Hostname: page-component-8448b6f56d-xtgtn Total loading time: 0 Render date: 2024-04-24T17:23:22.541Z Has data issue: false hasContentIssue false

Integrating stem cell-based experiments in clinical research

Published online by Cambridge University Press:  15 June 2020

Rakesh Karmacharya*
Affiliation:
Program in Neuroscience and Chemical Biology, Center for Genomic Medicine, Massachusetts General Hospital & McLean Hospital, Harvard University, Boston, MA, USA Chemical Biology and Therapeutic Science Program, Broad Institute of Harvard & MIT, Cambridge, MA, USA
Christian Kieling
Affiliation:
Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
Valeria Mondelli
Affiliation:
Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, and King's College London,London, United Kingdom
*
Rakesh Karmacharya, E-mail: karmacharya@mgh.harvard.edu

Extract

With the seminal discovery of somatic cell reprogramming with defined genetic factors, it is now a routine laboratory procedure to reprogram somatic cells to generate patient-specific induced pluripotent stem cells (iPSCs) [1] Patient-specific iPSCs can be differentiated to generate mature neurons as well as three-dimensional brain organoids that show appropriate functional activity in electrophysiological studies [2,3]. However, there is a significant gap in the thoughtful incorporation of patient-derived neuronal cells in clinical studies addressing disease risk.

Type
Viewpoint
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2020. Published by Cambridge University Press on behalf of European Psychiatric Association

With the seminal discovery of somatic cell reprogramming with defined genetic factors, it is now a routine laboratory procedure to reprogram somatic cells to generate patient-specific induced pluripotent stem cells (iPSCs) [Reference Takahashi and Yamanaka1] Patient-specific iPSCs can be differentiated to generate mature neurons as well as three-dimensional brain organoids that show appropriate functional activity in electrophysiological studies [Reference Watmuff, Berkovitch, Huang, Iaconelli, Toffel and Karmacharya2,Reference Kathuria, Lopez-Lengowski, Jagtap, Perlis and Cohen3]. However, there is a significant gap in the thoughtful incorporation of patient-derived neuronal cells in clinical studies addressing disease risk.

Recently, we have seen the implementation of well-designed clinical studies aimed at delineating biological and psychosocial risk factors for psychiatric disorders. One such example is the Identifying Depression in Adolescence, a multisite global study spanning four continents that has developed an empirically derived stratification tool for the identification of adolescents at high and low risk for developing depression [Reference Kieling, Adewuya, Fisher, Karmacharya, Kohrt and Swartz4]. These studies have the potential to undertake deep phenotyping of symptoms and risks while performing incisive biological studies. While such clinical cohort studies have been undertaken in the past with serum, fibroblasts, or peripheral blood mononuclear cells (PBMCs), it behooves the clinical and translational research community to utilize patient-derived neuronal cells, given the central role of the brain in psychiatric disorders.

The ability to generate specific neuronal and glial cell types as well as three-dimensional brain organoids with disease-specific genetic backgrounds provides new opportunities for targeted biological studies [Reference Kathuria, Lopez-Lengowski, Vater, Cohen and Karmacharya5]. For instance, given the role of microglia and neuroinflammation in psychiatric disorders [Reference Mondelli, Vernon, Turkheimer, Dazzan and Pariante6], iPSC-derived neurons and microglia can be used to investigate glia–neuron interaction in these disease contexts. Part of the historical challenge in psychiatry has been a lack of fruitful engagement between basic scientists and clinical researchers. Basic scientists often treat diagnostic labels as monolithic entities, with little understanding of the nuances of psychiatric disorders. On the other hand, most clinical researchers are not well-versed in cellular and molecular biology techniques that can inform on mechanistic bases of disease biology. There is a need for cross-fertilization of ideas and methodologies and active collaboration between these two groups of investigators.

Human neuronal cells with disease-specific genetic backgrounds provide novel ways to explore disease biology (Figure 1). We can pursue hypothesis-based studies to test specific theories about the biology underlying disease pathophysiology/risk, that is, cell-autonomous deficits in specific neuronal subtypes in a particular disorder [Reference Kathuria, Lopez-Lengowski, Watmuff, Cohen and Karmacharya7]. Another approach involves using discovery-based approaches to profile-specific neuronal cells with transcriptomic, proteomic, phospho-proteomic, and metabolomic experiments. Gene–environment interactions play a pivotal role in many psychiatric disorders. Profiling patient cells in the setting of environmental perturbations can be used to uncover biological features that may not be present under normal culture conditions [Reference Huang, Berkovitch, Iaconelli, Watmuff, Park and Chattopadhyay8,Reference Huang, Park, Iaconelli, Berkovitch and Watmuff9]. We can investigate how specific neuronal subtypes from disease subjects may behave differently in the setting of specific environmental stressors. For instance, do neural stem cells from youth at high risk for developing depression show differences in neurogenesis or show differential regulation of specific sets of genes when exposed to stress hormones, sex hormones, or pro-inflammatory cytokines? Such studies can help identify biological pathways that underlie the clinical manifestations of psychiatric disorders.

Figure 1. Framework for utilizing human system cell-derived neurons in clinical cohort studies.

There are subgroups of patients in the disease categories that respond to specific treatments while others do not. The differential treatment response is hypothesized to be due to different underlying biology in these subgroups [Reference Draganov, Arranz, Salazar, Diego-Adeliño, Gallego-Fabrega and Jubero10]. It would be tractable to generate neurons from treatment responders and nonresponders and undertake in vitro experiments to identify mechanisms underlying differential response to treatment. Identification of cellular features that correlate with treatment response not only provides face validity to the mechanistic basis for the disease biology, but it will also aid in the development of predictive and precision medicine methods. With the identification of a robust and reliable treatment-response signature, one can evaluate effects of potential medications in neurons from individual patients before making an educated decision to pick the medication that shows the most robust response in the in vitro assays for that particular patient.

We are at an opportune juncture for the application of cutting-edge stem cell approaches to study psychiatric disease biology. In order to successfully bring these new technologies to the service of psychiatric research, we need a more engaged collaboration between clinical researchers and basic scientists. It will also require funding agencies and academic journals to view this rich area of potential research with new lenses and involve translational researchers who understand the new technologies and at the same time can appreciate the intricacies involved in their application to the complex issues involved in clinical psychiatry research.

Conflict of Interest

The authors declare no conflicts of interest.

References

Takahashi, K, Yamanaka, S. Induced pluripotent stem cells in medicine and biology. Development. 2013;140:24572461. doi: 10.1242/dev.092551.CrossRefGoogle ScholarPubMed
Watmuff, B, Berkovitch, SS, Huang, JH, Iaconelli, J, Toffel, S, Karmacharya, R. Disease signatures for schizophrenia and bipolar disorder using patient-derived induced pluripotent stem cells. Mol Cell Neurosci. 2016;73:96103. doi: 10.1016/j.mcn.2016.01.003.CrossRefGoogle ScholarPubMed
Kathuria, A, Lopez-Lengowski, K, Jagtap, SS, McPhie D, Perlis, RH, Cohen, BM, et al.Transcriptomic landscape and functional characterization of induced pluripotent stem cell-derived cerebral organoids in schizophrenia. JAMA Psychiatry . 2020. doi:10.1001/jamapsychiatry.2020.0196.CrossRefGoogle Scholar
Kieling, C, Adewuya, A, Fisher, HL, Karmacharya, R, Kohrt, BA, Swartz, JR, et al.Identifying depression early in adolescence. Lancet Child Adolesc Health. 2019;3:211213. doi: 10.1016/S2352-4642(19)30059-8.CrossRefGoogle ScholarPubMed
Kathuria, A, Lopez-Lengowski, K, Vater, M, McPhie D, Cohen, BM, Karmacharya, R. Transcriptome analysis and functional characterization of cerebral organoids in bipolar disorder. Genome Med. 2020;12:34. doi:10.1186/s13073-020-00733-6.CrossRefGoogle ScholarPubMed
Mondelli, V, Vernon, AC, Turkheimer, F, Dazzan, P, Pariante, CM. Brain microglia in psychiatric disorders. Lancet Psychiatry. 2017;4:563572. doi: 10.1016/S2215-0366(17)30101-3.CrossRefGoogle ScholarPubMed
Kathuria, A, Lopez-Lengowski, K, Watmuff, B, McPhie D, Cohen, BM, Karmacharya, R. Synaptic deficits in iPSC-derived cortical interneurons in schizophrenia are mediated by NLGN2 and rescued by N-acetylcysteine. Transl Psychiatry. 2019;9:321. doi:10.1038/s41398-019-0660-x.CrossRefGoogle ScholarPubMed
Huang, JH, Berkovitch, SS, Iaconelli, J, Watmuff, B, Park, H, Chattopadhyay, S, et al.Perturbational profiling of metabolites in patient fibroblasts implicates α-aminoadipate as a potential biomarker for bipolar disorder. Mol Neuropsychiatry. 2016;2:97106. doi: 10.1159/000446654.CrossRefGoogle ScholarPubMed
Huang, JH, Park, H, Iaconelli, J, Berkovitch, SS, Watmuff, B, McPhie D, et al.Unbiased metabolite profiling of schizophrenia fibroblasts under stressful perturbations reveals dysregulation of plasmalogens and phosphatidylcholines. J Proteome Res. 2017;16:481–93. doi:10.1021/acs.jproteome.6b00628.CrossRefGoogle ScholarPubMed
Draganov, M, Arranz, MJ, Salazar, J, de Diego-Adeliño, J, Gallego-Fabrega, C, Jubero, M, et al.Association study of polymorphisms within inflammatory genes and methylation status in treatment response in major depression. Eur Psychiatry. 2019;60:713. doi:10.1016/j.eurpsy.2019.05.003.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Framework for utilizing human system cell-derived neurons in clinical cohort studies.

Submit a response

Comments

No Comments have been published for this article.