1
|
Carver AJ, Dunnwald M, Stevens HE. A head start: The relationship of placental factors to craniofacial and brain development. Dev Dyn 2025. [PMID: 40105397 DOI: 10.1002/dvdy.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
In recent years, the importance of placental function for fetal neurodevelopment has become increasingly studied. This field, known as neuroplacentology, has greatly expanded possible etiologies of neurodevelopmental disorders by exploring the influence of placental function on brain development. It is also well-established that brain development is influenced by craniofacial morphogenesis. However, there is less focus on the impact of the placenta on craniofacial development. Recent research suggests the functional influence of placental nutrients and hormones on craniofacial skeletal growth, such as prolactin, growth hormone, insulin-like growth factor 1, vitamin D, sulfate, and calcium, impacting both craniofacial and brain development. Therefore, interactions between the placenta and both fetal neurodevelopment and craniofacial development likely influence the growth and morphology of the head as a whole. This review discusses the role of placental hormone production and nutrient delivery in the development of the fetal head-defined as craniofacial and brain tissue together-expanding on the more established focus on brain development to also include the skull (or cranium) and face.
Collapse
Affiliation(s)
- Annemarie Jenna Carver
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Martine Dunnwald
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, Iowa City, Iowa, USA
| | - Hanna Elizabeth Stevens
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
- Psychiatry Department, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Hawk Intellectual and Developmental Disabilities Research Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Liu Y, Yu D, Ge X, Huang L, Pan PY, Shen H, Pettigrew RI, Chen SH, Mai J. Novel platinum therapeutics induce rapid cancer cell death through triggering intracellular ROS storm. Biomaterials 2025; 314:122835. [PMID: 39276409 PMCID: PMC11560510 DOI: 10.1016/j.biomaterials.2024.122835] [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: 05/22/2024] [Revised: 08/31/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Induction of reactive oxygen species (ROS) production in cancer cells plays a critical role for cancer treatment. However, therapeutic efficiency remains challenging due to insufficient ROS production of current ROS inducers. We designed a novel platinum (Pt)-based drug named "carrier-platin" that integrates ultrasmall Pt-based nanoparticles uniformly confined within a poly(amino acids) carrier. Carrier-platin dramatically triggered a burst of ROS in cancer cells, leading to cancer cell death as quick as 30 min. Unlike traditional Pt-based drugs which induce cell apoptosis through DNA intercalation, carrier-platin with superior ROS catalytic activities induces a unique pattern of cancer cell death that is neither apoptosis nor ferroptosis and operates independently of DNA damage. Importantly, carrier-platin demonstrates superior anti-tumor efficacy against a broad spectrum of cancers, particularly those with multidrug resistance, while maintaining minimal systemic toxicity. Our findings reveal a distinct mechanism of action of Pt in cancer cell eradication, positioning carrier-platin as a novel category of anti-cancer chemotherapeutics.
Collapse
Affiliation(s)
- Yongbin Liu
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA.
| | - Dongfang Yu
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Xueying Ge
- School of Engineering Medicine/ENMED, Texas A&M University and Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Lingyi Huang
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Ping-Ying Pan
- Center for Immunotherapy and Neal Cancer Center, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Roderic I Pettigrew
- School of Engineering Medicine/ENMED, Texas A&M University and Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Center for Immunotherapy and Neal Cancer Center, Houston Methodist Academic Institute, Houston, TX, 77030, USA; Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
den Bakker E, Smith DEC, Finken MJJ, Wamelink MMC, Salomons GS, van de Kamp JM, Bökenkamp A. Sulfate: a neglected (but potentially highly relevant) anion. Essays Biochem 2024; 68:391-399. [PMID: 38639060 DOI: 10.1042/ebc20230097] [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: 01/29/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
Sulfate is an important anion as sulfonation is essential in modulation of several compounds, such as exogens, polysaccharide chains of proteoglycans, cholesterol or cholesterol derivatives and tyrosine residues of several proteins. Sulfonation requires the presence of both the sulfate donor 3'-phosphoadenosine-5'-phosphosulfate (PAPS) and a sulfotransferase. Genetic disorders affecting sulfonation, associated with skeletal abnormalities, impaired neurological development and endocrinopathies, demonstrate the importance of sulfate. Yet sulfate is not measured in clinical practice. This review addresses sulfate metabolism and consequences of sulfonation defects, how to measure sulfate and why we should measure sulfate more often.
Collapse
Affiliation(s)
- Emil den Bakker
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Desiree E C Smith
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
| | - Martijn J J Finken
- Department of Pediatric Endocrinology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Gajja S Salomons
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jiddeke M van de Kamp
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Human Genetics, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arend Bökenkamp
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
4
|
Li X, Wu X, Lu T, Kuang C, Si Y, Zheng W, Li Z, Xue Y. Perineuronal Nets in the CNS: Architects of Memory and Potential Therapeutic Target in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3412. [PMID: 38542386 PMCID: PMC10970535 DOI: 10.3390/ijms25063412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The extracellular matrix (ECM) within the brain possesses a distinctive composition and functionality, influencing a spectrum of physiological and pathological states. Among its constituents, perineuronal nets (PNNs) are unique ECM structures that wrap around the cell body of many neurons and extend along their dendrites within the central nervous system (CNS). PNNs are pivotal regulators of plasticity in CNS, both during development and adulthood stages. Characterized by their condensed glycosaminoglycan-rich structures and heterogeneous molecular composition, PNNs not only offer neuroprotection but also participate in signal transduction, orchestrating neuronal activity and plasticity. Interfering with the PNNs in adult animals induces the reactivation of critical period plasticity, permitting modifications in neuronal connections and promoting the recovery of neuroplasticity following spinal cord damage. Interestingly, in the adult brain, PNN expression is dynamic, potentially modulating plasticity-associated states. Given their multifaceted roles, PNNs have emerged as regulators in the domains of learning, memory, addiction behaviors, and other neuropsychiatric disorders. In this review, we aimed to address how PNNs contribute to the memory processes in physiological and pathological conditions.
Collapse
Affiliation(s)
- Xue Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing 100191, China;
| | - Tangsheng Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Yue Si
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Zhonghao Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
| |
Collapse
|
5
|
Hurrion EM, Badawi N, Boyd RN, Morgan C, Gibbons K, Hennig S, Koorts P, Chauhan M, Bowling F, Flenady V, Kumar S, Dawson PA. SuPreme Study: a protocol to study the neuroprotective potential of sulfate among very/extremely preterm infants. BMJ Open 2023; 13:e076130. [PMID: 37451710 PMCID: PMC10351292 DOI: 10.1136/bmjopen-2023-076130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
INTRODUCTION Antenatal maternal magnesium sulfate (MgSO4) administration is a proven efficacious neuroprotective treatment reducing the risk of cerebral palsy (CP) among infants born preterm. Identification of the neuroprotective component with target plasma concentrations could lead to neonatal treatment with greater efficacy and accessibility. METHODS AND ANALYSIS This is a prospective observational cohort study, in three tertiary Australian centres. Participants are preterm infants, irrespective of antenatal MgSO4 exposure, born in 2013-2020 at 24+0 to 31+6 weeks gestation, and followed up to 2 years corrected age (CA) (to September 2023). 1595 participants are required (allowing for 17% deaths/loss to follow-up) to detect a clinically significant reduction (30% relative risk reduction) in CP when sulfate concentration at 7 days of age is 1 SD above the mean.A blood sample is collected on day 7 of age for plasma sulfate and magnesium measurement. In a subset of participants multiple blood and urine samples are collected for pharmacokinetic studies, between days 1-28, and in a further subset mother/infant blood is screened for genetic variants of sulfate transporter genes.The primary outcome is CP. Surviving infants are assessed for high risk of CP at 12-14 weeks CA according to Prechtl's Method to assess General Movements. Follow-up at 2 years CA includes assessments for CP, cognitive, language and motor development, and social/behavioural difficulties.Multivariate analyses will examine the association between day 7 plasma sulfate/magnesium concentrations with adverse neurodevelopmental outcomes. A population pharmacokinetic model for sulfate in the preterm infant will be created using non-linear mixed-effects modelling. ETHICS AND DISSEMINATION The study has been approved by Mater Misericordiae Ltd Human Research Ethics Committee (HREC/14/MHS/188). Results will be disseminated in peer-reviewed journal publications, and provided to the funding bodies. Using consumer input, a summary will be prepared for participants and consumer groups.
Collapse
Affiliation(s)
- Elizabeth M Hurrion
- Department of Newborn Services, Mater Mothers' Hospital, Brisbane, Queensland, Australia
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Nadia Badawi
- Cerebral Palsy Alliance, The University of Sydney, Sydney, New South Wales, Australia
| | - Roslyn N Boyd
- Queensland Cerebral Palsy and Rehabilitation Research Centre, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Catherine Morgan
- Cerebral Palsy Alliance, The University of Sydney, Sydney, New South Wales, Australia
| | - Kristen Gibbons
- Child Health Research Centre, Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Stefanie Hennig
- School of Clinical Sciences, Queensland University of Technology Faculty of Health, Kelvin Grove, Queensland, Australia
- Integrated Drug Development, Certara Strategic Consulting, Certara LP, Princeton, New Jersey, USA
| | - Pieter Koorts
- Grantley Stable Neonatal Unit, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Manbir Chauhan
- Department of Newborn Care, Gold Coast University Hospital, Southport, Queensland, Australia
| | - Francis Bowling
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Vicki Flenady
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Sailesh Kumar
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| | - Paul A Dawson
- Mater Research Institute The University of Queensland, South Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Panov J, Kaphzan H. An Association Study of DNA Methylation and Gene Expression in Angelman Syndrome: A Bioinformatics Approach. Int J Mol Sci 2022; 23:ijms23169139. [PMID: 36012404 PMCID: PMC9409443 DOI: 10.3390/ijms23169139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 12/01/2022] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of function of the E3-ligase UBE3A. Despite multiple studies, AS pathophysiology is still obscure and has mostly been explored in rodent models of the disease. In recent years, a growing body of studies has utilized omics datasets in the attempt to focus research regarding the pathophysiology of AS. Here, for the first time, we utilized a multi-omics approach at the epigenomic level and the transcriptome level, for human-derived neurons. Using publicly available datasets for DNA methylation and gene expression, we found genome regions in proximity to gene promoters and intersecting with gene-body regions that were differentially methylated and differentially expressed in AS. We found that overall, the genome in AS postmortem brain tissue was hypo-methylated compared to healthy controls. We also found more upregulated genes than downregulated genes in AS. Many of these dysregulated genes in neurons obtained from AS patients are known to be critical for neuronal development and synaptic functioning. Taken together, our results suggest a list of dysregulated genes that may be involved in AS development and its pathological features. Moreover, these genes might also have a role in neurodevelopmental disorders similar to AS.
Collapse
|
7
|
Clarke T, Fernandez FE, Dawson PA. Sulfation Pathways During Neurodevelopment. Front Mol Biosci 2022; 9:866196. [PMID: 35495624 PMCID: PMC9047184 DOI: 10.3389/fmolb.2022.866196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Sulfate is an important nutrient that modulates a diverse range of molecular and cellular functions in mammalian physiology. Over the past 2 decades, animal studies have linked numerous sulfate maintenance genes with neurological phenotypes, including seizures, impaired neurodevelopment, and behavioral abnormalities. Despite sulfation pathways being highly conserved between humans and animals, less than one third of all known sulfate maintenance genes are clinically reportable. In this review, we curated the temporal and spatial expression of 91 sulfate maintenance genes in human fetal brain from 4 to 17 weeks post conception using the online Human Developmental Biology Resource Expression. In addition, we performed a systematic search of PubMed and Embase, identifying those sulfate maintenance genes linked to atypical neurological phenotypes in humans and animals. Those findings, together with a search of the Online Mendelian Inheritance in Man database, identified a total of 18 candidate neurological dysfunction genes that are not yet considered in clinical settings. Collectively, this article provides an overview of sulfate biology genes to inform future investigations of perturbed sulfate homeostasis associated with neurological conditions.
Collapse
Affiliation(s)
- Taylor Clarke
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Banyo, QLD, Australia
| | - Francesca E. Fernandez
- School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Banyo, QLD, Australia
| | - Paul A. Dawson
- Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
- *Correspondence: Paul A. Dawson,
| |
Collapse
|
8
|
Winters C, Gorssen W, Ossorio-Salazar VA, Nilsson S, Golden S, D'Hooge R. Automated procedure to assess pup retrieval in laboratory mice. Sci Rep 2022; 12:1663. [PMID: 35102217 PMCID: PMC8803842 DOI: 10.1038/s41598-022-05641-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/17/2022] [Indexed: 11/25/2022] Open
Abstract
All mammalian mothers form some sort of caring bond with their infants that is crucial to the development of their offspring. The Pup Retrieval Test (PRT) is the leading procedure to assess pup-directed maternal care in laboratory rodents, used in a wide range of basic and preclinical research applications. Most PRT protocols require manual scoring, which is prone to bias and spatial and temporal inaccuracies. This study proposes a novel procedure using machine learning algorithms to enable reliable assessment of PRT performance. Automated tracking of a dam and one pup was established in DeepLabCut and was combined with automated behavioral classification of "maternal approach", "carrying" and "digging" in Simple Behavioral Analysis (SimBA). Our automated procedure estimated retrieval success with an accuracy of 86.7%, whereas accuracies of "approach", "carry" and "digging" were estimated at respectively 99.3%, 98.6% and 85.0%. We provide an open-source, step-by-step protocol for automated PRT assessment, which aims to increase reproducibility and reliability, and can be easily shared and distributed.
Collapse
Affiliation(s)
- Carmen Winters
- Laboratory of Biological Psychology, University of Leuven (KU Leuven), Leuven, Belgium.
- Leuven Experimental Attachment Research Lab, KU Leuven, Leuven, Belgium.
| | - Wim Gorssen
- Department of Biosystems, Center for Animal Breeding and Genetics, KU Leuven, Leuven, Belgium
| | | | - Simon Nilsson
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sam Golden
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, University of Leuven (KU Leuven), Leuven, Belgium.
| |
Collapse
|
9
|
Yang ML, Zhang JH, Li S, Zhu R, Wang L. SLC13A4 Might Serve as a Prognostic Biomarker and be Correlated with Immune Infiltration into Head and Neck Squamous Cell Carcinoma. Pathol Oncol Res 2021; 27:1609967. [PMID: 34840533 PMCID: PMC8610847 DOI: 10.3389/pore.2021.1609967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022]
Abstract
SLC13A4 is a sodium sulfate co-transporter, which is expressed in brains, placentas, thymes and other tissues, plays an essential role in maintaining the metabolic balance of sulfate in vivo. The TCGA database shows that it is differentially expressed in a variety of tumors, but its prognostic value in tumors has not been clarified. TCGA, Oncomine and Timer databases were used to analyze SLC13A4 mRNA expression in cancer tissues and normal tissues, and its correlation with clinical prognosis in head and neck tumor. The CIBERSORT database was used to analyze the correlation between SLC13A4 expression and the infiltration of immune cells. SLC13A4 enrichment analysis was carried out by GSEA. SLC13A4 mRNA levels were significantly lower in head and neck tumors than in paracancer tissues. SLC13A4 expression in Head and neck squamous cell carcinoma (HNSCC) was closely related to tumor pathological grade and clinical stage. Decreased SLC13A4 expression was associated with poor overall survival (OS), progression free survival (PFS), disease specific survival (DSS) and recurrence free survival (RFS) in HNSCC patients. The expression of SLC13A4 was negatively correlated with Monocytes, M1 macrophages, M2 macrophages, resting CD4+ memory T cells, resting NK cells and activated NK cells, but positively correlated with neutrophils, plasma cells, T follicular helper cells, gamma delta T cells, regulatory T cells and naive B cells. In addition, the genes in SLC13A4 low-expression group were mainly concentrated in immunity-related activities, viral diseases, typical tumor pathways and metabolism. The SLC13A4 high expression group was mainly enriched in metabolic pathways. These suggest that SLC13A4 may be a potential prognostic biomarker in HNSC and correlated with immune infiltrates.
Collapse
Affiliation(s)
- Meng-Ling Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Hua Zhang
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Li
- Department of General Surgery, Hospital of Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Harvey TJ, Davila RA, Vidovic D, Sharmin S, Piper M, Simmons DG. Genome-wide transcriptomic analysis of the forebrain of postnatal Slc13a4 +/- mice. BMC Res Notes 2021; 14:269. [PMID: 34256843 PMCID: PMC8276513 DOI: 10.1186/s13104-021-05687-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Sulfation is an essential physiological process that regulates the function of a wide array of molecules involved in brain development. We have previously shown expression levels for the sulfate transporter Slc13a4 to be elevated during postnatal development, and that sulfate accumulation in the brains of Slc13a4+/- mice is reduced, suggesting a role for this transporter during this critical window of brain development. In order to understand the pathways regulated by cellular sulfation within the brain, we performed a bulk RNA-sequencing analysis of the forebrain of postnatal day 20 (P20) Slc13a4 heterozygous mice and wild-type litter mate controls. DATA DESCRIPTION We performed an RNA transcriptomic based sequencing screen on the whole forebrain from Slc13a4+/- and Slc13a4+/+mice at P20. Differential expression analysis revealed 90 differentially regulated genes in the forebrain of Slc13a4+/- mice (a p-value of 0.1 was considered as significant). Of these, 55 were upregulated, and 35 were downregulated in the forebrain of heterozygous mice. Moreover, when we stratified further with a ± 1.2 fold-change, we observed 38 upregulated, and 16 downregulated genes in the forebrain of heterozygous mice. This resource provides a useful tool to interrogate which pathways may require elevated sulfate levels to drive normal postnatal development of the brain.
Collapse
Affiliation(s)
- Tracey J Harvey
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Raul Ayala Davila
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Diana Vidovic
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sazia Sharmin
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Piper
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David G Simmons
- School of Biomedical Sciences, The Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
11
|
Do Autophagy Enhancers/ROS Scavengers Alleviate Consequences of Mild Mitochondrial Dysfunction Induced in Neuronal-Derived Cells? Int J Mol Sci 2021; 22:ijms22115753. [PMID: 34072255 PMCID: PMC8197898 DOI: 10.3390/ijms22115753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/02/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
Mitochondrial function is at the nexus of pathways regulating synaptic-plasticity and cellular resilience. The involvement of brain mitochondrial dysfunction along with increased reactive oxygen species (ROS) levels, accumulating mtDNA mutations, and attenuated autophagy is implicated in psychiatric and neurodegenerative diseases. We have previously modeled mild mitochondrial dysfunction assumed to occur in bipolar disorder (BPD) using exposure of human neuronal cells (SH-SY5Y) to rotenone (an inhibitor of mitochondrial-respiration complex-I) for 72 and 96 h, which exhibited up- and down-regulation of mitochondrial respiration, respectively. In this study, we aimed to find out whether autophagy enhancers (lithium, trehalose, rapamycin, and resveratrol) and/or ROS scavengers [resveratrol, N-acetylcysteine (NAC), and Mn-Tbap) can ameliorate neuronal mild mitochondrial dysfunction. Only lithium (added for the last 24/48 h of the exposure to rotenone for 72/96 h, respectively) counteracted the effect of rotenone on most of the mitochondrial respiration parameters (measured as oxygen consumption rate (OCR)). Rapamycin, resveratrol, NAC, and Mn-Tbap counteracted most of rotenone's effects on OCR parameters after 72 h, possibly via different mechanisms, which are not necessarily related to their ROS scavenging and/or autophagy enhancement effects. The effect of lithium reversing rotenone's effect on OCR parameters is compatible with lithium's known positive effects on mitochondrial function and is possibly mediated via its effect on autophagy. By-and-large it may be summarized that some autophagy enhancers/ROS scavengers alleviate some rotenone-induced mild mitochondrial changes in SH-SY5Y cells.
Collapse
|
12
|
Sharmin S, Pradhan J, Zhang Z, Bellingham M, Simmons D, Piper M. Perineuronal net abnormalities in Slc13a4 +/- mice are rescued by postnatal administration of N-acetylcysteine. Exp Neurol 2021; 342:113734. [PMID: 33945789 DOI: 10.1016/j.expneurol.2021.113734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 03/30/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Disruptions to either sulfate supply or sulfation enzymes can affect brain development and have long-lasting effects on brain function, yet our understanding of the molecular mechanisms governing this are incomplete. Perineuronal nets (PNNs) are highly sulfated, specialized extracellular matrix structures that regulate the maturation of synaptic connections and neuronal plasticity. We have previously shown that mice heterozygous for the brain sulfate transporter Slc13a4 have abnormal social interactions, memory, exploratory behaviors, stress and anxiety of postnatal origin, pointing to potential deficits in PNN biology, and implicate SLC13A4 as a critical factor required for regulating normal synaptic connectivity and function. Here, we sought to investigate aberrant PNN formation as a potential mechanism contributing to the functional deficits displayed by Slc13a4+/- mice. Following social interactions, we reveal reduced neuronal activation in the somatosensory cortex of Slc13a4+/- mice, and altered inhibitory and excitatory postsynaptic currents. In line with this, we found a reduction in parvalbumin-expressing neurons decorated with PNNs, as well as reduced expression of markers for PNN maturation. Finally, we reveal that postnatal administration of N-acetylcysteine prevented PNN abnormalities from manifesting in Slc13a4+/- adult animals. Collectively, these data highlight a central role for postnatal SLC13A4 in normal PNN formation, circuit function and subsequent animal behavior.
Collapse
Affiliation(s)
- Sazia Sharmin
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jonu Pradhan
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhe Zhang
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Mark Bellingham
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David Simmons
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
13
|
Improvement of the skeletal phenotype in a mouse model of diastrophic dysplasia after postnatal treatment with N-acetylcysteine. Biochem Pharmacol 2021; 185:114452. [PMID: 33545117 DOI: 10.1016/j.bcp.2021.114452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022]
Abstract
Diastrophic dysplasia (DTD) is a recessive chondrodysplasia caused by mutations in the SLC26A2 gene encoding for a sulfate/chloride transporter. When SLC26A2 is impaired intracellular level of sulfate is reduced leading to the synthesis of undersulfated proteoglycans. In normal chondrocytes, the main source of intracellular sulfate is the extracellular uptake through SLC26A2, but a small amount comes from the catabolism of sulfur-containing amino acids and other thiols. Here N-acetylcysteine (NAC), an extensively used drug, is proposed as alternative source of intracellular sulfate in an animal model of DTD (dtd mouse). Mutant and wild type mice were treated twice a day with hypodermic injections of 250 mg NAC/kg body weight for one week after birth. At the end of the treatment, an improvement trend in cartilage proteoglycan sulfation and in the skeletal phenotype of treated dtd mice were observed. Thus, a longer treatment lasted three weeks starting from birth was performed. Treated mutant mice showed a significant increase of cartilage proteoglycan sulfation and a relevant improvement of the skeletal phenotype based on measurements of several bony elements and bone quality by DEXA and micro CT. Moreover, the amelioration of the overall growth plate morphology in treated dtd mice suggested a partial rescue of the endochondral ossification process. Overall, the results prove that NAC is an effective source of intracellular sulfate for dtd mice in the postnatal period. This finding paves the way for a potential pharmacological treatment of DTD patients taking advantage from a drug repositioning strategy.
Collapse
|
14
|
Zhang Z, Jhaveri D, Sharmin S, Harvey TJ, Dawson PA, Piper M, Simmons DG. Cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Biol Open 2020; 9:bio053132. [PMID: 32661132 PMCID: PMC7406315 DOI: 10.1242/bio.053132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/01/2020] [Indexed: 01/24/2023] Open
Abstract
Sulfate is a key anion required for a range of physiological functions within the brain. These include sulfonation of extracellular proteoglycans to facilitate local growth factor binding and to regulate the shape of morphogen gradients during development. We have previously shown that mice lacking one allele of the sulfate transporter Slc13a4 exhibit reduced sulfate transport into the brain, deficits in social behaviour, reduced performance in learning and memory tasks, and abnormal neurogenesis within the ventricular/subventricular zone lining the lateral ventricles. However, whether these mice have deficits in hippocampal neurogenesis was not addressed. Here, we demonstrate that adult Slc13a4+/- mice have increased neurogenesis within the subgranular zone (SGZ) of the hippocampal dentate gyrus, with elevated numbers of neural progenitor cells and intermediate progenitors. In contrast, by 12 months of age there were reduced numbers of neural stem cells in the SGZ of heterozygous mice. Importantly, we did not observe any changes in proliferation when we isolated and cultured progenitors in vitro in neurosphere assays, suggestive of a cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Collectively, these data demonstrate a requirement for sulfate transport during postnatal brain development to ensure normal adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Zhe Zhang
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Dhanisha Jhaveri
- Mater Research Institute, The University of Queensland, TRI Building, Woolloongabba, Brisbane, 4102, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - Sazia Sharmin
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Tracey J Harvey
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Paul A Dawson
- Mater Research Institute, The University of Queensland, TRI Building, Woolloongabba, Brisbane, 4102, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| |
Collapse
|