1
|
Adıgüzel Akman Ö, Esnafoglu E. Evaluation of procalcitonin and C-reactive protein levels in children with autism spectrum disorder and attention deficit hyperactivity disorder. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2023; 71:159-167. [PMID: 39882415 PMCID: PMC11774147 DOI: 10.1080/20473869.2023.2213923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 01/31/2025]
Abstract
Inflammation is thought to play a role in the etiopathogenesis of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). Procalcitonin (PCT) is an inflammatory biomarker released by thyroid parafollicular cells. CRP is systemic biomarker of inflammation. In this study, the hypothesis that PCT and C-reactive protein (CRP) might be indicators of increased inflammatory response was tested. For this purpose, PCT and CRP as well as other simple inflammatory markers were evaluated in children with ASD and ADHD. A total of 33 ASD, 36 ADHD, and 31 healthy controls were included in the study, who were similar in terms of gender and age. PCT, CRP, sedimentation and hemogram parameters (Neutrophil Lymphocyte Ratio) were measured in children aged 5-15 years. PCT and CRP values were found to be statistically significantly higher in ASD and ADHD children compared to healthy controls (p < 0.05). A positive correlation was found between PCT and CRP values (r: 0.358; p < 0.001). No significant correlations were found between disorder severity scale scores and PCT and CRP values (p > 0.05 for both). Accordingly, it is suggested that inflammation plays a role in the etiopathogenesis of these disorders. PCT and CRP may be used as inflammation markers.
Collapse
Affiliation(s)
- Öznur Adıgüzel Akman
- Department of Child and Adolescent Psychiatry, Black Sea Ereğli State Hospital, Zonguldak, Turkey
| | - Erman Esnafoglu
- Department of Child and Adolescent Psychiatry, Ordu University Faculty of Medicine, Ordu, Turkey
| |
Collapse
|
2
|
Shamim S, Khan N, Greene DL, Habiba UE, Umer A. The promise of autologous and allogeneic cellular therapies in the clinical trials of autism spectrum disorder. Regen Med 2023; 18:347-361. [PMID: 36935631 DOI: 10.2217/rme-2022-0176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/02/2023] [Indexed: 03/21/2023] Open
Abstract
Autism spectrum disorder (ASD) is a consortium of developmental conditions. As scientists have not yet identified the exact underlying cause for these disorders, it is not easy to narrow down a singular therapy to propose a reliable cure. The preponderance of research suggests that stem-cell therapy improves aspects of outcome measure scales in patients with ASD; therefore, future studies should give us more confidence in the results. This overview considers the data that have emerged from the small set of published trials conducted using different approaches in stem-cell therapy for ASD, evaluates their results and proposes additional steps that could be taken if this field of endeavor is to be pursued further.
Collapse
Affiliation(s)
- Sabiha Shamim
- Bello Bio Labs & Therapeutics (SMC) Pvt. Ltd, Jahangir Multiplex, Peshawar Road, Sector H-13 Islamabad, 44000, Pakistan
| | - Nasar Khan
- Bello Bio Labs & Therapeutics (SMC) Pvt. Ltd, Jahangir Multiplex, Peshawar Road, Sector H-13 Islamabad, 44000, Pakistan
- R3 Medical Research LLC, 10045 East Dynamite Boulevard Suite 260, Scottsdale, AZ 85262, United States of America
| | - David L Greene
- Bello Bio Labs & Therapeutics (SMC) Pvt. Ltd, Jahangir Multiplex, Peshawar Road, Sector H-13 Islamabad, 44000, Pakistan
- R3 Medical Research LLC, 10045 East Dynamite Boulevard Suite 260, Scottsdale, AZ 85262, United States of America
| | - Umm E Habiba
- Bello Bio Labs & Therapeutics (SMC) Pvt. Ltd, Jahangir Multiplex, Peshawar Road, Sector H-13 Islamabad, 44000, Pakistan
| | - Amna Umer
- Bello Bio Labs & Therapeutics (SMC) Pvt. Ltd, Jahangir Multiplex, Peshawar Road, Sector H-13 Islamabad, 44000, Pakistan
| |
Collapse
|
3
|
Zeng J, Liang Y, Sun R, Huang S, Wang Z, Xiao L, Lu J, Yu H, Yao P. Hematopoietic stem cell transplantation ameliorates maternal diabetes–mediated gastrointestinal symptoms and autism‐like behavior in mouse offspring. Ann N Y Acad Sci 2022; 1512:98-113. [PMID: 35220596 PMCID: PMC9307016 DOI: 10.1111/nyas.14766] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/16/2022] [Indexed: 01/16/2023]
Abstract
Epidemiological studies have shown that maternal diabetes is associated with autism spectrum disorder development, although the detailed mechanism remains unclear. We have previously found that maternal diabetes induces persistent epigenetic changes and gene suppression in neurons, subsequently triggering autism‐like behavior (ALB). In this study, we investigated the potential role and effect of hematopoietic stem cells (HSCs) on maternal diabetes–mediated gastrointestinal (GI) dysfunction and ALB in a mouse model. We show in vitro that transient hyperglycemia induced persistent epigenetic changes and gene suppression of tight junction proteins. In vivo, maternal diabetes–mediated oxidative stress induced gene suppression and inflammation in both peripheral blood mononuclear cells and intestine epithelial cells, subsequently triggering GI dysfunction with increased intestinal permeability and altered microbiota compositions, as well as suppressed gene expression in neurons and subsequent ALB in offspring; HSC transplantation (HSCT) ameliorates this effect by systematically reversing maternal diabetes–mediated oxidative stress. We conclude that HSCT can ameliorate maternal diabetes–mediated GI symptoms and autism‐like behavior in mouse offspring.
Collapse
Affiliation(s)
- Jiaying Zeng
- Department of Child HealthCare, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
| | - Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen Shenzhen Mental Health Center Shenzhen P. R. China
| | - Ruoyu Sun
- Department of Child HealthCare, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
| | - Saijun Huang
- Department of Child HealthCare, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen Shenzhen Mental Health Center Shenzhen P. R. China
| | - Li Xiao
- Department of Pediatrics, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
| | - Jianpin Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen Shenzhen Mental Health Center Shenzhen P. R. China
| | - Hong Yu
- Department of Child HealthCare, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
| | - Paul Yao
- Department of Child HealthCare, Affiliated Foshan Maternity & Child Healthcare Hospital The Second School of Clinical Medicine of Southern Medical University Foshan P. R. China
- Department of Child Psychiatry, Kangning Hospital of Shenzhen Shenzhen Mental Health Center Shenzhen P. R. China
| |
Collapse
|
4
|
Nguyen Thanh L, Nguyen H, Duy Ngo M, Bui VA, Dam PT, Thi Phuong Bui H, Van Ngo D, Tran KT, Thi Thanh Dang T, Duc Duong B, Anh Thi Nguyen P, Forstyth N, Heke M. In Reply. Stem Cells Transl Med 2021; 10:827-828. [PMID: 34010520 PMCID: PMC8133348 DOI: 10.1002/sctm.20-0452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/11/2020] [Accepted: 12/07/2020] [Indexed: 11/10/2022] Open
Affiliation(s)
- Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG)HanoiVietnam
| | - Hoang‐Phuong Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG)HanoiVietnam
| | - Minh Duy Ngo
- Vinmec Times City International HospitalHanoiVietnam
| | - Viet Anh Bui
- Vinmec Hightech Center, Vinmec Health Care SystemHanoiVietnam
| | - Phuong T.M. Dam
- Vinmec Hightech Center, Vinmec Health Care SystemHanoiVietnam
| | | | - Doan Van Ngo
- Vinmec Times City International HospitalHanoiVietnam
| | - Kien Trung Tran
- Vinmec Research Institute of Stem Cell and Gene Technology (VRISG)HanoiVietnam
| | | | | | | | | | - Michael Heke
- Department of BiologyStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
5
|
Healing autism spectrum disorder with cannabinoids: a neuroinflammatory story. Neurosci Biobehav Rev 2020; 121:128-143. [PMID: 33358985 DOI: 10.1016/j.neubiorev.2020.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/28/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with a multifactorial etiology. Latest researches are raising the hypothesis of a link between the onset of the main behavioral symptoms of ASD and the chronic neuroinflammatory condition of the autistic brain; increasing evidence of this connection is shedding light on new possible players in the pathogenesis of ASD. The endocannabinoid system (ECS) has a key role in neurodevelopment as well as in normal inflammatory responses and it is not surprising that many preclinical and clinical studies account for alterations of the endocannabinoid signaling in ASD. These findings lay the foundation for a better understanding of the neurochemical mechanisms underlying ASD and for new therapeutic attempts aimed at exploiting the renowned anti-inflammatory properties of cannabinoids to treat pathologies encompassed in the autistic spectrum. This review discusses the current preclinical and clinical evidence supporting a key role of the ECS in the neuroinflammatory state that characterizes ASD, providing hints to identify new biomarkers in ASD and promising therapies for the future.
Collapse
|
6
|
Larijani B, Foroughi Heravani N, Alavi-Moghadam S, Goodarzi P, Rezaei-Tavirani M, Payab M, Gholami M, Razi F, Arjmand B. Cell Therapy Targets for Autism Spectrum Disorders: Hopes, Challenges and Future Directions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1341:107-124. [PMID: 32072476 DOI: 10.1007/5584_2020_491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorders as a group of pediatric neurodevelopmental diseases is a crucial part of the worldwide disabilities which have influence in communication skills, social interactions, and ability to understand the concepts. The precise pathophysiology of autism spectrum disorders due to the abundance of involved mechanisms is unknown. Some of these involved mechanisms are related to genetic factors, chronic neuro inflammation, mitochondrial dysfunction, oxidative stress, immune dysregulation, hormonal imbalance, and environmental factors. Current main treatments for autisms are behavioral, nutritional and medical therapies, however there is not definitive treatment approach. Therein, more novel therapies are still required to improve the symptoms. Several preclinical and clinical evidence were shown that stem cell therapy is a potential treatment option for autism spectrum disorders individuals. Considering the significant factors which can affect the outcome of stem cell therapeutic effects including stem cell types, route and dosage of administration, and mechanism of activity along with selecting best animal models can be very important in performing clinical trials.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Foroughi Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy; Toxicology and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Siences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Lu J, Xiao M, Guo X, Liang Y, Wang M, Xu J, Liu L, Wang Z, Zeng G, Liu K, Li L, Yao P. Maternal Diabetes Induces Immune Dysfunction in Autistic Offspring Through Oxidative Stress in Hematopoietic Stem Cells. Front Psychiatry 2020; 11:576367. [PMID: 33101089 PMCID: PMC7495463 DOI: 10.3389/fpsyt.2020.576367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorders (ASD) have been found to be associated with immune dysfunction and elevated cytokines, although the detailed mechanism remains unknown. In this study, we aim to investigate the potential mechanisms through a maternal diabetes-induced autistic mouse model. We found that maternal diabetes-induced autistic offspring have epigenetic changes on the superoxide dismutase 2 (SOD2) promoter with subsequent SOD2 suppression in both hematopoietic stem cells (HSC) and peripheral blood mononuclear cells (PBMC). Bone marrow transplantation of normal HSC to maternal diabetes-induced autistic offspring transferred epigenetic modifications to PBMC and significantly reversed SOD2 suppression and oxidative stress and elevated inflammatory cytokine levels. Further, in vivo human study showed that SOD2 mRNA expression from PBMC in the ASD group was reduced to ~12% compared to typically developing group, and the SOD2 mRNA level-based ROC (Receiver Operating Characteristic) curve shows a very high sensitivity and specificity for ASD patients. We conclude that maternal diabetes induces immune dysfunction in autistic offspring through SOD2 suppression and oxidative stress in HSC. SOD2 mRNA expression in PBMC may be a good biomarker for ASD diagnosis.
Collapse
Affiliation(s)
- Jianping Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Meifang Xiao
- Hainan Women and Children's Medical Center, Haikou, China
| | - Xiaoling Guo
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, China
| | - Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Min Wang
- Hainan Women and Children's Medical Center, Haikou, China
| | - Jianchang Xu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Liyan Liu
- Hainan Women and Children's Medical Center, Haikou, China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Gang Zeng
- Hainan Women and Children's Medical Center, Haikou, China
| | - Kelly Liu
- Hainan Women and Children's Medical Center, Haikou, China
| | - Ling Li
- Hainan Women and Children's Medical Center, Haikou, China
| | - Paul Yao
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China.,Hainan Women and Children's Medical Center, Haikou, China
| |
Collapse
|
8
|
Pharmacological, non-pharmacological and stem cell therapies for the management of autism spectrum disorders: A focus on human studies. Pharmacol Res 2019; 152:104579. [PMID: 31790820 DOI: 10.1016/j.phrs.2019.104579] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/13/2019] [Accepted: 11/27/2019] [Indexed: 01/03/2023]
Abstract
In the last decade, the prevalence of autism spectrum disorders (ASD) has dramatically escalated worldwide. Currently available drugs mainly target some co-occurring symptoms of ASD, but are not effective on the core symptoms, namely impairments in communication and social interaction, and the presence of restricted and repetitive behaviors. On the other hand, transplantation of hematopoietic and mesenchymal stem cells in ASD children has been shown promising to stimulate the recruitment, proliferation, and differentiation of tissue-residing native stem cells, reducing inflammation, and improving some ASD symptoms. Moreover, several comorbidities have also been associated with ASD, such as immune dysregulation, gastrointestinal issues and gut microbiota dysbiosis. Non-pharmacological approaches, such as dietary supplementations with certain vitamins, omega-3 polyunsaturated fatty acids, probiotics, some phytochemicals (e.g., luteolin and sulforaphane), or overall diet interventions (e.g., gluten free and casein free diets) have been considered for the reduction of such comorbidities and the management of ASD. Here, interventional studies describing pharmacological and non-pharmacological treatments in ASD children and adolescents, along with stem cell-based therapies, are reviewed.
Collapse
|
9
|
Siniscalco D, Kannan S, Semprún-Hernández N, Eshraghi AA, Brigida AL, Antonucci N. Stem cell therapy in autism: recent insights. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:55-67. [PMID: 30425534 PMCID: PMC6204871 DOI: 10.2147/sccaa.s155410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorders (ASDs) are characterized by core domains: persistent deficits in social communication and interaction; restricted, repetitive patterns of behavior, interests, or activities. ASDs comprise heterogeneous and complex neurodevelopmental pathologies with well-defined inflammatory conditions and immune system dysfunction. Due to neurobiologic changes underlying ASD development, cell-based therapies have been proposed and applied to ASDs. Indeed, stem cells show specific immunologic properties, which make them promising candidates in ASD treatment. This comprehensive up-to-date review focuses on ASD cellular/molecular abnormalities, potentially useful stem cell types, animal models, and current clinical trials on the use of stem cells in treating autism. Limitations are also discussed.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, University of Campania, Napoli, Italy,
| | - Suresh Kannan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Neomar Semprún-Hernández
- Research Division, Autism Immunology Unit of Maracaibo, Catedra libre de Autismo, Universidad del Zulia, Maracaibo, Venezuela
| | - Adrien A Eshraghi
- Department of Otolaryngology, Hearing Research and Cochlear Implant Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Nicola Antonucci
- Biomedical Centre for Autism Research and Treatment, Bari, Italy
| |
Collapse
|
10
|
Siniscalco D, Schultz S, Brigida AL, Antonucci N. Inflammation and Neuro-Immune Dysregulations in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2018; 11:56. [PMID: 29867038 PMCID: PMC6027314 DOI: 10.3390/ph11020056] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 02/07/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is characterized by persistent deficits in social communication and interaction and restricted-repetitive patterns of behavior, interests, or activities. Strong inflammation states are associated with ASD. This inflammatory condition is often linked to immune system dysfunction. Several cell types are enrolled to trigger and sustain these processes. Neuro-inflammation and neuro-immune abnormalities have now been established in ASD as key factors in its development and maintenance. In this review, we will explore inflammatory conditions, dysfunctions in neuro-immune cross-talk, and immune system treatments in ASD management.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, University of Campania, 80138 Naples, Italy.
- Italian Group for Study Autism-GISA, 25018 Brescia, Italy.
- Centre for Autism-La Forza del Silenzio, 81036 Caserta, Italy.
| | - Stephen Schultz
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA.
| | | | - Nicola Antonucci
- Biomedical Centre for Autism Research and Treatment, 70124 Bari, Italy.
| |
Collapse
|
11
|
Ahmad SF, Nadeem A, Ansari MA, Bakheet SA, Al-Ayadhi LY, Attia SM. Upregulation of IL-9 and JAK-STAT signaling pathway in children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2017; 79:472-480. [PMID: 28802860 DOI: 10.1016/j.pnpbp.2017.08.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
Autism spectrum disorder (ASD) gradually develops predominantly neurodevelopmental disorders, which are socially diagnosed in early childhood. Though the etiopathology of ASD is not clear, immune alteration has been suggested as autism's pathophysiological mechanism. Previous studies found that several cytokines and transcription factor activation pathways were significantly increased in ASD. IL-9 has been confirmed to play a significant role in the central nervous system (CNS). The aim of the present study was to investigate the understudied role of pro- and anti-inflammatory cytokines and the JAK-STAT signaling pathway in ASD. We examined the IL-1β, IL-4, IFN-γ, and IL-9 positive immunostaining in all cells, and CD4+ T cells, in ASD and normally developing control children (TD), on peripheral blood mononuclear cells (PBMCs), using flow cytometry. We explored PBMC mRNA expression levels for IL-1β, IL-4, IFN-γ, IL-9, JAK1, and STAT5, by using real-time PCR (RT-PCR). We also explored PBMC protein expression levels for IL-1β, IL-4, IL-9, pJAK1, and pSTAT5 by using western blotting. We found that the children with ASD had increased IL-1β, IL-4, IFN-γ, and IL-9 positive immunostaining in all cells, and in CD4+ cells, relative to the TD controls. The mRNA and protein expression for IL-1β, IL-4, IFN-γ, IL-9, JAK1, pJAK1, STAT5, and pSTAT5 were also significantly elevated in ASD relative to TD controls. These results suggested that cytokines and JAK-STAT activation signaling have an essential role in immune dysfunction in ASD.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Laila Yousef Al-Ayadhi
- Autism Research and Treatment Center, AL-Amodi Autism Research Chair, Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
12
|
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]
|
13
|
Feng C, Chen Y, Pan J, Yang A, Niu L, Min J, Meng X, Liao L, Zhang K, Shen L. Redox proteomic identification of carbonylated proteins in autism plasma: insight into oxidative stress and its related biomarkers in autism. Clin Proteomics 2017; 14:2. [PMID: 28077936 PMCID: PMC5223466 DOI: 10.1186/s12014-017-9138-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/03/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Autism is a severe childhood neurological disorder with poorly understood etiology and pathology. Currently, there is no authentic laboratory test to confirm the diagnosis of autism. Oxidative damage may play a central role in the pathogenesis of autism. Present study is an effort to search for possible biomarkers of autism and further clarify the molecular changes associated with oxidative stress that occurs in the plasma of autistic children. METHODS We performed redox proteomics analysis to compare carbonylated proteins in the plasma of autistic subjects and healthy controls. Immunoprecipitation and Western blot analysis were used to validate carbonylated proteins identified by the redox proteomics. RESULTS Protein carbonylation levels in two proteins, complement component C8 alpha chain and Ig kappa chain C were found to be significantly increased in autistic patients compared with controls. These two proteins were successfully validated via immunoprecipitation and Western blot analysis. CONCLUSIONS The results further highlight the role of oxidative stress in the pathogenesis of autism and provide some information for the diagnosis and/or monitoring of autism.
Collapse
Affiliation(s)
- Chengyun Feng
- Early Childhood Development Center, Populations and Family Planning Hospital of Baoan, Shenzhen, 518101 People's Republic of China
| | - Youjiao Chen
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060 People's Republic of China
| | - Jintao Pan
- Early Childhood Development Center, Populations and Family Planning Hospital of Baoan, Shenzhen, 518101 People's Republic of China
| | - Aochu Yang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060 People's Republic of China
| | - Li Niu
- Early Childhood Development Center, Populations and Family Planning Hospital of Baoan, Shenzhen, 518101 People's Republic of China
| | - Jie Min
- Early Childhood Development Center, Populations and Family Planning Hospital of Baoan, Shenzhen, 518101 People's Republic of China
| | - Xianling Meng
- Early Childhood Development Center, Populations and Family Planning Hospital of Baoan, Shenzhen, 518101 People's Republic of China
| | - Liping Liao
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060 People's Republic of China
| | - Kaoyuan Zhang
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060 People's Republic of China
| | - Liming Shen
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060 People's Republic of China
| |
Collapse
|
14
|
Ardhanareeswaran K, Coppola G, Vaccarino F. The use of stem cells to study autism spectrum disorder. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2015; 88:5-16. [PMID: 25745370 PMCID: PMC4345539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autism spectrum disorder (ASD) affects as many as 1 in 68 children and is said to be the fastest-growing serious developmental disability in the United States. There is currently no medical cure or diagnostic test for ASD. Furthermore, the U.S. Food and Drug Administration has yet to approve a single drug for the treatment of autism's core symptoms. Despite numerous genome studies and the identification of hundreds of genes that may cause or predispose children to ASD, the pathways underlying the pathogenesis of idiopathic ASD still remain elusive. Post-mortem brain samples, apart from being difficult to obtain, offer little insight into a disorder that arises through the course of development. Furthermore, ASD is a disorder of highly complex, human-specific behaviors, making it difficult to model in animals. Stem cell models of ASD can be generated by performing skin biopsies of ASD patients and then dedifferentiating these fibroblasts into human-induced pluripotent stem cells (hiPSCs). iPSCs closely resemble embryonic stem cells and retain the unique genetic signature of the ASD patient from whom they were originally derived. Differentiation of these iPSCs into neurons essentially recapitulates the ASD patient's neuronal development in a dish, allowing for a patient-specific model of ASD. Here we review our current understanding of the underlying neurobiology of ASD and how the use of stem cells can advance this understanding, possibly leading to new therapeutic avenues.
Collapse
Affiliation(s)
- Karthikeyan Ardhanareeswaran
- Child Study Center, Yale School of Medicine, New Haven, Connecticut,Program in Neurodevelopment and Regeneration, Yale School of Medicine, New Haven, Connecticut,Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut
| | - Gianfilippo Coppola
- Child Study Center, Yale School of Medicine, New Haven, Connecticut,Program in Neurodevelopment and Regeneration, Yale School of Medicine, New Haven, Connecticut
| | - Flora Vaccarino
- Child Study Center, Yale School of Medicine, New Haven, Connecticut,Program in Neurodevelopment and Regeneration, Yale School of Medicine, New Haven, Connecticut,Yale Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut,Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut,To whom all correspondence should be addressed: Flora Vaccarino MD, Yale Child Study Center, 230 South Frontage Rd, New Haven, CT 06520; Tele: 203-737-4147; Fax: 203-737-3524;
| |
Collapse
|
15
|
Siniscalco D. Commentary: The Impact of Neuroimmune Alterations in Autism Spectrum Disorder. Front Psychiatry 2015; 6:145. [PMID: 26500565 PMCID: PMC4597004 DOI: 10.3389/fpsyt.2015.00145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 09/22/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Second University of Naples , Naples , Italy ; Centre for Autism - La Forza del Silenzio , Caserta , Italy ; Cancellautismo - No Profit Association for Autism Care , Florence , Italy
| |
Collapse
|
16
|
Bradstreet JJ, Sych N, Antonucci N, Klunnik M, Ivankova O, Matyashchuk I, Demchuk M, Siniscalco D. Efficacy of fetal stem cell transplantation in autism spectrum disorders: an open-labeled pilot study. Cell Transplant 2014; 23 Suppl 1:S105-12. [PMID: 25302490 DOI: 10.3727/096368914x684916] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorders (ASDs) are heterogeneous complex neurodevelopmental pathologies defined by behavioral symptoms, but which have well-characterized genetic, immunological, and physiological comorbidities. Despite extensive research efforts, there are presently no agreed upon therapeutic approaches for either the core behaviors or the associated comorbidities. In particular, the known autoimmune disorders associated with autism are appealing targets for potential stem cell therapeutics. Of the various stem cell populations, fetal stem cells (FSCs) offer the potent immunoregulatory functions found in primordial mesenchymal stem cells, while exhibiting rapid expansion capacity and recognized plasticity. These properties enhance their potential for clinical use. Furthermore, FSCs are potent and implantable "biopharmacies" capable of delivering trophic signals to the host, which could influence brain development. This study investigated the safety and efficacy of FSC transplantations in treating children diagnosed with ASDs. Subjects were monitored at pre, and then 6 and 12 months following the transplantations, which consisted of two doses of intravenously and subcutaneously administered FSCs. The Autism Treatment Evaluation Checklist (ATEC) test and Aberrant Behavior Checklist (ABC) scores were performed. Laboratory examinations and clinical assessment of adverse effects were performed in order to evaluate treatment safety. No adverse events of significance were observed in ASD children treated with FSCs, including no transmitted infections or immunological complications. Statistically significant differences (p < 0.05) were shown on ATEC/ABC scores for the domains of speech, sociability, sensory, and overall health, as well as reductions in the total scores when compared to pretreatment values. We recognize that the use of FSCs remains controversial for the present. The results of this study, however, warrant additional investigations into the mechanisms of cell therapies for ASDs, while prompting the exploration of FSCs as "biopharmacies" capable of manufacturing the full array of cell-signaling chemistry. This manuscript is published as part of the International Association of Neurorestoratology (IANR) special issue of Cell Transplantation.
Collapse
|
17
|
Siniscalco D, Bradstreet JJ, Sych N, Antonucci N. Mesenchymal stem cells in treating autism: Novel insights. World J Stem Cells 2014; 6:173-178. [PMID: 24772244 PMCID: PMC3999775 DOI: 10.4252/wjsc.v6.i2.173] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/19/2013] [Accepted: 03/18/2014] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASDs) are complex neurodevelopmental disorders characterized by dysfunctions in social interactions, abnormal to absent verbal communication, restricted interests, and repetitive stereotypic verbal and non-verbal behaviors, influencing the ability to relate to and communicate. The core symptoms of ASDs concern the cognitive, emotional, and neurobehavioural domains. The prevalence of autism appears to be increasing at an alarming rate, yet there is a lack of effective and definitive pharmacological options. This has created an increased sense of urgency, and the need to identify novel therapies. Given the growing awareness of immune dysregulation in a significant portion of the autistic population, cell therapies have been proposed and applied to ASDs. In particular, mesenchymal stem cells (MSCs) possess the immunological properties which make them promising candidates in regenerative medicine. MSC therapy may be applicable to several diseases associated with inflammation and tissue damage, where subsequent regeneration and repair is necessary. MSCs could exert a positive effect in ASDs through the following mechanisms: stimulation of repair in the damaged tissue, e.g., inflammatory bowel disease; synthesizing and releasing anti-inflammatory cytokines and survival-promoting growth factors; integrating into existing neural and synaptic network, and restoring plasticity. The paracrine mechanisms of MSCs show interesting potential in ASD treatment. Promising and impressive results have been reported from the few clinical studies published to date, although the exact mechanisms of action of MSCs in ASDs to restore functions are still largely unknown. The potential role of MSCs in mediating ASD recovery is discussed in light of the newest findings from recent clinical studies.
Collapse
|
18
|
Siniscalco D, Bradstreet JJ, Cirillo A, Antonucci N. The in vitro GcMAF effects on endocannabinoid system transcriptionomics, receptor formation, and cell activity of autism-derived macrophages. J Neuroinflammation 2014; 11:78. [PMID: 24739187 PMCID: PMC3996516 DOI: 10.1186/1742-2094-11-78] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/02/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immune system dysregulation is well-recognized in autism and thought to be part of the etiology of this disorder. The endocannabinoid system is a key regulator of the immune system via the cannabinoid receptor type 2 (CB2R) which is highly expressed on macrophages and microglial cells. We have previously published significant differences in peripheral blood mononuclear cell CB2R gene expression in the autism population. The use of the Gc protein-derived Macrophage Activating Factor (GcMAF), an endogenous glycosylated vitamin D binding protein responsible for macrophage cell activation has demonstrated positive effects in the treatment of autistic children. In this current study, we investigated the in vitro effects of GcMAF treatment on the endocannabinoid system gene expression, as well as cellular activation in blood monocyte-derived macrophages (BMDMs) from autistic patients compared to age-matched healthy developing controls. METHODS To achieve these goals, we used biomolecular, biochemical and immunocytochemical methods. RESULTS GcMAF treatment was able to normalize the observed differences in dysregulated gene expression of the endocannabinoid system of the autism group. GcMAF also down-regulated the over-activation of BMDMs from autistic children. CONCLUSIONS This study presents the first observations of GcMAF effects on the transcriptionomics of the endocannabinoid system and expression of CB2R protein. These data point to a potential nexus between endocannabinoids, vitamin D and its transporter proteins, and the immune dysregulations observed with autism.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Second University of Naples, via S, Maria di Costantinopoli, 16 - 80138 Naples, Italy.
| | | | | | | |
Collapse
|
19
|
Noriega DB, Savelkoul HFJ. Immune dysregulation in autism spectrum disorder. Eur J Pediatr 2014; 173:33-43. [PMID: 24297668 DOI: 10.1007/s00431-013-2183-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022]
Abstract
UNLABELLED Autism spectrum disorder (ASD) is a common and severe neuro-developmental disorder in early childhood which is defined by social and communication deficits and repetitive and stereotypic behaviours. The aetiology of ASD remains poorly understood. Susceptibility to development of ASD has significant environmental components, in addition to the profound genetic heritability. Few genes have been associated to the risk for ASD development. There is substantial evidence implicating chronic neurological inflammation and immune dysregulation leading to upregulation of inflammatory cytokines in the ASD brain, probably due to altered blood-brain barrier function. The immune system is characterized by excessive and skewed cytokine responses, modulated T cell reactivity, decreased regulation and production of immunosuppressive cytokines, modified NK function and increased autoantibody production. CONCLUSION The perinatal environment generates vulnerability to chronic neuro-inflammation in the brain associated with profound modulation and dysregulation in the immune system leading to the rapid development of ASD in genetically susceptible children.
Collapse
Affiliation(s)
- Daniela Briceno Noriega
- Cell Biology and Immunology Group, Wageningen University, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | | |
Collapse
|
20
|
Siniscalco D, Cirillo A, Bradstreet JJ, Antonucci N. Epigenetic findings in autism: new perspectives for therapy. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2013; 10:4261-73. [PMID: 24030655 PMCID: PMC3799534 DOI: 10.3390/ijerph10094261] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/14/2013] [Accepted: 09/06/2013] [Indexed: 12/22/2022]
Abstract
Autism and autism spectrum disorders (ASDs) are complex neurodevelopmental disorders characterized by dysfunctions in social interactions, communications, restricted interests, and repetitive stereotypic behaviors. Despite extensive genetic and biological research, significant controversy surrounds our understanding of the specific mechanisms of their pathogenesis. However, accumulating evidence points to the involvement of epigenetic modifications as foundational in creating ASD pathophysiology. Epigenetic modifications or the alteration of DNA transcription via variations in DNA methylation and histone modifications but without alterations in the DNA sequence, affect gene regulation. These alterations in gene expression, obtained through DNA methylation and/or histone modifications, result from transcriptional regulatory influences of environmental factors, such as nutritional deficiencies, various toxicants, immunological effects, and pharmaceuticals. As such these effects are epigenetic regulators which determine the final biochemistry and physiology of the individual. In contrast to psychopharmacological interventions, bettering our understanding of how these gene-environmental interactions create autistic symptoms should facilitate the development of therapeutic targeting of gene expression for ASD biomedical care.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Second University of Naples; via S. Maria di Costantinopoli, Napoli 16-80138, Italy
- Centre for Autism—La Forza del Silenzio, Caserta 81036, Italy
- Cancellautismo—Non-Profit Association for Autism Care, Florence 50132, Italy
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-0-81-566-5880; Fax: +39-0-81-566-7503
| | - Alessandra Cirillo
- Institute of Protein Biochemistry, National Research Council of Italy; Naples 80128, Italy; E-Mail:
| | | | - Nicola Antonucci
- Biomedical Centre for Autism Research and Treatment, Bari 70126, Italy; E-Mail:
| |
Collapse
|