1
|
Zivko C, Sagar R, Xydia A, Lopez-Montes A, Mintzer J, Rosenberg PB, Shade DM, Porsteinsson AP, Lyketsos CG, Mahairaki V. iPSC-derived hindbrain organoids to evaluate escitalopram oxalate treatment responses targeting neuropsychiatric symptoms in Alzheimer's disease. Mol Psychiatry 2024; 29:3644-3652. [PMID: 38840027 PMCID: PMC11541203 DOI: 10.1038/s41380-024-02629-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and the gradual deterioration of brain function eventually leads to death. Almost all AD patients suffer from neuropsychiatric symptoms (NPS), the emergence of which correlates with dysfunctional serotonergic systems. Our aim is to generate hindbrain organoids containing serotonergic neurons using human induced Pluripotent Stem Cells (iPSCs). Work presented here is laying the groundwork for the application of hindbrain organoids to evaluate individual differences in disease progression, NPS development, and pharmacological treatment response. Human peripheral blood mononuclear cells (PBMCs) from healthy volunteers (n = 3), an AD patient without NPS (n = 1), and AD patients with NPS (n = 2) were reprogrammed into iPSCs and subsequently differentiated into hindbrain organoids. The presence of serotonergic neurons was confirmed by quantitative reverse transcription PCR, flow cytometry, immunocytochemistry, and detection of released serotonin (5-HT). We successfully reprogrammed PBMCs into 6 iPSC lines, and subsequently generated hindbrain organoids from 6 individuals to study inter-patient variability using a precision medicine approach. To assess patient-specific treatment effects, organoids were treated with different concentrations of escitalopram oxalate, commonly prescribed for NPS. Changes in 5-HT levels before and after treatment with escitalopram were dose-dependent and variable across patients. Organoids from different people responded differently to the application of escitalopram in vitro. We propose that this 3D platform might be effectively used for drug screening purposes to predict patients with NPS most likely to respond to treatment in vivo and to understand the heterogeneity of treatment responses.
Collapse
Affiliation(s)
- Cristina Zivko
- Department of Genetic Medicine, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
| | - Ram Sagar
- Department of Genetic Medicine, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
| | - Ariadni Xydia
- Department of Genetic Medicine, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
| | - Alejandro Lopez-Montes
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA
| | - Jacobo Mintzer
- Department of Health Sciences, Medical University of South Carolina, 29425, Charleston, SC, USA
- Ralph H. Johnson VA Healthcare System, 29401, Charleston, SC, USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
| | - David M Shade
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 21205, Baltimore, MD, USA
| | - Anton P Porsteinsson
- Department of Psychiatry, University of Rochester School of Medicine and Dentistry, 14642, Rochester, NY, USA
| | - Constantine G Lyketsos
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA
- Johns Hopkins Alzheimer's Disease Research Center, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins School of Medicine, 21205, Baltimore, MD, USA.
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins School of Medicine, 21287, Baltimore, MD, USA.
| |
Collapse
|
2
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [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: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
3
|
Xu T, Cao L, Duan J, Li Y, Li Y, Hu Z, Li S, Zhang M, Wang G, Guo F, Lu J. Uncovering the role of FOXA2 in the Development of Human Serotonin Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303884. [PMID: 37679064 PMCID: PMC10646255 DOI: 10.1002/advs.202303884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/08/2023] [Indexed: 09/09/2023]
Abstract
Directed differentiation of serotonin neurons (SNs) from human pluripotent stem cells (hPSCs) provides a valuable tool for uncovering the mechanism of human SN development and the associated neuropsychiatric disorders. Previous studies report that FOXA2 is expressed by serotonergic progenitors (SNPs) and functioned as a serotonergic fate determinant in mouse. However, in the routine differentiation experiments, it is accidentally found that less SNs and more non-neuronal cells are obtained from SNP stage with higher percentage of FOXA2-positive cells. This phenomenon prompted them to question the role of FOXA2 as an intrinsic fate determinant for human SN differentiation. Herein, by direct differentiation of engineered hPSCs into SNs, it is found that the SNs are not derived from FOXA2-lineage cells; FOXA2-knockout hPSCs can still differentiate into mature and functional SNs with typical serotonergic identity; FOXA2 overexpression suppresses the SN differentiation, indicating that FOXA2 is not intrinsically required for human SN differentiation. Furthermore, repressing FOXA2 expression by retinoic acid (RA) and dynamically modulating Sonic Hedgehog (SHH) signaling pathway promotes human SN differentiation. This study uncovers the role of FOXA2 in human SN development and improves the differentiation efficiency of hPSCs into SNs by repressing FOXA2 expression.
Collapse
Affiliation(s)
- Ting Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Jinjin Duan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yingqi Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - You Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Zhangsen Hu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Shuanqing Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Meihui Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Guanhao Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Fei Guo
- Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Suzhou Institute of Tongji UniversitySuzhou215101China
| |
Collapse
|
4
|
Jamu IM, Okamoto H. Recent advances in understanding adverse effects associated with drugs targeting the serotonin receptor, 5-HT GPCR. Front Glob Womens Health 2022; 3:1012463. [PMID: 36619589 PMCID: PMC9812521 DOI: 10.3389/fgwh.2022.1012463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
It has been acknowledged that more women suffer from adverse effects of drugs than men globally. A group of drugs targeting serotonin [5-hydroxytryptamine] (5-HT) binding G-protein-coupled receptors (GPCRs) have been reported to preferentially affect women more than men, causing adverse effects such as breast cancer and infertility. 5-HT GPCR-targeted drugs in the central nervous system (CNS) manage psychiatric conditions, such as depression or bipolar and in the peripheral nervous system (PNS) treat migraines. Physiological characteristics such as specific types of hormones, higher body fat density and smaller body mass in women result in disparities in pharmacodynamics of drugs, thus explaining sex-related differences in the observed adverse effects. In this review, we discuss the side effects of drugs targeting 5-HT GPCRs based on serotonin's roles in the CNS and PNS. We have systematically reviewed adverse effects of drugs targeting 5-HT GPCR using information from the Food and Drug Administration and European Medicines Agency. Further information on drug side effects and receptor targets was acquired from the SIDER and DrugBank databases, respectively. These drugs bind to 5-HT GPCRs in the CNS, namely the brain, and PNS such as breasts, ovaries and testes, potentially causing side effects within these areas. Oestrogen affects both the biosynthesis of 5-HT and the densities of 5-HT GPCRs in given tissues and cells. 5-HT GPCR-targeting drugs perturb this process. This is likely a reason why women are experiencing more adverse effects than men due to their periodic increase and the relatively high concentrations of oestrogen in women and, thus a greater incidence of the oestrogen-mediated 5-HT system interference. In addition, women have a lower concentration of serotonin relative to men and also have a relatively faster rate of serotonin metabolism which might be contributing to the former. We discuss potential approaches that could mitigate at least some of the adverse effects experienced by women taking the 5-HT GPCR-targeting drugs.
Collapse
|
5
|
Hingorani M, Viviani AML, Sanfilippo JE, Janušonis S. High-resolution spatiotemporal analysis of single serotonergic axons in an in vitro system. Front Neurosci 2022; 16:994735. [PMID: 36353595 PMCID: PMC9638127 DOI: 10.3389/fnins.2022.994735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/28/2022] [Indexed: 12/04/2022] Open
Abstract
Vertebrate brains have a dual structure, composed of (i) axons that can be well-captured with graph-theoretical methods and (ii) axons that form a dense matrix in which neurons with precise connections operate. A core part of this matrix is formed by axons (fibers) that store and release 5-hydroxytryptamine (5-HT, serotonin), an ancient neurotransmitter that supports neuroplasticity and has profound implications for mental health. The self-organization of the serotonergic matrix is not well understood, despite recent advances in experimental and theoretical approaches. In particular, individual serotonergic axons produce highly stochastic trajectories, fundamental to the construction of regional fiber densities, but further advances in predictive computer simulations require more accurate experimental information. This study examined single serotonergic axons in culture systems (co-cultures and monolayers), by using a set of complementary high-resolution methods: confocal microscopy, holotomography (refractive index-based live imaging), and super-resolution (STED) microscopy. It shows that serotonergic axon walks in neural tissue may strongly reflect the stochastic geometry of this tissue and it also provides new insights into the morphology and branching properties of serotonergic axons. The proposed experimental platform can support next-generation analyses of the serotonergic matrix, including seamless integration with supercomputing approaches.
Collapse
|
6
|
Holmes J, Lau T, Saylor R, Fernández-Novel N, Hersey M, Keen D, Hampel L, Horschitz S, Ladewig J, Parke B, Reed MC, Nijhout HF, Best J, Koch P, Hashemi P. Voltammetric Approach for Characterizing the Biophysical and Chemical Functionality of Human Induced Pluripotent Stem Cell-Derived Serotonin Neurons. Anal Chem 2022; 94:8847-8856. [PMID: 35713335 DOI: 10.1021/acs.analchem.1c05082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Depression is quickly becoming one of the world's most pressing public health crises, and there is an urgent need for better diagnostics and therapeutics. Behavioral models in animals and humans have not adequately addressed the diagnosis and treatment of depression, and biomarkers of mental illnesses remain ill-defined. It has been very difficult to identify biomarkers of depression because of in vivo measurement challenges. While our group has made important strides in developing in vivo tools to measure such biomarkers (e.g., serotonin) in mice using voltammetry, these tools cannot be easily applied for depression diagnosis and drug screening in humans due to the inaccessibility of the human brain. In this work, we take a chemical approach, ex vivo, to introduce a human-derived system to investigate brain serotonin. We utilize human induced pluripotent stem cells differentiated into serotonin neurons and establish a new ex vivo model of real-time serotonin neurotransmission measurements. We show that evoked serotonin release responds to stimulation intensity and tryptophan preloading, and that serotonin release and reuptake kinetics resemble those found in vivo in rodents. Finally, after selective serotonin reuptake inhibitor (SSRI) exposure, we find dose-dependent internalization of the serotonin reuptake transporters (a signature of the in vivo response to SSRI). Our new human-derived chemical model has great potential to provide an ex vivo chemical platform as a translational tool for in vivo neuropsychopharmacology.
Collapse
Affiliation(s)
- Jordan Holmes
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Thorsten Lau
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Rachel Saylor
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Nadine Fernández-Novel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Melinda Hersey
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Pharmacology, Physiology, & Neuroscience, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Deanna Keen
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Lena Hampel
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Sandra Horschitz
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Julia Ladewig
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Brenna Parke
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Michael C Reed
- Department of Mathematics, Duke University, Durham, North Carolina 27708, United States
| | - H Frederik Nijhout
- Department of Biology, Duke University, Durham, North Carolina 27708, United States
| | - Janet Best
- Department of Mathematics, The Ohio State University, Columbus, Ohio 43210, United States
| | - Philipp Koch
- Department of Translational Brain Research, Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim, 68159 Mannheim, Germany.,German Cancer Research Center, 69120 Heidelberg, Germany.,HITBR Hector Institute for Translational Brain Research gGmbH, 68159 Mannheim, Germany
| | - Parastoo Hashemi
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States.,Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
7
|
Wang G, Li S, Li Y, Zhang M, Xu T, Li T, Cao L, Lu J. Corticosterone induces obesity partly via promoting intestinal cell proliferation and survival. Front Endocrinol (Lausanne) 2022; 13:1052487. [PMID: 36699046 PMCID: PMC9869250 DOI: 10.3389/fendo.2022.1052487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION A vicious cycle ensues whereby prolonged exposure to social stress causes increased production of glucocorticoids (GCs), leading to obesity even further. Understanding the role of GCs, the key element in the vicious circle, might be helpful to break the vicious circle. However, the mechanism by which GCs induce obesity remains elusive. METHODS Corticosterone (CORT) was administered to mice for 8 weeks. Food and water intake were recorded; obesity was analyzed by body-weight evaluation and magnetic resonance imaging (MRI); intestinal proliferation and survival were evaluated by H&E staining, EdU-progression test, TUNEL assay and immunofluorescence staining of Ki67 and CC3; RNA-seq was performed to analyze transcriptional alterations in small intestines and livers. RESULTS Chronic CORT treatment induced obesity, longer small intestines, hepatic steatosis and elevated levels of serum insulin and leptin in mice; CORT-treated mice showed increased cell proliferation and decreased apoptosis of small intestines; RNA-seq results indicate that differentially expressed genes (DEGs) were enriched in several cell growth/death-associated signaling pathways. DISCUSSION Herein we find that administration of CORT to mice promotes the proliferation and survival of intestinal cells, which might contribute to the longer small intestines and the elongated intestinal villi, thus leading to increased nutrient absorption and obesity in mice. Understanding CORT-induced alterations in intestines and associated signaling pathways might provide novel therapeutic clues for GCs or stress-associated obesity.
Collapse
Affiliation(s)
- Guanhao Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuanqing Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yingqi Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Meihui Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ting Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Tianming Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- *Correspondence: Jianfeng Lu, ; ; Lining Cao,
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Biomedical research center, Suzhou Institute of Tongji University, Suzhou, China
- *Correspondence: Jianfeng Lu, ; ; Lining Cao,
| |
Collapse
|
8
|
Unterholzner J, Millischer V, Wotawa C, Sawa A, Lanzenberger R. Making Sense of Patient-Derived iPSCs, Transdifferentiated Neurons, Olfactory Neuronal Cells, and Cerebral Organoids as Models for Psychiatric Disorders. Int J Neuropsychopharmacol 2021; 24:759-775. [PMID: 34216465 PMCID: PMC8538891 DOI: 10.1093/ijnp/pyab037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/30/2021] [Accepted: 07/02/2021] [Indexed: 11/17/2022] Open
Abstract
The improvement of experimental models for disorders requires a constant approximation towards the dysregulated tissue. In psychiatry, where an impairment of neuronal structure and function is assumed to play a major role in disease mechanisms and symptom development, this approximation is an ongoing process implicating various fields. These include genetic, animal, and post-mortem studies. To test hypotheses generated through these studies, in vitro models using non-neuronal cells such as fibroblasts and lymphocytes have been developed. For brain network disorders, cells with neuronal signatures would, however, represent a more adequate tissue. Considering the limited accessibility of brain tissue, research has thus turned towards neurons generated from induced pluripotent stem cells as well as directly induced neurons, cerebral organoids, and olfactory neuroepithelium. Regarding the increasing importance and amount of research using these neuronal cells, this review aims to provide an overview of all these models to make sense of the current literature. The development of each model system and its use as a model for the various psychiatric disorder categories will be laid out. Also, advantages and limitations of each model will be discussed, including a reflection on implications and future perspectives.
Collapse
Affiliation(s)
- Jakob Unterholzner
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Vincent Millischer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria,Neurogenetics Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Wotawa
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Akira Sawa
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA,Departments of Psychiatry, Neuroscience, Biomedical Engineering and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria,Correspondence: Prof. Rupert Lanzenberger, MD, PD, NEUROIMAGING LABS (NIL) - PET, MRI, EEG, TMS & Chemical Lab, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria ()
| |
Collapse
|
9
|
Kaswan NK, Mohammed Izham NAB, Tengku Mohamad TAS, Sulaiman MR, Perimal EK. Cardamonin Modulates Neuropathic Pain through the Possible Involvement of Serotonergic 5-HT1A Receptor Pathway in CCI-Induced Neuropathic Pain Mice Model. Molecules 2021; 26:3677. [PMID: 34208700 PMCID: PMC8234694 DOI: 10.3390/molecules26123677] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardamonin, a naturally occurring chalcone isolated from Alpinia species has shown to possess strong anti-inflammatory and anti-nociceptive activities. Previous studies have demonstrated that cardamonin exerts antihyperalgesic and antiallodynic properties in chronic constriction injury (CCI)-induced neuropathic pain animal model. However, the mechanisms underlying cardamonin's effect have yet to be fully understood. The present study aims to investigate the involvement of the serotonergic system in cardamonin induced antihyperalgesic and antiallodynic effects in CCI-induced neuropathic pain mice model. The neuropathic pain symptoms in the CCI mice model were assessed using Hargreaves Plantar test and von-Frey filament test on day 14 post-surgery. Central depletion of serotonin along the descending serotonergic pathway was done using ρ-chlorophenylalanine (PCPA, 100 mg/kg, i.p.), an inhibitor of serotonin synthesis for four consecutive days before cardamonin treatment, and was found to reverse the antihyperalgesic and antiallodynic effect produced by cardamonin. Pretreatment of the mice with several 5-HT receptor subtypes antagonists: methiothepin (5-HT1/6/77 receptor antagonist, 0.1 mg/kg), WAY 100635 (5-HT1A receptor antagonist, 1 mg/kg), isamoltane (5-HT1B receptor antagonist, 2.5 mg/kg), ketanserin (5-HT2A receptor antagonist, 0.3 mg/kg), and ondansetron (5-HT3 receptor antagonist, 0.5 mg/kg) were shown to abolish the effect of cardamonin induced antihyperalgesic and antiallodynic effects. Further evaluation of the 5-HT1A receptor subtype protein expressions reveals that cardamonin significantly upregulated its expression in the brainstem and spinal cord. Our results suggest that the serotonergic pathway is essential for cardamonin to exert its antineuropathic effect in CCI mice through the involvement of the 5-HT1A receptor subtype in the central nervous system.
Collapse
Affiliation(s)
- Nur Khalisah Kaswan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (N.K.K.); (N.A.B.M.I.); (T.A.S.T.M.); (M.R.S.)
| | - Noor Aishah Binti Mohammed Izham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (N.K.K.); (N.A.B.M.I.); (T.A.S.T.M.); (M.R.S.)
| | - Tengku Azam Shah Tengku Mohamad
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (N.K.K.); (N.A.B.M.I.); (T.A.S.T.M.); (M.R.S.)
| | - Mohd Roslan Sulaiman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (N.K.K.); (N.A.B.M.I.); (T.A.S.T.M.); (M.R.S.)
| | - Enoch Kumar Perimal
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (N.K.K.); (N.A.B.M.I.); (T.A.S.T.M.); (M.R.S.)
- Centre of Excellence for Nanoscale BioPhotonics, Australian Research Council, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
10
|
Liu HK, He SJ, Zhang JG. A bioinformatic study revealed serotonergic neurons are involved in the etiology and therapygenetics of anxiety disorders. Transl Psychiatry 2021; 11:297. [PMID: 34011923 PMCID: PMC8134630 DOI: 10.1038/s41398-021-01432-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Genetic factors contribute to the susceptibility of anxiety disorders (ADs) and responses to associated cognitive-behavioral therapy (CBT). However, the type of brain cell affected by the related genes remains unclear. Previous studies have indicated various important brain neurons associated with psychiatric disorders, highlighting the necessity to study the cellular basis of anxiety. We assembled 37 AD-related genes and 23 CBT-related genes from recent large-scale genome-wide association studies, and then investigated their cell-type specificity in single-cell transcriptome data via an expression weighted cell type enrichment method. Additionally, to investigate the cellular differences between ADs and other psychiatric disorders, we excluded the genes associated with major depressive disorder, bipolar disorder, and neuroticism, resulting in 29 AD-specific genes. Remarkably, results indicate that serotonergic neurons are significantly associated with both AD-related and CBT-related genes, despite the two gene sets showing no overlap. These observations provide evidence that serotonergic neurons are involved in the etiology and therapygenetics of ADs. Moreover, results also showed that serotonergic neurons are associated with AD-specific genes, providing a supplementary finding that is in opposition to previous studies that found no evidence for the association between serotonergic neurons and psychiatric disorders via the same strategy. In summary, the current study found that serotonergic neurons are involved in the etiology and therapygenetics of ADs, providing insights into their genetic and cellular basis. Further, this cellular difference study may deepen our understanding of ADs and other psychiatric disorders.
Collapse
Affiliation(s)
- Han-Kui Liu
- grid.21155.320000 0001 2034 1839BGI-Shenzhen, Shenzhen, China
| | - Si-Jie He
- Shijiazhuang BGI Genomics Co., Ltd, Shijiazhuang, China
| | | |
Collapse
|
11
|
Brady MV, Vaccarino FM. Role of SHH in Patterning Human Pluripotent Cells towards Ventral Forebrain Fates. Cells 2021; 10:cells10040914. [PMID: 33923415 PMCID: PMC8073580 DOI: 10.3390/cells10040914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 12/03/2022] Open
Abstract
The complexities of human neurodevelopment have historically been challenging to decipher but continue to be of great interest in the contexts of healthy neurobiology and disease. The classic animal models and monolayer in vitro systems have limited the types of questions scientists can strive to answer in addition to the technical ability to answer them. However, the tridimensional human stem cell-derived organoid system provides the unique opportunity to model human development and mimic the diverse cellular composition of human organs. This strategy is adaptable and malleable, and these neural organoids possess the morphogenic sensitivity to be patterned in various ways to generate the different regions of the human brain. Furthermore, recapitulating human development provides a platform for disease modeling. One master regulator of human neurodevelopment in many regions of the human brain is sonic hedgehog (SHH), whose expression gradient and pathway activation are responsible for conferring ventral identity and shaping cellular phenotypes throughout the neural axis. This review first discusses the benefits, challenges, and limitations of using organoids for studying human neurodevelopment and disease, comparing advantages and disadvantages with other in vivo and in vitro model systems. Next, we explore the range of control that SHH exhibits on human neurodevelopment, and the application of SHH to various stem cell methodologies, including organoids, to expand our understanding of human development and disease. We outline how this strategy will eventually bring us much closer to uncovering the intricacies of human neurodevelopment and biology.
Collapse
Affiliation(s)
| | - Flora M. Vaccarino
- Child Study Center, Yale University, New Haven, CT 06520, USA;
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
- Yale Kavli Institute for Neuroscience, New Haven, CT 06520, USA
- Correspondence:
| |
Collapse
|
12
|
Current State-of-the-Art and Unresolved Problems in Using Human Induced Pluripotent Stem Cell-Derived Dopamine Neurons for Parkinson's Disease Drug Development. Int J Mol Sci 2021; 22:ijms22073381. [PMID: 33806103 PMCID: PMC8037675 DOI: 10.3390/ijms22073381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Human induced pluripotent stem (iPS) cells have the potential to give rise to a new era in Parkinson's disease (PD) research. As a unique source of midbrain dopaminergic (DA) neurons, iPS cells provide unparalleled capabilities for investigating the pathogenesis of PD, the development of novel anti-parkinsonian drugs, and personalized therapy design. Significant progress in developmental biology of midbrain DA neurons laid the foundation for their efficient derivation from iPS cells. The introduction of 3D culture methods to mimic the brain microenvironment further expanded the vast opportunities of iPS cell-based research of the neurodegenerative diseases. However, while the benefits for basic and applied studies provided by iPS cells receive widespread coverage in the current literature, the drawbacks of this model in its current state, and in particular, the aspects of differentiation protocols requiring further refinement are commonly overlooked. This review summarizes the recent data on general and subtype-specific features of midbrain DA neurons and their development. Here, we review the current protocols for derivation of DA neurons from human iPS cells and outline their general weak spots. The associated gaps in the contemporary knowledge are considered and the possible directions for future research that may assist in improving the differentiation conditions and increase the efficiency of using iPS cell-derived neurons for PD drug development are discussed.
Collapse
|
13
|
Serotonin Neuronal Function from the Bed to the Bench: Is This Really a Mirrored Way? eNeuro 2019; 6:ENEURO.0021-19.2019. [PMID: 31118207 PMCID: PMC6553573 DOI: 10.1523/eneuro.0021-19.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 11/21/2022] Open
|
14
|
Gong L, Cao L, Shen Z, Shao L, Gao S, Zhang C, Lu J, Li W. Materials for Neural Differentiation, Trans-Differentiation, and Modeling of Neurological Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705684. [PMID: 29573284 DOI: 10.1002/adma.201705684] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Indexed: 05/02/2023]
Abstract
Neuron regeneration from pluripotent stem cells (PSCs) differentiation or somatic cells trans-differentiation is a promising approach for cell replacement in neurodegenerative diseases and provides a powerful tool for investigating neural development, modeling neurological diseases, and uncovering the mechanisms that underlie diseases. Advancing the materials that are applied in neural differentiation and trans-differentiation promotes the safety, efficiency, and efficacy of neuron regeneration. In the neural differentiation process, matrix materials, either natural or synthetic, not only provide a structural and biochemical support for the monolayer or three-dimensional (3D) cultured cells but also assist in cell adhesion and cell-to-cell communication. They play important roles in directing the differentiation of PSCs into neural cells and modeling neurological diseases. For the trans-differentiation of neural cells, several materials have been used to make the conversion feasible for future therapy. Here, the most current applications of materials for neural differentiation for PSCs, neuronal trans-differentiation, and neurological disease modeling is summarized and discussed.
Collapse
Affiliation(s)
- Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lining Cao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenmin Shen
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Shao
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shaorong Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianfeng Lu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|