum disorders? Biomark Med 2014; 8:313-5. [PMID: 24712420 DOI: 10.2217/bmm.14.11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
“…it is quite possible that impairment of the metabolism of tryptophan … provides the unifying model that explains the heterogeneity of autism spectrum disorder…”
Collapse
Affiliation(s)
- Charles E Schwartz
- JC Self Research Institute, Greenwood Genetic Center, 113 Gregor Mendel Circle, Greenwood, SC 29646, USA.
| |
Collapse
|
36
|
Mazur-Kolecka B, Cohen IL, Gonzalez M, Jenkins EC, Kaczmarski W, Brown WT, Flory M, Frackowiak J. Autoantibodies against neuronal progenitors in sera from children with autism. Brain Dev 2014; 36:322-9. [PMID: 23838310 DOI: 10.1016/j.braindev.2013.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/09/2013] [Accepted: 04/30/2013] [Indexed: 12/13/2022]
Abstract
The pathological role of autoantibodies in development of CNS disorders is a new idea with growing interest among neuroscientists. The involvement of autoimmune response in the pathogenesis of autism spectrum disorders (ASD) has been suggested by the presence of multiple brain-specific autoantibodies in children with ASD and in their mothers. The possibility of the effect of autoimmunity on neurogenesis and postnatal brain plasticity has not been determined. The presence of autoantibodies against human neuronal progenitor cells (NPCs) stimulated for neuronal differentiation in culture was tested in sera from children with autism (n=20) and age-matched controls (n=18) by immunoblotting and immunocytochemistry. Immunoreactivity against multiple NPCs proteins of molecular sizes of approximately 55 kDa, 105 kDa, 150 kDa, and 210 kDa in sera from individuals with autism had a higher incidence and was stronger than in control sera which immunoreacted mainly with a 150 kDa protein. The sera from children with autism immunoreacted the strongest with NPCs expressing neuronal markers Tuj1 and doublecortin, but not astrocyte marker GFAP. The epitopes recognized by antibodies from sera were not human-specific because they detected also NPCs in situ in murine hippocampus. The autoimmune reactions against NPCs suggest an impaired tolerance to neural antigens in autism. These autoantibodies may be symptomatic for autism and furthermore, their presence suggests that autoimmunity may affect postnatal neuronal plasticity particularly after impairment of blood-brain barrier. Future studies will determine the diagnostic value of the presence of autoantibodies in autism and the therapeutic value of prevention of autoimmunity in autism.
Collapse
Affiliation(s)
- Bozena Mazur-Kolecka
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA.
| | | | | | | | - Wojciech Kaczmarski
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA
| | - W Ted Brown
- Department of Human Genetics, NYS IBRDD, USA
| | - Michael Flory
- Laboratory of Research Design and Analysis, NYS IBRDD, USA
| | - Janusz Frackowiak
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, USA
| |
Collapse
|
37
|
Mitchell RHB, Goldstein BI. Inflammation in children and adolescents with neuropsychiatric disorders: a systematic review. J Am Acad Child Adolesc Psychiatry 2014; 53:274-96. [PMID: 24565356 DOI: 10.1016/j.jaac.2013.11.013] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/21/2013] [Accepted: 12/13/2013] [Indexed: 11/29/2022]
Abstract
OBJECTIVE There has been rapid growth in research regarding inflammation in neuropsychiatric disorders as it relates to youth. We therefore set out to systematically review the literature on inflammation and neuropsychiatric disorders in children and adolescents. METHOD A systematic review of the literature was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Studies were included if proinflammatory markers (PIMs) in children and/or adolescents with neuropsychiatric disorders were measured. RESULTS Sixty-seven studies were included, involving 3,952 youth. Evidence for a proinflammatory state is strongest for autism spectrum disorders (ASD). PIMs are elevated in children and adolescents with major depressive disorder (MDD), bipolar disorder (BD), post-traumatic stress disorder (PTSD), obsessive-compulsive disorder (OCD), Tourette's disorder (TD), attention-deficit/hyperactivity disorder (ADHD), and schizophrenia (SZ). However, the data are inconsistent. Evidence for specific PIMs is equivocal at this stage, although the findings in youth with MDD, BD, and PTSD converge with the extant adult literature in these areas. Definitive conclusions are limited by methodologic factors including cross-sectional and retrospective study design, between-study differences in specific markers and methods of analysis, small sample size, and other sources of heterogeneity. CONCLUSION The literature regarding inflammation among children and adolescents with neuropsychiatric disorders represents nearly 4,000 youth. There is preliminary evidence for elevated markers of inflammation in this population. Larger, prospective studies are needed to realize the goal of inflammatory markers informing clinical practice. In the interim, present findings suggest that further examination of this topic is warranted.
Collapse
|
38
|
Wei H, Alberts I, Li X. Brain IL-6 and autism. Neuroscience 2013; 252:320-5. [PMID: 23994594 DOI: 10.1016/j.neuroscience.2013.08.025] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 08/14/2013] [Accepted: 08/15/2013] [Indexed: 12/17/2022]
Abstract
Autism is a severe neurodevelopmental disorder characterized by impairments in social interaction, deficits in verbal and non-verbal communication, and repetitive behavior and restricted interests. Emerging evidence suggests that aberrant neuroimmune responses may contribute to phenotypic deficits and could be appropriate targets for pharmacologic intervention. Interleukin (IL)-6, one of the most important neuroimmune factors, has been shown to be involved in physiological brain development and in several neurological disorders. For instance, findings from postmortem and animal studies suggest that brain IL-6 is an important mediator of autism-like behaviors. In this review, a possible pathological mechanism behind autism is proposed, which suggests that IL-6 elevation in the brain, caused by the activated glia and/or maternal immune activation, could be an important inflammatory cytokine response involved in the mediation of autism-like behaviors through impairments of neuroanatomical structures and neuronal plasticity. Further studies to investigate whether IL-6 could be used for therapeutic interventions in autism would be of great significance.
Collapse
Affiliation(s)
- H Wei
- Central Laboratory, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China.
| | | | | |
Collapse
|
39
|
Pineda E, Shin D, You SJ, Auvin S, Sankar R, Mazarati A. Maternal immune activation promotes hippocampal kindling epileptogenesis in mice. Ann Neurol 2013; 74:11-9. [PMID: 23907982 DOI: 10.1002/ana.23898] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 03/02/2013] [Accepted: 03/15/2013] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Maternal immune activation (MIA) triggered by infections has been identified as a cause of autism in offspring. Considering the involvement of perturbations in innate immunity in epilepsy, we examined whether MIA represents a risk factor for epilepsy as well. The role of specific MIA components interleukin (IL)-6 and IL-1β was also addressed. METHODS MIA was induced in C57BL/6 mice by polyinosinic-polycytidylic acid (PIC) injected during embryonic days 12 to 16. Beginning from postnatal day 40, the propensity of the offspring to epilepsy was examined using hippocampal kindling; autismlike behavior was studied using the sociability test. The involvement of IL-6 and IL-1β in PIC-induced effects was studied by the coadministration of the cytokine antibodies with PIC, and by delivering recombinant cytokines in lieu of PIC. RESULTS The offspring of PIC-exposed mice exhibited increased hippocampal excitability, accelerated kindling rate, prolonged increase of seizure susceptibility after kindling, and diminished sociability. Epileptic impairments were abolished by antibodies to IL-6 or IL-1β. Neither of the recombinant cytokines alone increased the propensity to seizures; however, when combined, they produced effects similar to those induced by PIC. PIC-induced behavioral deficits were abolished by IL-6 antibodies and were mimicked by recombinant IL-6; IL-1β was not involved. INTERPRETATION In addition to confirming the previously established critical role of IL-6 in the development of autismlike behavior following MIA, the present study shows that concurrent involvement of IL-6 and IL-1β is required for priming the offspring for epilepsy. These data shed light on mechanisms of comorbidity between autism and epilepsy.
Collapse
Affiliation(s)
- Eduardo Pineda
- Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | | | | | | | | | | |
Collapse
|
40
|
Essa MM, Subash S, Braidy N, Al-Adawi S, Lim CK, Manivasagam T, Guillemin GJ. Role of NAD(+), Oxidative Stress, and Tryptophan Metabolism in Autism Spectrum Disorders. Int J Tryptophan Res 2013; 6:15-28. [PMID: 23922500 PMCID: PMC3729335 DOI: 10.4137/ijtr.s11355] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a pervasive neuro-developmental disorder characterized by impaired social interaction, reduced/absent verbal and non-verbal communication, and repetitive behavior during early childhood. The etiology of this developmental disorder is poorly understood, and no biomarkers have been identified. Identification of novel biochemical markers related to autism would be advantageous for earlier clinical diagnosis and intervention. Studies suggest that oxidative stress-induced mechanisms and reduced antioxidant defense, mitochondrial dysfunction, and impaired energy metabolism (NAD(+), NADH, ATP, pyruvate, and lactate), are major causes of ASD. This review provides renewed insight regarding current autism research related to oxidative stress, mitochondrial dysfunction, and altered tryptophan metabolism in ASD.
Collapse
Affiliation(s)
- Musthafa Mohamed Essa
- Dept of Food Science and Nutrition, College of Agriculture and Marine Sciences, Sultan Qaboos University, Oman. ; School of Medical Sciences, Department of Pharmacology, Faculty of Medicine, University of NSW, Sydney, Australia
| | | | | | | | | | | | | |
Collapse
|
41
|
Siniscalco D, Bradstreet JJ, Antonucci N. Therapeutic role of hematopoietic stem cells in autism spectrum disorder-related inflammation. Front Immunol 2013; 4:140. [PMID: 23772227 PMCID: PMC3677147 DOI: 10.3389/fimmu.2013.00140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/26/2013] [Indexed: 12/24/2022] Open
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous, severe neuro-developmental disorders with core symptoms of dysfunctions in social interactions and communication skills, restricted interests, repetitive – stereotypic verbal and non-verbal behaviors. Biomolecular evidence points to complex gene-environmental interactions in ASDs. Several biochemical processes are associated with ASDs: oxidative stress (including endoplasmic reticulum stress), decreased methylation capacity, limited production of glutathione; mitochondrial dysfunction, intestinal dysbiosis, increased toxic metal burden, and various immune abnormalities. The known immunological disorders include: T-lymphocyte populations and function, gene expression changes in monocytes, several autoimmune-related findings, high levels of N-acetylgalactosaminidase (which precludes macrophage activation), and primary immune deficiencies. These immunological observations may result in minicolumn structural changes in the brain, as well as, abnormal immune mediation of synaptic functions. Equally, these immune dysregulations serve as the rationale for immune-directed interventions such as hematopoietic stem cells (HSCs), which are pivotal in controlling chronic inflammation and in the restoration of immunological balance. These properties make them intriguing potential agents for ASD treatments. This prospective review will focus on the current state-of-the-art knowledge and challenges intrinsic in the application of HSCs for ASD-related immunological disorders.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Second University of Naples , Naples , Italy ; Centre for Autism - La Forza del Silenzio , Caserta , Italy ; Cancellautismo , Florence , Italy
| | | | | |
Collapse
|
42
|
Altered cytokine and BDNF levels in autism spectrum disorder. Neurotox Res 2013; 24:491-501. [PMID: 23604965 DOI: 10.1007/s12640-013-9393-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 01/08/2023]
Abstract
The contribution of neuroimmune functioning and brain-derived neurotrophic factor (BDNF) to functional dysregulation in autism spectrum disorder was assessed in 29 patients under treatment in two specialized centers of Basilicata (Chiaromonte and Matera), Southern Italy, through analysis of serum levels of cytokines and BDNF. Elevated levels of the pro-inflammatory cytokine, including interleukin-1, interleukin-6, interleukin-12, interleukin-23, tumor necrosis factor-α and BDNF were observed, regardless of age and gender. Comparisons were made with age- and gender-related healthy controls. The present findings reinforce current notions regarding immunoexcitotoxic mechanisms contributing to the pathophysiology of autistic disorder.
Collapse
|
43
|
Sato K. Placenta-derived hypo-serotonin situations in the developing forebrain cause autism. Med Hypotheses 2013; 80:368-72. [PMID: 23375670 DOI: 10.1016/j.mehy.2013.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/13/2012] [Accepted: 01/03/2013] [Indexed: 11/16/2022]
Abstract
Autism is a pervasive developmental disorder that is characterized by the behavioral traits of impaired social cognition and communication, and repetitive and/or obsessive behavior and interests. Although there are many theories and speculations about the pathogenetic causes of autism, the disruption of the serotonergic system is one of the most consistent and well-replicated findings. Recently, it has been reported that placenta-derived serotonin is the main source in embryonic day (E) 10-15 mouse forebrain, after that period, the serotonergic fibers start to supply serotonin into the forebrain. E 10-15 is the very important developing period, when cortical neurogenesis, migration and initial axon targeting are processed. Since all these events have been considered to be involved in the pathogenesis of autism and they are highly controlled by serotonin signals, the paucity of placenta-derived serotonin should have potential importance when the pathogenesis of autism is considered. I, thus, postulate a hypothesis that placenta-derived hypo-serotonin situations in the developing forebrain cause autism. The hypothesis is as follows. Various factors, such as inflammation, dysfunction of the placenta, together with genetic predispositions cause a decrease of placenta-derived serotonin levels. The decrease of placenta-derived serotonin levels leads to hypo-serotonergic situations in the forebrain of the fetus. The paucity of serotonin in the forebrain leads to mis-wiring in important regions which are responsible for the theory of mind. The paucity of serotonin in the forebrain also causes over-growth of serotonergic fibers. These disturbances result in network deficiency and aberration of the serotonergic system, leading to the autistic phenotypes.
Collapse
Affiliation(s)
- Kohji Sato
- Department of Anatomy & Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
44
|
Thiazolidine-2,4-diones: progress towards multifarious applications. Bioorg Med Chem 2013; 21:1599-620. [PMID: 23419324 DOI: 10.1016/j.bmc.2013.01.029] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 11/24/2022]
Abstract
The promising activity shown by compounds containing thiazolidine-2,4-dione nucleus in numerous categories such as anti-hyperglycaemics, aldose reductase inhibitors, anti-cancer, anti-inflammatory, anti-arthritics, anti-microbials, etc. has made it an indispensable anchor for development of new therapeutic agents. Varied substituents on the thiazolidine-2,4-dione nucleus have provided a wide spectrum of biological activities. Importance of this nucleus in some activities like, peroxisome proliferator activated receptor γ (PPARγ) agonism and PPARγ-dependent and -independent anti-cancer activities are reviewed separately in literature. Short reviews on biological importance of this nucleus are also known in literature. However, owing to fast development of new drugs possessing thiazolidine-2,4-dione nucleus many research reports are generated in short span of time. So, there is a need to couple the latest information with the earlier information to understand the current status of thiazolidine-2,4-dione nucleus in medicinal chemistry research. In the present review, various derivatives of thiazolidine-2,4-diones with different pharmacological activities are described on the basis of substitution pattern around the nucleus combined with the docking studies performed in the active site of the corresponding receptors with an aim to help medicinal chemists for developing an SAR on thiazolidine-2,4-dione derived compounds for each activity. This discussion will further help in the development of novel thiazolidine-2,4-dione compounds.
Collapse
|
45
|
Nousen EK, Franco JG, Sullivan EL. Unraveling the mechanisms responsible for the comorbidity between metabolic syndrome and mental health disorders. Neuroendocrinology 2013; 98:254-66. [PMID: 24080959 PMCID: PMC4121390 DOI: 10.1159/000355632] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/10/2013] [Indexed: 12/25/2022]
Abstract
The increased prevalence and high comorbidity of metabolic syndrome (MetS) and mental health disorders (MHDs) have prompted investigation into the potential contributing mechanisms. There is a bidirectional association between MetS and MHDs including schizophrenia, bipolar disorder, depression, anxiety, attention-deficit/hyperactivity disorder, and autism spectrum disorders. Medication side effects and social repercussions are contributing environmental factors, but there are a number of shared underlying neurological and physiological mechanisms that explain the high comorbidity between these two disorders. Inflammation is a state shared by both disorders, and it contributes to disruptions of neuroregulatory systems (including the serotonergic, dopaminergic, and neuropeptide Y systems) as well as dysregulation of the hypothalamic-pituitary-adrenal axis. MetS in pregnant women also exposes the developing fetal brain to inflammatory factors that predispose the offspring to MetS and psychopathologies. Due to the shared nature of these conditions, treatment should address aspects of both mental health and metabolic disorders. Additionally, interventions that can interrupt the transfer of increased risk of the disorders to the next generation need to be developed. © 2013 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Elizabeth K. Nousen
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Juliana G. Franco
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Elinor L. Sullivan
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR, USA
- Department of Biology, University of Portland, Portland, OR, USA
| |
Collapse
|
46
|
Sullivan EL, Nousen EK, Chamlou KA. Maternal high fat diet consumption during the perinatal period programs offspring behavior. Physiol Behav 2012; 123:236-42. [PMID: 23085399 DOI: 10.1016/j.physbeh.2012.07.014] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 07/20/2012] [Accepted: 07/30/2012] [Indexed: 12/16/2022]
Abstract
The environment that a developing offspring experiences during the perinatal period is markedly influenced by maternal health and diet composition. Evidence from both epidemiological studies and animal models indicates that maternal diet and metabolic status play a critical role in programming the neural circuitry that regulates behavior, resulting in long-term consequences for offspring behavior. Maternal diet and metabolic state influence the behavior of offspring directly by impacting the intrauterine environment and indirectly by modulating maternal behavior. The mechanisms by which maternal diet and metabolic profile shape the perinatal environment remain largely unknown, but recent research has found that increases in inflammatory cytokines, nutrients (glucose and fatty acids), and hormones (insulin and leptin) affect the environment of the developing offspring. Offspring exposed to maternal obesity and high fat diet consumption during development are more susceptible to developing mental health and behavioral disorders such as anxiety, depression, attention deficit hyperactivity disorder, and autism spectrum disorders. Recent evidence suggests that this increased risk for behavioral disorders is driven by modifications in the development of neural pathways involved in behavioral regulation. In particular, research indicates that the development of the serotonergic system is impacted by exposure to maternal obesity and high fat diet consumption, and this disruption may underlie many of the behavioral disturbances observed in these offspring. Given the high rates of obesity and high fat diet consumption in pregnant women, it is vital to examine the influence that maternal nutrition and metabolic profile have on the developing offspring.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Department of Biology, University of Portland, Portland, OR, USA; Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA.
| | | | | |
Collapse
|
47
|
Boekelheide K, Blumberg B, Chapin RE, Cote I, Graziano JH, Janesick A, Lane R, Lillycrop K, Myatt L, States JC, Thayer KA, Waalkes MP, Rogers JM. Predicting later-life outcomes of early-life exposures. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:1353-61. [PMID: 22672778 PMCID: PMC3491941 DOI: 10.1289/ehp.1204934] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 06/06/2012] [Indexed: 05/17/2023]
Abstract
BACKGROUND In utero exposure of the fetus to a stressor can lead to disease in later life. Epigenetic mechanisms are likely mediators of later-life expression of early-life events. OBJECTIVES We examined the current state of understanding of later-life diseases resulting from early-life exposures in order to identify in utero and postnatal indicators of later-life diseases, develop an agenda for future research, and consider the risk assessment implications of this emerging knowledge. METHODS This review was developed based on our participation in a National Research Council workshop titled "Use of in Utero and Postnatal Indicators to Predict Health Outcomes Later in Life: State of the Science and Research Recommendations." We used a case study approach to highlight the later-life consequences of early-life malnutrition and arsenic exposure. DISCUSSION The environmental sensitivity of the epigenome is viewed as an adaptive mechanism by which the developing organism adjusts its metabolic and homeostatic systems to suit the anticipated extrauterine environment. Inappropriate adaptation may produce a mismatch resulting in subsequent increased susceptibility to disease. A nutritional mismatch between the prenatal and postnatal environments, or early-life obesogen exposure, may explain at least some of the recent rapid increases in the rates of obesity, type 2 diabetes, and cardiovascular diseases. Early-life arsenic exposure is also associated with later-life diseases, including cardiovascular disease and cancer. CONCLUSIONS With mounting evidence connecting early-life exposures and later-life disease, new strategies are needed to incorporate this emerging knowledge into health protective practices.
Collapse
Affiliation(s)
- Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Division of Biology and Medicine, Brown University, Providence, Rhode Island 02912, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wei H, Mori S, Hua K, Li X. Alteration of brain volume in IL‐6 overexpressing mice related to autism. Int J Dev Neurosci 2012; 30:554-9. [DOI: 10.1016/j.ijdevneu.2012.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/17/2012] [Accepted: 08/17/2012] [Indexed: 10/28/2022] Open
Affiliation(s)
- Hongen Wei
- Central LaboratoryShanxi Provincial People's HospitalTaiyuanChina
| | - Susumu Mori
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kegang Hua
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Xiaohong Li
- Department of NeurochemistryNY State Institute for Basic Research in Developmental DisabilitiesNew YorkNYUSA
| |
Collapse
|
49
|
Neonatal levels of cytokines and risk of autism spectrum disorders: an exploratory register-based historic birth cohort study utilizing the Danish Newborn Screening Biobank. J Neuroimmunol 2012; 252:75-82. [PMID: 22917523 DOI: 10.1016/j.jneuroim.2012.07.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 07/21/2012] [Accepted: 07/23/2012] [Indexed: 01/07/2023]
Abstract
The aim of the study was to analyze cytokine profiles in neonatal dried blood samples (n-DBSS) retrieved from The Danish Newborn Screening Biobank of children developing Autism Spectrum Disorders (ASD) later in life and controls. Samples of 359 ASD cases and 741 controls were analyzed using Luminex xMAP technology and clinical data were retrieved from nationwide registers. Findings showed that children developing ASD were more likely to have decreased levels of both T helper-1(Th-1)-like cytokines (i.e. IFN-γ) and Th-2like cytokines (i.e. IL-4, IL-10) which may suggest a depressed or hypoactive immune cell activity during neonatal period in ASD.
Collapse
|
50
|
Abstract
Microglia are enigmatic non-neuronal cells that infiltrate and take up residence in the brain during development and are thought to perform a surveillance function. An established literature has documented how microglia are activated by pathogenic stimuli and how they contribute to and resolve injuries to the brain. However, much less work has been aimed at understanding their function in the uninjured brain. A series of recent in vivo imaging studies shows that microglia in their resting state are highly motile and actively survey their neuronal surroundings. Furthermore, new data suggest that microglia in their resting state are able to phagocytose unwanted synapses and in this way contribute to synaptic pruning and maturation during development. Coupled with their exquisite sensitivity to pathogenic stimuli, these data suggest that microglia form a link that couples changes in brain environment to changes in brain wiring. Here we discuss this hypothesis and propose a model for the role of microglia during development in sculpting brain connectivity.
Collapse
|